39 results on '"Meiying Zhang"'
Search Results
2. KAT6A, a novel regulator of β-catenin, promotes tumorigenicity and chemoresistance in ovarian cancer by acetylating COP1
- Author
-
Qiqi Shi, Mingda Zhang, Yanxin Li, Wenxue Liu, Zhiyan Zhan, Xiuying Xiao, Haizhong Feng, Bowen Sun, Weiwei Zhang, Fei Luo, Yanli Hou, and Meiying Zhang
- Subjects
0301 basic medicine ,Medicine (miscellaneous) ,Apoptosis ,medicine.disease_cause ,Metastasis ,Gene Knockout Techniques ,0302 clinical medicine ,Protein Interaction Mapping ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Tumor Stem Cell Assay ,beta Catenin ,Histone Acetyltransferases ,Ovarian Neoplasms ,biology ,COP1 ,Prognosis ,Neoplasm Proteins ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,ovarian cancer ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Disease Progression ,Heterografts ,Female ,Research Paper ,Signal Transduction ,medicine.drug ,Ubiquitin-Protein Ligases ,Mice, Nude ,KAT6A ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Antineoplastic Agents, Alkylating ,acetylation ,Cisplatin ,Oncogene ,business.industry ,Ubiquitination ,β-catenin ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Catenin ,biology.protein ,Cancer research ,Ovarian cancer ,Carcinogenesis ,business ,Protein Processing, Post-Translational - Abstract
Background: Ovarian cancer is a fatal gynecologic malignancy that is found worldwide and exhibits an insidious onset and a lack of early warning symptoms. Despite ongoing studies, the mechanistic basis of the aggressive phenotypes of ovarian cancer remains unclear. Lysine acetyltransferase 6A (KAT6A) is a MYST-type histone acetyltransferase (HAT) enzyme identified as an oncogene in breast cancer, glioblastoma and leukemia. However, the specific functions of KAT6A in ovarian cancer remain unclear. Methods: Immunohistochemistry (IHC) staining and western blotting were performed to characterize KAT6A protein expression in ovarian cancer tissues and cell lines. The biological functions of KAT6A in ovarian cancer were evaluated by cell proliferation, wound healing and transwell invasion assays in vitro. Tumorigenesis and metastasis assays were performed in nude mice to detect the role of KAT6A in vivo. Mass spectrometry and immunoprecipitation assays were performed to detect the KAT6A-COP1 interaction. An in vivo ubiquitination assay was performed to determine the regulation of β-catenin by KAT6A. Results: In the present study, we revealed that KAT6A expression is upregulated in ovarian cancer and is associated with patient overall survival. Downregulation of KAT6A markedly inhibited the proliferation and migration abilities of ovarian cancer cells in vivo and in vitro. Additionally, the inhibition of KAT6A induced apoptosis and enhanced the sensitivity of ovarian cancer cells to cisplatin. Furthermore, KAT6A bound to and acetylated COP1 at K294. The acetylation of COP1 impaired COP1 function as an E3 ubiquitin ligase and led to the accumulation and enhanced activity of β-catenin. Conclusions: Our findings suggest that the KAT6A/COP1/β-catenin signaling axis plays a critical role in ovarian cancer progression and that targeting the KAT6A/COP1/β-catenin signaling axis could be a novel strategy for treating ovarian cancer.
- Published
- 2021
3. Intratumor Epigenetic Heterogeneity—A Panel Gene Methylation Study in Thyroid Cancer
- Author
-
Chaofan Zhu, Meiying Zhang, Qian Wang, Jin Jen, Baoguo Liu, and Mingzhou Guo
- Subjects
DNA methylation ,CDH1 ,Genetic heterogeneity ,epigenetic heterogeneity ,Bisulfite sequencing ,Cancer ,Methylation ,RASSF1A ,Biology ,QH426-470 ,medicine.disease ,Malignancy ,DACT2 ,Cancer research ,medicine ,Genetics ,Molecular Medicine ,Epigenetics ,Thyroid cancer ,Genetics (clinical) ,AP2 ,HIN1 ,Original Research - Abstract
BackgroundThyroid cancer (TC) is the most common endocrine malignancy, and the incidence is increasing very fast. Surgical resection and radioactive iodine ablation are major therapeutic methods, however, around 10% of differentiated thyroid cancer and all anaplastic thyroid carcinoma (ATC) are failed. Comprehensive understanding the molecular mechanisms may provide new therapeutic strategies for thyroid cancer. Even though genetic heterogeneity is rigorously studied in various cancers, epigenetic heterogeneity in human cancer remains unclear.MethodsA total of 405 surgical resected thyroid cancer samples were employed (three spatially isolated specimens were obtained from different regions of the same tumor). Twenty-four genes were selected for methylation screening, and frequently methylated genes in thyroid cancer were used for further validation. Methylation specific PCR (MSP) approach was employed to detect the gene promoter region methylation.ResultsFive genes (AP2, CDH1, DACT2, HIN1, and RASSF1A) are found frequently methylated (>30%) in thyroid cancer. The five genes panel is used for further epigenetic heterogeneity analysis. AP2 methylation is associated with gender (P < 0.05), DACT2 methylation is associated with age, gender and tumor size (all P < 0.05), HIN1 methylation is associated to tumor size (P < 0.05) and extra-thyroidal extension (P < 0.01). RASSF1A methylation is associated with lymph node metastasis (P < 0.01). For heterogeneity analysis, AP2 methylation heterogeneity is associated with tumor size (P < 0.01), CDH1 methylation heterogeneity is associated with lymph node metastasis (P < 0.05), DACT2 methylation heterogeneity is associated with tumor size (P < 0.01), HIN1 methylation heterogeneity is associated with tumor size and extra-thyroidal extension (all P < 0.01). The multivariable analysis suggested that the risk of lymph node metastasis is 2.5 times in CDH1 heterogeneous methylation group (OR = 2.512, 95% CI 1.135, 5.557, P = 0.023). The risk of extra-thyroidal extension is almost 3 times in HIN1 heterogeneous methylation group (OR = 2.607, 95% CI 1.138, 5.971, P = 0.023).ConclusionFive of twenty-four genes were found frequently methylated in human thyroid cancer. Based on 5 genes panel analysis, epigenetic heterogeneity is an universal event. Epigenetic heterogeneity is associated with cancer development and progression.
- Published
- 2021
4. Proactive risk assessment of intrahospital transport of critically ill patients from emergency department to intensive care unit in a teaching hospital and its implications
- Author
-
Meiying Zhang, Xueyan Li, Weiying Zhang, Runv Zhou, Hongming Gu, Jianhong Lv, Xiaoyan Ma, and Jin Zhao
- Subjects
Safety Management ,Critical Illness ,Risk Assessment ,law.invention ,Teaching hospital ,law ,Medicine ,Humans ,Hospitals, Teaching ,General Nursing ,business.industry ,Critically ill ,General Medicine ,Emergency department ,medicine.disease ,Intensive care unit ,Project team ,Checklist ,Intensive Care Units ,Cross-Sectional Studies ,Transportation of Patients ,Action plan ,Medical emergency ,Risk assessment ,business ,Emergency Service, Hospital - Abstract
AIMS AND OBJECTIVES To explore the effects of the health failure mode and effect analysis (HFMEA) used in intrahospital transport (IHT) of critically ill patients from emergency department (ED) to the intensive care unit (ICU). BACKGROUND Patients who were transported from ED to ICU is highly critical. IHT of these patients is potentially risky, which may associate with adverse events (AEs). The concern of safe IHT can be addressed by performing proactive risk assessments using HFMEA and implementing the findings after the ED of our hospital being reconstructed. DESIGN A qualitative action research study combined with a quantitative cross-sectional method. METHODS According to the HFMEA method, the failure modes of IHT were identified and analysed, and the effect of alterations was verified. We built a project team, drawn up a IHT flow chart, defined steps of IHT, classified the failure modes, calculated risk priority number and analysed by the decision tree, then formulated an action plan and verified the effects of the alterations. Incidence of AEs of transport was compared before and after HFMEA.SQUIRE 2.0 checklist was chosen on reporting the study process. RESULTS The HFMEA outlined a total of 5 major steps and 16 sub-steps in the IHT process. From this, 64 potential failure modes were identified, with 17 modes having a RPN score higher than 8. Determined by the decision tree, there were 20 priority control failure modes, of which 16 involved 8 IHT alterations. Notable work-flow alterations included use of a three-stage hierarchical transport strategy based on patients' condition assisted by the intelligent assessment system. Incidence of AEs was significantly decreased from 19.64% to 7.14% after the implementation of HFMEA (p
- Published
- 2021
5. Characterization of LRP4/Agrin Antibodies From a Patient With Myasthenia Gravis
- Author
-
Zheng Yu, Wen Cheng Xiong, Hongyang Jing, Brandy Quarles, Jin-Xiu Pan, Meiying Zhang, Michael H. Rivner, Rangjuan Cao, Lin Mei, Peng Chen, Yue Yu, and Bin Luo
- Subjects
medicine.medical_specialty ,animal structures ,Neuromuscular transmission ,Neuromuscular Junction ,Neuromuscular junction ,Epitope ,Antibodies ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,Myasthenia Gravis ,Medicine ,Animals ,Humans ,Agrin ,LDL-Receptor Related Proteins ,030304 developmental biology ,Acetylcholine receptor ,0303 health sciences ,business.industry ,Muscle weakness ,medicine.disease ,Myasthenia gravis ,3. Good health ,Compound muscle action potential ,medicine.anatomical_structure ,Endocrinology ,nervous system ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Background and ObjectiveTo determine whether human anti-LRP4/agrin antibodies are pathogenic in mice and to investigate underpinning pathogenic mechanisms.MethodsImmunoglobulin (Ig) was purified from a patient with myasthenia gravis (MG) with anti-LRP4/agrin antibodies and transferred to mice. Mice were characterized for body weight, muscle strength, twitch and tetanic force, neuromuscular junction (NMJ) functions including compound muscle action potential (CMAP) and endplate potentials, and NMJ structure. Effects of the antibodies on agrin-elicited muscle-specific tyrosine kinase (MuSK) activation and AChR clustering were studied and the epitopes of these antibodies were identified.ResultsPatient Ig-injected mice had MG symptoms, including weight loss and muscle weakness. Decreased CMAPs, reduced twitch and tetanus force, compromised neuromuscular transmission, and NMJ fragmentation and distortion were detected in patient Ig-injected mice. Patient Ig inhibited agrin-elicited MuSK activation and AChR clustering. The patient Ig recognized the β3 domain of LRP4 and the C-terminus of agrin and reduced agrin-enhanced LRP4–MuSK interaction.DiscussionAnti-LRP4/agrin antibodies in the patient with MG is pathogenic. It impairs the NMJ by interrupting agrin-dependent LRP4–MuSK interaction.
