Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Med Virol ; 96(1): e29386, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38235919

ABSTRACT

Human papillomavirus (HPV) is a major causative factor of head and neck squamous cell carcinoma (HNSCC), and the incidence of HPV- associated HNSCC is increasing. The role of tumor microenvironment in viral infection and metastasis needs to be explored further. We studied the molecular characteristics of primary tumors (PTs) and lymph node metastatic tumors (LNMTs) by stratifying them based on their HPV status. Eight samples for single-cell RNA profiling and six samples for spatial transcriptomics (ST), composed of matched primary tumors (PT) and lymph node metastases (LNMT), were collected from both HPV- negative (HPV- ) and HPV-positive (HPV+ ) patients. Using the 10x Genomics Visium platform, integrative analyses with single-cell RNA sequencing were performed. Intracellular and intercellular alterations were analyzed, and the findings were confirmed using experimental validation and publicly available data set. The HPV+ tissues were composed of a substantial amount of lymphoid cells regardless of the presence or absence of metastasis, whereas the HPV- tissue exhibited remarkable changes in the number of macrophages and plasma cells, particularly in the LNMT. From both single-cell RNA and ST data set, we discovered a central gene, pyruvate kinase muscle isoform 1/2 (PKM2), which is closely associated with the stemness of cancer stem cell-like populations in LNMT of HPV- tissue. The consistent expression was observed in HPV- HNSCC cell line and the knockdown of PKM2 weakened spheroid formation ability. Furthermore, we found an ectopic lymphoid structure morphology and clinical effects of the structure in ST slide of the HPV+ patients and verified their presence in tumor tissue using immunohistochemistry. Finally, the ephrin-A (EPHA2) pathway was detected as important signals in angiogenesis for HPV- patients from single-cell RNA and ST profiles, and knockdown of EPHA2 declined the cell migration. Our study described the distinct cellular composition and molecular alterations in primary and metastatic sites in HNSCC patients based on their HPV status. These results provide insights into HNSCC biology in the context of HPV infection and its potential clinical implications.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Papillomavirus Infections , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , Carcinoma, Squamous Cell/pathology , Human Papillomavirus Viruses , Papillomaviridae/genetics , Head and Neck Neoplasms/genetics , Gene Expression Profiling/methods , RNA , Tumor Microenvironment/genetics
2.
Allergy ; 78(12): 3166-3177, 2023 12.
Article in English | MEDLINE | ID: mdl-37415527

ABSTRACT

BACKGROUND: Anti-heat shock protein (HSP) autoantibodies are detected in autoimmune diseases. We sought to ascertain whether anti-HSP10 IgG is present in patients with CSU and to elucidate the role of HSP10 in CSU pathogenesis. METHOD: Using a human proteome microarray, six potential autoantibodies had higher expression in 10 CSU samples compared with 10 normal controls (NCs). Among them, HSP10 IgG autoantibody was quantified by immune dot-blot assay in sera from 86 CSU patients and 44 NCs. The serum levels of HSP10 and microRNA-101-5p were measured in CSU patients and NCs. The effects of HSP10 and miR-101-5p on mast cell degranulation in response to IgE, compound 48/80, and platelet-activating factor (PAF) were investigated. RESULTS: CSU patients had higher IgG positivity to HSP10 (40.7% vs. 11.4%, p = .001), lower serum HSP10 levels (5.8 ± 3.6 vs. 12.2 ± 6.6 pg/mL, p < .001) than in NCs, and their urticaria severity was associated with anti-HSP10 IgG positivity, while HSP10 levels were related to urticaria control status. MiR-101-5p was increased in CSU patients. PAF enhanced IL4 production in PBMCs from CSU patients. IL-4 upregulated miR-101-5p and reduced HSP10 expression in keratinocytes. Transfection of miR-101-5p reduced HSP10 expression in keratinocytes. MiR-101-5p promoted PAF-induced mast cell degranulation, while HSP10 specifically prevented it. CONCLUSION: A new autoantibody, anti-HSP10 IgG was detected in CSU patients, which showed a significant correlation with UAS7 scores. A decreased serum HSP10 level was associated with upregulation of miR-101-5p due to increased IL-4 and PAF in CSU patients. Modulation of miR-101-5p and HSP10 may be a novel therapeutic approach for CSU.


