Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nat Commun ; 13(1): 3057, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35650195

ABSTRACT

Desmoplastic small round cell tumor (DSRCT) is an aggressive, usually incurable sarcoma subtype that predominantly occurs in post-pubertal young males. Recent evidence suggests that the androgen receptor (AR) can promote tumor progression in DSRCTs. However, the mechanism of AR-induced oncogenic stimulation remains undetermined. Herein, we demonstrate that enzalutamide and AR-directed antisense oligonucleotides (AR-ASO) block 5α-dihydrotestosterone (DHT)-induced DSRCT cell proliferation and reduce xenograft tumor burden. Gene expression analysis and chromatin immunoprecipitation sequencing (ChIP-seq) were performed to elucidate how AR signaling regulates cellular epigenetic programs. Remarkably, ChIP-seq revealed novel DSRCT-specific AR DNA binding sites adjacent to key oncogenic regulators, including WT1 (the C-terminal partner of the pathognomonic fusion protein) and FOXF1. Additionally, AR occupied enhancer sites that regulate the Wnt pathway, neural differentiation, and embryonic organ development, implicating AR in dysfunctional cell lineage commitment. Our findings have direct clinical implications given the widespread availability of FDA-approved androgen-targeted agents used for prostate cancer.


Subject(s)
Androgen Receptor Antagonists , Desmoplastic Small Round Cell Tumor , Receptors, Androgen , Androgen Receptor Antagonists/pharmacology , Androgens , Animals , Cell Line, Tumor , Desmoplastic Small Round Cell Tumor/genetics , Humans , Male , Oligonucleotides, Antisense/pharmacology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays
2.
Gut ; 71(5): 938-949, 2022 05.
Article in English | MEDLINE | ID: mdl-34059508

ABSTRACT

OBJECTIVE: Enhancer aberrations are beginning to emerge as a key epigenetic feature of colorectal cancers (CRC), however, a comprehensive knowledge of chromatin state patterns in tumour progression, heterogeneity of these patterns and imparted therapeutic opportunities remain poorly described. DESIGN: We performed comprehensive epigenomic characterisation by mapping 222 chromatin profiles from 69 samples (33 colorectal adenocarcinomas, 4 adenomas, 21 matched normal tissues and 11 colon cancer cell lines) for six histone modification marks: H3K4me3 for Pol II-bound and CpG-rich promoters, H3K4me1 for poised enhancers, H3K27ac for enhancers and transcriptionally active promoters, H3K79me2 for transcribed regions, H3K27me3 for polycomb repressed regions and H3K9me3 for heterochromatin. RESULTS: We demonstrate that H3K27ac-marked active enhancer state could distinguish between different stages of CRC progression. By epigenomic editing, we present evidence that gains of tumour-specific enhancers for crucial oncogenes, such as ASCL2 and FZD10, was required for excessive proliferation. Consistently, combination of MEK plus bromodomain inhibition was found to have synergistic effects in CRC patient-derived xenograft models. Probing intertumour heterogeneity, we identified four distinct enhancer subtypes (EPIgenome-based Classification, EpiC), three of which correlate well with previously defined transcriptomic subtypes (consensus molecular subtypes, CMSs). Importantly, CMS2 can be divided into two EpiC subgroups with significant survival differences. Leveraging such correlation, we devised a combinatorial therapeutic strategy of enhancer-blocking bromodomain inhibitors with pathway-specific inhibitors (PARPi, EGFRi, TGFßi, mTORi and SRCi) for EpiC groups. CONCLUSION: Our data suggest that the dynamics of active enhancer underlies CRC progression and the patient-specific enhancer patterns can be leveraged for precision combination therapy.


Subject(s)
Chromatin , Colorectal Neoplasms , Basic Helix-Loop-Helix Transcription Factors , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Enhancer Elements, Genetic/genetics , Humans , Nuclear Proteins , Transcription Factors/genetics
3.
Mol Cell Oncol ; 8(5): 1984827, 2021.
Article in English | MEDLINE | ID: mdl-34859145

ABSTRACT

We reported that histone H3 lysine (K) 4 methyltransferase, KMT2D, serves as a potent tumor-suppressor in melanoma, which was identified via in vivo epigenome-focused RNA interference (RNAi) screen. KMT2D-deficient tumors show substantial reprogramming of key metabolic pathways including glycolysis via reduction of H3K4me1 (Histone H3K4 mono-methylation)-marked active enhancers, conferring sensitivity to inhibitors of glycolysis and IGFR (Insulin Growth Factor Receptor) pathway.

