Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Cell Signal ; 113: 110971, 2024 01.
Article En | MEDLINE | ID: mdl-37979898

Bladder cancer, the most common malignant tumor in the urinary system, exhibits significantly up-regulated expression of P3H4, which is associated with pathological factors. The objective of this study was to elucidate the underlying mechanism of P3H4 in bladder cancer. Initially, we analyzed P3H4 gene expression using the TCGA database and evaluated P3H4 levels in clinical samples and various bladder cell lines. P3H4 was found to be markedly overexpressed in bladder cancer samples. Subsequently, bladder cancer cells were transfected with shRNA targeting P3H4 (sh-P3H4), sh-METTL3, and P3H4 overexpression vectors (P3H4 OE). Viability, migration, and invasion of bladder cancer cells were assessed using CCK-8, wound healing, and transwell assays. Western blot analysis was performed to determine the levels of EMT-associated proteins, while RNA stability assays determined the half-life of P3H4. Knockdown of P3H4 resulted in inhibition of bladder cancer cell proliferation, migration, invasion, and EMT progression. Mechanistically, METTL3 was found to regulate the mRNA stability of P3H4 in bladder cancer. Moreover, overexpression of P3H4 reversed the inhibitory effects of METTL3 knockdown on bladder cancer cell behaviors. Stable cell lines were established by infecting EJ cells with lentiviral vectors containing sh-METTL3 or P3H4 OE. These cells were then implanted into the skin of BALB/c nude mice, and IHC analysis was used to analyze the expression levels of EMT-associated proteins. In vivo studies demonstrated that inhibition of METTL3 suppressed bladder cancer growth and EMT through P3H4. In conclusion, our findings suggest that METTL3 regulates the proliferation, metastasis, and EMT progression of bladder cancer through P3H4, highlighting its potential as a therapeutic target.


Urinary Bladder Neoplasms , Animals , Mice , Cell Line, Tumor , Mice, Nude , Cell Proliferation/genetics , Urinary Bladder Neoplasms/pathology , Cell Movement/genetics , Gene Expression Regulation, Neoplastic
3.
Exp Cell Res ; 417(1): 113210, 2022 08 01.
Article En | MEDLINE | ID: mdl-35597298

Cancer cells acquire immunoediting ability to evade immune surveillance and thus escape eradication. It is widely known that mutant proteins encoded from tumor suppressor TP53 exhibit gain-of-function in cancer cells, thereby promoting progression; however, how mutant p53 contributes to the sheltering of cancer cells from host anticancer immunity remains unclear. Herein, we report that murine p53 missense mutation G242A (corresponding to human G245A) suppresses the activation of host natural killer (NK) cells, thereby enabling breast cancer cells to avoid immune assault. We found that serial injection of EMT6 breast cancer cells that carry wild-type (wt) Trp53, like normal fibroblasts, promoted NK activity in mice, while SVTneg2 cells carrying Trp53 G242A+/+ mutation decreased NK cell numbers and increased CD8+ T lymphocyte numbers in spleen. Innate immunity based on NK cells and CD8 T cells was reduced in p53 mutant-carrying transgenic mice (Trp53 R172H/+, corresponding to human R175H/+). Further, upon co-culture with isolated NK cells, EMT6 cells substantively activated NK cells and proliferation thereof, increasing interferon-gamma (IFN-γ) production; however, SVTneg2 cells suppressed NK cell activation. Further mechanistic study elucidated that p53 can modulate expression by cancer cells of Mult-1 and H60a, which are activating and inhibitory ligands for NKG2D receptors of NK cells, respectively, to enhance immune surveillance against cancer. Our findings demonstrate that wt p53 is requisite for NK cell-based immune recognition and elimination of cancerous cells, and perhaps more importantly, that p53 missense mutant presence in cancer cells impairs NK cell-attributable responses, thus veiling cancerous cells from host immunity and enabling cancer progression.


