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.
FASEB J ; 36(11): e22597, 2022 11.
Article in English | MEDLINE | ID: mdl-36197688

ABSTRACT

Estrogen signaling has been extensively studied, especially in cancers that express estrogen receptor alpha (ERα). However, little is known regarding the effect of estrogen on cancer-associated fibroblasts (CAFs). Here, we explored the role of estrogen signaling of CAFs in gastric cancer (GC) progression. We investigated the phenotypic changes in CAFs upon 17ß-estradiol (E2) treatment using ERα-negative/positive CAFs, and the conditioned media (CM) collected from these were compared with regard to cancer cell proliferation, migration, and invasion. A paracrine factor was found using a cytokine array and was confirmed using qRT-PCR, western blotting, and enzyme-linked immunosorbent assays. ERα-CD147-matrix metalloproteinase (MMP) axis was confirmed by knockdown experiments using specific siRNAs. We found that a subset of CAFs expressed ERα. ERα-positive CAFs were responsive to E2, inducing ERα expression in a dose-dependent manner. Although E2 did not induce the proliferation of ERα-positive CAFs, the CM from E2-bound ERα-positive CAFs significantly promoted cancer cell migration and invasion. Cytokine array revealed that CD147 was induced in ERα-positive CAFs upon E2 treatment; this was mediated via ERα. Increased CD147 upregulated MMP2 and MMP9 in CAFs, and also influenced cancer cells in a paracrine manner to increase MMPs and CD147 in cancer cells. High CD147 expression in tumor tissue was associated with a worse prognosis in GC patients. Our data suggest that estrogen signaling activation in CAFs and the byproduct CD147 are among the critical mediators between the interplay of CAFs and cancer cells to facilitate cancer progression.


Subject(s)
Basigin/metabolism , Cancer-Associated Fibroblasts , Stomach Neoplasms , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Proliferation , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Stomach Neoplasms/pathology
3.
Cancers (Basel) ; 14(9)2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35565206

ABSTRACT

Pancreatic cancer is a devastating disease and is highly resistant to anticancer drugs because of its complex microenvironment. Cancer-associated fibroblasts (CAFs) are an important source of extracellular matrix (ECM) components, which alter the physical and chemical properties of pancreatic tissue, thus impairing effective intratumoral drug delivery and resulting in resistance to conventional chemotherapy. The objective of this study was to develop a new cancer organoid model, including a fibrous tumor microenvironment (TME) using CAFs. The CAF-integrated pancreatic cancer organoid (CIPCO) model developed in this study histologically mimicked human pancreatic cancer and included ECM production by CAFs. The cancer cell-CAF interaction in the CIPCO promoted epithelial-mesenchymal transition of cancer cells, which was reversed by CAF inhibition using all-trans retinoic acid. Deposition of newly synthesized collagen I in the CIPCO disturbed the delivery of gemcitabine to cancer cells, and treatment with collagenase increased the cytotoxic effect of gemcitabine. This model may lead to the development of next-generation cancer organoid models recapitulating the fibrous TME.

4.
Biochem Biophys Res Commun ; 613: 180-186, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35597125

ABSTRACT

miRNA (miR)-4742-5p is a recently identified microRNA regarding progression and metastasis in gastric cancer (GC). However, the biological function of this novel miRNA is largely unknown. We identified that the miR-4742-5p expression level was variably increased in GC cell lines. Suppression of miR-4742-5p using miR-inhibitor reduced the proliferation, migration, and invasion of GC cells with high miR-4742-5p expression, whereas overexpression of miR-4742-5p-mimic enhanced the aforementioned properties in GC cells with low miR-4742-5p expression. miR-4742-5p expression induced the decreases of Zo-1 and E-cadherin expression as well as the increases of vimentin and N-cadherin expression, leading to epithelial-mesenchymal transition (EMT) of cancer cells. RNA sequencing results indicated Ras-related GTP-binding protein 43 (Rab43) as a potential target gene. We identified that the expression of Rab43 is associated with activation of AKT and nuclear factor-kappa B (NF-κB) which are key oncogenic pathways in cancer cells. Our results demonstrate a new component in GC progression, promising a potential therapeutic strategy.