4.
Cell Rep ; 36(3): 109410, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289358

ABSTRACT

The dynamic evolution of chromatin state patterns during metastasis, their relationship with bona fide genetic drivers, and their therapeutic vulnerabilities are not completely understood. Combinatorial chromatin state profiling of 46 melanoma samples reveals an association of NRAS mutants with bivalent histone H3 lysine 27 trimethylation (H3K27me3) and Polycomb repressive complex 2. Reprogramming of bivalent domains during metastasis occurs on master transcription factors of a mesenchymal phenotype, including ZEB1, TWIST1, and CDH1. Resolution of bivalency using pharmacological inhibition of EZH2 decreases invasive capacity of melanoma cells and markedly reduces tumor burden in vivo, specifically in NRAS mutants. Coincident with bivalent reprogramming, the increased expression of pro-metastatic and melanocyte-specific cell-identity genes is associated with exceptionally wide H3K4me3 domains, suggesting a role for this epigenetic element. Overall, we demonstrate that reprogramming of bivalent and broad domains represents key epigenetic alterations in metastatic melanoma and that EZH2 plus MEK inhibition may provide a promising therapeutic strategy for NRAS mutant melanoma patients.


Subject(s)
Chromatin/metabolism , GTP Phosphohydrolases/genetics , Melanoma/genetics , Membrane Proteins/genetics , Mutation/genetics , Polycomb Repressive Complex 2/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , GTP Phosphohydrolases/metabolism , Histones/metabolism , Humans , Melanocytes/metabolism , Membrane Proteins/metabolism , Mesoderm/metabolism , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Metastasis , Polycomb Repressive Complex 2/metabolism , Transcription, Genetic , Tumor Burden
5.
Cell Rep ; 33(3): 108293, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086062

ABSTRACT

Histone methyltransferase KMT2D harbors frequent loss-of-function somatic point mutations in several tumor types, including melanoma. Here, we identify KMT2D as a potent tumor suppressor in melanoma through an in vivo epigenome-focused pooled RNAi screen and confirm the finding by using a genetically engineered mouse model (GEMM) based on conditional and melanocyte-specific deletion of KMT2D. KMT2D-deficient tumors show substantial reprogramming of key metabolic pathways, including glycolysis. KMT2D deficiency aberrantly upregulates glycolysis enzymes, intermediate metabolites, and glucose consumption rates. Mechanistically, KMT2D loss causes genome-wide reduction of H3K4me1-marked active enhancer chromatin states. Enhancer loss and subsequent repression of IGFBP5 activates IGF1R-AKT to increase glycolysis in KMT2D-deficient cells. Pharmacological inhibition of glycolysis and insulin growth factor (IGF) signaling reduce proliferation and tumorigenesis preferentially in KMT2D-deficient cells. We conclude that KMT2D loss promotes tumorigenesis by facilitating an increased use of the glycolysis pathway for enhanced biomass needs via enhancer reprogramming, thus presenting an opportunity for therapeutic intervention through glycolysis or IGF pathway inhibitors.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Melanoma/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genes, Tumor Suppressor , Glucose/metabolism , Glycolysis/genetics , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Humans , Insulin/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptor, IGF Type 1/metabolism , Regulatory Sequences, Nucleic Acid , Signal Transduction , Xenograft Model Antitumor Assays/methods
6.
Cancer Cell ; 37(4): 599-617.e7, 2020 04 13.
Article in English | MEDLINE | ID: mdl-32243837