Breast Neoplasms , Killer Cells, Natural , Tumor Suppressor Protein p53 , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Female , Killer Cells, Natural/metabolism , Mice , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Transl Androl Urol ; 11(1): 91-103, 2022 Jan.
Article En | MEDLINE | ID: mdl-35242644

BACKGROUND: Melatonin is a hormone naturally produced by the pineal gland in the brain. In addition to modulating circadian rhythms, it has pleiotropic biological effects including antioxidant, immunomodulatory, and anti-cancer effects. Herein, we report that melatonin has the ability to decrease the growth and metastasis of androgen-dependent prostate cancer. METHODS: To evaluate the anti-cancer effect of melatonin on androgen-sensitive prostate cancer in vitro or in vivo, the effects of cell proliferation, apoptosis, migration and invasion were analyzed by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation, flow cytometry, Transwell assay, and immunohistochemistry (IHC), respectively. Next, the interaction between androgen receptor (AR) and SUMO specific protease 1 (SENP1) was detected by real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blotting, and confirmed by luciferase reporter assay. Furthermore, the Small Ubiquitin-like Modifier (SUMO) proteins are a group of small proteins that are covalently attached to and detached from other proteins in cells to modify their function. (SUMOylation) of histone deacetylases 1 (HDAC1) was measured by proximity ligation assay (PLA). RESULTS: The treatment of melatonin cripples the transcriptional activity of AR, which is essential for the growth of the androgen-dependent prostate cancer cell, LNCaP. The lower activity of AR was dependent on melatonin induced SUMOylation of HDAC1, which has been established as a key factor for the transcriptional activity of AR. Mechanistically, the effect of melatonin on AR was due to the decreased SENP1 protein level and the subsequent increased HDAC1 SUMOylation level. The overexpression of SENP1 abrogated the anti-cancer ability of melatonin on LNCaP cells. CONCLUSIONS: These findings indicate that melatonin is a suppressor of androgen-dependent prostate cancer tumorigenesis.

5.
BMC Cancer ; 21(1): 1135, 2021 Oct 23.
Article En | MEDLINE | ID: mdl-34688260

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is one of the most lethal urological malignancies, but the pathogenesis and prognosis of ccRCC remain obscure, which need to be better understand. METHODS: Differentially expressed genes were identified and function enrichment analyses were performed using three publicly available ccRCC gene expression profiles downloaded from the Gene Expression Omnibus database. The protein-protein interaction and the competing endogenous RNA (ceRNA) networks were visualized by Cytoscape. Multivariate Cox analysis was used to predict an optimal risk mode, and the survival analysis was performed with the Kaplan-Meier curve and log-rank test. Protein expression data were downloaded from Clinical Proteomic Tumor Analysis Consortium database and Human Protein Atlas database, and the clinical information as well as the corresponding lncRNA and miRNA expression data were obtained via The Cancer Genome Atlas database. The co-expressed genes and potential function of candidate genes were explored using data exacted from the Cancer Cell Line Encyclopedia database. RESULTS: Of the 1044 differentially expressed genes shared across the three datasets, 461 were upregulated, and 583 were downregulated, which significantly enriched in multiple immunoregulatory-related biological process and tumor-associated pathways, such as HIF-1, PI3K-AKT, P53 and Rap1 signaling pathways. In the most significant module, 36 hub genes were identified and were predominantly enriched in inflammatory response and immune and biotic stimulus pathways. Survival analysis and validation of the hub genes at the mRNA and protein expression levels suggested that these genes, particularly complement component 3 (C3) and fibronectin 1 (FN1), were primarily responsible for ccRCC tumorigenesis and progression. Increased expression of C3 or FN1 was also associated with advanced clinical stage, high pathological grade, and poor survival in patients with ccRCC. Univariate and multivariate Cox regression analysis qualified the expression levels of the two genes as candidate biomarkers for predicting poor survival. FN1 was potentially regulated by miR-429, miR-216b and miR-217, and constructed a bridge to C3 and C3AR1 in the ceRNA network, indicating a critical position of FN1. CONCLUSIONS: The biomarkers C3 and FN1 could provide theoretical support for the development of a novel prognostic tool to advance ccRCC diagnosis and targeted therapy.


Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/genetics , Fibronectins/metabolism , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/mortality , Disease Progression , Humans , Kidney Neoplasms/mortality , Survival Analysis
6.
AAPS PharmSciTech ; 21(6): 202, 2020 Jul 21.
Article En | MEDLINE | ID: mdl-32696338