- Published
- 2020
6. IncRNA MAPKAPK5-AS1 promotes proliferation and migration of thyroid cancer cell lines by targeting miR-519e-5p/YWHAH
- Author
-
Yan Luo, Yan Xiong, Xueling Jiang, Yan Zhou, Meiying Zhang, and Shanshan Liu
- Subjects
0301 basic medicine ,Histology ,Biophysics ,Down-Regulation ,Apoptosis ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,YWHAH ,Cell Movement ,Cell Line, Tumor ,medicine ,Humans ,Endocrine system ,Thyroid Neoplasms ,Tyrosine ,lcsh:QH301-705.5 ,Thyroid cancer ,Cell Proliferation ,Mechanism (biology) ,Cell growth ,Cell Biology ,Transfection ,medicine.disease ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,lcsh:Biology (General) ,thyroid cancer cell ,14-3-3 Proteins ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,LncRNA MAPKAPK5-AS1 ,MiR-519e-5p ,RNA, Long Noncoding ,IncRNA MAPKAPK5-AS1 - Abstract
Thyroid cancer is a common malignant tumour of the endocrine system and ranks ninth in cancer incidence worldwide. An extensive body of evidence has demonstrated that lncRNAs play a critical role in the progression of thyroid cancer. The lncRNA MAPKAPK5-AS1 has been reported to be abnormally expressed and to play a role in the development of various human cancers. However, MAPKAPK5-AS1’s potential role in thyroid cancer progression remains unknown. The objective of our study was to explore the role and mechanism of MAPKAPK5-AS1 in thyroid cancer cells and provide a potential target for its biological diagnosis and treatment. We transfected sh-MAPKAPK5-AS1 and sh-NC into BCPAP and TPC-1 cells for loss-of-function assays. Results of RT-qPCR analysis demonstrated that MAPKAPK5-AS1 was more highly expressed in thyroid cancer cells compared to normal cells. Functional assays demonstrated that interfering with the expression of MAPKAPK5-AS1 notably repressed proliferation and invasion and accelerated apoptosis of BCPAP and TPC-1 cells. Mechanistically, we found that miR-519e-5p was negatively regulated by MAPKAPK5-AS1 and that tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein eta (YWHAH) was a target of miR-519e-5p. Additionally, rescue assays demonstrated that downregulation of MAPKAPK5-AS1 expression inhibited cell proliferation, migration, and invasion and promoted apoptosis by sponging miR-519e-5p, thereby increasing YWHAH expression. Ultimately, our study revealed that MAPKAPK5-AS1 promotes proliferation and migration of thyroid cancer cells by targeting the miR-519e-5p/YWHAH axis, which provides novel insight into the development and progression of thyroid cancer.
- Published
- 2020
7. Lrp4 in hippocampal astrocytes serves as a negative feedback factor in seizures
- Author
-
Yue Yu, Zheng Yu, Bin Luo, Shunqi Wang, Shiwen Luo, Hongyang Jing, and Meiying Zhang
- Subjects
Low-density lipoprotein receptor-related protein 4 ,Epilepsy ,microRNA ,business.industry ,Research ,Astrocytic NMDA receptor ,Glutamate receptor ,Hippocampus ,Hippocampal formation ,medicine.disease ,Epileptogenesis ,General Biochemistry, Genetics and Molecular Biology ,nervous system ,Pilocarpine ,Medicine ,NMDA receptor ,Pentylenetetrazol ,business ,The threshold of seizure ,Neuroscience ,medicine.drug - Abstract
Background Epilepsy is characterized by the typical symptom of seizure, and anti-seizure medications are the main therapeutic method in clinical, but the effects of these therapy have not been satisfactory. To find a better treatment, it makes sense to further explore the regulatory mechanisms of seizures at genetic level. Lrp4 regionally expresses in mice hippocampus where is key to limbic epileptogenesis. It is well known that neurons release a high level of glutamate during seizures, and it has been reported that Lrp4 in astrocytes down-regulates glutamate released from neurons. However, it is still unclear whether there is a relationship between Lrp4 expression level and seizures, and whether Lrp4 plays a role in seizures. Results We found that seizures induced by pilocarpine decreased Lrp4 expression level and increased miR-351-5p expression level in mice hippocampus. Glutamate reduced Lrp4 expression and enhanced miR-351-5p expression in cultured hippocampal astrocytes, and these effects can be partially attenuated by AP5. Furthermore, miR-351-5p inhibitor lessened the reduction of Lrp4 expression in glutamate treated hippocampal astrocytes. Local reduction of Lrp4 in hippocampus by sh Lrp4 lentivirus injection in hippocampus increased the threshold of seizures in pilocarpine or pentylenetetrazol (PTZ) injected mice. Conclusions These results indicated that high released glutamate induced by seizures down-regulated astrocytic Lrp4 through increasing miR-351-5p in hippocampal astrocytes via activating astrocytic NMDA receptor, and locally reduction of Lrp4 in hippocampus increased the threshold of seizures. Lrp4 in hippocampal astrocytes appears to serve as a negative feedback factor in seizures. This provides a new potential therapeutic target for seizures regulation.
- Published
- 2020
8. Clonality, Heterogeneity, and Evolution of Synchronous Bilateral Ovarian Cancer
- Author
-
Xia Yin, Guanglei Zhuang, Yi Zhang, Meiying Zhang, Mei-Chun Cai, Peng-Fei Ma, Ying Jing, Wen Di, and Cong Xu
- Subjects
0301 basic medicine ,Cancer Research ,DNA Mutational Analysis ,Carcinoma, Ovarian Epithelial ,Biology ,medicine.disease_cause ,Bioinformatics ,Neoplasms, Multiple Primary ,03 medical and health sciences ,Carcinoma ,medicine ,Humans ,Exome ,Neoplasms, Glandular and Epithelial ,Retrospective Studies ,Ovarian Neoplasms ,Mutation ,Base Sequence ,Genome, Human ,Genetic heterogeneity ,High-Throughput Nucleotide Sequencing ,Cancer ,Sequence Analysis, DNA ,Neoplastic Cells, Circulating ,medicine.disease ,Human genetics ,Clone Cells ,030104 developmental biology ,Oncology ,Cancer cell ,Cancer research ,Female ,Ovarian cancer ,Carcinogenesis - Abstract
Synchronous bilateral ovarian cancer (SBOC) represents a relatively frequent occurrence and clinically relevant diagnostic dilemma. Delineation of its clonal architecture, genetic heterogeneity, and evolutionary trajectories may have important implications for prognosis and management of patients with SBOC. Here, we describe the results of next-generation whole-exome or whole-genome sequencing of specimens from 12 SBOC cases and report that bilateral tumors from each individual display a comparable number of genomic abnormalities and similar mutational signatures of single-nucleotide variations. Clonality indices based on tumor-specific alterations supported monoclonal origins of SBOC. Each of the ovarian lesions was nevertheless oligoclonal, with inferred metastatic tumors harboring more subclones than their primary counterparts. The phylogenetic structure of SBOC indicated that most cancer cell dissemination occurred early, when the primary carcinoma was still relatively small (
- Published
- 2017
9. ZNF545 suppresses human hepatocellular carcinoma growth by inhibiting NF-kB signaling
- Author
-
Mingzhou Guo, Meiying Zhang, Weili Yang, Shuai Yang, Tao He, and Dan Gao
- Subjects
0301 basic medicine ,Cancer Research ,Cell ,NF-kB signaling ,Biology ,Loss of heterozygosity ,03 medical and health sciences ,Genetics ,medicine ,Epigenetics ,neoplasms ,DNA methylation ,epigenetics ,Cell growth ,Cancer ,hepatocellular carcinoma ,medicine.disease ,digestive system diseases ,030104 developmental biology ,medicine.anatomical_structure ,Apoptosis ,Hepatocellular carcinoma ,Cancer research ,ZNF545 ,Research Paper - Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers and the second leading cause of cancer related death worldwide. ZNF545 is located in the chromosome 19q13.13, which is frequent loss of heterozygosity in human astrocytoma. Methylation of ZNF545 was found frequently in a few kinds of cancers. While the function of ZNF545 in human HCC remains unclear. The purpose of this study is to explore the function and mechanism of ZNF545 in human HCC. Restoration of ZNF545 expression suppressed cell proliferation, migration and invasion, induced G1/S arrest and apoptosis in SNU449 and Huh7 cells. Further study suggested that ZNF545 suppressed HCC cell growth by inhibiting NF-kB signaling. These results were further validated by siRNA knocking down technique in ZNF545 highly expressed HXBF344 cells. In vivo, ZNF545 suppressed tumor growth in SNU449 cell xenograft mice. In conclusion, ZNF545 suppresses human HCC growth by inhibiting NF-kB signaling.
- Published
- 2017
10. Methylation of DIRAS1 promotes colorectal cancer progression and may serve as a marker for poor prognosis
- Author
-
Meiying Zhang, Mingzhou Guo, Dan Gao, Xiaomei Zhang, Tao He, Ruipan Zheng, Lixin Wei, and Enqiang Linghu
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,Colorectal cancer ,Bisulfite sequencing ,lcsh:Medicine ,Mouse model of colorectal and intestinal cancer ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Epigenetics ,Cancer epigenetics ,Molecular Biology ,Genetics (clinical) ,DNA methylation ,lcsh:R ,Cancer ,Methylation ,medicine.disease ,DIRAS1 ,lcsh:Genetics ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Developmental Biology - Abstract
DIRAS1 is a new member of the Ras gene family. It was described as a potential tumor suppressor in human glioblastomas and esophageal cancer. The role of DIRAS1 in colorectal cancer remains unclear. To explore the epigenetic changes and function of DIRAS1 in human colorectal cancer, we studied ten colorectal cancer cell lines and 146 primary colorectal cancer samples and 50 matched adjacent samples using semi-quantitative reverse transcription PCR, immunohistochemistry, methylation-specific PCR and bisulfite sequencing, western blot, flow cytometry, and transwell assays. DIRAS1 expression was found in DKO and HCT116 cells, while reduced expression was detected in LoVo, SW48, LS180, and SW620 cells, and there was no expression detected in DLD1, HT29, RKO, and SW480 cells. Complete methylation was found in the promoter region of DLD1, HT29, RKO, and SW480 cells. Partial methylation was detected in LoVo, LS180, SW48, and SW620 cells, and unmethylation was found in DKO and HCT116 cells. These results indicate that promoter region methylation correlated with loss of/reduced expression of DIRAS1. Re-expression of DIRAS1 was induced by 5-aza-2′-deoxycytidine, suggesting that the expression of DIRAS1 is regulated by promoter region methylation. DIRAS1 was methylated in 47.3% (69/146) of primary colorectal cancer samples, no methylation was found in non-cancerous colonic tissue samples. Methylation of DIRAS1 was significantly associated with TNM stage (P
- Published
- 2017
11. Epigenetic silencing of IGFBPL1 promotes esophageal cancer growth by activating PI3K-AKT signaling
- Author
-
Lidong Wang, Lirong Zhang, Yingge Liu, Mingzhou Guo, Meiying Zhang, Tao He, and Weili Yang
- Subjects
Adult ,Male ,Esophageal Neoplasms ,medicine.medical_treatment ,Cell ,Esophageal cancer ,Biology ,Decitabine ,Epigenesis, Genetic ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Gene Silencing ,Enzyme Inhibitors ,Molecular Biology ,Genetics (clinical) ,Aged ,Neoplasm Staging ,Aged, 80 and over ,DNA methylation ,Cell growth ,PI3K-AKT ,Growth factor ,Tumor Suppressor Proteins ,Research ,IGFBPL1 ,Methylation ,Middle Aged ,medicine.disease ,Insulin-Like Growth Factor Binding Proteins ,medicine.anatomical_structure ,Cell culture ,Apoptosis ,Models, Animal ,Cancer research ,Heterografts ,Female ,Esophageal Squamous Cell Carcinoma ,Proto-Oncogene Proteins c-akt ,Developmental Biology ,Signal Transduction - Abstract
Background There are seven insulin-like growth factor binding proteins (IGFBPs) that bind insulin-like growth factors (IGFs). IGFBP like protein1 (IGFBPL1) is a new member of this family. The function and mechanism of IGFBPL1 in esophageal cancer remains to be elucidated. Methods Eight esophageal cancer cell lines, 114 cases of esophageal dysplasia, and 501 cases of primary esophageal cancer samples were examined in this study. Methylation-specific polymerase chain reaction (MSP), immunohistochemistry, Western blot, flow cytometry, RNA interference assay, and xenograft mouse models were employed. Results The expression of IGFBPL1was lost and complete methylation was found in KYSE150 and KYSE410 cells. Reduced expression and partial methylation of IGFBPL1 was found in Bic1, KYSE140, KYSE450, KYSE520, and COLO680N cells. High expression and unmethylation was detected in KYSE510 cells. Restoration of IGFBPL1 expression was found in KYSE150 and KYSE410 cells and the expression of IGFBPL1 was increased in Bic1, KYSE140, KYSE450, KYSE520, and COLO680N cells, after 5-AZA-2′-deoxycytidine treatment. IGFBPL1 was methylated in 47.3% (53/114) of esophageal dysplasia and 49.1% (246/501) of human primary esophageal squamous cell carcinoma (ESCC). Methylation of IGFBPL1 was significantly associated with TNM stage (p = 0.012), and tumor size (p = 0.009). IGFBPL1 inhibited esophageal cancer cell clonal formation and proliferation and induced cell apoptosis and G1/S phase arrest. Further study found that IGFBPL1 is involved in PI3K-AKT signaling and IGFBPL1 suppressed human ESCC xenografts growth in mice. Conclusion IGFBPL1 suppresses esophageal cancer cell growth by inhibiting PI3K-AKT signaling in vitro and in vivo. IGFBPL1 is a novel tumor suppressor in human esophageal cancer.