ABSTRACT

Epigenetic modifiers frequently harbor loss-of-function mutations in lung cancer, but their tumor-suppressive roles are poorly characterized. Histone methyltransferase KMT2D (a COMPASS-like enzyme, also called MLL4) is among the most highly inactivated epigenetic modifiers in lung cancer. Here, we show that lung-specific loss of Kmt2d promotes lung tumorigenesis in mice and upregulates pro-tumorigenic programs, including glycolysis. Pharmacological inhibition of glycolysis preferentially impedes tumorigenicity of human lung cancer cells bearing KMT2D-inactivating mutations. Mechanistically, Kmt2d loss widely impairs epigenomic signals for super-enhancers/enhancers, including the super-enhancer for the circadian rhythm repressor Per2. Loss of Kmt2d decreases expression of PER2, which regulates multiple glycolytic genes. These findings indicate that KMT2D is a lung tumor suppressor and that KMT2D deficiency confers a therapeutic vulnerability to glycolytic inhibitors.


Subject(s)
Adenocarcinoma of Lung/pathology , DNA-Binding Proteins/antagonists & inhibitors , Deoxyglucose/pharmacology , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Glycolysis , Histone-Lysine N-Methyltransferase/physiology , Myeloid-Lymphoid Leukemia Protein/physiology , Neoplasm Proteins/antagonists & inhibitors , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/metabolism , Animals , Antimetabolites/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histones/genetics , Histones/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mice, Nude , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Nat Commun ; 10(1): 1398, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30923315

ABSTRACT

The roles of Plant Homeodomain (PHD) fingers in catalysis of histone modifications are unknown. We demonstrated that the PHD finger of Ubiquitin Protein Ligase E3 Component N-Recognin7 (UBR7) harbors E3 ubiquitin ligase activity toward monoubiquitination of histone H2B at lysine120 (H2BK120Ub). Purified PHD finger or full-length UBR7 monoubiquitinated H2BK120 in vitro, and loss of UBR7 drastically reduced H2BK120Ub genome-wide binding sites in MCF10A cells. Low UBR7 expression was correlated with occurrence of triple-negative breast cancer and metastatic tumors. Consistently, UBR7 knockdown enhanced the invasiveness, induced epithelial-to-mesenchymal transition and promoted metastasis. Conversely, ectopic expression of UBR7 restored these cellular phenotypes and reduced tumor growth. Mechanistically, UBR7 loss reduced H2BK120Ub levels on cell adhesion genes, including CDH4, and upregulated the Wnt/ß-Catenin signaling pathway. CDH4 overexpression could partially revert UBR7-dependent cellular phenotypes. Collectively, our results established UBR7 as a histone H2B monoubiquitin ligase that suppresses tumorigenesis and metastasis of triple-negative breast cancer.


Subject(s)
Carcinogenesis/genetics , Histone Code/genetics , Histones/metabolism , PHD Zinc Fingers/genetics , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Cadherins/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Transplantation, Heterologous , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics , Wnt Signaling Pathway
9.
J Vis Exp ; (134)2018 04 05.
Article in English | MEDLINE | ID: mdl-29683440

ABSTRACT

Histone modifications constitute a major component of the epigenome and play important regulatory roles in determining the transcriptional status of associated loci. In addition, the presence of specific modifications has been used to determine the position and identity non-coding functional elements such as enhancers. In recent years, chromatin immunoprecipitation followed by next generation sequencing (ChIP-seq) has become a powerful tool in determining the genome-wide profiles of individual histone modifications. However, it has become increasingly clear that the combinatorial patterns of chromatin modifications, referred to as Chromatin States, determine the identity and nature of the associated genomic locus. Therefore, workflows consisting of robust high-throughput (HT) methodologies for profiling a number of histone modification marks, as well as computational analyses pipelines capable of handling myriads of ChIP-Seq profiling datasets, are needed for comprehensive determination of epigenomic states in large number of samples. The HT-ChIP-Seq workflow presented here consists of two modules: 1) an experimental protocol for profiling several histone modifications from small amounts of tumor samples and cell lines in a 96-well format; and 2) a computational data analysis pipeline that combines existing tools to compute both individual mark occupancy and combinatorial chromatin state patterns. Together, these two modules facilitate easy processing of hundreds of ChIP-Seq samples in a fast and efficient manner. The workflow presented here is used to derive chromatin state patterns from 6 histone mark profiles in melanoma tumors and cell lines. Overall, we present a comprehensive ChIP-seq workflow that can be applied to dozens of human tumor samples and cancer cell lines to determine epigenomic aberrations in various malignancies.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/genetics , Chromosome Mapping/methods , High-Throughput Nucleotide Sequencing/methods , Humans
10.
Cell Rep ; 19(4): 875-889, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28445736