Subject(s)
MicroRNAs , Stomach Neoplasms , rab GTP-Binding Proteins , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
5.
Gastric Cancer ; 24(3): 577-588, 2021 May.
Article in English | MEDLINE | ID: mdl-33164142

ABSTRACT

BACKGROUND: Despite the promising preclinical antitumor activity of MET-targeting therapies, most clinical trials have failed. We introduced a new concept of quantitation of stroma-induced hepatocyte growth factor (HGF) to assess the actual MET signalling activity in gastric cancer (GC). METHODS: We treated serially diluted HGF and conditioned media (CM) from cancer-associated fibroblasts (CAFs) on low MET-expressing cancer cells and investigated the phenotypical and signalling changes. Stromal proportion and MET expression in GC samples were assessed, and gene set enrichment analysis (GSEA) from the public database was performed. The antitumor effect of anti-MET treatment was examined, especially when cancer cells were activated in a ligand-dependent manner. RESULTS: Relatively high doses of HGF or high-concentrated CM fully activated MET signalling cascades and promoted cell proliferation/invasion. High stromal proportion denoted worse patient survival in MET-positive GCs than in MET-negative ones. GSEA showed that the gene sets regarding proliferation, migration, and CAF as well as MET pathway signature were enriched in simultaneously MET- and HGF-positive samples. Sufficient ligand-dependent MET signalling activation increased the sensitivity to crizotinib. CONCLUSIONS: We conclude that patients whose tumours have a high stromal proportion and at least low MET expression may benefit more from MET-targeted therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Crizotinib/therapeutic use , Proto-Oncogene Proteins c-met/metabolism , Stomach Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Crizotinib/chemistry , Crizotinib/pharmacology , Humans , Signal Transduction , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Structure-Activity Relationship
6.
Mod Pathol ; 34(3): 672-683, 2021 03.
Article in English | MEDLINE | ID: mdl-32973329

ABSTRACT

Although a certain proportion of intramucosal carcinomas (IMCs) of the stomach does metastasize, the majority of patients are currently treated with endoscopic resection without lymph node dissection, and this potentially veils any existing metastasis and may put some patients in danger. In this regard, biological markers from the resected IMC that can predict metastasis are warranted. Here, we discovered unique miRNA expression profiles that consist of 21 distinct miRNAs that are specifically upregulated (miR-628-5p, miR-1587, miR-3175, miR-3620-5p, miR-4459, miR-4505, miR-4507, miR-4720-5p, miR-4742-5p, and miR-6779-5p) or downregulated (miR-106b-3p, miR-125a-5p, miR-151b, miR-181d-5p, miR-486-5p, miR-500a-3p, miR-502-3p, miR-1231, miR-3609, and miR-6831-5p) in metastatic (M)-IMC compared to nonmetastatic (N)-IMC, or nonneoplastic gastric mucosa. Intriguingly, most of these selected miRNAs showed stepwise increased or decreased expression from nonneoplastic tissue to N-IMC to M-IMC. This suggests that common oncogenic mechanisms are gradually intensified during the metastatic process. Using a machine-learning algorithm, we demonstrated that such miRNA signatures could distinguish M-IMC from N-IMC. Gene ontology and pathway analysis revealed that TGF-ß signaling was enriched from upregulated miRNAs, whereas E2F targets, apoptosis-related, hypoxia-related, and PI3K/AKT/mTOR signaling pathways, were enriched from downregulated miRNAs. Immunohistochemical staining of samples from multiple institutions indicated that PI3K/AKT/mTOR pathway components, MAPK1, phospho-p44/42 MAPK, and pS6 were highly expressed and the expression of SMAD7, a TGF-ß pathway component, was decreased in M-IMC, which could aid in distinguishing M-IMC from N-IMC. The miRNA signature discovered in this study is a valuable biological marker for identifying metastatic potential of IMCs, and provides novel insights regarding the metastatic progression of IMC.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , Transcriptome , Biomarkers, Tumor/analysis , Gastric Mucosa/enzymology , Gastric Mucosa/pathology , Gene Expression Profiling , Gene Regulatory Networks , Humans , Immunohistochemistry , Lymphatic Metastasis , Machine Learning , Predictive Value of Tests , Reproducibility of Results , Republic of Korea , Signal Transduction/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology
7.
J Exp Clin Cancer Res ; 38(1): 484, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31805999