The adverse side effects and toxicity caused by the non-targeted delivery of doxorubicin has emphasized the demand of emerging a targeted delivery system. The goal of this study is to enhance the delivery of doxorubicin by formulating an aptamer-labeled liposomal nanoparticle delivery system that will carry and deliver doxorubicin specifically into Her-2+ breast cancer cells. Twelve liposomal batches were prepared using different saturated (HSPC and DPPC) and unsaturated (POPC and DOPC) lipids by thin film hydration. The liposomes were characterized for their particle size, zeta potential, and drug encapsulation efficiency. The particles were also assessed for in vitro toxicity and DOX delivery into the breast cancer cells. The formulations, F1 through F12, had a small particle size of less than 200 nm and a high entrapment efficiency of about 88 ± 5%. The best formulation, F5, had a particle size of 101 ± 14nm, zeta potential of + 5.63 ± 0.46 mV, and entrapment efficiency of ≈ 93%. The cytotoxicity studies show that the DOX-loaded liposomal formulations are more effective in killing cancer cells than the free DOX in both MCF-7 and SKBR-3 cells. The uptake studies show a significant increase in the uptake of the aptamer-labeled liposomes (i.e., F5) by more than 60% into Her-2+ MCF-7 and SKBR-3 breast cancer cells compare to non-aptamer-labeled nanoparticles. F5 also shows ≈ 1.79-fold increase in uptake of DOX in the Her-2+ cells compared to the Her-2- cells. This preliminary study indicates that aptamer-labeled F5 nanoparticles among several batches showed the highest uptake as well as the targeted delivery of doxorubicin into Her-2+ breast cancer cells. Thus, aptamer targeted approach results in substantial reduction in the dose of DOX and improves the therapeutic benefits by promoting the target specificity.


Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Drug Delivery Systems , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Doxorubicin/administration & dosage , Female , Humans , Liposomes , Nanoparticles , Polyethylene Glycols/administration & dosage
7.
Tumour Biol ; 37(8): 11147-11162, 2016 Aug.
Article En | MEDLINE | ID: mdl-26935058

Patients with advanced epithelial ovarian cancer often experience disease recurrence after standard therapies, a critical factor in determining their five-year survival rate. Recent reports indicated that long-term or short-term survival is associated with varied gene expression of cancer cells. Thus, identification of novel prognostic biomarkers should be considered. Since the mouse genome is similar to the human genome, we explored potential prognostic biomarkers using two groups of mouse ovarian cancer cell lines (group 1: IG-10, IG-10pw, and IG-10pw/agar; group 2: IG-10 clones 2, 3, and 11) which display highly and moderately aggressive phenotypes in vivo. Mice injected with these cell lines have different survival time and rates, capacities of tumor, and ascites formations, reflecting different prognostic potentials. Using an Affymetrix Mouse Genome 430 2.0 Array, a total of 181 genes were differentially expressed (P < 0.01) by at least twofold between two groups of the cell lines. Of the 181 genes, 109 and 72 genes were overexpressed in highly and moderately aggressive cell lines, respectively. Analysis of the 109 and 72 genes using Ingenuity Pathway Analysis (IPA) tool revealed two cancer-related gene networks. One was associated with the highly aggressive cell lines and affiliated with MYC gene, and another was associated with the moderately aggressive cell lines and affiliated with the androgen receptor (AR). Finally, the gene enrichment analysis indicated that the overexpressed 89 genes (out of 109 genes) in highly aggressive cell lines had a function annotation in the David database. The cancer-relevant significant gene ontology (GO) terms included Cell cycle, DNA metabolic process, and Programmed cell death. None of the genes from a set of the 72 genes overexpressed in the moderately aggressive cell lines had a function annotation in the David database. Our results suggested that the overexpressed MYC and 109 gene set represented highly aggressive ovarian cancer potential biomarkers while overexpressed AR and 72 gene set represented moderately aggressive ovarian cancer potential biomarkers. Based on our knowledge, the current study is first time to report the potential biomarkers relevant to different aggressive ovarian cancer. These potential biomarkers provide important information for investigating human ovarian cancer prognosis.


Biomarkers, Tumor/genetics , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Transcriptome , Animals , Carcinoma, Ovarian Epithelial , Disease Models, Animal , Female , Gene Expression Profiling , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
8.
PLoS One ; 9(11): e108192, 2014.
Article En | MEDLINE | ID: mdl-25383875

The association of B7-1/CD28 between antigen presenting cells (APCs) and T-cells provides a second signal to proliferate and activate T-cell immunity at the induction phase. Many reports indicate that tumor cells transfected with B7-1 induced augmented antitumor immunity at the induction phase by mimicking APC function; however, the function of B7-1 on antitumor immunity at the effector phase is unknown. Here, we report direct evidence of enhanced T-cell antitumor immunity at the effector phase by the B7-1 molecule. Our experiments in vivo and in vitro indicated that reactivity of antigen-specific monoclonal and polyclonal T-cell effectors against a Lass5 epitope presented by RMA-S cells is increased when the cells expressed B7-1. Use of either anti-B7-1 or anti-CD28 antibodies to block the B7-1/CD28 association reduced reactivity of the T effectors against B7-1 positive RMA-S cells. Transfection of Lass5 cDNA into or pulse of Lass5 peptide onto B7-1 positive RMA-S cells overcomes the requirement of the B7-1/CD28 signal for T effector response. To our knowledge, the data offers, for the first time, strong evidence that supports the requirement of B7-1/CD28 secondary signal at the effector phase of antitumor T-cell immunity being dependent on the density of an antigenic peptide.