ABSTRACT

The extent and nature of epigenomic changes associated with melanoma progression is poorly understood. Through systematic epigenomic profiling of 35 epigenetic modifications and transcriptomic analysis, we define chromatin state changes associated with melanomagenesis by using a cell phenotypic model of non-tumorigenic and tumorigenic states. Computation of specific chromatin state transitions showed loss of histone acetylations and H3K4me2/3 on regulatory regions proximal to specific cancer-regulatory genes in important melanoma-driving cell signaling pathways. Importantly, such acetylation changes were also observed between benign nevi and malignant melanoma human tissues. Intriguingly, only a small fraction of chromatin state transitions correlated with expected changes in gene expression patterns. Restoration of acetylation levels on deacetylated loci by histone deacetylase (HDAC) inhibitors selectively blocked excessive proliferation in tumorigenic cells and human melanoma cells, suggesting functional roles of observed chromatin state transitions in driving hyperproliferative phenotype. Through these results, we define functionally relevant chromatin states associated with melanoma progression.


Subject(s)
Chromatin/metabolism , Epigenomics , Histones/metabolism , Acetylation , Cell Line , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Disease-Free Survival , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Kaplan-Meier Estimate , Melanoma/metabolism , Melanoma/mortality , Melanoma/pathology , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Principal Component Analysis , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Vorinostat
11.
Bioinformatics ; 32(4): 533-41, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26515818

ABSTRACT

MOTIVATION: A major goal of biomedical research is to identify molecular features associated with a biological or clinical class of interest. Differential expression analysis has long been used for this purpose; however, conventional methods perform poorly when applied to data with high within class heterogeneity. RESULTS: To address this challenge, we developed EMDomics, a new method that uses the Earth mover's distance to measure the overall difference between the distributions of a gene's expression in two classes of samples and uses permutations to obtain q-values for each gene. We applied EMDomics to the challenging problem of identifying genes associated with drug resistance in ovarian cancer. We also used simulated data to evaluate the performance of EMDomics, in terms of sensitivity and specificity for identifying differentially expressed gene in classes with high within class heterogeneity. In both the simulated and real biological data, EMDomics outperformed competing approaches for the identification of differentially expressed genes, and EMDomics was significantly more powerful than conventional methods for the identification of drug resistance-associated gene sets. EMDomics represents a new approach for the identification of genes differentially expressed between heterogeneous classes and has utility in a wide range of complex biomedical conditions in which sample classes show within class heterogeneity. AVAILABILITY AND IMPLEMENTATION: The R package is available at http://www.bioconductor.org/packages/release/bioc/html/EMDomics.html.


Subject(s)
Biomarkers, Tumor/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling/methods , Ovarian Neoplasms/genetics , Software , Antineoplastic Agents/pharmacology , Databases, Factual , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Humans , Ovarian Neoplasms/drug therapy , Sensitivity and Specificity
12.
Oncol Lett ; 7(4): 1102-1108, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24944676

ABSTRACT

Cisplatin is regularly used in the treatment of ovarian cancer. However, the drug only provides a modest survival advantage, primarily due to chemoresistance and the upregulation of antiapoptotic machineries in ovarian cancer cells. Therefore, targeting the mechanisms responsible for cisplatin resistance in ovarian cancer cells may improve the therapeutic outcomes. Twist basic helix-loop-helix transcription factor 2 (Twist2) is a novel zinc finger transcription factor that has been indicated to be an important inducer of epithelial-mesenchymal transition, which has been shown to be involved in various phases of tumorigenicity and progression. However, whether Twist2 suppression increases the chemosensitivity of ovarian cancer cells to chemotherapeutic agents remains unclear. In the present study, Twist2 expression was found to differ between human ovarian cisplatin-sensitive cancer cell line, OV2008, and the resistant variant, C13K cells. Twist2 plasmids or RNA interference were then utilized to alter Twist2 expression in OV2008 or C13K cells, respectively, to further assess apoptosis, cell viability and cell growth, as well as a possible mechanism. The results of the present study indicated that Twist2 plays a crucial role in the chemoresistance of ovarian cancer. In addition, the downregulation of Twist2 expression may facilitate apoptosis and recover the sensitivity of chemoresistant ovarian cancer through the protein kinase B/glycogen synthase kinase-3ß pathway. Therefore, Twist2 depletion may be a promising approach to ovarian cancer therapy.

