15 results on '"Wei-Wei Pan"'
Search Results
2. PD-L1 expression level in different thymoma stages and thymic carcinoma: a meta-analysis
- Author
-
Xiao-Feng Li, Chunwei Xu, Wei-Wei Pan, Kai-qi Du, Hua-Fei Chen, Li-Xin Wu, Min-Hua Wu, Jian-Hui Huang, Wenxian Wang, and You-cai Zhu
- Subjects
Adult ,Male ,Cancer Research ,Thymoma ,Programmed Cell Death 1 Receptor ,030204 cardiovascular system & hematology ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,PD-L1 ,Programmed cell death 1 ,Biomarkers, Tumor ,medicine ,Humans ,Thymic carcinoma ,Aged ,Neoplasm Staging ,biology ,Chemistry ,Thymus Neoplasms ,General Medicine ,Middle Aged ,Ligand (biochemistry) ,medicine.disease ,Immune checkpoint ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Pd l1 expression - Abstract
Background: The immune checkpoint ligand, programmed cell death 1 ligand 1 (PD-L1), is expressed in various tumors and associated with response to drugs that target programmed cell death protein 1. Previous studies have estimated the level of PD-L1 expression among different stages of thymoma and thymic carcinoma to evaluate its potential use as a diagnostic factor; however, its varying expression level has been problematic. We conducted this meta-analysis of published literature to evaluate PD-L1 expression in thymomas and thymic carcinomas. Methods: We analyzed 12 studies that included 320 patients with type A/AB/B1 thymoma, 225 patients with type B2/B3 thymoma, and 180 patients with thymic carcinoma. Results: No difference in PD-L1 expression level was found between the B2/B3 vs C groups (odds ratio [OR], 0.67; 95% confidence interval [CI], 0.26, 1.76; p = 0.42). However, the heterogeneity was very high ( I2 = 78%), and a significant difference was found between groups A/AB/B1 and B2/B3 (OR, 0.22; 95% CI, 0.12, 0.41; p < 0.001), with a relatively low heterogeneity ( I2 = 55%). Conclusion: PD-L1 positivity might be a useful factor to differentiate type A/AB/B1 thymoma from type B2/B3 and thymic carcinoma. This result might be valuable for potential anti PD-L1 treatment in thymoma and thymic carcinoma.
- Published
- 2020
3. Effect of icotinib on advanced lung adenocarcinoma patients with sensitive EGFR mutation detected in ctDNA by ddPCR
- Author
-
Lei Lei, Wen-Xian Wang, Quxia Zhang, Kai-qi Du, Hua-Fei Chen, Yong-Hua Min, Min Wang, Wei-Wei Pan, You-cai Zhu, Li-Xin Wu, Xiao-Feng Li, and Chunwei Xu
- Subjects
Cancer Research ,Lung ,business.industry ,Therapeutic effect ,medicine.disease ,Disease control ,Predictive value ,medicine.anatomical_structure ,Oncology ,Egfr mutation ,Icotinib ,medicine ,Cancer research ,Adenocarcinoma ,Radiology, Nuclear Medicine and imaging ,business ,L858r mutation - Abstract
Background: Whether or not EGFR mutation status detected by ddPCR in plasma predicts the effect of icotinib on patients with advanced lung adenocarcinoma was determined. Methods: Plasma and matched tissue specimens from patients with advanced lung adenocarcinoma were collected prior to icotinib treatment. The ARMS method was used to detect EGFR mutation status in DNA extracted from tissue specimens, while the EGFR mutation status in ctDNA extracted from plasma specimens was determined by ddPCR. The therapeutic effects of icotinib were compared between patients with EGFR - activating mutations detected by ddPCR in ctDNA and ARMS in tissue DNA. Results: EGFR mutation status was detected in 96 tissue and 100 plasma specimens. The sensitivity and positive predictive value of 19del detected in ctDNA by ddPCR was 70.97% (22/31) and 44.90% (22/49), respectively. The positive predictive value was 84.62% (22/26) and the sensitivity was 53.66% (22/41) for the L858R mutation. For the common sensitive EGFR mutations, ddPCR had a positive predictive value of 77.19% (44/57) and a sensitivity of 48.89% (44/90). Patients with sensitive EGFR mutations in ctDNA had objective response and disease control rates (DCR) similar to patients who had sensitive EGFR mutations in tissues detected by ARMS when treated with icotinib (57.14% vs . 51.51% and 92.86% vs . 90.91%, respectively). Conclusions: Patients with sensitive EGFR mutations in plasma specimens detected with ddPCR had a higher ORR and DCR compared with patients with sensitive EGFR mutations in tissue detected with the ARMS method.