Subject(s)
Chronic Urticaria , MicroRNAs , Urticaria , Humans , MicroRNAs/genetics , Platelet Activating Factor , Interleukin-4 , Chronic Disease , Autoantibodies , Immunoglobulin G
3.
Int Arch Allergy Immunol ; 182(11): 1066-1071, 2021.
Article in English | MEDLINE | ID: mdl-34139696

ABSTRACT

INTRODUCTION: Chronic spontaneous urticaria (CSU) is a common cutaneous disease caused by mast-cell degranulation. Human ß-defensin 2 (HBD2) is a well-known antimicrobial peptide that is also a pruritogen inducing vascular permeability via non-IgE-mediated mast-cell degranulation. OBJECTIVE: We investigated the associations between serum HBD2 levels and the clinical characteristics of CSU patients. METHODS: Serum samples from 124 CSU patients and 56 healthy controls were screened for the levels of HBD2 and translationally controlled tumor protein (TCTP)_ by using ELISA. The urticaria activity score over 7 days (UAS7) was used to measure disease activity in CSU patients. Accompanying angioedema was self-reported. RESULTS: Serum HBD2 levels were higher in the CSU group than in healthy subjects (median [interquartile range], 84.1 [43.5, 142.5] vs. 59.5 [26.7, 121.5], p = 0.034). In CSU patients, serum HBD2 level was negatively correlated with the peripheral basophil percentages (Spearman's rho = -0.229, p = 0.01) and vitamin D levels (-0.262, p = 0.02), but positively correlated with TCTP levels (0.252, p = 0.006). In CSU patients, HBD2 level was higher in those with than without angioedema (101.7 [50.9, 184.2] vs. 66.7 [37.9, 132.0], p = 0.019). It did not differ by aspirin hypersensitivity or atopy status, or autologous serum skin test positivity. CONCLUSION: A known mast-cell degranulator, HBD2 was elevated in the sera from CSU patients compared to healthy controls and may be involved in the pathogenesis of accompanying angioedema.


Subject(s)
Angioedema/blood , Chronic Urticaria/blood , beta-Defensins/blood , Adult , Biomarkers, Tumor/blood , Female , Humans , Male , Middle Aged , Tumor Protein, Translationally-Controlled 1
4.
Allergy Asthma Immunol Res ; 13(3): 498-506, 2021 May.
Article in English | MEDLINE | ID: mdl-33733642

ABSTRACT

Mas-related G-protein coupled receptor-X2 (MRGPRX2), a receptor on mast cells, basophils, and eosinophils associated with immunoglobulin E (IgE)-independent degranulation, has been reported to be highly expressed on cutaneous mast cells in patients with severe chronic spontaneous urticaria (CSU). We sought to investigate whether MRGPRX2 levels in the sera from CSU patients differ from those in healthy control subjects and to evaluate the clinical utility of MRGPRX2 levels in CSU patients. Severe CSU was defined as urticaria activity score over 7 days (UAS7) ≥ 28. Serum samples from 116 (73 severe and 43 non-severe) CSU patients and 50 healthy subjects were screened for MRGPRX2 using enzyme-linked immunosorbent assay. Serum MRGPRX2 levels were significantly higher in patients with severe CSU (median [interquartile range], 16.5 [10.8-24.8]) than in healthy controls (11.7 [6.5-21.2], P = 0.036) and in non-severe CSU patients (8.7 [4.5-18.8], P = 0.002), although they did not differ between healthy subjects and non-severe CSU patients. Serum MRGPRX2 levels in CSU patients showed positive correlations with UAS7 and specific IgE against Dermatophagoides farinae in CSU subjects, whereas no correlations were observed for age, sex, urticaria duration, atopy, combined angioedema, autologous serum skin test positivity, or total IgE levels. Logistic regression analysis identified serum MRGPRX2 ≥ 12 ng/mL (odds ratio, 6.421; P = 0.002) as an independent risk factor for severe CSU, along with increased serum total IgE levels, peripheral basophil percentage, and angioedema. In conclusion, we suggest that serum MRGPRX2 could help indicate severe CSU.