ABSTRACT

BACKGROUND: Protein tyrosine phosphatase receptor delta (PTPRD) is frequently inactivated in various types of cancers. Here, we explored the underlying mechanism of PTPRD-loss-induced cancer metastasis and investigated an efficient treatment option for PTPRD-inactivated gastric cancers (GCs). METHODS: PTPRD expression was evaluated by immunohistochemistry. Microarray analysis was used to identify differentially expressed genes in PTPRD-inactivated cancer cells. Quantitative reverse transcription (qRT-PCR), western blotting, and/or enzyme-linked immunosorbent assays were used to investigate the PTPRD-CXCL8 axis and the expression of other related genes. An in vitro tube formation assay was performed using HUVECs. The efficacy of metformin was assessed by MTS assay. RESULTS: PTPRD was frequently downregulated in GCs and the loss of PTPRD expression was associated with advanced stage, worse overall survival, and a higher risk of distant metastasis. Microarray analysis revealed a significant increase in CXCL8 expression upon loss of PTPRD. This was validated in various GC cell lines using transient and stable PTPRD knockdown. PTPRD-loss-induced angiogenesis was mediated by CXCL8, and the increase in CXCL8 expression was mediated by both ERK and STAT3 signaling. Thus, specific inhibitors targeting ERK or STAT3 abrogated the corresponding signaling nodes and inhibited PTPRD-loss-induced angiogenesis. Additionally, metformin was found to efficiently inhibit PTPRD-loss-induced angiogenesis, decrease cell viability in PTPRD-inactivated cancers, and reverse the decrease in PTPRD expression. CONCLUSIONS: Thus, the PTPRD-CXCL8 axis may serve as a potential therapeutic target, particularly for the suppression of metastasis in PTPRD-inactivated GCs. Hence, we propose that the therapeutic efficacy of metformin in PTPRD-inactivated cancers should be further investigated.


Subject(s)
Interleukin-8/genetics , Metformin/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Stomach Neoplasms/blood supply , Cell Line, Tumor , Down-Regulation , Gene Silencing , Humans , Hypoglycemic Agents/pharmacology , Interleukin-8/metabolism , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/deficiency , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transfection
8.
Int J Cancer ; 145(4): 916-926, 2019 08 15.
Article in English | MEDLINE | ID: mdl-30664822