- Published
- 2019
4. Attritional evaluation of lipophilic and hydrophilic metallated phthalocyanines for oncological photodynamic therapy
- Author
-
Lionel Mendes Dias, José E. Cavaco, Lianne R. de Haan, Emilie C B Desclos, Daniel J de Klerk, Jakub Kochan, Michal Heger, Leonardo Pereira Franchi, Przemek M. Krawczyk, Wei-wei Pan, Baoyue Ding, Farangis Sharifi, Barbara Mesquita, Xuan Huang, Enzo M. Scutigliani, Antonio Claudio Tedesco, Albert C. van Wijk, Daniël Ernst, Mark J de Keijzer, Medical Biology, Adult Psychiatry, APH - Mental Health, Amsterdam Neuroscience - Complex Trait Genetics, Amsterdam Neuroscience - Mood, Anxiety, Psychosis, Stress & Sleep, Graduate School, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, Amsterdam Neuroscience - Neurodegeneration, CCA - Cancer biology and immunology, and Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
Programmed cell death ,Cell Membrane Permeability ,Indoles ,Time Factors ,medicine.medical_treatment ,Cell ,Biophysics ,Sulforhodamine B ,Photodynamic therapy ,Antineoplastic Agents ,Apoptosis ,Isoindoles ,Photosensitizers, cell death ,Photosensitizers ,TERAPIA FOTODINÂMICA ,Cell survival ,Phototoxicity ,chemistry.chemical_compound ,Structure-Activity Relationship ,Cell Line, Tumor ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Drug Carriers ,Radiation ,Photosensitizing Agents ,Radiological and Ultrasound Technology ,Zinc phthalocyanine ,Dose-Response Relationship, Radiation ,Drug Liberation ,medicine.anatomical_structure ,cell death ,chemistry ,Photochemotherapy ,Dark toxicity ,Liposomes ,Cancer research ,A431 cells ,Intracellular ,Aluminum phthalocyanine - Abstract
Background and aim Oncological photodynamic therapy (PDT) relies on photosensitizers (PSs) to photo-oxidatively destroy tumor cells. Currently approved PSs yield satisfactory results in superficial and easy-to-access tumors but are less suited for solid cancers in internal organs such as the biliary system and the pancreas. For these malignancies, second-generation PSs such as metallated phthalocyanines are more appropriate. Presently it is not known which of the commonly employed metallated phtahlocyanines, namely aluminum phthalocyanine (AlPC) and zinc phthalocyanine (ZnPC) as well as their tetrasulfonated derivatives AlPCS4 and ZnPCS4, is most cytotoxic to tumor cells. This study therefore employed an attritional approach to ascertain the best metallated phthalocyanine for oncological PDT in a head-to-head comparative analysis and standardized experimental design. Methods ZnPC and AlPC were encapsulated in PEGylated liposomes. Analyses were performed in cultured A431 cells as a template for tumor cells with a dysfunctional P53 tumor suppressor gene and EGFR overexpression. First, dark toxicity was assessed as a function of PS concentration using the WST-1 and sulforhodamine B assay. Second, time-dependent uptake and intracellular distribution were determined by flow cytometry and confocal microscopy, respectively, using the intrinsic fluorescence of the PSs. Third, the LC50 values were established for each PS at 671 nm and a radiant exposure of 15 J/cm2 following 1-h PS exposure. Finally, the mode of cell death as a function of post-PDT time and cell cycle arrest at 24 h after PDT were analyzed. Results In the absence of illumination, AlPC and ZnPC were not toxic to cells up to a 1.5-μM PS concentration and exposure for up to 72 h. Dark toxicity was noted for AlPCS4 at 5 μM and ZnPCS4 at 2.5 μM. Uptake of all PSs was observed as early as 1 min after PS addition to cells and increased in amplitude during a 2-h incubation period. After 60 min, the entire non-nuclear space of the cell was photosensitized, with PS accumulation in multiple subcellular structures, especially in case of AlPC and AlPCS4. PDT of cells photosensitized with ZnPC, AlPC, and