- Published
- 2019
12. Prevalence of and independent risk factors for metabolic syndrome in adults: a population-based, cross-sectional, epidemiological survey in Jiangxi province, China
- Author
-
liting wu, YunFeng Shen, Lei Hu, XiaoYang Lai, and Meiying Zhang
- Subjects
medicine.medical_specialty ,business.industry ,Environmental health ,Epidemiology ,medicine ,Population based ,Metabolic syndrome ,China ,medicine.disease ,business - Abstract
Background Metabolic syndrome (MS) has abruptly increased in China in the past two decades, gradually representing an important public health threat over the years. Here, we firstly reported the prevalence of and independent risk factors for metabolic syndrome in Jiangxi province, China. Methods A population-based cross-sectional survey was performed in Jiangxi province, China, from April to August 2015. MS was diagnosed by International Diabetes Federation (IDF) and Chinese Diabetes Society (CDS) criteria, respectively. Independent risk factors for MS were investigated by multivariate logistic regression. Results A total of 2665 residents aged over 18 years were enrolled, and 2580 effectively participated. According to IDF and CDS criteria, age-standardized prevalence rates of MS were 21.1% and 15.2% in all participants, respectively; prevalence rates were 19.6% and 17.1% in men, and 22.7% or 13.0% in women, based on these respective criteria. Rural participants had a significantly higher prevalence than urban individuals, so did rural females. Prevalence in males did not differ between rural and urban participants. Furthermore, both low education level and menopausal state were independent risk factors for MS in adults. Conclusions MS is highly prevalent in adults in Jiangxi province, China. Low education level and menopausal state are independent risk factors for MS.
- Published
- 2019
13. PAX8 regulon in human ovarian cancer links lineage dependency with epigenetic vulnerability to HDAC inhibitors
- Author
-
Peng-Fei Ma, Ying Yan, Mei-Chun Cai, Shi Kaixuan, Xia Yin, Wen Di, Chenqiang Jia, Zhenfeng Zhang, Zhenyu Gu, Guanglei Zhuang, Shengzhe Zhang, and Meiying Zhang
- Subjects
0301 basic medicine ,Mouse ,Epigenesis, Genetic ,Ovarian tumor ,0302 clinical medicine ,Cell Movement ,Biology (General) ,Cancer Biology ,Ovarian Neoplasms ,biology ,General Neuroscience ,Acetylation ,General Medicine ,Gene Expression Regulation, Neoplastic ,Histone ,ovarian cancer ,030220 oncology & carcinogenesis ,Medicine ,Female ,Research Article ,Human ,QH301-705.5 ,Science ,Histone Deacetylases ,General Biochemistry, Genetics and Molecular Biology ,PAX8 Transcription Factor ,03 medical and health sciences ,HDAC inhibitor ,Cell Line, Tumor ,super-enhancer ,medicine ,Humans ,lineage-survival oncogene ,Cell Lineage ,Epigenetics ,Autocrine signalling ,Cell Proliferation ,General Immunology and Microbiology ,Oncogene ,E. coli ,Cancer ,Cell Biology ,medicine.disease ,PAX8 ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Regulon ,Tissue Array Analysis ,regulon ,biology.protein ,Cancer research ,Ovarian cancer - Abstract
PAX8 is a prototype lineage-survival oncogene in epithelial ovarian cancer. However, neither its underlying pro-tumorigenic mechanisms nor potential therapeutic implications have been adequately elucidated. Here, we identified an ovarian lineage-specific PAX8 regulon using modified cancer outlier profile analysis, in which PAX8-FGF18 axis was responsible for promoting cell migration in an autocrine fashion. An image-based drug screen pinpointed that PAX8 expression was potently inhibited by small-molecules against histone deacetylases (HDACs). Mechanistically, HDAC blockade altered histone H3K27 acetylation occupancies and perturbed the super-enhancer topology associated with PAX8 gene locus, resulting in epigenetic downregulation of PAX8 transcripts and related targets. HDAC antagonists efficaciously suppressed ovarian tumor growth and spreading as single agents, and exerted synergistic effects in combination with standard chemotherapy. These findings provide mechanistic and therapeutic insights for PAX8-addicted ovarian cancer. More generally, our analytic and experimental approach represents an expandible paradigm for identifying and targeting lineage-survival oncogenes in diverse human malignancies.
- Published
- 2019
14. Author response: PAX8 regulon in human ovarian cancer links lineage dependency with epigenetic vulnerability to HDAC inhibitors
- Author
-
Shengzhe Zhang, Zhenyu Gu, Shi Kaixuan, Chenqiang Jia, Meiying Zhang, Zhenfeng Zhang, Xia Yin, Wen Di, Guanglei Zhuang, Mei-Chun Cai, Peng-Fei Ma, and Ying Yan
- Subjects
Genetics ,Regulon ,Dependency (UML) ,Lineage (genetic) ,medicine ,Vulnerability ,Epigenetics ,Biology ,Ovarian cancer ,medicine.disease ,PAX8 - Published
- 2019
15. P07.04 Using ctDNA to Detect Minimal Residual Disease after Surgery in Resectable Lung Cancer
- Author
-
Meiying Zhang, Runzhe Chen, Y. Mou, X. Xia, and J. Huang
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Oncology ,business.industry ,Medicine ,Radiology ,business ,Lung cancer ,medicine.disease ,Minimal residual disease - Published
- 2021
16. FERMT1 mediates epithelial–mesenchymal transition to promote colon cancer metastasis via modulation of β-catenin transcriptional activity
- Author
-
Dongwang Yan, Senlin Zhao, Chen-Ying Liu, Xiaoqiang Zhu, Ben Yue, Cai Dl, Meiying Zhang, Wu Xs, Sun F, Peng Zh, and Zehua Wu
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Transcription, Genetic ,Biology ,Molecular oncology ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Growth factor receptor ,Cell Movement ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Epithelial–mesenchymal transition ,Intestinal Mucosa ,Neoplasm Metastasis ,Wnt Signaling Pathway ,Molecular Biology ,beta Catenin ,Cell Proliferation ,Neoplasm Staging ,Regulation of gene expression ,Glycogen Synthase Kinase 3 beta ,Wnt signaling pathway ,Membrane Proteins ,Cadherins ,medicine.disease ,Neoplasm Proteins ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Catenin ,Colonic Neoplasms ,Immunology ,Cancer research ,Female ,Neoplasm Grading ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Biomarkers - Abstract
We previously demonstrated that fermitin family member 1 (FERMT1) was significantly overexpressed in colon cancer (CC) and associated with poor metastasis-free survival. This study aimed to investigate the precise role of FERMT1 in CC metastasis and the mechanism by which FERMT1 is involved in the epithelial-mesenchymal transition (EMT). Correlations between FERMT1 and EMT markers (E-cadherin, Slug, N-cadherin and β-catenin) were examined via immunohistochemistry in a cohort of CC tissues and adjacent normal colon mucosae. A series of in vitro and in vivo assays were performed to elucidate the function of FERMT1 in CC metastasis and underlying mechanisms. The upregulated expression of FERMT1 in CC tissues correlated positively with that of Slug, N-cadherin and β-catenin, but correlated inversely with E-cadherin expression. Altered FERMT1 expression led to marked changes in the proliferation, migration, invasion and EMT markers of CC cells both in vitro and in vivo. Investigations of underlying mechanisms found that FERMT1 interacted directly with β-catenin and activated the Wnt/β-catenin signaling pathway by decreasing the phosphorylation level of β-catenin, enhancing β-catenin nuclear translocation and increasing the transcriptional activity of β-catenin/TCF/LEF. Activation of the Wnt/β-catenin pathway by CHIR99021 reversed the effect of FERMT1 knockdown, whereas inhibition of the Wnt/β-catenin pathway by XAV939 impaired the effect of FERMT1 overexpression on EMT and cell motility. In conclusion, findings of this study suggest that FERMT1 activates the β-catenin transcriptional activity to promote EMT in CC metastasis.