ABSTRACT

Notwithstanding remarkable treatment success with anti-PD-1 monoclonal antibody, oncogenic mechanism of PD-L1 regulation in gastric cancer (GC) remains poorly understood. We hypothesized that ARID1A might be related to tumor PD-L1 expression in GC. We found that tumor PD-L1 positivity was associated with loss of ARID1A and showed trend toward better survival of patients with various molecular subtypes of GC (experimental set, n = 273). Considering heterogeneous ARID1A expression, we validated this using whole tissue sections (n = 159) and found that loss of ARID1A was correlated with microsatellite instability-high (MSI-H), Epstein-Barr virus (EBV), and PD-L1 positivity. Furthermore, for patients with MSI-H tumors, the degree of PD-L1 expression was significantly higher in ARID1A-deficient tumors. After ARID1A knockdown in GC cell lines, total and membranous PD-L1 protein, and PD-L1 mRNA levels were increased based on Western blot, flow cytometry, and qRT-PCR, respectively. With IFN-γ treatment, PD-L1 expression was significantly increased both in ARID1A-deficient cancer cells and controls, but the increase was not more pronounced in the former. Loss of ARID1A increased PD-L1 via activating AKT signaling, while LY294002 (PI3K inhibitor) decreased PD-L1 levels. Furthermore, we found that 3 MSI-H tumors showing highest expression of PD-L1 had simultaneous KRAS mutation and loss of ARID1A, suggesting a possible synergistic role boosting PD-L1. Our results strongly indicate that loss of ARID1A is tightly associated with high PD-L1 expression in GC. These results would increase our understanding of the oncogenic mechanism of PD-L1 regulation in GC, and also help to find the optimal candidates for immunotherapy.


Subject(s)
B7-H1 Antigen/metabolism , Nuclear Proteins/metabolism , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Carcinogenesis/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Female , Herpesvirus 4, Human/pathogenicity , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Microsatellite Instability , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Stomach Neoplasms/virology
9.
Exp Mol Med ; 50(4): 1-11, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29622765

ABSTRACT

Diffuse-type gastric cancer (DGC) is a GC subtype with heterogeneous clinical outcomes. Lymph node metastasis of DGC heralds a dismal progression, which hampers the curative treatment of patients. However, the genomic heterogeneity of DGC remains unknown. To identify genomic variations associated with lymph node metastasis in DGC, we performed whole exome sequencing on 23 cases of DGC and paired non-tumor tissues and compared the mutation profiles according to the presence (N3, n = 13) or absence (N0, n = 10) of regional lymph node metastasis. Overall, we identified 185 recurrently mutated genes in DGC, which included a significant novel mutation at CMTM2, as well as previously known mutations at CDH1, RHOA, and TP53. Noticeably, CMTM2 expression could predict the prognostic outcomes of DGC but not intestinal-type GC (IGC), indicating pivotal roles of CMTM2 in DGC progression. In addition, we identified a recurrent loss of heterozygosity (LOH) of DNA copy numbers at the 3p12-pcen locus in DGC. A comparison of N0 and N3 tumors showed that N3 tumors exhibited more frequent DNA copy number aberrations, including copy-neutral LOH and mutations of CpTpT trinucleotides, than N0 tumors (P = 0.2 × 10-3). In conclusion, DGCs have distinct profiles of somatic mutations and DNA copy numbers according to the status of lymph node metastasis, and this might be helpful in delineating the pathobiology of DGC.


Subject(s)
Genetic Variation , Genomics/methods , Lymph Nodes/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Adult , Aged , DNA Copy Number Variations , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Mutation , Neoplasm Staging , Exome Sequencing
10.
Oncol Rep ; 25(1): 33-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21109954

ABSTRACT

The presence of active immunity within the brain supports the possibility of effective immunotherapy for glioblastoma (GBM). To provide a clinically-relevant adoptive immunotherapy for GBM using ex vivo expanded cytokine-induced killer (CIK) cells, the treatment capability of CIK cells, either alone or in combination with temozolomide (TMZ) were evaluated. Human CIK (hCIK) cells were cultured from PBMC using activating anti-CD3 antibody and IL-2, which were 99% CD3+, 91% CD3+CD8+ and 29% CD3+CD56+. In vitro, hCIK cells showed tumor-specific cytotoxicity against U-87MG human GBM cells. When hCIK cells were injected into tail veins of immune-compromised mice bearing U-87MG tumors in their brains, numerous CIK cells infiltrated into the brain tumors. CIK treatments (1x10(5), 1x10(6) or 1x10(7), once a week for four weeks) inhibited the tumor growth significantly in a dose-dependent manner; 44, 54 and 72% tumor volume reduction, respectively, compared with the control group (P<0.05). Moreover, hCIK cells (1x10(7), once a week for four weeks) and TMZ (2.5 mg/kg, daily for 5 days) combination treatment further increased tumor cell apoptosis and decreased tumor cell proliferation and vessel density (P<0.05), creating a more potent therapeutic effect (95% reduction in tumor volume) compared with either hCIK cells or TMZ single therapy (72% for both, P<0.05). Taken together, CIK cell-immunotherapy and TMZ chemotherapy have synergistic therapeutic effects and could be combined for a successful treatment of GBM.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Cytokine-Induced Killer Cells/transplantation , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , Animals , Combined Modality Therapy , Dacarbazine/therapeutic use , Humans , Mice , Mice, Nude , Temozolomide , Xenograft Model Antitumor Assays
11.
Phytother Res ; 22(7): 883-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18521967