AlPCS4 yielded LC50 values of 0.13 μM, 0.04 μM, and 0.81 μM, respectively, 24 h post-PDT (based on sulforhodamine B assay). ZnPCS4 did not induce notable phototoxicity, which was echoed in the mode of cell death and cell cycle arrest data. At 4 h post-PDT, the mode of cell death comprised mainly apoptosis for ZnPC and AlPC, the extent of which was gradually exacerbated in AlPC-photosensitized cells during 8 h. ZnPC-treated cells seemed to recover at 8 h post-PDT compared to 4 h post-PDT, which had been observed before in another cell line. AlPCS4 induced considerable necrosis in addition to apoptosis, whereby most of the cell death had already manifested at 2 h after PDT. During the course of 8 h, necrotic cell death transitioned into mainly late apoptotic cell death. Cell death signaling coincided with a reduction in cells in the G0/G1 phase (ZnPC, AlPC, AlPCS4) and cell cycle arrest in the S-phase (ZnPC, AlPC, AlPCS4) and G2 phase (ZnPC and AlPC). Cell cycle arrest was most profound in cells that had been photosensitized with AlPC and subjected to PDT. Conclusions Liposomal AlPC is the most potent PS for oncological PDT, whereas ZnPCS4 was photodynamically inert in A431 cells. AlPC did not induce dark toxicity at PS concentrations of up to 1.5 μM, i.e., > 37 times the LC50 value, which is favorable in terms of clinical phototoxicity issues. AlPC photosensitized multiple intracellular loci, which was associated with extensive, irreversible cell death signaling that is expected to benefit treatment efficacy and possibly immunological long-term tumor control, granted that sufficient AlPC will reach the tumor in vivo. Given the differential pharmacokinetics, intracellular distribution, and cell death dynamics, liposomal AlPC may be combined with AlPCS4 in a PS cocktail to further improve PDT efficacy.
- Published
- 2021
5. WISP2 promotes cell proliferation via targeting ERK and YAP in ovarian cancer cells
- Author
-
Yi Zhang, Liu-qing Yang, Zi-Qing Shi, Yao Han, Wei-Wei Pan, Zi-Yan Chen, Han Zhang, Meng-Dan Lyu, and Heng-Yan Zhu
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,MAP Kinase Signaling System ,Proliferation ,Clone (cell biology) ,Mice, Nude ,Apoptosis ,Cell Cycle Proteins ,lcsh:Gynecology and obstetrics ,CCN Intercellular Signaling Proteins ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Movement ,Ovarian cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,lcsh:RG1-991 ,Cell Proliferation ,Ovarian Neoplasms ,WISP2 ,Cell growth ,Kinase ,Chemistry ,Research ,Cell Cycle ,Ovary ,Obstetrics and Gynecology ,Cell cycle ,medicine.disease ,Repressor Proteins ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Signal Transduction ,Transcription Factors - Abstract
Background Wnt-inducible signaling pathway protein 2 (WISP2) is a wnt1-induced signaling pathway protein 2. Although studies indicate that WISP2 may promote the development of various tumors, its role in ovarian cancer remains unclear. The objective of the current study was to analyze the effects of WISP2 on the proliferation and migration of ovarian cancer cells in vitro and in vivo. Results Immunohistochemistry and western blotting indicated that WISP2 was highly expressed in various ovarian cancer tissues and cell lines, but weakly expressed in normal ovary tissue. WISP2 deletion inhibited cell growth, clone formation, and migration of ovarian cancer cells while promoting cell apoptosis and affecting the cell cycle. This growth inhibitory effect caused by WISP2 loss is due to the inhibition of phosphorylated extracellular signal-related kinase (p-ERK)1/2, as well as CCAAT/enhancer-binding protein α (CEBPα) and CEPBβ. In addition, WISP2 deletion also activated the Yes-associated protein (YAP). Conclusion WISP2 deletion inhibits ovarian cancer cell proliferation by affecting ERK signaling pathways.