- Published
- 2016
17. Transcriptional profiling analysis and functional prediction of long noncoding RNAs in cancer
- Author
-
Rui Xing, Runsheng Chen, Mingzhou Guo, Meiying Zhang, Jianjun Luo, Lihui Liu, Dongdong Zhang, Tengfei Xiao, Xiaowei Chen, Youyong Lu, Jiao Yuan, Haiyan Yue, Wei Wu, Nan Wu, Xin Tong, Xiaomin Chen, and Jian Zhao
- Subjects
0301 basic medicine ,Colorectal cancer ,Computational biology ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Neoplasms ,expression ,Gene expression ,Biomarkers, Tumor ,medicine ,Humans ,Gene ,Oligonucleotide Array Sequence Analysis ,business.industry ,gastric cancer ,Gene Expression Profiling ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,030104 developmental biology ,Real-time polymerase chain reaction ,colon cancer ,Oncology ,IncRNA ,Case-Control Studies ,biomarker ,Biomarker (medicine) ,RNA, Long Noncoding ,Cancer biomarkers ,business ,Biogenesis ,Research Paper - Abstract
Long noncoding RNAs (lncRNAs), which are noncoding RNAs (ncRNAs) with length more than 200 nucleotides (nt), have been demonstrated to be involved in various types of cancer. Consequently, it has been frequently discussed that lncRNAs with aberrant expression in cancer serve as potential diagnostic biomarkers and therapeutic targets. However, one major challenge of developing cancer biomarkers is tumor heterogeneity which means that tumor cells show different cellular morphology, metastatic potential as well as gene expression. In this study, a custom designed microarray platform covering both mRNAs and lncRNAs was applied to tumor tissues of gastric, colon, liver and lung. 316 and 157 differentially expressed (DE-) protein coding genes and lncRNAs common to these four types of cancer were identified respectively. Besides, the functional roles of common DE-lncRNAs were inferred based on their expression and genomic position correlation with mRNAs. Moreover, mRNAs and lncRNAs with tissue specificity were also identified, suggesting their particular roles with regard to specific biogenesis and functions of different organs. Based on the large-scale survey of mRNAs and lncRNAs in four types of cancer, this study may offer new biomarkers common or specific for various types of cancer.
- Published
- 2016
18. BET Bromodomain Inhibition as a Therapeutic Strategy in Ovarian Cancer by Downregulating FoxM1
- Author
-
Wen Di, Ying Jing, Zhenfeng Zhang, Zhenyu Gu, Guanglei Zhuang, Wei-Qiang Gao, Meiying Zhang, Huixin Peng, Peng-Fei Ma, Ying Yan, Shengzhe Zhang, Zhiliang Ji, and Mei-Chun Cai
- Subjects
0301 basic medicine ,BRD4 ,Down-Regulation ,Mice, Nude ,Medicine (miscellaneous) ,Antineoplastic Agents ,Bioinformatics ,Heterocyclic Compounds, 4 or More Rings ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Downregulation and upregulation ,Cell Line, Tumor ,Ovarian carcinoma ,medicine ,Animals ,Humans ,Epigenetics ,BET inhibitors ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Ovarian Neoplasms ,Mice, Inbred BALB C ,business.industry ,Pteridines ,Carcinoma ,Forkhead Box Protein M1 ,Nuclear Proteins ,Volasertib ,Azepines ,Triazoles ,medicine.disease ,Bromodomain ,ovarian cancer ,030104 developmental biology ,chemistry ,FoxM1 ,Cancer research ,FOXM1 ,Female ,Ovarian cancer ,business ,Research Paper ,Transcription Factors - Abstract
Ovarian cancer is responsible for the highest mortality among all gynecologic malignancies, and novel therapies are urgently needed to improve patient outcome. Here we performed an integrative genomic analysis and identified the bromodomain and extraterminal domain (BET) protein BRD4 as a potential therapeutic target in ovarian cancer. Suppression of BRD4 using small-molecule BET inhibitors JQ1 and I-BET151, or dual kinase-bromodomain inhibitor volasertib, led to robust and broad antitumor effects across all subclasses of ovarian cancer. In contrast to many other cancers which are susceptible to BET inhibition due to downregulation of super-enhancer-dependent MYC transcript, we discovered that JQ1-sensitive ovarian cancer cells exhibited marked disruption of Forkhead box protein M1 (FoxM1) pathway, a key driver of ovarian carcinoma. These in vitro findings were further supported by in vivo efficacies of JQ1 targeting both cell line-based and patient-derived xenograft models. Our data establish a new treatment strategy against ovarian cancer by employing epigenetic vulnerabilities, and provide a mechanistic rationale for the clinical investigation of BET bromodomain inhibitors in this deadly disease.
- Published
- 2016
19. Combination of asprosin and adiponectin as a novel marker for diagnosing non-alcoholic fatty liver disease
- Author
-
Meiying Zhang, Guohui Xue, Ling Gao, Xueling Jiang, Fangfang Li, Xiaoyang Lai, Fan Ke, and Yunfeng Shen
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Fibrillin-1 ,Immunology ,Serum albumin ,Adipokine ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Insulin resistance ,Non-alcoholic Fatty Liver Disease ,Internal medicine ,Diabetes mellitus ,medicine ,Humans ,Immunology and Allergy ,Molecular Biology ,Aged ,biology ,Adiponectin ,Triglyceride ,business.industry ,Incidence ,Fatty liver ,nutritional and metabolic diseases ,Hematology ,Middle Aged ,medicine.disease ,030104 developmental biology ,Endocrinology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Biomarker (medicine) ,Female ,business ,Biomarkers - Abstract
Background and objective Patients with non-alcoholic fatty liver disease (NAFLD) have insulin resistance and are at an increased risk of diabetes. Recent evidence suggests that asprosin–a novel hormone secreted by white adipose tissue–may play a role in the pathogenesis of insulin resistance. However, the role of asprosin in NAFLD remains unclear. This study aimed to determine whether serum asprosin level could be used as a biochemical marker for NAFLD diagnosis. Methods Forty-three untreated NAFLD patients and 50 sex- and age-matched healthy controls were included. Circulating serum asprosin and adiponectin (another adipokine) levels were detected by ELISA. Other metabolic parameters related to NAFLD were also determined. Results Increased circulating serum asprosin and decreased serum adiponectin levels were found in NAFLD patients unlike in healthy controls. A positive correlation was observed between asprosin and platelet counts (PLT) (r = 0.3653, p = 0.015), fasting blood glucose (FBG) (r = 0.3592, p = 0.017), triglyceride (TG) levels (r = 0.3383, p = 0.025), serum albumin (ALB) levels (r = 0.3273, p = 0.030), and insulin resistance (HOMA-IR) (r = 0.4799, p = 0.001), whereas a negative correlation existed between adiponectin and TG levels in the NAFLD group. Multivariate linear regression showed that FBG and HOMA-IR were independently related to asprosin levels. Receiver operating characteristic (ROC) curves showed that asprosinAUC and adiponectinAUC were 0.735 (95%CI 0.633–0.836, P Conclusion High serum asprosin and low adiponectin level might be associated with the presence of insulin resistance in NAFLD, and the combination of asprosin and adiponectin could be a novel biomarker for diagnosing NAFLD. These data needed to be confirmed and extended in further large-population, well-designed clinical studies.
- Published
- 2020
20. Alpha-Lipoic Acid Preconditioning and Ischaemic Postconditioning Synergistically Protect Rats from Cerebral Injury Induced by Ischemia and Reperfusion Partly via Inhibition TLR4/MyD88/ NF-κB Signaling Pathway
- Author
-
Jing Zhang, Xifeng Wang, Yunfeng Shen, Haili Lang, Peng Yu, Lieliang Zhang, Bin Zhou, Meiying Zhang, Fuzhou Hua, Xiaoyang Lai, and Fan Xiao
- Subjects
0301 basic medicine ,Male ,Physiology ,Anti-Inflammatory Agents ,Inflammation ,Cerebral ischaemia/reperfusion injury ,Pharmacology ,medicine.disease_cause ,Neuroprotection ,lcsh:Physiology ,Brain Ischemia ,lcsh:Biochemistry ,Rats, Sprague-Dawley ,03 medical and health sciences ,medicine ,Animals ,lcsh:QD415-436 ,TLR4 ,Ischemic Postconditioning ,Neuroinflammation ,chemistry.chemical_classification ,Reactive oxygen species ,TUNEL assay ,lcsh:QP1-981 ,Thioctic Acid ,business.industry ,NF-kappa B ,Brain ,medicine.disease ,Rats ,Toll-Like Receptor 4 ,Alpha-lipoic acid preconditioning ,030104 developmental biology ,Neuroprotective Agents ,chemistry ,Apoptosis ,Reperfusion Injury ,Ischaemic postconditioning ,Myeloid Differentiation Factor 88 ,medicine.symptom ,business ,Reperfusion injury ,Oxidative stress ,Signal Transduction - Abstract
Background/Aims: A combination of alpha-lipoic acid preconditioning (ALAP) and ischaemic preconditioning (IPC) has not been tested in an in vivo rat cerebral ischaemia/reperfusion injury (I/RI) model, and the potential protective mechanisms have not been well elucidated. The aim of this study was to investigate the role of the TLR4/ MyD88/ NF-κB signaling pathway in the synergistically neuroprotective and anti-inflammatory effects of ALAP and IPC. Methods: One hundred and fifty male Sprague-Dawley rats, weighing 180-230 g, were randomly divided into the following 5 groups: 1) sham-operated control; 2) I/R; 3) I/R+ALAP; 4) I/R+IPC; 5) I/R+IPC+ALAP. After 2 h of reperfusion, the infarct size, neurological deficit scores, brain oedema, oxidative stress, and inflammatory and apoptotic biomarkers were assessed. In addition, reactive oxygen species (ROS) and cell apoptosis were detected by DHE staining and TUNEL staining, respectively. Results: Both ALAP and IPC treatment attenuated the I/RI-induced neuronal injury, reflected by reductions in the infarct size, neurological deficit scores, brain oedema, lactate dehydrogenase (LDH) release and the inflammatory response, as well as decreased HMGB1, TLR4, MyD88, p65, C-Caspase 3 and Bax expression and increased IKB-α, HO-1, SOD-2 and Bcl-2 expression compared to that in the I/R group. Furthermore, the combination of the two strategies had synergistic anti-inflammatory effects and antioxidant benefits, ultimately limiting neuronal apoptosis. Conclusion: The ‘cocktail’ strategy exhibited a significant neuroprotection against I/RI by attenuating neuroinflammation via inhibition of the TLR4/MyD88/NF-κB signaling pathway.
