1.Chinese agarwood petroleum ether extract suppressed gastric cancer progression via up-regulation of DNA damage-induced G0/G1 phase arrest and HO-1-mediated ferroptosis.
Lishan OUYANG ; Xuejiao WEI ; Fei WANG ; Huiming HUANG ; Xinyu QIU ; Zhuguo WANG ; Peng TAN ; Yufeng GAO ; Ruoxin ZHANG ; Jun LI ; Zhongdong HU
Chinese Journal of Natural Medicines (English Ed.) 2025;23(10):1210-1220
Gastric cancer (GC) is characterized by high morbidity and mortality rates. Chinese agarwood comprises the resin-containing wood of Aquilaria sinensis (Lour.) Gilg., traditionally utilized for treating asthma, cardiac ischemia, and tumors. However, comprehensive research regarding its anti-GC effects and underlying mechanisms remains limited. In this study, Chinese agarwood petroleum ether extract (CAPEE) demonstrated potent cytotoxicity against human GC cells, with half maximal inhibitory concentration (IC50) values for AGS, HGC27, and MGC803 cells of 2.89, 2.46, and 2.37 μg·mL-1, respectively, at 48 h. CAPEE significantly induced apoptosis in these GC cells, with B-cell lymphoma-2 (BCL-2) associated X protein (BAX)/BCL-2 antagonist killer 1 (BAK) likely mediating CAPEE-induced apoptosis. Furthermore, CAPEE induced G0/G1 phase cell cycle arrest in human GC cells via activation of the deoxyribonucleic acid (DNA) damage-p21-cyclin D1/cyclin-dependent kinase 4 (CDK4) signaling axis, and increased Fe2+, lipid peroxides and reactive oxygen species (ROS) levels, thereby inducing ferroptosis. Ribonucleic acid (RNA) sequencing, real-time quantitative polymerase chain reaction (RT-qPCR), and Western blotting analyses revealed CAPEE-mediated upregulation of heme oxygenase-1 (HO-1) in human GC cells. RNA interference studies demonstrated that HO-1 knockdown reduced CAPEE sensitivity and inhibited CAPEE-induced ferroptosis in human GC cells. Additionally, CAPEE administration exhibited robust in vivo anti-GC activity without significant toxicity in nude mice while inhibiting tumor cell growth and promoting apoptosis in tumor tissues. These findings indicate that CAPEE suppresses human GC cell growth through upregulation of the DNA damage-p21-cyclin D1/CDK4 signaling axis and HO-1-mediated ferroptosis, suggesting its potential as a candidate drug for GC treatment.
Animals
;
Humans
;
Mice
;
Antineoplastic Agents, Phytogenic
;
Apoptosis/drug effects*
;
Cell Line, Tumor
;
Cyclin D1/genetics*
;
Cyclin-Dependent Kinase 4/genetics*
;
DNA Damage/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
;
Ferroptosis/drug effects*
;
G1 Phase Cell Cycle Checkpoints/drug effects*
;
Heme Oxygenase-1/genetics*
;
Mice, Inbred BALB C
;
Mice, Nude
;
Plant Extracts/pharmacology*
;
Stomach Neoplasms/physiopathology*
;
Thymelaeaceae/chemistry*
;
Up-Regulation/drug effects*
2.CUDC-101 as a dual-target inhibitor of EGFR and HDAC enhances the anti-myeloma effects of bortezomib by regulating G2/M cell cycle arrest.
Wen CAO ; Shunnan YAO ; Anqi LI ; Haoguang CHEN ; Enfan ZHANG ; Liqin CAO ; Jinna ZHANG ; Yifan HOU ; Zhenfeng DAI ; Jing CHEN ; Xi HUANG ; Li YANG ; Zhen CAI
Journal of Zhejiang University. Science. B 2023;24(5):442-454
CUDC-101, an effective and multi-target inhibitor of epidermal growth factor receptor (EGFR), histone deacetylase (HDAC), and human epidermal growth factor receptor 2 (HER2), has been reported to inhibit many kinds of cancers, such as acute promyelocytic leukemia and non-Hodgkin's lymphoma. However, no studies have yet investigated whether CUDC-101 is effective against myeloma. Herein, we proved that CUDC-101 effectively inhibits the proliferation of multiple myeloma (MM) cell lines and induces cell apoptosis in a time- and dose-dependent manner. Moreover, CUDC-101 markedly blocked the signaling pathway of EGFR/phosphoinositide-3-kinase (PI3K) and HDAC, and regulated the cell cycle G2/M arrest. Moreover, we revealed through in vivo experiment that CUDC-101 is a potent anti-myeloma drug. Bortezomib is one of the important drugs in MM treatment, and we investigated whether CUDC-101 has a synergistic or additive effect with bortezomib. The results showed that this drug combination had a synergistic anti-myeloma effect by inducing G2/M phase blockade. Collectively, our findings revealed that CUDC-101 could act on its own or in conjunction with bortezomib, which provides insights into exploring new strategies for MM treatment.