Antigen-Presenting Cells/immunology , B7-1 Antigen/immunology , CD28 Antigens/immunology , Immunity, Cellular/immunology , Membrane Proteins/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Flow Cytometry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction
9.
PLoS One ; 7(7): e41226, 2012.
Article En | MEDLINE | ID: mdl-22815976

Human albumin has recently been demonstrated to protect brain neurons from injury in rat ischemic brain. However, there is no information available about whether human albumin can prevent loss of tyrosine hydroxylase (TH) expression of dopaminergic (DA) neurons induced by 6-hydroxydopamine (6-OHDA) toxicity that is most commonly used to create a rat model of Parkinson's disease (PD). In the present study, two microliters of 1.25% human albumin were stereotaxically injected into the right striatum of rats one day before or 7 days after the 6-OHDA lesion in the same side. D-Amphetamine-induced rotational asymmetry was measured 7 days, 3 and 10 weeks after 6-OHDA lesion. We observed that intrastriatal administration of human albumin significantly reduced the degree of rotational asymmetry. The number of TH-immunoreactive neurons present in the substantia nigra was greater in 6-OHDA lesioned rats following human albumin-treatment than non-human albumin treatment. TH-immunoreactivity in the 6-OHDA-lesioned striatum was also significantly increased in the human albumin-treated rats. To examine the mechanisms underlying the effects of human albumin, we challenged PC12 cells with 6-OHDA as an in vitro model of PD. Incubation with human albumin prevented 6-OHDA-induced reduction of cell viability in PC12 cell cultures, as measured by MTT assay. Furthermore, human albumin reduced 6-OHDA-induced formation of reactive oxygen species (ROS) and apoptosis in cultured PC12 cells, as assessed by flow cytometry. Western blot analysis showed that human albumin inhibited 6-OHDA-induced activation of JNK, c-Jun, ERK, and p38 mitogen-activated protein kinases (MAPK) signaling in PC12 cultures challenged with 6-OHDA. Human albumin may protect against 6-OHDA toxicity by influencing MAPK pathway followed by anti-ROS formation and anti-apoptosis.


Albumins/chemistry , Oxidopamine/pharmacology , Parkinson Disease/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Caspase 3/metabolism , Cell Line, Tumor , Dextroamphetamine/pharmacology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Female , Gene Expression Regulation, Enzymologic , Humans , In Vitro Techniques , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Time Factors
10.
Int J Biochem Cell Biol ; 44(11): 1770-8, 2012 Nov.
Article En | MEDLINE | ID: mdl-22728310

Myelosuppression and drug resistance are common adverse effects in cancer patients with chemotherapy, and those severely limit the therapeutic efficacy and lead treatment failure. It is unclear by which cellular mechanism anticancer drugs suppress bone marrow, while drug-resistant tumors survive. We report that due to the difference of glucosylceramide synthase (GCS), catalyzing ceramide glycosylation, doxorubicin (Dox) eliminates bone marrow stem cells (BMSCs) and expands breast cancer stem cells (BCSCs). It was found that Dox decreased the numbers of BMSCs (ABCG2(+)) and the sphere formation in a dose-dependent fashion in isolated bone marrow cells. In tumor-bearing mice, Dox treatments (5mg/kg, 6 days) decreased the numbers of BMSCs and white blood cells; conversely, those treatments increased the numbers of BCSCs (CD24(-)/CD44(+)/ESA(+)) more than threefold in the same mice. Furthermore, therapeutic-dose of Dox (1mg/kg/week, 42 days) decreased the numbers of BMSCs while it increased BCSCs in vivo. Breast cancer cells, rather than bone marrow cells, highly expressed GCS, which was induced by Dox and correlated with BCSC pluripotency. These results indicate that Dox may have opposite effects, suppressing BMSCs versus expanding BCSCs, and GCS is one determinant of the differentiated responsiveness of bone marrow and cancer cells.


Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Breast Neoplasms/pathology , Doxorubicin/toxicity , Glucosyltransferases/metabolism , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Blood Cell Count , Bone Marrow Cells/drug effects , Breast Neoplasms/blood , Breast Neoplasms/enzymology , Dose-Response Relationship, Drug , Female , Humans , Mice , Neoplastic Stem Cells/drug effects , Paclitaxel/toxicity , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tumor Cells, Cultured
11.
Methods Mol Biol ; 716: 179-91, 2011.
Article En | MEDLINE | ID: mdl-21318907

Flow cytometry can sensitively detect and efficiently sort cells based on fluorescent signals integrated into cellular markers of proteins or DNA. It has been broadly applied to assess cell division, apoptosis and to isolate cells including stem cells. As the seeds for tumorigenesis and metastasis, cancer stem cells (CSCs) are often more resistant to cytotoxins and anticancer agents than other heterogeneous cells in tumors. Analyzing CSCs under treatments is an effective way to evaluate new therapeutic agents for cancers. We introduce a method using flow cytometry to assess breast CSCs (CD44(+)/CD24(-)/(low)) in human MCF-7/Dox breast cancer cells, after the treatment of mixed-backbone oligonucleotide against glucosylceramide synthase. Flow cytometry analysis of CSCs is a reliable, effective, and easy-handling approach to screen agents targeting CSCs.


Breast Neoplasms/drug therapy , Drug Screening Assays, Antitumor/methods , Flow Cytometry/methods , Neoplastic Stem Cells/drug effects , Antineoplastic Agents/pharmacology , CD24 Antigen/immunology , Cell Line, Tumor , Female , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Humans , Hyaluronan Receptors/immunology , Neoplastic Stem Cells/immunology , Oligonucleotides/pharmacology
12.
J Nanosci Nanotechnol ; 11(11): 9997-10002, 2011 Nov.
Article En | MEDLINE | ID: mdl-22413338

A monolayer of a ferrocenyl hemicyanine was covalently self-assembled on an indium tin oxide (ITO)-coated glass substrate, and was characterized by UV/Vis absorption and X-ray photoelectron spectroscopy, and cyclic voltammetry. The photoelectrochemical properties and mechanism of photocurrent generation have also been studied. This monolayer film was found to exhibit a large anodic photocurrent density of 0.13 microA/cm2 with the highest photoelectric yield of 3.32% under irradiation of white light (730 nm > lambda > 325 nm) at a bias potential of +0.4 V versus saturated calomel electrode.

13.
J Lipid Res ; 51(4): 866-74, 2010 Apr.
Article En | MEDLINE | ID: mdl-19826105

Glucosylceramide synthase (GCS or GlcT-1), converting ceramide to glucosylceramide, is a key enzyme for the synthesis of glycosphingolipids. Due to its diverse roles in physiology and diseases, GCS may be a disease marker and drug target. Current assays for enzymes including GCS are based on reactions conducted in a test tube using enzyme preparations. Measurement of enzyme activity in laboratory-made conditions cannot directly evaluate the role of GCS in cells. Here, we introduce a new approach to determine GCS cellular activity using fluorescent NBD C6-ceramide in vivo. Cellular GCS transfers UDP-glucose to NBD C6-ceramide and produces NBD C6-glucosylceramide. C6-glucosylceramide is then separated from C6-ceramide by thin-layer chromatography and both are then quantitated by spectrophotometer. This cell-based method is able to quantitate glucosylceramide in pmol range, produced by approximately 50,000 cells or 1.0 mg tissue. This method has been used successfully to evaluate the degrees of GCS enzyme in cells and in tumors subjected to gene manipulation and chemical inhibition. These data indicate that this cell-based fluorescent method is direct, reproducible, and simple for assessing ceramide glycosylation. It is applicable to validate GCS activity in drug-resistant cancers and in other disorders.