- Published
- 2020
6. A model to predict the onset of non-alcoholic fatty liver disease within 2 years in elderly adults
- Author
-
Zhen-Zhen Yu, Shuai Gao, Xi-Mei Gao, Ya-Jie Lin, Xiao-Peng Fan, Ping Xu, and Wei-Wei Pan
- Subjects
medicine.medical_specialty ,Framingham Risk Score ,Cirrhosis ,Hepatology ,business.industry ,Fatty liver ,Gastroenterology ,nutritional and metabolic diseases ,Disease ,medicine.disease ,digestive system diseases ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Internal medicine ,medicine ,030211 gastroenterology & hepatology ,Analysis of variance ,Age of onset ,business ,Body mass index - Abstract
Background and Aim Non-alcoholic fatty liver disease (NAFLD) is a common cause of chronic hepatitis, which leads to cirrhosis and hepatocellular carcinoma. However, it is difficult to identify subjects at high risk for NAFLD onset. This study aims to construct a model to predict the onset of NAFLD within 2 years in elderly adults. Methods This study included and followed 3378 initial NAFLD-free subjects aged 60 years or over for 2 years, which were randomly divided into a training set and a validation set. NAFLD was diagnosed on ultrasound. Clinical and laboratory data were recorded at baseline. A model was constructed in the training set to predict the onset of NAFLD and validated in the validation set. Results Body mass index, hemoglobin, fasting blood glucose, and triglycerides were identified as predictors for the onset of NAFLD. A risk score (R) was calculated by them. It classified the subjects into low-risk group (R ≤ −2.88), moderate-risk group (−2.88 −1.26). In the training set, 4.68% of the participants in the low-risk group, 11.59% of the participants in the moderate-risk group, and 31.02% of the participants in the high-risk group developed NAFLD. In the validation set, 5.84% of the participants in the low-risk group, 10.57% of the participants in the moderate-risk group, and 29.44% of the participants in the high-risk group developed NAFLD. Conclusions This study developed a model to predict the onset of NAFLD in elderly adults, which might provide indications for intervention to these subjects.
- Published
- 2017
7. Abstract 130: CpG binding protein promotes cell proliferation through H3K4 methylation in ovarian cancer
- Author
-
Lei Ao, Li-Zhong Wang, Liu-qing Yang, Ya-Bo Jiang, Yao Han, Xuan Huang, Heng-Yan Zhu, Ying Xu, Chun-wei Xu, Wei-wei Pan, Michal Heger, Xiao-min Wang, Shu-Qun Cheng, Han-yin Hu, and Xuan Che
- Subjects
Cancer Research ,biology ,Cell growth ,Chemistry ,medicine.disease ,Ubiquitin ligase ,Histone H3 ,Oncology ,Apoptosis ,biology.protein ,Cancer research ,medicine ,Cytotoxic T cell ,Neddylation ,Ovarian cancer ,Cullin - Abstract
E3 ubiquitin ligase CRL4 complex activation requires cullin neddylation. MLN4924, a NEDD8-activating enzyme (NAE) inhibitor, reportedly blocked cullin neddylation and inactivated cullin (CUL)-RING E3 ligases (CRLs). Studies have shown that MLN4924 inhibits cell proliferation and survival. We examined the oncostatic and cytotoxic properties of MLN4924 in ovarian cancer. MLN4924 blocked human cancer cell proliferation in nude mice by inhibiting CpG binding protein CFP1 expression. Loss of CFP1 reduced cell growth, promoted apoptosis, and increased senescence. CFP1-dependent H3K4 trimethylation is necessary to maintain the expression of target genes in ovarian cancer cells. We identified bone marrow stromal cell antigen 2 (BST2) and noggin (NOG) as direct targets of CFP1. Their expression was selectively induced by CFP1 deletion. Furthermore, deletion of NOG and overexpression of BST2 prevented the growth-inhibitory effect of CFP1 loss. Our study further demonstrated that CRL4 inactivation reduced CFP1 transcription and trimethylation on lysine 4 of histone H3, which in turn inhibited cell proliferation and induced apoptosis. Therefore, CRL4 constitutes a druggable target in ovarian cancer. Citation Format: Liu-qing Yang, Han-yin Hu, Yao Han, Heng-Yan Zhu, Xiao-min Wang, Lei Ao, Ying Xu, Xuan Huang, Xuan Che, Li-Zhong Wang, Ya-Bo Jiang, Chun-wei Xu, Shu-Qun Cheng, Michal Heger, Wei-wei Pan. CpG binding protein promotes cell proliferation through H3K4 methylation in ovarian cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 130.