13.
Reprod Biomed Online ; 26(1): 93-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23177411

ABSTRACT

Endometriosis is a steroid-dependent complex disease. The oestrogen receptor plays an important role by mediating oestrogen action and eutopic or ectopic endometrium development. This study investigated whether single-nucleotide polymorphisms in the genes for oestrogen receptor 1 (ESR1) and oestrogen receptor 2 (ESR2) are associated with endometriosis and endometriosis-related infertility. The participants included 157 infertile and 155 fertile endometriosis women as well as 92 women with primary infertility and 265 fertile women as controls. The iPLEX Gold system (MassARRAY system, Sequenom) was used for genotyping of ESR1 and ESR2. Statistical analysis showed that ESR1 (rs3798573 A/G) was significantly associated with endometriosis and endometriosis-related infertility (P=0.011, P=0.009). No association was found with ESR1 (rs1159327 A/G, rs3020348 A/C) and ESR2 (rs17179740 A/G) either for endometriosis or endometriosis-related infertility. According to the revised American Fertility Society classification, all of the detected single-nucleotide polymorphisms had no association with endometriosis in stage I-II or in stage III-IV. The results suggest that the ESR1 polymorphism rs3798573 A/G is associated with increased risk of endometriosis and endometriosis-related infertility in Han women from central China. Endometriosis is an oestrogen-dependent complex disease, which is one of the most common causes of infertility. Oestrogen receptors (ESR), which mediate oestrogen actions, are considered to play an essential role in the pathogenesis of endometriosis. Therefore, ESR may also play an important role in endometriosis-related infertility. To investigate the association between ESR and endometriosis or endometriosis-related infertility, detection of ESR polymorphisms have been carried out in several populations by other researchers; however, the results remain controversial. In a previous study of ours, through a pooling-based genome-wide scan of endometriosis and controls, we obtained two highly ranked single-nucleotide polymorphisms (SNP) that were individually located in introns of the genes ESR1 and ESR2. To find more evidence of a relationship between ESR and endometriosis or endometriosis-related infertility, the current study selected for genotyping another two ESR1 SNP from a Japanese genome-wide association study in endometriosis. According to the genotypes and the patients' histories, we found that the ESR1 polymorphism rs3798573 A/G was associated with risk of endometriosis and infertile endometriosis in Han women from central China.


Subject(s)
Endometriosis/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Infertility, Female/genetics , Polymorphism, Single Nucleotide , China , Endometriosis/complications , Female , Genetic Association Studies , Genotype , Humans , Infertility, Female/etiology
14.
J Immunother ; 34(8): 581-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21904218

ABSTRACT

LIGHT is a tumor necrosis factor superfamily ligand that is considered as a promising candidate for cancer therapy. It has a potent antitumor activity through establishing lymphoid-like tissues inside tumor sites and recruiting naive T cells into the tumor. In this study, we examined the possibility of antitumor activity by expressing LIGHT in cervical cancer (CC) model. A recombinant adeno-associated virus (AAV) vector was chosen for the transfer, based on its transfection efficiency and lack of detectable pathology. In vitro transfer of recombinant AAV vector expressing LIGHT (AAV-LIGHT) stimulated T-lymphocyte proliferation and activation. AAV-mediated gene transfer of LIGHT by intratumoral injection exerted a very potent antitumor effect against preexisting TC-1 cell CC in C57BL/6 mice. This study confirmed that AAV-LIGHT regressed tumor growth by activating cytotoxic T lymphocyte, enhancing infiltration of inflammatory cells in tumor and increasing stimulatory cytokine expression in tumor microenvironment. Therefore, AAV-LIGHT therapy might have potential utility for the treatment of CC.