- Published
- 2018
21. Epigenetic silencing of NKD2, a major component of Wnt signaling, promotes breast cancer growth
- Author
-
James G. Herman, Yali Zhao, Meiying Zhang, Weidong Han, Baoping Cao, Yang Dong, Mingzhou Guo, and François Fuks
- Subjects
NKD2 ,Bisulfite sequencing ,Mice, Nude ,Breast Neoplasms ,Cell Growth Processes ,Mice ,breast cancer ,Breast cancer ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Gene Silencing ,Cancer epigenetics ,Epigenetics ,Promoter Regions, Genetic ,skin and connective tissue diseases ,Wnt Signaling Pathway ,Adaptor Proteins, Signal Transducing ,Mice, Inbred BALB C ,DNA methylation ,epigenetics ,business.industry ,Calcium-Binding Proteins ,Wnt signaling pathway ,Cancer ,Methylation ,medicine.disease ,Wnt signaling ,Oncology ,Immunology ,Cancer research ,Heterografts ,Female ,Carrier Proteins ,business ,Research Paper - Abstract
// Yan Dong 1, 2 , Baoping Cao 1, 3 , Meiying Zhang 1, 3 , Weidong Han 2 , James G. Herman 4 , Francois Fuks 5 , Yali Zhao 6 , Mingzhou Guo 1 1 Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing 100853, China 2 Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China 3 Medical College of NanKai University, Tianjin 300071, China 4 The Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, U.S.A 5 Laboratory of Cancer Epigenetics, Free University of Brussels (U.L.B.), 808 route de Lennik, Brussels 1070, Belgium 6 Central Laboratory, The Affiliated Hainan Hospital of Chinese PLA General Hospital, Hai Tang Wan, Sanya 572013, China Correspondence to: Mingzhou Guo, e-mail: mzguo1@gmail.com Yali Zhao, e-mail: zhaoyl301@163.com Keywords: NKD2, DNA methylation, Wnt signaling, epigenetics, breast cancer Received: February 27, 2015 Accepted: June 08, 2015 Published: June 19, 2015 ABSTRACT Naked cuticle homolog 2 (NKD2) has been reported to antagonize Wnt signaling in zebrafish, mouse and mammals. The aim of this study is to investigate the epigenetic changes and mechanisms of NKD2 in human breast cancer development. Six breast cancer cell lines (MCF-7, ZR75–1, MDA-MB-468, MDA-MB-231, T47D and BT474) and 68 cases of primary human breast cancer were studied using methylation specific PCR, immunohistochemistry, western blot, flow cytometry techniques and a xenograft mouse model. The expression of NKD1 and NKD2 was regulated by promoter region methylation in breast cancer cells. No NKD1 methylation was found in primary human breast cancer. NKD2 was methylated in 51.4% (35/68) of human primary breast cancer samples. NKD2 methylation was significantly associated with reduction of NKD2 expression, and tumor stage ( p < 0.05). NKD2 suppressed breast cancer cell proliferation both in vitro and in vivo . NKD2 induced G1/S arrest and inhibited Wnt signaling in breast cancer cells. In conclusion, NKD2 is frequently methylated in human breast cancer, and the expression of NKD2 is regulated by promoter region methylation. NKD2 suppresses breast cancer proliferation by inhibiting Wnt signaling.
- Published
- 2015
22. RASSF10 suppresses hepatocellular carcinoma growth by activating P53 signaling and methylation of RASSF10 is a docetaxel resistant marker
- Author
-
Miao Yu, James G. Herman, Meiying Zhang, Qimin Zhan, Mingzhou Guo, Yongshuai Jin, and Baoping Cao
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,DNA methylation ,epigenetics ,Colorectal cancer ,Cancer ,hepatocellular carcinoma ,Methylation ,Biology ,medicine.disease ,digestive system diseases ,P53 signaling ,Loss of heterozygosity ,Docetaxel ,Hepatocellular carcinoma ,Genetics ,Cancer research ,medicine ,RASSF10 ,Epigenetics ,neoplasms ,Research Paper ,medicine.drug - Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignances and the second leading cause of cancer related death worldwide. RASSF10 is located on chromosome 11p15.2, a region that shows frequent loss of heterozygosity (LOH) in several cancer types. Our previous study found that RASSF10 suppresses colorectal cancer growth by activating P53 signaling. To explore the epigenetic changes and the mechanism of RASSF10 in human HCC, 69 cases of primary HCC, twenty cases of normal liver tissue samples and 17 HCC cell lines were involved in this study. We found that RASSF10 was methylated in 82.6% (57/69) of human primary HCC and methylation of RASSF10 was significantly associated with tumor size (P < 0.05) and TNM stage (P < 0.05). The expression of RASSF10 was regulated by promoter region methylation. Restoration of RASSF10 expression suppressed cell proliferation, induced apoptosis and G2/M phase arrest, as well as sensitized cells to docetaxel and activated P53 signaling in HepG2 and QGY7703 cells. In conclusion, we demonstrated that RASSF10 is frequently methylated in human HCC and its methylation is a potential docetaxel resistant marker. Our data also indicate that RASSF10 suppresses human HCC growth by activating P53 signaling.
- Published
- 2015
23. Silencing HOXD10 by promoter region hypermethylation activates ERK signaling in hepatocellular carcinoma
- Author
-
James G. Herman, Meiying Zhang, Yulin Guo, Mingzhou Guo, Enqiang Linghu, Yaojun Peng, Guanglong Dong, François Fuks, Weili Yang, and Dan Gao
- Subjects
0301 basic medicine ,Génétique du développement ,Génétique clinique ,Hepatocellular carcinoma ,Cell ,lcsh:Medicine ,Mice ,0302 clinical medicine ,Cell Movement ,Promoter Regions, Genetic ,Genetics (clinical) ,DNA methylation ,ERK1/2 ,Liver Neoplasms ,Cell Differentiation ,Methylation ,Hep G2 Cells ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Carcinoma, Hepatocellular ,lcsh:QH426-470 ,MAP Kinase Signaling System ,Biology ,03 medical and health sciences ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Gene Silencing ,Molecular Biology ,Homeodomain Proteins ,Cell growth ,Research ,lcsh:R ,Cancer ,Biologie moléculaire ,medicine.disease ,Survival Analysis ,HOXD10 ,digestive system diseases ,lcsh:Genetics ,030104 developmental biology ,Cell culture ,Apoptosis ,IGFBP3 ,Cancer research ,théorie et applications [Econométrie et méthodes statistiques] ,Developmental Biology ,Transcription Factors - Abstract
Background: Hepatocellular carcinoma is the fifth most common malignancy and the third leading cause of cancer-related death worldwide. Dysregulation of HomeoboxD10 (HOXD10) was found to suppress or promote cancer progression in different cancer types. The function and regulation of HOXD10 remain unclear in human hepatocellular carcinoma (HCC). Methods: Primary HCC samples (117), normal liver tissue samples (15), and 13 HCC cell lines (SNU182, SNU449, HBXF344, SMMC7721, Huh7, HepG2, LM3, PLC/PRF/5, BEL7402, SNU387, SNU475, QGY7703, and Huh1) were included in this study. Methylation-specific PCR, flow cytometry, western blot, transwell, siRNA, and chromatin immunoprecipitation assays were employed. Results: HOXD10 was methylated in 76.9% (90/117) of human primary HCC samples. HOXD10 methylation was significantly associated with vessel cancerous embolus, tumor cell differentiation, and the 3-year overall survival rate (all P < 0.05). The expression of HOXD10 was regulated by promoter region methylation. HOXD10 suppressed colony formation, cell proliferation, cell invasion and migration, and induced G2/M phase arrest and apoptosis in HCC cells. HOXD10 suppressed HCC cell xenograft growth in mice. HOXD10 suppresses HCC growth by inhibiting ERK signaling. Conclusion: HOXD10 is frequently methylated in human HCC, and the expression of HOXD10 is regulated by promoter region methylation. HOXD10 suppresses HCC cell growth both in vitro and in vivo. HOXD10 suppresses human HCC by inhibiting ERK signaling., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2017
24. Methylation of DACT2 promotes breast cancer development by activating Wnt signaling
- Author
-
Lili Zheng, Tingting Cao, Mingzhou Guo, Jingyi Li, Hongxia Li, Tao He, and Meiying Zhang
- Subjects
0301 basic medicine ,Oncology ,Carcinogenesis ,medicine.disease_cause ,0302 clinical medicine ,Cell Movement ,Promoter Regions, Genetic ,skin and connective tissue diseases ,Wnt Signaling Pathway ,Multidisciplinary ,Cell Cycle ,Wnt signaling pathway ,Middle Aged ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,DNA methylation ,Medicine ,Female ,Adult ,medicine.medical_specialty ,Science ,Mice, Nude ,CA 15-3 ,Breast Neoplasms ,Biology ,Article ,03 medical and health sciences ,Breast cancer ,Cancer stem cell ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Gene Silencing ,Adaptor Proteins, Signal Transducing ,Aged ,Cell Proliferation ,Cancer ,Cell Cycle Checkpoints ,DNA Methylation ,medicine.disease ,030104 developmental biology ,Cancer cell ,Cancer research ,Carrier Proteins - Abstract
Breast cancer is the most common malignant tumor in women worldwide. To explore the role of DACT2 in breast cancer, 5 cell lines and 153 cases of primary cancer were studied. The expression of DACT2 was detected in BT474, MDA-MB-231 and BT549 cells, while no expression was found in MDA-MB-468 and HBL100 cells. Complete methylation of DACT2 was found in MDA-MB-468 and HBL100 cells, partial methylation was observed in BT474 and BT549 cells, and no methylation was detected in MDA-MB-231 cells. Restoration of DACT2 expression was induced by 5-Aza in MDA-MB-468 and HBL100 cells. DACT2 was methylated in 49.7% (76/153) of primary breast cancer samples. Methylation of DACT2 was significantly associated with tumor size (P P in vitro and in vivo.
- Published
- 2017
25. Methylation of SLFN11 is a marker of poor prognosis and cisplatin resistance in colorectal cancer
- Author
-
Mingzhou Guo, Ruipan Zheng, Shufang Zheng, Tao He, Meiying Zhang, James G Herman, and Enqiang Linghu
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Colorectal cancer ,Antineoplastic Agents ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Epigenetics ,Promoter Regions, Genetic ,neoplasms ,Cells, Cultured ,Cisplatin ,Mice, Inbred BALB C ,Cell growth ,Nuclear Proteins ,Promoter ,Methylation ,DNA Methylation ,Middle Aged ,medicine.disease ,Molecular biology ,digestive system diseases ,030104 developmental biology ,Cell culture ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Female ,Colorectal Neoplasms ,medicine.drug - Abstract
Aim: The expression of human SLFN11 was reported to sensitize cancer cells to DNA damaging agents. This study is to explore the epigenetic change and the function of SLFN11 in human colorectal cancer (CRC). Materials & methods: Six CRC cell lines and 128 primary CRC samples were used. Results: SLFN11 was methylated in 55.47% (71/128) of primary CRC. The expression of SLFN11 was regulated by promoter region methylation. Methylation of SLFN11 was significantly associated with age, poor 5-year overall survival and 5-year relapse-free survival (all p < 0.05). SLFN11 suppressed CRC cell growth both in vitro and in vivo and sensitized CRC cells to cisplatin. Conclusion: SLFN11 is frequently methylated in human CRC, and the expression of SLFN11 is regulated by promoter region methylation. Methylation of SLFN11 reduced the sensitivity of CRC cells to cisplatin.