ABSTRACT

The present study reports the antiinflammatory activity of a methanol extract isolated from the stem bark of Magnolia kobus (MK). MK potently inhibited lipopolysaccharide (LPS)-induced production of nitric oxide and interleukin-1beta (IL-1beta) in RAW 264.7 cells, a murine macrophage-like cell line. The secretion of tumor necrosis factor-alpha (TNF-alpha) was also suppressed in LPS-stimulated RAW 264.7 cells although the magnitude of inhibition was weaker than that of nitric oxide and IL-1beta. The mRNA expressions of inducible nitric oxide synthase (iNOS), IL-1beta and TNF-alpha were also suppressed by MK in LPS-stimulated RAW 264.7 cells. Further study demonstrated that LPS-induced DNA binding of AP-1 and phosphorylation of c-jun N-terminal kinase (JNK) were inhibited by MK treatment in RAW 264.7 cells, whereas phosphorylation of p38 mitogen-activated protein kinase was unaffected. Moreover, topical application of MK suppressed ear swelling in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced skin inflammation model. Collectively, these results suggest that MK exerts antiinflammatory effects in vitro and in vivo and this might be mediated, at least in part, by blocking AP-1 and JNK activation.


Subject(s)
Anti-Inflammatory Agents/isolation & purification , Magnolia/chemistry , Phytotherapy , Plant Bark/chemistry , Plant Extracts/isolation & purification , Administration, Topical , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Cell Line , Cell Survival/drug effects , DNA/chemistry , DNA/metabolism , Ear, External/drug effects , Ear, External/pathology , Edema/chemically induced , Edema/pathology , Gene Expression/drug effects , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Methanol/chemistry , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Protein Binding/drug effects , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Int Immunopharmacol ; 7(13): 1793-801, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17996690

ABSTRACT

Cytokine-induced killer (CIK) cells are ex vivo expanded T cells with natural killer cell phenotypes and functions. In this study, the anti-tumor activity of CIK cells against hepatocellular carcinoma was evaluated in vitro and in vivo. In the presence of anti-CD3 antibody and IL-2 for 14 days, human peripheral blood mononuclear cell population changed to heterogeneous CIK cell population, which comprised 96% CD3(+), 3% CD3( inverted exclamation mark(c))CD56(+), 32% CD3(+)CD56(+), 11% CD4(+), 75% CD8(+), and 30% CD8(+)CD56(+). CIK cells produced significant amounts of IFN-gamma and TNF-alpha; however, produced only slight amounts of IL-2, IL-4, and IL-5. At an effector-target cell ratio of 30:1, CIK cells destroyed 33% of SNU-354 human hepatocellular carcinoma cells, which was determined by the (51)Cr-release assay. In addition, a dose of 1x10(6) CIK cells per mouse inhibited 60% of SNU-354 tumor growth in irradiated nude mice. This study suggests that CIK cells may be used as an adoptive immunotherapy for patients with hepatocellular carcinoma.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Liver Neoplasms, Experimental/therapy , Animals , CD3 Complex/analysis , CD56 Antigen/analysis , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Interferon-gamma/biosynthesis , Liver Neoplasms, Experimental/immunology , Mice
13.
Int Immunopharmacol ; 7(1): 78-87, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17161820