5.
Clin Transl Allergy ; 9: 33, 2019.
Article in English | MEDLINE | ID: mdl-31346408

ABSTRACT

BACKGROUND: Platelet activating factor (PAF) is an endogenous, active phospholipid released from inflammatory cells, platelets, and endothelial cells, and is involved in the regulation of immune responses. Degradation of PAF by PAF acetylhydrolase (PAF-AH) has been shown to be associated with anaphylaxis, asthma, and peanut allergy. The purpose of this study was to investigate relationships among clinical parameters, including urticaria severity and treatment responsiveness, and PAF and PAF-AH levels in sera from patients with chronic spontaneous urticaria (CSU). METHODS: Serum PAF and PAF-AH levels were measured by enzyme-linked immunosorbent assay in 283 CSU patients and 111 age- and sex-matched, healthy normal controls (NCs). Urticaria severity was evaluated by urticaria activity score over 7 days (UAS7). Within 3 months after measuring PAF levels, patients whose urticaria was not controlled by antihistamine treatment were classified as histamine receptor 1 antagonist (H1RA) non-responders. RESULTS: Serum PAF levels were significantly higher in CSU patients than in NCs (median 4368.9 vs. 3256.4 pg/ml, p = 0.015), while serum PAF-AH levels were significantly lower in CSU patients (105.6 vs. 125.7 ng/ml, p = 0.001). H1RA non-responders had higher levels of PAF in their sera than H1RA responders. A generalized linear model revealed that a higher UAS7 score (odds ratio 1.023, p = 0.024) and a PAF level ≥ 5000 pg/ml (1.409, p < 0.001) were significant predictors of a poor response to H1RA treatment. CONCLUSIONS: Compared with NCs, CSU patients, particularly those with H1RA refractoriness, showed significant increases in serum PAF levels and decreases in PAF-AH. Therapies modulating PAF and PAF-AH levels could be effective in patients with CSU refractory to antihistamines.

6.
Ann Surg Oncol ; 23(6): 2046-53, 2016 06.
Article in English | MEDLINE | ID: mdl-26812910

ABSTRACT

PURPOSE: It has been reported that the abnormal activation of receptor tyrosine kinases is associated with the development of many human carcinomas and the high activation of EGFR and Met mediates the tumorigenicity of laryngeal carcinoma. In this study, we have done the therapeutic efficacy of ME22S (a novel EGFR/Met bispecific antibody) in laryngeal carcinoma in vitro and in vivo was thoroughly evaluated. METHODS: The effects of ME22S on cell viability was assessed through MTT assays, and then Western blotting and immunocytochemistry were used to determine the expression of EGFR and Met. Also, wound healing and invasion assays were performed to observe the inhibitory effects of ME22S. RESULTS: We found the ability of ME22S reducing the expression of both EGFR and Met and significantly inhibiting the cell migration, invasion, and proliferation of SNU899 and HN3 in vitro. Also, the notably reduced levels of p-Met, p-ERK, and p-AKT were found when the cells were treated with only ME22S alone or with HGF together. Meanwhile, ME22S, interestingly enough, caused caspase-3-dependent apoptotic cell death when HN3 cells were treated with ME22S for 72 h, decreased the HGF-induced Slug expression, and also inhibited the tumor growth of HN3 cells in a xenograft model in vivo. CONCLUSIONS: Taken together, our findings suggest that the dual inhibition of EGFR and Met through ME22S largely suppresses the invasion and growth of laryngeal carcinoma both in vitro and in vivo, hence, can be a practical approach as a novel therapeutic strategy for the treatment of laryngeal carcinoma.