Ceramides/analysis , Enzyme Assays/methods , Glucosyltransferases/metabolism , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/metabolism , Animals , Cell Line, Tumor , Cell Membrane Permeability , Ceramides/metabolism , Chromatography, Thin Layer , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Female , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , Glycosylation , Humans , Injections, Intralesional , Mice , Mice, Nude , Morpholines/administration & dosage , Morpholines/therapeutic use , Oligonucleotides/administration & dosage , Oligonucleotides/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Random Allocation , Reproducibility of Results , Xenograft Model Antitumor Assays
14.
PLoS One ; 4(9): e6938, 2009 Sep 09.
Article En | MEDLINE | ID: mdl-19742320

Enhanced ceramide glycosylation catalyzed by glucosylceramide synthase (GCS) limits therapeutic efficiencies of antineoplastic agents including doxorubicin in drug-resistant cancer cells. Aimed to determine the role of GCS in tumor response to chemotherapy, a new mixed-backbone oligonucleotide (MBO-asGCS) with higher stability and efficiency has been generated to silence human GCS gene. MBO-asGCS was taken up efficiently in both drug-sensitive and drug-resistant cells, but it selectively suppressed GCS overexpression, and sensitized drug-resistant cells. MBO-asGCS increased doxorubicin sensitivity by 83-fold in human NCI/ADR-RES, and 43-fold in murine EMT6/AR1 breast cancer cells, respectively. In tumor-bearing mice, MBO-asGCS treatment dramatically inhibited the growth of multidrug-resistant NCI/ADR-RE tumors, decreasing tumor volume to 37%, as compared with scrambled control. Furthermore, MBO-asGCS sensitized multidrug-resistant tumors to chemotherapy, increasing doxorubicin efficiency greater than 2-fold. The sensitization effects of MBO-asGCS relied on the decreases of gene expression and enzyme activity of GCS, and on the increases of C(18)-ceramide and of caspase-executed apoptosis. MBO-asGCS was accumulation in tumor xenografts was greater in other tissues, excepting liver and kidneys; but MBO-asGCS did not exert significant toxic effects on liver and kidneys. This study, for the first time in vivo, has demonstrated that GCS is a promising therapeutic target for cancer drug resistance, and MBO-asGCS has the potential to be developed as an antineoplastic agent.


Apoptosis , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Glucosyltransferases/antagonists & inhibitors , Oligonucleotides/pharmacology , Animals , Cell Line, Tumor , Cell Survival , Female , Glucosyltransferases/genetics , Humans , Kidney/metabolism , Liver/metabolism , Mice , Mice, Nude , Neoplasm Transplantation
15.
PLoS One ; 4(7): e6385, 2009 Jul 24.
Article En | MEDLINE | ID: mdl-19629186

Tumors deficient in expression of the transporter associated with antigen processing (TAP) usually fail to induce T-cell-mediated immunity and are resistant to T-cell lysis. However, we have found that introduction of the B7.1 gene into TAP-negative (TAP(-)) or TAP1-transfected (TAP1(+)) murine lung carcinoma CMT.64 cells can augment the capacity of the cells to induce a protective immune response against wild-type tumor cells. Differences in the strength of the protective immune responses were observed between TAP(-) and TAP1(+) B7.1 expressing CMT.64 cells depending on the doses of gamma-irradiated cell immunization. While mice immunized with either high or low dose of B7.1-expressing TAP1(+) cells rejected TAP(-) tumors, only high dose immunization with B7.1-expressing TAP(-) cells resulted in tumor rejection. The induced protective immunity was T-cell dependent as demonstrated by dramatically reduced antitumor immunity in mice depleted of CD8 or CD4 cells. Augmentation of T-cell mediated immune response against TAP(-) tumor cells was also observed in a virally infected tumor cell system. When mice were immunized with a high dose of gamma-irradiated CMT.64 cells infected with vaccinia viruses carrying B7.1 and/or TAP1 genes, we found that the cells co-expressing B7.1 and TAP1, but not those expressing B7.1 alone, induced protective immunity against CMT.64 cells. In addition, inoculation with live tumor cells transfected with several different gene(s) revealed that only B7.1- and TAP1-coexpressing tumor cells significantly decreased tumorigenicity. These results indicate that B7.1-provoked antitumor immunity against TAP(-) cancer is facilitated by TAP1-expression, and thus both genes should be considered for cancer therapy in the future.