Humans
;
Antineoplastic Agents/therapeutic use*
;
Apoptosis
;
Bortezomib/pharmacology*
;
Cell Line, Tumor
;
Cell Proliferation
;
ErbB Receptors/antagonists & inhibitors*
;
G2 Phase Cell Cycle Checkpoints
;
Histone Deacetylase Inhibitors/pharmacology*
;
Histone Deacetylases/metabolism*
;
M Cells
;
Multiple Myeloma/drug therapy*
3.miR-18a enhances the radiosensitivity of nasopharyngeal carcinoma cells through inducing autophagy.
Li Hong CHANG ; Zhou Zhou YAO ; Hong Wei BAO ; Yue LI ; Xiao Hong CHEN ; Xiao Ping LAI ; Zi Zhen HUANG ; Ge Hua ZHANG
Chinese Journal of Otorhinolaryngology Head and Neck Surgery 2021;56(7):736-745
Objective: To explore the impacts of miR-18a overexpression or depression on the radiosensitivities of nasopharyngeal carcinoma cell line CNE1 and CNE2 and underlying mechanisms. Methods: CNE1 and CNE2 were transfected with miR-18a mimics, inhibitor and the corresponding control vectors. qRT-PCR and western blot were used to determine the ataxia telangiectasia mutated (ATM) expressions in CNE1 and CNE2. CNE1 and CNE2 with stably expressing miR-18a and miR-18a siRNA were constructed. Methyl thiazolyl tetrazolium (MTT) assay was used to detect the impacts of the miR-18a overexpression or depression combined with irradiation on the cell growth. Flow cytometry was used to detect the cell apoptosis and cell cycle. Colony formation assay was used to evaluate the raodiosensitivities of cells. Acridine orange (AO) staining and western blot were used respectively to test the autophagy and the expressions of related proteins. Independent samples t test was used to compare the mean value between groups by using SPSS 16.0. Results: ATM mRNA was decreased significantly in CNE1 and CNE2 cells transfected with 100 or 200 nmol/L miR-18a mimics for 48 hours (CNE1: RQ=0.174±0.139 and 0.003±0.001, t=9.939 and 19 470.783;CNE2: RQ=0.024±0.008 and 0.019±0.012, t=270.230 and 137.746, respectively, all P<0.001). ATM proteins were also decreased after transfected with 100 or 200 nmol/L miR-18a mimics for 72 hours. While in the cells transfected with 100 and 200 nmol/L miR-18a inhibitor for 48 hours, the expressions of ATM mRNA were upregulated significantly (CNE1: RQ=9.419±2.495 and 2.500±1.063, t=-4.427 and -41.241; CNE2: RQ=7.210±0.171 and 115.875±15.805, t=-62.789 and -12.589, all P<0.05), and the expressions of ATM proteins increased after transfected for 72 hours. The growth of cells with miR-18a overexpression plus 4 Gy irradiation were obviously inhibited compared to that of cells with the 4Gy irradiation alone; while the growth of miR-18a-inhibited cells increased compared to that of cells with 4 Gy irradiation alone (all P<0.05). CNE1 transfected with 100 nmol/L miR-18a mimics plus 4 Gy irradiation showed the higher apoptosis rate than the cells with 4 Gy irradiation alone ((22.9±2.1)% vs. (16.3±1.0)%, t=-4.870, P<0.01). Compared to the cells with 4 Gy irradiation alone, miR-18a-overexpressed cells plus 4 Gy irradiation decreased their percentages in G1 phases ((20.2±3.0)% vs. (29.8±4.4)%, t=3.119) and G2/M phases ((21.5±0.9)% vs. (33.4±3.1)%, t=6.410, P<0.05 for both), and increased their percentages in S phases ((56.7±4.9)% vs. (36.8±6.4)%, t=-4.246, P<0.05), and these cells possessed less colony number after exposure to different doses of irradiation, more autophagy-lysosome number, and more expressions of LC3 proteins (all P<0.05). There were no significant differences in the expressions of p62 expressions between different groups of cells. Conclusion: Overexpression of miR-18a can enhance the radiosensitivities of NPC cells by targeting ATM to abrogate G1/S, G2/M arrest and to induce autophagy and apoptosis.