Subject(s)
Antibodies, Bispecific/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Laryngeal Neoplasms/prevention & control , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Biomarkers, Tumor/metabolism , Humans , Laryngeal Neoplasms/drug therapy , Laryngeal Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Cancer Lett ; 372(1): 65-74, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26721202

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents an effective agent for the treatment of many cancers, though the majority of thyroid cancers are found to be resistant. Therefore it would be necessary to identify agents capable of increasing the sensitivity of these cancers to TRAIL-mediated cell death. Here, we examined the therapeutic effect and its underlying mechanism of combination treatment of TRAIL and histone deacetylase inhibitor, Valproic acid (VPA) in vitro using human papillary thyroid cancer (PTC) cells and in vivo using an orthotopic mouse model of PTC. TRAIL-VPA combination therapy synergistically induced apoptotic cell death in TRAIL-resistant PTC through caspase activation. In addition, downregulation of antioxidant transcription factor, Nrf2 by co-treatment of TRAIL-VPA induces cell death via suppression of Bcl-xL in vitro and in vivo; these effects were further enhanced following siRNA inhibition of these proteins in combination with TRAIL or TRAIL-VPA. Taken together, VPA sensitized TRAIL-resistant PTC cells to apoptotic cell death through involvement of Nrf2 and Bcl-xL. Thus, the combination of VPA and TRAIL may be a promising therapy for TRAIL-resistant PTC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Carcinoma/drug therapy , Drug Resistance, Neoplasm , Histone Deacetylase Inhibitors/pharmacology , NF-E2-Related Factor 2/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thyroid Neoplasms/drug therapy , Valproic Acid/pharmacology , bcl-X Protein/metabolism , Animals , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Papillary , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Drug Synergism , Humans , Male , Mice, Inbred BALB C , Mice, Nude , NF-E2-Related Factor 2/genetics , RNA Interference , Signal Transduction/drug effects , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Time Factors , Transfection , bcl-X Protein/genetics
8.
Cancer Res Treat ; 48(2): 738-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26511816

ABSTRACT

PURPOSE: The purpose of this study is to determine whether luminacin, a marine microbial extract from the Streptomyces species, has anti-tumor effects on head and neck squamous cell carcinoma (HNSCC) cell lines via autophagic cell death. MATERIALS AND METHODS: Inhibition of cell survival and increased cell death was measured using cell viability, colony forming, and apoptosis assays. Migration and invasion abilities of head and cancer cells were evaluated using wound healing, scattering, and invasion assays. Changes in the signal pathway related to autophagic cell death were investigated. Drug toxicity of luminacin was examined in in vitro HaCaT cells and an in vivo zebrafish model. RESULTS: Luminacin showed potent cytotoxicity in HNSCC cells in cell viability, colony forming, and fluorescence-activated cell sorting analysis. In vitro migration and invasion of HNSCC cells were attenuated by luminacin treatment. Combined with Beclin-1 and LC3B, Luminacin induced autophagic cell death in head and neck cancer cells. In addition, in a zebrafish model and human keratinocyte cell line used for toxicity testing, luminacin treatment with a cytotoxic concentration to HNSCC cells did not cause toxicity. CONCLUSION: Taken together, these results demonstrate that luminacin induces the inhibition of growth and cancer progression via autophagic cell death in HNSCC cell lines, indicating a possible alternative chemotherapeutic approach for treatment of HNSCC.