- Published
- 2020
8. DAXX promotes ovarian cancer ascites cell proliferation and migration by activating the ERK signaling pathway
- Author
-
Xue-Ping Lin, Wei-Wei Pan, Zhong-Fei Shen, Sheng-Bing Liu, and Ying Xu
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,MAP Kinase Signaling System ,Cell ,Mice, Nude ,lcsh:Gynecology and obstetrics ,Ascites cell ,03 medical and health sciences ,Promyelocytic leukemia protein ,0302 clinical medicine ,Death-associated protein 6 ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cell migration ,Cell proliferation ,lcsh:RG1-991 ,Ovarian Neoplasms ,biology ,Cell growth ,Kinase ,Chemistry ,Research ,Intracellular Signaling Peptides and Proteins ,Ascites ,Nuclear Proteins ,Obstetrics and Gynecology ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,CCAAT-Enhancer-Binding Proteins ,DAXX ,Cancer research ,biology.protein ,Female ,Carrier Proteins ,Co-Repressor Proteins ,Molecular Chaperones - Abstract
Background The death-domain-associated protein (DAXX) was originally identified as a protein that binds to the transmembrane death receptor FAS and enhances both FAS-induced and transforming growth factor-β-dependent apoptosis. In a previous study, we found that nude mice injected with DAXX-overexpressing cells (ES-2-DAXX) accumulated large concentrations of first-generation ascites cells (I ascites cells). The role of DAXX in the development of ascites is unknown. The aim of this study was to analyze the effect of DAXX on proliferation and migration of ascites cells in ovarian cancer in vitro and in vivo. Methods Nude mice were housed in cages with a 14:10 h light:dark cycle; water and food were provided ad libitum. ES-2-DAXX cells (1×106) were injected intraperitoneally into athymic nude mice (8-week-old female mice). After 4 weeks, I ascites cells were collected. The I ascites cells were injected intraperitoneally into athymic nude mice (8-week-old female mice). After 4 weeks, II ascites cells were collected and cultured. Ascites cell survival, migration, and colony formation were measured using colony formation and cell growth assays. Immunofluorescent staining revealed the co-localization of DAXX and promyelocytic leukemia protein (PML) in ascites cell nuclei. Western blotting and immunohistochemistry showed that extracellular signal-related kinase (p-ERK) 1/2 and CEBP-β were highly expressed in tumor tissues formed by II ascites cells. Through immunoprecipitation, we also found that DAXX can interact with CEBP-β. Results DAXX enhanced ascites cell survival, migration, and colony formation. DAXX and PML nuclear foci dramatically increased in a passage-dependent manner in ascites cells, DAXX promoted the tumor growth of ascites cells in vivo, increased ascites cell proliferation in vivo, and enhanced ascites cell survival and migration by activating the ERK signalling pathway and integrating with CEBP-β. Conclusions DAXX can interact with CEBP-β. DAXX can induce ovarian cancer ascites formation by activating the ERK signal pathway and binding to CEBP-β.
- Published
- 2018
9. Cell type-dependent function of LATS1/2 in cancer cell growth
- Author
-
Toshiro Moroishi, Ja Hyun Koo, Wei-Wei Pan, and Kun-Liang Guan
- Subjects
0301 basic medicine ,Transcriptional Activation ,Cancer Research ,Cell type ,Transcription, Genetic ,Carcinogenesis ,Mice, Nude ,LATS ,Biology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Article ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Hippo ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,WISP2 ,Hippo signaling pathway ,Cell growth ,Tumor Suppressor Proteins ,HEK 293 cells ,Wnt signaling pathway ,Cell biology ,Repressor Proteins ,030104 developmental biology ,HEK293 Cells ,MC38 ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Intercellular Signaling Peptides and Proteins ,Female ,YAP ,Signal Transduction ,Transcription Factors - Abstract
The Hippo pathway controls organ size and tissue homeostasis, and its dysregulation often contributes to tumorigenesis. Extensive studies have shown that the Hippo pathway inhibits cell proliferation, and survival in a cell-autonomous manner. We examined the function of the Hippo pathway kinases LATS1/2 (large tumor suppressor 1 and 2) in cancer cells. As expected, loss of LATS1/2 promotes cancer cell growth in most cell lines. Surprisingly, however, LATS1/2 deletion inhibits the growth of murine MC38 colon cancer cells, especially under detachment conditions. This growth inhibitory effect caused by LATS1/2 deletion is due to uncontrolled activation of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), the key downstream transcriptional coactivators inhibited by LATS1/2. We identified Wnt inducible signaling pathway protein 2 (Wisp2) and coiled-coil domain containing 80 (Ccdc80) as direct targets of YAP/TAZ. Their expression is selectively induced by LATS1/2 deletion in MC38 cells. Furthermore, deletion of WISP2 and CCDC80 prevents the growth inhibitory effect of LATS1/2 loss in MC38 cells. Our study demonstrates that the function of LATS1/2 in cell growth is cell context dependent, suggesting that LATS1/2 inhibition can be a therapeutic approach for some cancer types.