ABSTRACT

During the evolution of neoplastic diseases, dendritic cell (DC) functions are usually attenuated, and this presents a problem to DC-based immunotherapies against cancer. Here, we investigated the effects of angelan, an acidic polysaccharide isolated from Angelica gigas Nakai, on DC maturation. Angelan efficiently increased the maturation of tlr4(+/+) DCs from C57BL/6 and C3H/HeN mice, but not tlr4(-/-) DCs from C3H/HeJ mice. Phenotypic maturation was confirmed by the elevated expressions of CD80, CD86, and MHC-class II molecules, and functional maturation by increased IL-12 production, enhanced allogenic T cell stimulation, and decreased endocytosis. Angelan was found to activate ERK and NF-kappaB, which are signaling molecules down-stream of toll-like receptor-4 (TLR4) receptors. Angelan-treated mature DC more effectively inhibited B16F10 tumor growth than immature DCs in syngenic murine tumor model. These results indicate that angelan induces DC maturation via TLR4 signaling pathways and suggest the possible use of angelan in DC-based immunotherapies.


Subject(s)
Dendritic Cells/drug effects , Polysaccharides/pharmacology , Toll-Like Receptor 4/immunology , Angelica/chemistry , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Endocytosis/drug effects , Female , Lymphocyte Culture Test, Mixed , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Polysaccharides/isolation & purification , Spleen/cytology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
14.
Int Immunopharmacol ; 6(10): 1616-22, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16919834

ABSTRACT

Mite antigen has been considered to play important roles in the development of atopic eczema/dermatitis syndrome (AEDS). In the present study, we attempted to induce an AEDS-like skin lesion in mice using Dermatophagoides pteronyssinus crude extract (DPE) as an antigen and performed pathophysiological evaluations. Ears of mice were tape-stripped and DPE was painted 3 times a week. Eczematous skin lesion and ear swelling were apparent in NC/Nga mice treated with DPE after 2 weeks, whereas neither skin lesion nor ear swelling were observed in BALB/c mice even after 30 days. Histological evaluation demonstrated that edema, epidermal hyperplasia and the accumulation of inflammatory cells were apparent in the ears of DPE-treated NC/Nga mice. In contrast to skin lesion and ear swelling, total serum IgE levels were increased in both NC/Nga and BALB/c mice. Treatment with DPE also increased auricular lymph node weight in both NC/Nga mice and BALB/c mice. To further characterize, we analyzed cytokine mRNA expression in ears and lymph nodes of DPE-treated NC/Nga mice. Increased expression of IL-4 and TNF-alpha mRNA was observed in both ears and lymph nodes of NC/Nga mice treated with DPE. Additionally, there was no change in the responsiveness of BALB/c mice to DPE treatment by adaptive transfer of serum from DPE-treated NC/Nga mice to BALB/c mice. Taken together, our results indicate that eczematous skin lesion and ear swelling caused by repeated application of DPE in NC/Nga mice has a Th2-dominant background and that inflammation is involved in this process. The animal model of AEDS established in this report may be used to investigate the pathogenesis of AEDS and evaluate the potential therapeutic agents for AEDS.


Subject(s)
Antigens, Dermatophagoides/immunology , Dermatitis, Allergic Contact/etiology , Dermatitis, Atopic/chemically induced , Dermatophagoides pteronyssinus/immunology , Disease Models, Animal , Mice , Administration, Topical , Animals , Cytokines/biosynthesis , Cytokines/immunology , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/pathology , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Edema/immunology , Edema/pathology , Female , Immunoglobulin E/blood , Immunoglobulin E/immunology , Mice, Inbred BALB C , Mice, Inbred Strains , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...