Subject(s)
Autophagy/drug effects , Benzaldehydes/pharmacology , Benzaldehydes/therapeutic use , Carcinoma, Squamous Cell/pathology , Cell Extracts/pharmacology , Cell Extracts/therapeutic use , Head and Neck Neoplasms/pathology , Spiro Compounds/pharmacology , Spiro Compounds/therapeutic use , Animals , Benzaldehydes/adverse effects , Carcinoma, Squamous Cell/drug therapy , Cell Extracts/adverse effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Disease Progression , Head and Neck Neoplasms/drug therapy , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Neoplasm Invasiveness/prevention & control , Spiro Compounds/adverse effects , Zebrafish/embryology
9.
Exp Mol Med ; 46: e110, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25145391

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (MSCs) have been reported to be beneficial for the treatment of liver fibrosis. Here, we investigated the use of genetically engineered MSCs that overexpress hepatocyte growth factor (HGF) as a means to improve their therapeutic effect in liver fibrosis. Liver fibrosis was induced by intraperitoneal injection of dimethylnitrosamine. HGF-secreting MSCs (MSCs/HGF) were prepared by transducing MSCs with an adenovirus carrying HGF-encoding cDNA. MSCs or MSCs/HGF were injected directly into the spleen of fibrotic rats. Tissue fibrosis was assessed by histological analysis 12 days after stem cell injection. Although treatment with MSCs reduced fibrosis, treatment with MSCs/HGF produced a more significant reduction and was associated with elevated HGF levels in the portal vein. Collagen levels in the liver extract were decreased after MSC/HGF therapy, suggesting recovery from fibrosis. Furthermore, liver function was improved in animals receiving MSCs/HGF, indicating that MSC/HGF therapy resulted not only in reduction of liver fibrosis but also in improvement of hepatocyte function. Assessment of cell and biochemical parameters revealed that mRNA levels of the fibrogenic cytokines PDGF-bb and TGF-ß1 were significantly decreased after MSC/HGF therapy. Subsequent to the decrease in collagen, expression of matrix metalloprotease-9 (MMP-9), MMP-13, MMP-14 and urokinase-type plasminogen activator was augmented following MSC/HGF, whereas tissue inhibitor of metalloprotease-1 (TIMP-1) expression was reduced. In conclusion, therapy with MSCs/HGF resulted in an improved therapeutic effect compared with MSCs alone, probably because of the anti-fibrotic activity of HGF. Thus, MSC/HGF represents a promising approach toward a cell therapy for liver fibrosis.


Subject(s)
Genetic Engineering , Hepatocyte Growth Factor/genetics , Liver Cirrhosis/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Up-Regulation , Animals , Cell Engineering , Cells, Cultured , Hepatocyte Growth Factor/analysis , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Male , Rats , Rats, Sprague-Dawley
10.
Ann Surg Oncol ; 20 Suppl 3: S716-24, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23982257

ABSTRACT

BACKGROUND: Anaplastic thyroid carcinoma (ATC) is an aggressive human tumor associated with a median survival of 2-6 months. TRAIL, as a ligand of death receptors, is known to induce apoptotic cell death in several cancer cells. However, TRAIL treatment alone is not effective against TRAIL-resistant cancer cells. This study was designed to investigate whether valproic acid (VPA) enhances apoptotic cell death of TRAIL-resistant ATC cells and to identify the mechanism of cell death of ATC cells by combination treatment with VPA and TRAIL. METHODS: To evaluate the cytotoxic effect of TRAIL and/or VPA on ATC cells, we used the MTT assay. The effects of VPA and TRAIL on apoptosis were assessed using FACS analysis (Annexin-V/PI stain) and Western blotting. RESULTS: The combination of VPA with TRAIL significantly induced apoptotic cell death compared with 8505C and ARO cells treated with TRAIL alone. The protein levels of cleaved caspase-8, -3, and PARP were increased in VPA and TRAIL co-treated ARO cells. The combination induced the activation of JNK and the phosphorylation of FADD and c-Jun but not p38. However, pretreatment with caspase inhibitors reduced the expression of cleaved caspase-8, -3, and PARP in co-treated ARO cells. SP600125 remarkably reduced the expression of cleaved caspase-8, -3, and PARP and the phosphorylation of FADD and c-Jun, as well as apoptotic cell death. CONCLUSIONS: VPA sensitized TRAIL-resistant ATC cells to apoptotic cell death through involvement of the JNK pathway. Thus, the combination of VPA and TRAIL may be a promising therapy for ATC.