- Published
- 2017
26. TP53 mutation-mediated genomic instability induces the evolution of chemoresistance and recurrence in epithelial ovarian cancer
- Author
-
Wenjing Wang, Guanglei Zhuang, Xiangjun Sun, Yan-Ying Shen, Qing Li, Wen Di, and Meiying Zhang
- Subjects
Genomic instability ,Adult ,0301 basic medicine ,Genome instability ,Histology ,endocrine system diseases ,DNA Mutational Analysis ,Mutant ,Gene Dosage ,Aneuploidy ,Kaplan-Meier Estimate ,Carcinoma, Ovarian Epithelial ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,medicine ,Humans ,Neoplasms, Glandular and Epithelial ,neoplasms ,In Situ Hybridization, Fluorescence ,Aged ,Ovarian Neoplasms ,Chromosome 7 (human) ,Mutation ,medicine.diagnostic_test ,Research ,TP53 mutation ,MDR1 copy number ,General Medicine ,Epithelial ovarian cancer ,Middle Aged ,Flow Cytometry ,medicine.disease ,Immunohistochemistry ,Molecular biology ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Female ,Neoplasm Recurrence, Local ,Tumor Suppressor Protein p53 ,Ovarian cancer ,Fluorescence in situ hybridization - Abstract
Background Genomic instability caused by mutation of the checkpoint molecule TP53 may endow cancer cells with the ability to undergo genomic evolution to survive stress and treatment. We attempted to gain insight into the potential contribution of ovarian cancer genomic instability resulted from TP53 mutation to the aberrant expression of multidrug resistance gene MDR1. Methods TP53 mutation status was assessed by performing nucleotide sequencing and immunohistochemistry. Ovarian cancer cell DNA ploidy was determined using Feulgen-stained smears or flow cytometry. DNA copy number was analyzed by performing fluorescence in situ hybridization (FISH). Results In addition to performing nucleotide sequencing for 5 cases of ovarian cancer, TP53 mutations were analyzed via immunohistochemical staining for P53. Both intensive P53 immunohistochemical staining and complete absence of signal were associated with the occurrence of TP53 mutations. HE staining and the quantification of DNA content indicated a significantly higher proportion of polyploidy and aneuploidy cells in the TP53 mutant group than in the wild-type group (p 6 MDR1 copies and chromosome 7 amplication in the TP53 mutant group than in the wild-type group [11.7 ± 2.3% vs. 3.0 ± 0.7% and 2.1 ± 0.7% vs. 0.3 ± 0.05%, (p
- Published
- 2017
27. Preclinical Efficacy and Molecular Mechanism of Targeting CDK7-Dependent Transcriptional Addiction in Ovarian Cancer
- Author
-
Shi Kaixuan, Wei-Qiang Gao, Meiying Zhang, Xiaojie Wang, Huixin Peng, Jin Liu, Guanglei Zhuang, Shengzhe Zhang, Mei-Chun Cai, Xia Yin, Wen Di, Zhenfeng Zhang, Peng-Fei Ma, and Ying Jing
- Subjects
0301 basic medicine ,Drug ,Cancer Research ,Transcription, Genetic ,media_common.quotation_subject ,Antineoplastic Agents ,Apoptosis ,Biology ,Pharmacology ,Phenylenediamines ,03 medical and health sciences ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epigenetics ,Cytotoxicity ,Transcription factor ,media_common ,Cell Proliferation ,Regulation of gene expression ,Ovarian Neoplasms ,Cell Cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,Cyclin-Dependent Kinases ,Bromodomain ,E2F Transcription Factors ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Pyrimidines ,Oncology ,Cancer research ,Female ,Cyclin-dependent kinase 7 ,Drug Screening Assays, Antitumor ,Ovarian cancer ,Cyclin-Dependent Kinase-Activating Kinase - Abstract
Ovarian cancer remains a significant cause of gynecologic cancer mortality, and novel therapeutic strategies are urgently needed in clinic as new treatment options. We previously showed that BET bromodomain inhibitors displayed promising efficacy for the treatment of epithelial ovarian cancer by downregulating pivot transcription factors. However, the potential antitumor activities and molecular mechanisms of other epigenetic or transcriptional therapies have not been systematically determined. Here, by performing an unbiased high-throughput drug screen to identify candidate compounds with antineoplastic effects, we identified THZ1, a recently developed covalent CDK7 inhibitor, as a new transcription-targeting compound that exerted broad cytotoxicity against ovarian tumors. Mechanistically, CDK7 represented a previously unappreciated actionable vulnerability in ovarian cancer, and CDK7 inhibition led to a pronounced dysregulation of gene transcription, with a preferential repression of E2F-regulated genes and transcripts associated with super-enhancers. Our findings revealed the molecular underpinnings of THZ1 potency and established pharmaceutically targeting transcriptional addiction as a promising therapeutic strategy in aggressive ovarian cancer. Mol Cancer Ther; 16(9); 1739–50. ©2017 AACR.
- Published
- 2017
28. Stroma-associated master regulators of molecular subtypes predict patient prognosis in ovarian cancer
- Author
-
Wei-Qiang Gao, Meiying Zhang, Shi Kaixuan, Shengzhe Zhang, Guanglei Zhuang, Ying Jing, Huixin Peng, Zhenfeng Zhang, and Peng-Fei Ma
- Subjects
Stromal cell ,endocrine system diseases ,Gene regulatory network ,Bioinformatics ,Article ,Ovarian carcinoma ,Biomarkers, Tumor ,Tumor Microenvironment ,Humans ,Medicine ,Gene Regulatory Networks ,Ovarian Neoplasms ,Tumor microenvironment ,Multidisciplinary ,business.industry ,Gene Expression Profiling ,Mesenchymal stem cell ,Prognosis ,medicine.disease ,female genital diseases and pregnancy complications ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Gene expression profiling ,Serous fluid ,Female ,Stromal Cells ,business ,Ovarian cancer ,Algorithms - Abstract
High-grade serous ovarian carcinoma (HGS-OvCa) has the lowest survival rate among all gynecologic cancers and is hallmarked by a high degree of heterogeneity. The Cancer Genome Atlas network has described a gene expression-based molecular classification of HGS-OvCa into Differentiated, Mesenchymal, Immunoreactive and Proliferative subtypes. However, the biological underpinnings and regulatory mechanisms underlying the distinct molecular subtypes are largely unknown. Here we showed that tumor-infiltrating stromal cells significantly contributed to the assignments of Mesenchymal and Immunoreactive clusters. Using reverse engineering and an unbiased interrogation of subtype regulatory networks, we identified the transcriptional modules containing master regulators that drive gene expression of Mesenchymal and Immunoreactive HGS-OvCa. Mesenchymal master regulators were associated with poor prognosis, while Immunoreactive master regulators positively correlated with overall survival. Meta-analysis of 749 HGS-OvCa expression profiles confirmed that master regulators as a prognostic signature were able to predict patient outcome. Our data unraveled master regulatory programs of HGS-OvCa subtypes with prognostic and potentially therapeutic relevance and suggested that the unique transcriptional and clinical characteristics of ovarian Mesenchymal and Immunoreactive subtypes could be, at least partially, ascribed to tumor microenvironment.
- Published
- 2015
29. Risk prediction model for epithelial ovarian cancer using molecular markers and clinical characteristics
- Author
-
Meiying Zhang, Aimin Zhao, Guanglei Zhuang, Yan-Ying Shen, Wen Di, and Xiangjun Sun
- Subjects
Adult ,Oncology ,medicine.medical_specialty ,Pathology ,Kaplan-Meier Estimate ,Carcinoma, Ovarian Epithelial ,medicine.disease_cause ,Risk Assessment ,Breast cancer ,Internal medicine ,Obstetrics and Gynaecology ,Biomarkers, Tumor ,medicine ,Carcinoma ,Humans ,Overall survival ,Neoplasms, Glandular and Epithelial ,Epidermal growth factor receptor ,Early Detection of Cancer ,Aged ,Retrospective Studies ,Ovarian Neoplasms ,biology ,Proportional hazards model ,business.industry ,Research ,Obstetrics and Gynecology ,Retrospective cohort study ,Cytoreduction Surgical Procedures ,Middle Aged ,Epithelial ovarian cancer ,medicine.disease ,Debulking ,Immunohistochemistry ,Chemotherapy, Adjuvant ,Molecular classification ,biology.protein ,Female ,KRAS ,Ovarian cancer ,business ,Risk prediction model - Abstract
Background A high-quality risk prediction model is urgently needed for the clinical management of ovarian cancer. However most existing models are solely based on clinical parameters, and molecular classifications in recent reports are still being debated. This study aimed to establish a risk prediction model by using both clinicopathological and molecular factors (the synthetic model) for epithelial ovarian cancer. Methods A retrospective cohort study was conducted in epithelial ovarian cancer patients (n = 161) treated with primary debulking surgery and adjuvant chemotherapy. The expression level of 15 selected molecular markers were measured using immunohistochemistry. A risk model was developed using COX regression analysis with overall survival as the primary outcome. A simplified scoring system for each prognostic factor was based on its coefficient. Independent validation (n = 40) was conducted to evaluate the performance of the model. Results A total of 10 out of 15 molecular markers were significantly associated with clinical characteristics and overall survival. The synthetic model performed better than the clinicopathological risk model or the molecular risk model alone, as assessed by analysis of the receiver-operating characteristics curve area and the Youden index. The synthetic model included parity (>3), peritoneal metastasis, stage, tumor type, residual disease, and expression of human epidermal growth factor receptor 2 (HER2), epidermal growth factor receptor (EGFR), breast cancer 1 (BRCA1), murine sarcoma viral oncogene homolog B (BRAF) and Kirsten rat sarcoma viral oncogene homolog (KRAS). Conclusions Our synthetic risk model may more accurately predict survival of epithelial ovarian cancer patients than current models. Electronic supplementary material The online version of this article (doi:10.1186/s13048-015-0195-6) contains supplementary material, which is available to authorized users.
- Published
- 2015
30. Methylation of DACT2 accelerates esophageal cancer development by activating Wnt signaling
- Author
-
James G. Herman, Malcolm V. Brock, Meiying Zhang, Baoping Cao, Enqiang Linghu, Tao He, Qimin Zhan, Mingzhou Guo, and Rong Xiang
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,Esophageal Neoplasms ,Molecular oncology ,Metastasis ,03 medical and health sciences ,Mice ,DACT2 ,0302 clinical medicine ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,metastasis ,esophageal cancer ,Wnt Signaling Pathway ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,business.industry ,Wnt signaling pathway ,Cancer ,wnt signaling ,Esophageal cancer ,DNA Methylation ,Middle Aged ,medicine.disease ,Squamous carcinoma ,Neoplasm Proteins ,Survival Rate ,030104 developmental biology ,HEK293 Cells ,Oncology ,Dysplasia ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,Heterografts ,Female ,methylation ,business ,Carrier Proteins ,Research Paper - Abstract
// Meiying Zhang 1, 2 , Enqiang Linghu 1 , Qimin Zhan 3 , Tao He 1 , Baoping Cao 1 , Malcolm V. Brock 4 , James G. Herman 5 , Rong Xiang 2 , Mingzhou Guo 1 1 Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing 100853, China 2 Medical College of NanKai University, Tianjin 300071, China 3 State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R.China 4 Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA 5 The Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA Correspondence to: Mingzhou Guo, e-mail: mzguo@hotmail.com Rong Xiang, e-mail: rxiang@nankai.edu.cn Keywords: DACT2, esophageal cancer, methylation, metastasis, Wnt signaling Received: August 14, 2015 Accepted: February 06, 2016 Published: February 23, 2016 ABSTRACT Esophageal cancer is one of the most common malignancies worldwide. DACT2 is frequently methylated in human lung, hepatic, gastric and thyroid cancers. The methylation status and function of DACT2 remain to be elucidated in human esophageal cancer. Ten esophageal cancer cell lines, 42 cases of dysplasia and 126 cases of primary esophageal cancer samples were analyzed in this study. The expression of DACT2 was detected in YES2 cells, while reduced DACT2 expression levels were found in TE8 and KYSE70 cells, and complete loss of DACT2 expression was found in KYSE30, KYSE140, KYSE150, KYSE410, KYSE450, TE3 and TE7 cells. Loss of expression or reduced expression of DACT2 correlated with promoter region hypermethylation in esophageal cancer cells. Restoration of DACT2 expression was induced by 5-aza-2′-deoxycytidine. In human primary esophageal squamous carcinoma, 69% (87/126) of samples were methylated. Methylation of DACT2 was significantly associated with tumor stage and metastasis ( P < 0.01, P < 0.05). DACT2 suppressed colony formation, cell migration and invasion in esophageal cancer cells, and it also suppressed esophageal cancer cell xenograft growth. DACT2 inhibited Wnt signaling in human esophageal cancer cells. In conclusion, DACT2 is frequently methylated in human esophageal cancer and its expression is regulated by promoter region methylation. DACT2 suppresses esophageal cancer growth by inhibiting Wnt signaling.