Subject(s)
Cell Transformation, Neoplastic , Dependovirus/genetics , Genetic Vectors/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Uterine Cervical Neoplasms/therapy , Animals , CHO Cells , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cricetinae , Disease Models, Animal , Female , Gene Transfer Techniques , HEK293 Cells , Humans , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Spleen/immunology , Spleen/metabolism , Transduction, Genetic , Tumor Burden/genetics , Tumor Burden/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Uterine Cervical Neoplasms/immunology , Xenograft Model Antitumor Assays
15.
J Immunol ; 183(12): 7842-50, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19923459

ABSTRACT

B and T lymphocyte attenuator (BTLA)-herpesvirus entry mediator (HVEM) signaling coinhibitory pathway is believed to impair antitumor immune competences. An intriguing unresolved question is whether blockade of BTLA-HVEM guides an effective therapeutic tool against established tumors. To address this issue, we constructed a eukaryotic expression plasmid (psBTLA) that expressed the extracellular domain of murine BTLA (soluble form of BTLA), which could bind HVEM, the ligand of BTLA, and block BTLA-HVEM interactions. The data in this study showed that treatment by injection of psBTLA resulted in down-regulation of IL-10 and TGF-beta and promotion of dendritic cell function by increasing the expression of B7-1 and IL-12, but the adaptive antitumor immune responses achieved by psBTLA administration alone were limited and could not eradicate the tumor effectively. Next, we evaluated the immunotherapeutic efficacy and mechanism of combination therapy of heat shock protein 70 (HSP70) vaccine/psBTLA by using murine TC-1 cervical cancer mice as an ectopic tumor model. Our in vivo studies revealed that treatment with HSP70 vaccine alone did not lead to satisfactory tumor growth inhibition, whereas cotreatment with psBTLA significantly improved antitumor immunity and compensated the deficiency of HSP70 vaccine by increasing the expression of Th1 cytokines, IL-2, and IFN-gamma and decreasing transcription levels of IL-10, TGF-beta, and Foxp3 in the tumor microenvironment. Taken together, our findings indicate that blocking the BTLA-HVEM interaction with sBTLA enhances antitumor efficacy and results in a significant synergistic effect against existent tumor cells in vivo when combined with the HSP70 vaccine.


Subject(s)
B-Lymphocyte Subsets/immunology , HSP70 Heat-Shock Proteins/immunology , Receptors, Immunologic/physiology , Receptors, Tumor Necrosis Factor, Member 14/physiology , T-Lymphocyte Subsets/immunology , Uterine Cervical Neoplasms/prevention & control , Vaccines, DNA/immunology , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Disease Models, Animal , Extracellular Space/immunology , Extracellular Space/metabolism , Female , HSP70 Heat-Shock Proteins/administration & dosage , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/prevention & control , Mice , Protein Structure, Tertiary , Receptors, Immunologic/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors , Signal Transduction/genetics , Signal Transduction/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/metabolism , Vaccines, DNA/administration & dosage
16.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-404844

ABSTRACT

Objective To construct eukaryotic expressing vector of mouse soluble CD160 and stably transfect into CHO cells for eukaryotic expression.Methods Recombinant soluble CD160(rsCD160) was constructed by gene recombination.Total RNA was extracted from the spleen of C57BL/6 mice.cDNA was amplified for the soluble form of CD160.Then,the PCR product was cloned tO pcDNA3.1 and pEGFP-N1.The recombinant plasmid was identified by restriction map and sequence analy-sis.The soluble CDl 60 expression in CHO cells transfected with recombinant psCDl 60 was verified by RT-PCR and Westernblot.The binding ability of psCD160 tO its ligand was detected by FACS.Results 520 bp mouse soluble CD160 gene was obtained.Recombinant mouse psCD160 was successfully constructed.After transfection,soluble CD160 expression in the culture supernatant of CHO cells was successfully detected.FACS analysis indicated that soluble CD160 could bind tO its ligand.Conclusion Recombinant mouse psCD160 is successfully constructed,which will benefit our further study on soluble CD160 for immune therapy against tumor in the future experiments.

SELECTION OF CITATIONS
SEARCH DETAIL
...