Subject(s)
Anticonvulsants/pharmacology , Apoptosis/drug effects , Drug Resistance, Neoplasm/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Valproic Acid/pharmacology , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation/drug effects , Drug Synergism , Drug Therapy, Combination , Humans , Immunoenzyme Techniques , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms/metabolism , Tumor Cells, Cultured
11.
Endocr Relat Cancer ; 20(3): 283-91, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23429749

ABSTRACT

Anaplastic thyroid carcinoma (ATC) is an aggressive human tumor with a median survival of 6 months. We previously developed an agonistic anti-death receptor 4 MAB, AY4, and demonstrated the antitumor effects of AY4 in head and neck cancer cells. Presently, we show that ATC cells are sensitive to AY4 and that the sensitivity correlates with the reduced expression level of Bcl-xL and reactive oxygen species (ROS) generation. AY4 induced death of C-643, U-HTH 7, HTH83, and SW1736 cells. To elucidate the role of ROS generation in AY4-induced apoptosis of ATC cells, U-HTH 7 and SW1736 cells were pretreated with an antioxidant (N-acetyl cysteine, NAC) followed by AY4 treatment. The cell death was blocked by NAC. AY4-induced cell death was accompanied by the downregulation of the anti-apoptotic protein, Bcl-xL (BCL2L1). To examine the link between the apoptotic response and Bcl-xL protein expression, U-HTH 7 cells were transfected with Bcl-xL plasmid. The consequence of the overexpression of Bcl-xL appeared to decrease AY4-mediated cell death by blocking ROS generation in U-HTH 7 cells. By contrast, Bcl-xL knockdown using small interfering RNA of Bcl-xL enhanced AY4 sensitivity in HTH83 and C-643 cells and rendered the cells sensitive to AY4-induced cell death. The results support the conclusion that the expression level of Bcl-xL is important in the AY4-induced apoptosis of ATC cells through ROS generation. AY4 may be a promising tool for ATC therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Thyroid Neoplasms/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Down-Regulation , Humans , Reactive Oxygen Species/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Thyroid Carcinoma, Anaplastic
12.
Exp Mol Med ; 43(2): 111-20, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21209554

ABSTRACT

Aberrant activation of hepatocyte growth factor/scatter factor (HGF/SF) and its receptor, Met, is involved in the development and progression of many human cancers. In the cell-based screening assay, (-)epigallocatechin-3-gallate (EGCG) inhibited HGF/SF-Met signaling as indicated by its inhibitory activity on HGF/SF-induced cell scattering and uPA activation (IC50=15.8 microgram/ml). Further analysis revealed that EGCG at low doses specifically inhibited HGF/SF-induced tyrosine phosphorylation of Met but not epidermal growth factor (EGF)-induced phosphorylation of EGF receptor (EGFR). On the other hand, high-dose EGCG decreased both Met and EGFR proteins. We also found that EGCG did not act on the intracellular portion of Met receptor tyrosine kinase, i.e., it inhibited InlB-dependent activation of Met but not NGF-induced activation of Trk-Met hybrid receptor. This inhibition decreased HGF-induced migration and invasion by parental or HGF/SF-transfected B16F10 melanoma cells in vitro in either a paracrine or autocrine manner. Furthermore, EGCG inhibited the invasion/metastasis of HGF/SF-transfected B16F10 melanoma cells in mice. Our data suggest the possible use of EGCG in human cancers associated with dysregulated paracrine or autocrine HGF/SF-Met signaling.