ATP-Binding Cassette Transporters/immunology , B7-1 Antigen/immunology , T-Lymphocytes/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , Animals , Cell Line, Tumor , Mice
16.
Immunology ; 128(3): 420-8, 2009 Nov.
Article En | MEDLINE | ID: mdl-20067541

We previously showed that introduction of transporter associated with antigen processing (TAP) 1 into TAP-negative CMT.64, a major histocompatibility complex class I (MHC-I) down-regulated mouse lung carcinoma cell line, enhanced T-cell immunity against TAP-deficient tumour cells. Here, we have addressed two questions: (1) whether such immunity can be further augmented by co-expression of TAP1 with B7.1 or H-2K(b) genes, and (2) which T-cell priming mechanism (tumour direct priming or dendritic cell cross-priming) plays the major role in inducing an immune response against TAP-deficient tumours. We introduced the B7.1 or H-2K(b) gene into TAP1-expressing CMT.64 cells and determined which gene co-expressed with TAP1 was able to provide greater protective immunity against TAP-deficient tumour cells. Our results show that immunization of mice with B7.1 and TAP1 co-expressing but not H-2K(b) and TAP1 co-expressing CMT.64 cells dramatically augments T-cell-mediated immunity, as shown by an increase in survival of mice inoculated with live CMT.64 cells. In addition, our results suggest that induction of T-cell-mediated immunity against TAP-deficient tumour cells could be mainly through tumour direct priming rather than dendritic cell cross-priming as they show that T cells generated by tumour cell-lysate-loaded dendritic cells recognized TAP-deficient tumour cells much less than TAP-proficient tumour cells. These data suggest that direct priming by TAP1 and B7.1 co-expressing tumour cells is potentially a major mechanism to facilitate immune responses against TAP-deficient tumour cells.


ATP-Binding Cassette Transporters/genetics , B7-1 Antigen/metabolism , Carcinoma, Lewis Lung/immunology , H-2 Antigens/metabolism , Transgenes/genetics , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cross-Priming , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , H-2 Antigens/genetics , H-2 Antigens/immunology , Immunotherapy, Adoptive , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Transfection
17.
Zhongguo Gu Shang ; 22(12): 914-6, 2009 Dec.
Article Zh | MEDLINE | ID: mdl-20112574

OBJECTIVE: To compare therapeutic effects between moxibustion and infrared therapy for the treatment of knee osteoarthritis. METHODS: From January 2007 to June 2008 period, 65 patients with knee osteoarthritis were divided into treatment and control groups randomly uniform random number table generated from SAS statistical software. Among 35 patients in the treatment group, 17 patients were male and 18 patients were female, ranging in age from 45 to 75 years, with an average of (61.2+/-6.4) years; the course of disease ranged from 9 to 43 months, with a mean of (23.6+/-13.8) months; the preoperative Lysholm score ranged from 19 to 28 scores, averaged (24.3+/-3.3) scores. In the control group, there were 30 patients, including 13 males and 17 females, ranging in age from 47 to 79 years, with an average of (62.5+/-9.3) years; the course of disease ranged from 8 to 45 months, with a mean of (24.6+/-16.6) months; the preoperative Lysholm score ranged from 20 to 29 scores, averaged (25.9+/-3.0) scores. The patients in the treatment group were treated with moxibustion, and the patients in control group were treated with infrared therapy. All the patients were followed up for 4 weeks. The Lysholm scores were compared between the two groups. RESULTS: According to Lysholm score for clinical efficacy, treatment group got (87.5+/-5.6) scores and the control group were (85.9+/-3.5) scores, the Lysholm score of the treatment group was higher than that of the control group (P<0.05). Among pain score, joint flexion and extension score, joint stability score, and up and down stairs score, the pain and joint stability scores of patients in the treatment group were higher than those of control group (P<0.05). CONCLUSION: Compared with infrared therapy, moxibustion treatment for knee osteoarthritis can get better joint function, which is effect to alleviate the patient's pain, improve joint stability, improve the efficacy, and is valued to be promoted.


Moxibustion/methods , Osteoarthritis, Knee/therapy , Aged , Female , Humans , Male , Middle Aged , Multicenter Studies as Topic , Treatment Outcome
18.
Zhongguo Gu Shang ; 22(11): 813-5, 2009 Nov.
Article Zh | MEDLINE | ID: mdl-20084933