- Published
- 2018
10. TET1 inhibits cell proliferation by inducing RASSF5 expression
- Author
-
Wei-Wei Pan, Bo-Tai Li, Ying Xu, Heng-Yu Fan, Sheng-Bing Liu, and Chao Yu
- Subjects
0301 basic medicine ,DNA methylation ,Cell growth ,proliferation ,Biology ,medicine.disease ,TET1 ,law.invention ,03 medical and health sciences ,030104 developmental biology ,ovarian cancer ,Oncology ,CpG site ,law ,Cell culture ,Ubiquitin ligase complex ,Immunology ,Cancer research ,medicine ,Suppressor ,RASSF5 ,Ectopic expression ,Ovarian cancer ,Research Paper - Abstract
Tet methylcytosine dioxygenases (TETs) catalyze the oxidative reactions of 5-methylcytosine to 5-hydroxymethylcytosine (5hmC). However, TET1 roles in ovarian cancer cell growth are unknown. Here, we show that ectopic expression of TET1 increased 5hmC levels, and inhibited proliferation and colony formation in ovarian cancer cell lines. Furthermore, in vitro and in vivo functional studies demonstrated that TET1 overexpression is necessary for the suppression of ovarian cancer growth, whereas depletion of TET1 expression had the opposite effect. Furthermore, the results of RNA-seq and qRT-PCR analyses identified a tumor suppressor, Ras association domain family member 5 (RASSF5), as the key downstream target of TET1. TET1 promotes RASSF5 expression by demethylating a CpG site within RASSF5 promoter. Up-regulated RASSF5 expression leads to the suppression of ovarian cancer cells growth. Additionally, we demonstrated that inhibition of CUL4-DDB1 ubiquitin ligase complex decrease 5hmC levels in ovarian cancer cells. These results provide new insights into the understanding of how ovarian cancers develop and grow, and identify TET1 as a key player in this process.
- Published
- 2017
11. The Hippo Pathway Kinases LATS1/2 Suppress Cancer Immunity
- Author
-
Yu Fujita, Kun-Liang Guan, Jun Qin, Matthew V. Holt, Dennis A. Carson, Toshiro Moroishi, Wei-Wei Pan, and Tomoko Hayashi
- Subjects
0301 basic medicine ,TAZ ,medicine.medical_treatment ,LATS ,Inbred C57BL ,Medical and Health Sciences ,Mice ,Cancer immunotherapy ,Hippo ,Interferon ,Neoplasms ,2.1 Biological and endogenous factors ,Aetiology ,Inbred BALB C ,Cancer ,Toll-like receptor ,Immunogenicity ,Toll-Like Receptors ,interferon ,Biological Sciences ,Inbred C3H ,B16 melanoma ,YAP ,immunotherapy ,medicine.drug ,Signal Transduction ,chemical and pharmacologic phenomena ,Biology ,Protein Serine-Threonine Kinases ,Cancer Vaccines ,General Biochemistry, Genetics and Molecular Biology ,Article ,Vaccine Related ,03 medical and health sciences ,Immune system ,cancer immunity ,medicine ,Immune Tolerance ,Genetics ,Animals ,exosome ,Hippo signaling pathway ,Tumor Suppressor Proteins ,Immunotherapy ,030104 developmental biology ,TRIF ,Immunology ,Cancer research ,Immunization ,Gene Deletion ,Developmental Biology - Abstract
Poorly immunogenic tumor cells evade host immunity and grow even in the presence of an intact immune system, but the complex mechanisms regulating tumor immunogenicity have not been elucidated. Here, we discovered an unexpected role of the Hippo pathway in suppressing anti-tumor immunity. We demonstrate that, in three different murine syngeneic tumor models (B16, SCC7, and 4T1), loss of the Hippo pathway kinases LATS1/2 (large tumor suppressor 1 and 2) in tumor cells inhibits tumor growth. Tumor regression by LATS1/2 deletion requires adaptive immune responses, and LATS1/2 deficiency enhances tumor vaccine efficacy. Mechanistically, LATS1/2-null tumor cells secrete nucleic-acid-rich extracellular vesicles, which induce a typeI interferon response via the Toll-like receptors-MYD88/TRIF pathway. LATS1/2 deletion in tumors thus improves tumor immunogenicity, leading to tumor destruction by enhancing anti-tumor immune responses. Our observations uncover a key role of the Hippo pathway in modulating tumor immunogenicity and demonstrate a proof of concept for targeting LATS1/2 in cancer immunotherapy.