Subject(s)
Autocrine Communication/drug effects , Catechin/analogs & derivatives , Hepatocyte Growth Factor , Neoplasms, Experimental/metabolism , Paracrine Communication/drug effects , Animals , Catechin/metabolism , Catechin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/metabolism , Signal Transduction
13.
Cell Signal ; 21(3): 419-27, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19071214

ABSTRACT

Met receptor tyrosine kinase mediates pleiotropic cellular responses following its activation by hepatocyte growth factor or scatter factor (HGF/SF). STAT3 was reported to be one of direct downstream molecules in HGF/SF-Met signaling. In the present study, however, we observed that Tyr705 of STAT3 was phosphorylated from 2 h or 6 h in NIH3T3 and Chang liver cells, respectively, after HGF/SF treatment. Blocking of the phosphorylation by cycloheximide or actinomycin D and the rapid STAT3 phosphorylation with the conditioned medium from HGF/SF-treated NIH3T3 cells suggested that a newly synthesized secretory protein was responsible for the delayed STAT3 phosphorylation. Among the known mediators to induce STAT3 phosphorylation, interleukin-6 (IL-6) mRNA and protein were induced by HGF/SF, and the released IL-6 was accumulated in the conditioned medium after HGF/SF treatment. Furthermore, the neutralizing IL-6 antibody abolished the STAT3 phosphorylation. Treatment with LY294002, a PI3 kinase inhibitor, but not with other signal inhibitors, resulted in the loss of delayed STAT3 phosphorylation by HGF/SF, showing the involvement of PI3 kinase pathway. Collectively, these results demonstrate that HGF/SF-Met signal cascade stimulates IL-6 production via PI3 kinase pathway, leading to STAT3 phosphorylation as a secondary effect.


Subject(s)
Hepatocyte Growth Factor/metabolism , Interleukin-6/metabolism , Phosphatidylinositol 3-Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Culture Media, Conditioned/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Hepatocyte Growth Factor/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Interleukin-6/genetics , Mice , NIH 3T3 Cells , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , STAT3 Transcription Factor/drug effects , Signal Transduction/drug effects
14.
Exp Mol Med ; 35(2): 118-24, 2003 Apr 30.
Article in English | MEDLINE | ID: mdl-12754416

ABSTRACT

Pervanadate, a complex of vanadate and H(2)O(2), has an insulin mimetic effect, and acts as an inhibitor of protein tyrosine phosphatase. Pervanadate-induced phospholipase D (PLD) activation is known to be dependent on the tyrosine phosphorylation of cellular proteins and protein kinase C (PKC) activation, and yet underlying molecular mechanisms are not clearly understood. Here, we investigated the signaling pathway of pervanadate-induced PLD activation in Rat2 fibroblasts. Pervanadate increased PLD activity in dose- and time- dependent manner. Protein tyrosine kinase inhibitor, genistein, blocked PLD activation. Interestingly, AG-1478, a specific inhibitor of the tyrosine kinase activity of epidermal growth factor receptor (EGFR) blocked not only the PLD activation completely but also phosphorylation of p38 mitogen-activated protein kinase (MAPK). However, AG-1295, an inhibitor specific for the tyrosine kinase activity of pletlet drived growth factor receptor (PDGFR) did not show any effect on the PLD activation by pervanadate. We further found that pervanadate increased phosphorylation levels of p38, extracellular signal-regulated kinase (ERK) and c-Jun NH(2)-terminal kinase (JNK). SB203580, a p38 MAPK inhibitor, blocked the PLD activation completely. However, the inhibitions of ERK by the treatment of PD98059 or of JNK by the overexpression of JNK interacting peptide JBD did not show any effect on pervanadate-induced PLD activation. Inhibition or down-regulation of PKC did not alter the pervanadate-induced PLD activation in Rat2 cells. Thus, these results suggest that pervanadate-induced PLD activation is coupled to the transactivation of EGFR by pervanadate resulting in the activation of p38 MAP kinase.


Subject(s)
ErbB Receptors/agonists , ErbB Receptors/metabolism , Phospholipase D/metabolism , Vanadates/pharmacology , Animals , Cell Line , Enzyme Activation/drug effects , Fibroblasts , Mitogen-Activated Protein Kinases/metabolism , Rats , p38 Mitogen-Activated Protein Kinases , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...