- Published
- 2015
31. Effect of Hepatitis B Virus Genotypes on the Efficacy of Adefovir Dipivoxil Antiviral Therapy
- Author
-
Zhili Wen, Qinghua Li, Hua Zhang, Haiming Zhang, Deming Tan, Le Deng, Meiying Zhang, and Ping Wu
- Subjects
Hepatitis B virus ,Genotype ,Hepatology ,business.industry ,medicine.drug_class ,virus diseases ,Lamivudine ,Hepatitis B ,Adefovir Dipivoxil ,medicine.disease_cause ,medicine.disease ,Placebo ,Virology ,digestive system diseases ,Infectious Diseases ,medicine ,Adefovir ,Antiviral drug ,business ,Nested polymerase chain reaction ,Research Article ,medicine.drug - Abstract
Background: Hepatitis B is a common infectious disease in China. Many studies have shown that the genotype of hepatitis B virus (HBV) is probably associated with the efficacy of some antiviral drugs such as interferon α (IFN-α) and Lamivudine (LAM). However, the association between HBV genotype and adefovir dipivoxil (ADV) is controversial. ADV is the most popular antiviral drug in China due to its low price, good antiviral efficacy, few side effects, and convenient of administration. Objectives: This study focused on the effect of HBV genotypes on antiviral efficacy of ADV in patients with chronic hepatitis B infection (CHB). Patients and Methods: A total of 526 HBeAg-positive patients with CHB were randomly allocated into two groups. One group took ADV and another group took placebo. Nested Polymerase Chain Reaction (PCR) with multiple pairs of genotype-specific primers (nPCR-MPP) was used to analyze genotypes of HBV in these patients. Antiviral efficacy after treatment for three, six, 12 months was compared among the patients with different HBV genotypes. Results: Genotype B (73.6%) and genotype C (26.4%) were detected in these patients. After treatment for 12 months, the rate of HBV DNA seroclearance, ALT normalization, and HBeAg seroconversion were significantly higher in ADV group than in placebo group (P < 0.05). However, there were no significant differences in these three rates between patients infected with genotype B and C (P > 0.05). Conclusions: HBV genotypes B and C have no significant difference in virologic, biochemical, and immunologic response to ADV.
- Published
- 2014
32. Methylation of DACT2 promotes papillary thyroid cancer metastasis by activating Wnt signaling
- Author
-
James G. Herman, Malcolm V. Brock, Meiying Zhang, Jindong Sheng, Mingzhou Guo, Zhiyan Zhao, and Baoguo Liu
- Subjects
Adult ,Male ,medicine.medical_specialty ,lcsh:Medicine ,Biology ,Biochemistry ,Metastasis ,Papillary thyroid cancer ,Thyroid carcinoma ,Internal medicine ,Cell Line, Tumor ,medicine ,Genetics ,Humans ,Thyroid Neoplasms ,Neoplasm Metastasis ,lcsh:Science ,Promoter Regions, Genetic ,Thyroid cancer ,Wnt Signaling Pathway ,Adaptor Proteins, Signal Transducing ,Regulation of gene expression ,Multidisciplinary ,DNA methylation ,Biology and life sciences ,Thyroid ,lcsh:R ,Carcinoma ,Wnt signaling pathway ,DNA, Neoplasm ,DNA ,Middle Aged ,medicine.disease ,Carcinoma, Papillary ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Endocrinology ,medicine.anatomical_structure ,Thyroid Cancer, Papillary ,Cancer research ,lcsh:Q ,Female ,Carrier Proteins ,DNA modification ,Research Article - Abstract
Thyroid cancer is the most common endocrine malignant disease and the incidence is increasing. DACT2 was found frequently methylated in human lung cancer and hepatocellular carcinoma. To explore the epigenetic change and the role of DACT2 in thyroid cancer, 7 thyroid cancer cell lines, 10 cases of non-cancerous thyroid tissue samples and 99 cases of primary thyroid cancer samples were involved in this study. DACT2 was expressed and unmethylated in K1, SW579, FTC-133, TT, W3 and 8505C cell lines. Loss of expression and complete methylation was found in TPC-1 cells. Restoration of DACT2 expression was induced by 5-aza-2′deoxycytidine treatment. It demonstrates that the expression of DACT2 was regulated by promoter region methylation. In human primary papillary thyroid cancer, 64.6% (64/99) was methylated and methylation of DACT2 was related to lymph node metastasis (p
- Published
- 2014
33. Metabolism of cystathionine, N-monoacetylcystathione, perhydro-1,4-thiazepine-3,5-dicarboxylic acid, and cystathionine ketimine in the liver and kidney of d,l-propargylglycine-treated rats
- Author
-
Jianying Zhang, Meiying Zhang, Kazunori Sugahara, Hiroyuki Kodama, and Deshun Ma
- Subjects
Male ,medicine.medical_specialty ,Thiazepines ,Endocrinology, Diabetes and Metabolism ,Glycine ,Atmospheric-pressure chemical ionization ,Kidney ,Cystathionine ,Endocrinology ,Internal medicine ,medicine ,Animals ,Rats, Wistar ,Amino Acid Metabolism, Inborn Errors ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,biology ,Metabolism ,medicine.disease ,Cystathionine beta synthase ,Rats ,Dicarboxylic acid ,medicine.anatomical_structure ,Liver ,chemistry ,Biochemistry ,Cystathioninuria ,Alkynes ,Toxicity ,biology.protein ,Kidney disease - Abstract
Experimental cystathioninuria was induced by injection of d,l -propargylglycine in rats. The novel cystathionine metabolites, N -monoacetylcystathionine (NAc-cysta), perhydro-1,4-thiazepine-3,5-dicarboxylic acid (PHTZDC), and cystathionine ketimine (CK), were identified previously in the urine of patients with cystathioninuria and d,l -propargylglycine-treated rats. In this study, we identified these compounds in the liver and kidney of d,l -propargylglycine-treated rats using liquid chromatography-mass spectrometry with an atmospheric pressure chemical ionization interface system (LC/APCI-MS) and an amino acid analyzer. The metabolism of these compounds in the liver and kidney of d,l -propargylglycine-treated rats was also studied. PHTZDC, NAc-cysta, and CK were accumulated in the rat tissues in proportion to the content of cystathionine after d,l -propargylglycine administration. The concentrations of these compounds in the liver were higher than those in the kidney, and these compounds reached maxima earlier in the liver than in the kidney.
- Published
- 1998
34. Abstract 4450: Silencing NKD2 by promoter region hypermethylation promotes esophageal cancer progression by activating Wnt signaling
- Author
-
Weili Yang, James G. Herman, Meiying Zhang, Baoping Cao, Qimin Zhan, Tao He, Mingzhou Guo, Yongshuai Jin, and Guanglin Zhong
- Subjects
Cancer Research ,business.industry ,Bisulfite sequencing ,Wnt signaling pathway ,Cancer ,Esophageal cancer ,medicine.disease ,medicine.disease_cause ,Oncology ,DNA methylation ,medicine ,Cancer research ,Gene silencing ,Epigenetics ,Carcinogenesis ,business - Abstract
Background: Despite the use of surgery or chemoradiotherapy to treat, esophageal cancer, its prognosis still remains poor, with a 5-year survival below 15%. Recent insights into the epigenetic mechanisms associated with multi-step carcinogenesis provide new opportunities to develop novel effective targeted therapies for esophageal squamous cell carcinoma. Naked cuticle homolog 2 (NKD2) was found frequently methylated in human breast cancer. The epigenetic changes and mechanisms of NKD2 in human esophageal cancer remain unclear. Methods: Nine esophageal cancer cell lines and 154 cases of primary esophageal cancer samples were analyzed using methylation specific PCR, immunohistochemistry, western blot, flow cytometry techniques and a xenograft mouse model. Results: Loss of NKD2 expression and complete methylation were found in KYSE150 and TE1 cells. Reduced expression and partial methylation were observed in KYSE30, KYSE70, KYSE410, KYSE140 and COLO680 cells. High level expression and unmethylation were detected in KYSE450 and TE8 cells. Re-expression of NKD2 was induced by 5-aza-2’-deoxycytidine in NKD2 unexpressed or reduced cells. NKD2 was methylated in 53.2% (82/154) of human primary esophageal cancer samples, and promoter region hypermethylation was associated with reduced expression of NKD2 significantly (p Citation Format: Mingzhou Guo, Baoping Cao, Weili Yang, Yongshuai Jin, Meiying Zhang, Tao He, Qimin Zhan, James G. Herman, Guanglin Zhong. Silencing NKD2 by promoter region hypermethylation promotes esophageal cancer progression by activating Wnt signaling. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4450.