OBJECTIVE: To investigate and research WHOQOL-BREF evaluation of the clinical effect of application of moxibustion for treatment of knee osteoarthritis, to provide clinical according for the treatment. METHODS: From January 2007 to June 2008, 90 cases of knee osteoarthritis were randomly divided into treatment group and control group. In treatment group there were 45 cases included 16 males and 29 females with an average age of (62.5 +/- 7.4) years; the average course was (26.5 +/- 14.6) months; Lysholm score of knee function before treatment was (65.5 +/- 3.5) hours on average. In control group, there were 45 cases included 11 males and 34 females with an average age of (62.5 +/- 9.3) years;the average course was (24.6 +/- 16.6) months; Lysholm score of knee function before treatment was (66.3 +/- 2.3) hours on average. Applied Quality of Life Scale WHOQOL-BREF in line with the inclusion criteria of the study evaluation. While applied Lysholm scoring of knee joint for supporting the evaluation criteria. All data were statistical analyzed by package SPSS 11.5. RESULTS: Moxibustion was applied to treat the knee osteoarthritis and the quality of life was evaluated according to the WHOQOL-BREF scale score. The scores of the physical state, psychological state, as well as their own general health status of subjective feeling, and so on in treatment was significantly higher than that in control group (P < 0.05). According to Lysholm scoring, the score in treatment group was significantly higher than that of control group, too (t = 0.65, P < 0.05). CONCLUSION: Application of moxibustion for treatment of knee osteoarthritis is a simple, economical and practical, and can avoid the further development of the disease, but also to improve the joint function and improve quality of life.


Moxibustion , Osteoarthritis, Knee/therapy , Quality of Life , World Health Organization , Aged , Female , Humans , Male , Middle Aged , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/physiopathology , Treatment Outcome
19.
Tumour Biol ; 29(6): 359-70, 2008.
Article En | MEDLINE | ID: mdl-19052485

Ovarian cancer is the fifth most common cause of cancer death in women. Due to a lack of appropriate animal models, studies involving tumorigenicity, tumor progression and immune response at the molecular level are limited. We isolated many clones derived from thesurvivors of a transformed mouse ovarian epithelial cell line IG-10 in immune- competent mice and found that the clones displayed diverse phenotypes. Most clones were deficient in components of the MHC-I antigen presentation pathway. Soft-agarose colony assays showed different growth rates among clones. However, this did not completely correlate with each clone's in vivo tumorigenicity regarding growth, tumor mass and ascites formation, suggesting the possibility that the clones may display contrasting intrinsic gene expression. We therefore performed two types of arrays to evaluate gene expression at transcriptional and translational levels. The results showed differences in expression of COL4alpha5, NOS-2, and SOCS-1 genes at the transcriptional level, MIP-2 gene at the protein level and CCL5, CXCL-10, IL-1alpha genes at both transcriptional and protein levels between low and high tumorigenic clones. Thus, our animal cell model together with the identified genes may provide a useful tool to study ovarian cancer immune response, tumorigenicity and tumor-host cell interactions in the tumor microenvironment.


Cell Transformation, Neoplastic , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Animals , Cell Line, Transformed , Disease Models, Animal , Female , Genes, MHC Class I , Humans , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Phenotype
20.
PLoS One ; 3(8): e3097, 2008 Aug 29.
Article En | MEDLINE | ID: mdl-18769733

The transporter associated with antigen processing (TAP) and the major histocompatibility complex class I (MHC-I), two important components of the MHC-I antigen presentation pathway, are often deficient in tumor cells. The restoration of their expression has been shown to restore the antigenicity and immunogenicity of tumor cells. However, it is unclear whether TAP and MHC-I expression in tumor cells can affect the induction phase of the T cell response. To address this issue, we expressed viral antigens in tumors that are either deficient or proficient in TAP and MHC-I expression. The relative efficiency of direct immunization or immunization through cross-presentation in promoting adaptive T cell responses was compared. The results demonstrated that stimulation of animals with TAP and MHC-I proficient tumor cells generated antigen specific T cells with greater killing activities than those of TAP and MHC-I deficient tumor cells. This discrepancy was traced to differences in the ability of dendritic cells (DCs) to access and sample different antigen reservoirs in TAP and MHC-I proficient versus deficient cells and thereby stimulate adaptive immune responses through the process of cross-presentation. In addition, our data suggest that the increased activity of T cells is caused by the enhanced DC uptake and utilization of MHC-I/peptide complexes from the proficient cells as an additional source of processed antigen. Furthermore, we demonstrate that immune-escape and metastasis are promoted in the absence of this DC 'arming' mechanism. Physiologically, this novel form of DC antigen sampling resembles trogocytosis, and acts to enhance T cell priming and increase the efficacy of adaptive immune responses against tumors and infectious pathogens.


Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Infections/immunology , Major Histocompatibility Complex , Neoplasms/immunology , T-Lymphocytes/immunology , Adaptation, Biological/immunology , Animals , Antigen-Presenting Cells/immunology , Cross-Priming/immunology , Gamma Rays , Histocompatibility Antigens Class I/radiation effects , Humans , Mice , T-Lymphocytes/radiation effects , T-Lymphocytes, Cytotoxic/immunology
...