- Published
- 2016
12. CRL4 Complex Regulates Mammalian Oocyte Survival and Reprogramming by Activation of TET Proteins
- Author
-
Jian-Jie Zhou, Xiao-Meng Li, Zhao-Jia Ge, Wei-Wei Pan, Qing-Yuan Sun, Heng-Yu Fan, Chao Yu, Yin-Li Zhang, Zhong-Wei Wang, Chao Tong, and Yong Cang
- Subjects
Cell Survival ,Protein Serine-Threonine Kinases ,Gonadal Dysgenesis ,Bioinformatics ,Dioxygenases ,Mixed Function Oxygenases ,Mice ,DDB1 ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Gene silencing ,Gene Silencing ,Gene ,Mice, Knockout ,Multidisciplinary ,Zygote ,biology ,Ovary ,Cellular Reprogramming ,Cullin Proteins ,Oocyte ,Ubiquitin ligase ,Cell biology ,DNA-Binding Proteins ,Fertility ,medicine.anatomical_structure ,Oocytes ,biology.protein ,Female ,Carrier Proteins ,Reprogramming ,Germ cell ,HeLa Cells - Abstract
Ubiquitin Fertility Insurance The female mammal's reproductive lifespan is determined by a pool of ovarian primordial follicles that are generated early in life. Yu et al. (p. 1518 ) found that in mice, the ubiquitin E3 ligase complex CRL4 is essential for oocyte survival within primordial follicles and for development after fertilization. CRL4 binds to and activates an adaptor protein that mediates ubiquitination, but if any component is deleted, the genes required for oocyte maintenance and early embryo development are silenced and the female mice become infertile.
- Published
- 2013
13. Ubiquitin E3 Ligase CRL4CDT2/DCAF2 as a Potential Chemotherapeutic Target for Ovarian Surface Epithelial Cancer
- Author
-
Lian-Jun Guo, Jian-Jie Zhou, Heng-Yu Fan, Ying Xu, Chao Yu, Dawang Zhou, Wei-Wei Pan, Fang-Zhou Song, and Hai-Yi Zhang
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Programmed cell death ,endocrine system diseases ,Cell Survival ,Ubiquitin-Protein Ligases ,Blotting, Western ,Mice, Nude ,Apoptosis ,Cell Cycle Proteins ,macromolecular substances ,Cyclopentanes ,Carcinoma, Ovarian Epithelial ,Biochemistry ,Mice ,Ovarian tumor ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasms, Glandular and Epithelial ,Molecular Biology ,Cells, Cultured ,Ovarian Neoplasms ,Dose-Response Relationship, Drug ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Cell growth ,fungi ,Nuclear Proteins ,Cell Biology ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Ubiquitin ligase ,Cell biology ,Gene Expression Regulation, Neoplastic ,enzymes and coenzymes (carbohydrates) ,Pyrimidines ,embryonic structures ,biology.protein ,Cancer research ,Female ,RNA Interference ,Neddylation ,CUL4A ,Ovarian cancer ,Cyclin-Dependent Kinase Inhibitor p27 ,Cullin ,DNA Damage - Abstract
Cullin-RING ubiquitin ligases (CRLs) are the largest family of E3 ligases and require cullin neddylation for their activation. The NEDD8-activating enzyme inhibitor MLN4924 reportedly blocked cullin neddylation and inactivated CRLs, which resulted in apoptosis induction and tumor suppression. However, CRL roles in ovarian cancer cell survival and the ovarian tumor repressing effects of MLN4924 are unknown. We show here that CRL4 components are highly expressed in human epithelial ovarian cancer tissues. MLN4924-induced DNA damage, cell cycle arrest, and apoptosis in ovarian cancer cells in a time- and dose-dependent manner. In addition, MLN4924 sensitized ovarian cancer cells to other chemotherapeutic drug treatments. Depletion of CRL4 components Roc1/2, Cul4a, and DDB1 had inhibitory effects on ovarian cancer cells similar to MLN4924 treatment, which suggested that CRL4 inhibition contributed to the chemotherapeutic effect of MLN4924 in ovarian cancers. We also investigated for key CRL4 substrate adaptors required for ovarian cancer cells. Depleting Vprbp/Dcaf1 did not significantly affect ovarian cancer cell growth, even though it was expressed by ovarian cancer tissues. However, depleting Cdt2/Dcaf2 mimicked the pharmacological effects of MLN4924 and caused the accumulation of its substrate, CDT1, both in vitro and in vivo. MLN4924-induced DNA damage and apoptosis were partially rescued by Cdt1 depletion, suggesting that CRL4(CDT2) repression and CDT1 accumulation were key biochemical events contributing to the genotoxic effects of MLN4924 in ovarian cancer cells. Taken together, these results indicate that CRL4(CDT2) is a potential drug target in ovarian cancers and that MLN4924 may be an effective anticancer agent for targeted ovarian cancer therapy.