- Published
- 2016
35. A high M1/M2 ratio of tumor-associated macrophages is associated with extended survival in ovarian cancer patients
- Author
-
Yifeng He, Meiying Zhang, Wenjing Wang, Aimin Zhao, Qing-qing Li, Wen Di, and Xiangjun Sun
- Subjects
Oncology ,Pathology ,Time Factors ,M1 ,Fluorescent Antibody Technique ,Kaplan-Meier Estimate ,M2 ,Tumor-associated macrophage ,Obstetrics and Gynaecology ,skin and connective tissue diseases ,Ovarian Neoplasms ,Obstetrics and Gynecology ,Cell Differentiation ,Middle Aged ,Prognosis ,Flow Cytometry ,Immunohistochemistry ,Serous fluid ,Phenotype ,Female ,hormones, hormone substitutes, and hormone antagonists ,Adult ,medicine.medical_specialty ,stomatognathic system ,Ovarian cancer ,Predictive Value of Tests ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Aged ,Neoplasm Staging ,Proportional Hazards Models ,Chi-Square Distribution ,Proportional hazards model ,business.industry ,Research ,Macrophages ,Cancer ,Reproducibility of Results ,medicine.disease ,Multivariate Analysis ,Neoplasm Grading ,business ,Chi-squared distribution ,Clear cell - Abstract
Background Tumor-associated macrophages (TAMs) are classified into two major phenotypes, M1 and M2. M1 TAMs suppress cancer progression, while M2 TAMs promote it. However, little is known regarding the role of TAMs in the development of ovarian cancer. Here, we investigated the relationship between TAM distribution patterns (density, microlocalization, and differentiation) and ovarian cancer histotypes, and we explored whether altered TAM distribution patterns influence long-term outcomes in ovarian cancer patients. Methods A total of 112 ovarian cancer patients were enrolled in this study, and the subjects were divided into two groups according to their survival (< 5 years vs. ≥ 5 years). Immunohistochemistry and immunofluorescence were used to determine the density, microlocalization, and differentiation status of TAMs in ovarian cancer tissues for each histotype. Kaplan-Meier survival and multivariate Cox regression analyses were used to evaluate the prognostic significance of TAM-related parameters in ovarian cancer. Results TAMs most frequently infiltrated into the cancer tissue of the serous histotype, followed by mucinous, undifferentiated, endometrioid, and clear cell histotypes (p = 0.049). The islet/stroma ratio of total TAMs varied among the cancer histotypes, with mucinous and undifferentiated cancers displaying the lowest and highest ratios, respectively (p = 0.005). The intratumoral TAM density significantly increased with increasing cancer stage and grade (p = 0.023 and 0.006, respectively). However, the overall M1/M2 TAM ratio decreased as the cancer stage increased (p = 0.012). In addition, the intra-islet M1/M2 ratio inversely correlated with the residual site size (p = 0.004). Among the TAM-related parameters, only the increased overall and intra-islet M1/M2 TAM ratios displayed prognostic significance in both the Kaplan-Meier survival and multivariate Cox regression analyses; however, the values of these two parameters did not differ significantly among the cancer histotypes. Conclusions The patients with increased overall or intra-islet M1/M2 TAM ratios presented with an improved 5-year prognosis. Nevertheless, the TAM distribution patterns did not influence the overall outcomes of different ovarian cancer histotypes.
- Published
- 2014
36. High MUC2 Expression in Ovarian Cancer Is Inversely Associated with the M1/M2 Ratio of Tumor-Associated Macrophages and Patient Survival Time
- Author
-
Wen Di, Xin Wu, Ting Xu, Meiying Zhang, Yifeng He, Qizhi He, and Xiangjun Sun
- Subjects
Pathology ,medicine.medical_specialty ,Colorectal cancer ,lcsh:Medicine ,Dinoprostone ,Metastasis ,Prostate cancer ,stomatognathic system ,medicine ,Humans ,lcsh:Science ,skin and connective tissue diseases ,Survival analysis ,Aged ,Neoplasm Staging ,Proportional Hazards Models ,Feedback, Physiological ,Ovarian Neoplasms ,Mucin-2 ,Multidisciplinary ,business.industry ,Macrophages ,lcsh:R ,Cancer ,Middle Aged ,Prognosis ,medicine.disease ,Adenocarcinoma, Mucinous ,Immunohistochemistry ,Survival Analysis ,Cystadenocarcinoma, Serous ,Gene Expression Regulation, Neoplastic ,Cyclooxygenase 2 ,Cancer cell ,Cancer research ,Adenocarcinoma ,lcsh:Q ,Female ,Ovarian cancer ,business ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,Research Article - Abstract
Mucin 2 (MUC2) is a mucin molecule aberrantly expressed by ovarian cancer cells. Previous in vitro studies have indicated that MUC2 promotes cancer growth and metastasis through a tumor-associated macrophage (TAM)-dependent mechanism. However, this mechanism has never been linked to clinical oncology, and its prognostic significance needed to be clarified. Here, we collected 102 consecutive ovarian cancer specimens and used the multiple immuno-histo-chemical/-fluorescent technique to determine the correlations between the MUC2 expression status, the ratio of M1/M2 TAMs and the densities of cyclooxygenase-2 (COX-2)(+) TAMs and COX-2(+) cancer cells. The Kaplan-Meier survival analysis and multivariate Cox regression analysis were used to evaluate the prognostic influences of these parameters. As a result, we found that the MUC2 overexpression (immunostaining ++/+++) was significantly correlated with a reduced ratio of M1/M2 TAMs (p
- Published
- 2013
37. Effect of six steroidal saponins isolated from anemarrhenae rhizoma on platelet aggregation and hemolysis in human blood
- Author
-
Zhiyun Meng, Suixu Xu, Meiying Zhang, Hiroyuki Kodama, Jianying Zhang, and Deshun Ma
- Subjects
Erythrocytes ,Platelet Aggregation ,Clinical Biochemistry ,Saponin ,Drug Evaluation, Preclinical ,Biochemistry ,Hemolysis ,Antithrombins ,Anemarrhena asphodeloides ,Antithrombotic ,Thromboplastin ,Medicine ,Humans ,Platelet ,chemistry.chemical_classification ,Plants, Medicinal ,Traditional medicine ,biology ,Molecular Structure ,business.industry ,Liliaceae ,Biochemistry (medical) ,General Medicine ,Saponins ,biology.organism_classification ,medicine.disease ,In vitro ,chemistry ,Partial Thromboplastin Time ,Steroids ,business ,Platelet Aggregation Inhibitors - Abstract
Six steroidal saponins were isolated from Anemarrhena asphodeloides Bunge (Liliaceae), a traditional chinese medicine, and named anemarrhenasaponin I (An-I), anemarrhenasaponin Ia (An-Ia), timosaponin B-I (TB-I), timosaponin B-II (TB-II), timosaponin B-III (TB-III), and timosaponin A-III (TA-III). The effects of these six compounds on platelet aggregation and hemolysis in human blood were studied. All these compounds provoked remarkable inhibiting effect on platelet aggregation, and activated partial thromboplastin times (APTT) are sensitive to the presence of these six compounds. Using an in vitro system, APTT was delayed with the increment of the concentrations of these six compounds. In these six compounds, only timosaponin A-III appeared a strong effect on hemolysis, and anemarrhenasaponin Ia had a slight effect on hemolysis, other had no effect on hemolysis. These results suggested that these steroidal saponins isolated from Anemarrhena asphodeloides Bunge (Liliaceae) might be used as a novel antithrombotic therapeutic agents in post-myocardial infarction.
- Published
- 1999
38. Priming effect of N-acetyl-S-(3-oxo-3-carboxy-n-propyl)cysteine in human neutrophils and tyrosyl phosphorylation of 45 kDa protein
- Author
-
Noriyoshi Masuoka, Yasuhiro Sagara, Jianying Zhang, Hiroyuki Kodama, Osamu Ito, Meiying Zhang, and Toshihiko Ubuka
- Subjects
Neutrophils ,Clinical Biochemistry ,Carboxylic Acids ,Biochemistry ,Phosphorylation Process ,chemistry.chemical_compound ,Superoxides ,medicine ,Staurosporine ,Humans ,Amino Acids ,Enzyme Inhibitors ,Phosphorylation ,Protein kinase A ,Cells, Cultured ,Protein Kinase C ,Chemistry ,Superoxide ,Biochemistry (medical) ,Drug Synergism ,General Medicine ,Blood Proteins ,Protein-Tyrosine Kinases ,medicine.disease ,N-Formylmethionine Leucyl-Phenylalanine ,Kinetics ,Cystathioninuria ,Tyrosine ,Tyrosine kinase ,Cysteine ,medicine.drug - Abstract
Human peripheral blood polymorphonuclear leukocytes were preincubated with N-acetylcystathionine and N-acetyl-S-(3-oxo-3-carboxy-n-propyl)cysteine (NAc-OCPC) found in the urine of a patient with cystathioninuria. NAc-OCPC significantly enhanced the N-formyl-methionyl-leucyl-phenylalanine-induced superoxide generation, whereas N-acetylcystathionine did not enhance the superoxide generation. When the cells were incubated with NAc-OCPC, the tyrosyl phosphorylation of 45 kDa protein of the cell was markedly increased with time. The phosphorylation process was dependent on the concentration of NAc-OCPC. Both the superoxide generation and the tyrosyl phosphorylation of 45 kDa protein increased by NAc-OCPC were inhibited by genistein and herbimycin A, the inhibitors of protein tyrosine kinase, but were rather enhanced by staurosporine, an inhibitor of protein kinase C.
- Published
- 1998
39. Silencing NKD2 by Promoter Region Hypermethylation Promotes Esophageal Cancer Progression by Activating Wnt Signaling
- Author
-
Yongshuai Jin, Guanglin Zhong, Meiying Zhang, Mingzhou Guo, Tao He, Qimin Zhan, Baoping Cao, James G. Herman, and Weili Yang
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,NKD2 ,Esophageal Neoplasms ,Esophageal cancer ,Mice, Nude ,Biology ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Gene Silencing ,Epigenetics ,Promoter Regions, Genetic ,Wnt Signaling Pathway ,Adaptor Proteins, Signal Transducing ,Aged ,Aged, 80 and over ,Mice, Inbred BALB C ,DNA methylation ,Cell growth ,Calcium-Binding Proteins ,Wnt signaling pathway ,Methylation ,Middle Aged ,medicine.disease ,Molecular biology ,Wnt signaling ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Disease Progression ,Heterografts ,Female ,Carrier Proteins - Abstract
IntroductionNaked cuticle homolog 2 (NKD2) was found to be frequently methylated in human breast and gastric cancers. However, the epigenetic changes and mechanisms of NKD2 in human esophageal cancer remain unclear.MethodsNine esophageal cancer cell lines and 154 cases of primary esophageal cancer samples were analyzed using methylation-specific polymerase chain reaction, immunohistochemical analysis, Western blot, and xenograft mouse models.ResultsLoss of NKD2 expression and complete methylation were found in KYSE150 and TE1 cells. Reduced NKD2 expression and partial methylation of the promoter region were observed in KYSE30, KYSE70, KYSE410, KYSE140, and COLO680 cells. High levels of NKD2 expression and unmethylation were detected in KYSE450 and TE8 cells. Reexpression of NKD2 was induced by 5-aza-2′-deoxycytidine in cells in which NKD2 was not expressed or cells in which NKD2 expression was reduced. NKD2 was methylated in 53.2% of human primary esophageal cancer samples (82 of 154), and promoter region hypermethylation was significantly associated with reduced expression of NKD2 (p < 0.01). NKD2 methylation was associated with tumor, node, and metastasis stage and lymph node metastasis (p < 0.01). Our results suggest that NKD2 is regulated by promoter region methylation and that methylation of NKD2 may serve as a prognostic marker in esophageal cancer. Our further studies demonstrate that NKD2 suppresses cell proliferation, colony formation, cell invasion, and migration and also induces G1/S checkpoint arrest in esophageal cancer cells. NKD2 suppressed xenograft tumor growth and inhibited Wnt signaling in human esophageal cancer cells.ConclusionsNKD2 is frequently methylated in human esophageal cancer, and the expression of NKD2 is regulated by promoter region methylation. NKD2 suppresses esophageal cancer progression by inhibiting Wnt signaling both in vitro and in vivo.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.