- Published
- 2013
14. Isoquinoline alkaloids from Zanthoxylum simulans and their biological evaluation
- Author
-
Wen-Bing Sheng, Sheng-Hui Yang, Yue-Hu Wang, Wei-wei Pan, Xiao-Jiang Zhou, Junfeng Wang, Rui Cai, and Yan-Qun Liu
- Subjects
Pharmacology ,Zanthoxylum ,biology ,business.industry ,Spectrum Analysis ,Arthritis ,Zanthoxylum simulans ,biology.organism_classification ,medicine.disease ,Isoquinolines ,Rats ,Arthritis, Rheumatoid ,chemistry.chemical_compound ,Alkaloids ,chemistry ,Antirheumatic Agents ,Drug Discovery ,medicine ,Animals ,Isoquinoline ,Spectrum analysis ,business ,Biological evaluation - Published
- 2014
15. Death Domain-associated Protein DAXX Promotes Ovarian Cancer Development and Chemoresistance*
- Author
-
Xiao-Man Liu, Wei-Wei Pan, Lian-Jun Guo, Chao Yu, Qinghua Shi, Jian-Jie Zhou, Ying Xu, and Heng-Yu Fan
- Subjects
endocrine system ,endocrine system diseases ,Cell Survival ,Gene Expression ,Mice, Nude ,Promyelocytic Leukemia Protein ,medicine.disease_cause ,Biochemistry ,Radiation Tolerance ,Metastasis ,Promyelocytic leukemia protein ,Mice ,Death-associated protein 6 ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Nuclear protein ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Ovarian Neoplasms ,Oncogene ,biology ,Cell growth ,Tumor Suppressor Proteins ,Cystadenoma, Serous ,Ovary ,Nuclear Proteins ,Epithelial Cells ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,female genital diseases and pregnancy complications ,Cell biology ,Cell Transformation, Neoplastic ,Drug Resistance, Neoplasm ,biology.protein ,Female ,Ovarian cancer ,Carcinogenesis ,Co-Repressor Proteins ,DNA Damage ,Molecular Chaperones ,Transcription Factors - Abstract
The role of DAXX in ovarian cancer development and metastasis has not been investigated before now.Overexpression of DAXX enhanced ovarian cancer cell proliferation, colony formation, and migration, whereas Daxx depletion had the opposite effects.DAXX promotes ovarian cancer cell proliferation and chemoresistance.ModulatingDAXXmay be an effective strategy for preventing the recurrence and chemoresistance of ovarian cancers. Understanding the genes involved in apoptosis and DNA damage responses may improve therapeutic strategies for ovarian cancer. The death domain-associated protein DAXX can be either a pro-apoptotic or an anti-apoptotic factor, depending on the cell type and context. In this study, we found that DAXX was highly expressed in human ovarian surface epithelial tumors but not in granulosa cell tumors. In cultured ovarian cancer cells, DAXX interacted with promyelocytic leukemia protein (PML) and localized to subnuclear domains (so-called PML nuclear bodies). A role for DAXX in ovarian cancer cell proliferation, metastasis, and radio/chemoresistance was examined. Overexpression of DAXX enhanced multiple ovarian cancer cell lines' proliferation, colony formation, and migration, whereas Daxx depletion by RNA interference had the opposite effects. When transplanted into nude mice, ovarian cancer cells that overexpressed DAXX displayed enhanced tumorigenesis capability in vivo, whereas Daxx depletion inhibited tumor development. Importantly, Daxx induced tumorigenic transformation of normal ovarian surface epithelial cells. Daxx also protected ovarian cancer cells against x-irradiation- and chemotherapy-induced DNA damage by interacting with PML. Taken together, our results suggest that DAXX is a novel ovarian cancer oncogene that promotes ovarian cancer cell proliferation and chemoresistance in ovarian cancer cells. Thus, modulating DAXX-PML nuclear body activity may be an effective strategy for preventing the recurrence and chemoresistance of ovarian cancers.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.