Apoptosis
;
Autophagy
;
Cell Line, Tumor
;
Cell Proliferation
;
G2 Phase Cell Cycle Checkpoints
;
Humans
;
MicroRNAs/genetics*
;
Nasopharyngeal Carcinoma/genetics*
;
Nasopharyngeal Neoplasms/genetics*
;
Radiation Tolerance
4.In Vitro and In Vivo Study on the Effect of Lysosome-associated Protein Transmembrane 4 Beta on the Progression of Breast Cancer
Deyou TAO ; Junqing LIANG ; Yihong PAN ; Yanting ZHOU ; Ying FENG ; Lin ZHANG ; Jingjing XU ; Hui WANG ; Ping HE ; Jie YAO ; Yang ZHAO ; Qinjie NING ; Wen WANG ; Wei JIANG ; Jing ZHENG ; Xia WU
Journal of Breast Cancer 2019;22(3):375-386
PURPOSE: Although the effect of lysosome-associated protein transmembrane 4 beta (LAPTM4B) on the proliferation, migration, and invasion of breast cancer (BC) cells has already been studied, its specific role in BC progression is still elusive. Here, we evaluated the effect of different levels of LAPTM4B expression on the proliferation, invasion, adhesion, and tumor formation abilities of BC cells in vitro, as well as on breast tumor progression in vivo. METHODS: We investigated the influence of LAPTM4B expression on MCF-7 cell proliferation, invasion, adhesion, and tube formation abilities in vitro through its overexpression or knockdown and on breast tumor progression in vivo. RESULTS: Cell growth curves and colony formation assays showed that LAPTM4B promoted the proliferation of breast tumor cells. Cell cycle analysis results revealed that LAPTM4B promoted the entry of cells from the G1 into the S phase. Transwell invasion and cell extracellular matrix adhesion assays showed that LAPTM4B overexpression increased the invasion and adhesion capabilities of MCF-7 cells. More branches were observed in MCF-7 cells overexpressing LAPTM4B under an electron microscope. In comparison with LAPTM4B overexpression, LAPTM4B knockdown decreased the expression of vascular endothelial growth factor-A and significantly inhibited the vasculogenic tube formation ability of tumors. These results were also verified with western blot analysis. CONCLUSION: LAPTM4B promoted the proliferation of MCF-7 cells through the downregulation of p21 (WAF1/CIP1) and caspase-3, and induced cell invasion, adhesion, and angiogenesis through the upregulation of hypoxia-inducible factor 1 alpha, matrix metalloproteinase 2 (MMP2), and MMP9 expression. This specific role deems LAPTM4B as a potential therapeutic target for BC treatment.
Blotting, Western
;
Breast Neoplasms
;
Breast
;
Caspase 3
;
Cell Cycle
;
Disease Progression
;
Down-Regulation
;
Extracellular Matrix
;
Hypoxia-Inducible Factor 1
;
In Vitro Techniques
;
Matrix Metalloproteinase 2
;
MCF-7 Cells
;
S Phase
;
Up-Regulation
;
Vascular Endothelial Growth Factor A
5.Helicobacter pylori inhibited cell proliferation in human periodontal ligament fibroblasts through the Cdc25C/CDK1/cyclinB1 signaling cascade
Huanying LI ; Dongsheng LIANG ; Naiming HU ; Xingzhu DAI ; Jianing HE ; Hongmin ZHUANG ; Wanghong ZHAO
Journal of Periodontal & Implant Science 2019;49(3):138-147
PURPOSE: Several studies have shown that the oral cavity is a secondary location for Helicobacter pylori colonization and that H. pylori is associated with the severity of periodontitis. This study investigated whether H. pylori had an effect on the periodontium. We established an invasion model of a standard strain of H. pylori in human periodontal ligament fibroblasts (hPDLFs), and evaluated the effects of H. pylori on cell proliferation and cell cycle progression. METHODS: Different concentrations of H. pylori were used to infect hPDLFs, with 6 hours of co-culture. The multiplicity of infection in the low- and high-concentration groups was 10:1 and 100:1, respectively. The Cell Counting Kit-8 method and Ki-67 immunofluorescence were used to detect cell proliferation. Flow cytometry, quantitative real-time polymerase chain reaction, and western blots were used to detect cell cycle progression. In the high-concentration group, the invasion of H. pylori was observed by transmission electron microscopy. RESULTS: It was found that H. pylori invaded the fibroblasts, with cytoplasmic localization. Analyses of cell proliferation and flow cytometry showed that H. pylori inhibited the proliferation of periodontal fibroblasts by causing G2 phase arrest. The inhibition of proliferation and G2 phase arrest were more obvious in the high-concentration group. In the low-concentration group, the G2 phase regulatory factors cyclin dependent kinase 1 (CDK1) and cell division cycle 25C (Cdc25C) were upregulated, while cyclin B1 was inhibited. However, in the high-concentration group, cyclin B1 was upregulated and CDK1 was inhibited. Furthermore, the deactivated states of tyrosine phosphorylation of CDK1 (CDK1-Y15) and serine phosphorylation of Cdc25C (Cdc25C-S216) were upregulated after H. pylori infection. CONCLUSIONS: In our model, H. pylori inhibited the proliferation of hPDLFs and exerted an invasive effect, causing G2 phase arrest via the Cdc25C/CDK1/cyclin B1 signaling cascade. Its inhibitory effect on proliferation was stronger in the high-concentration group.
Blotting, Western
;
CDC2 Protein Kinase
;
Cell Count
;
Cell Cycle
;
Cell Proliferation
;
Coculture Techniques
;
Colon
;
Cyclin B1
;
Cytoplasm
;
Fibroblasts
;
Flow Cytometry
;
Fluorescent Antibody Technique
;
G2 Phase
;
Helicobacter pylori
;
Helicobacter
;
Humans
;
Methods
;
Microscopy, Electron, Transmission
;
Mouth
;
Periodontal Ligament
;
Periodontitis
;
Periodontium
;
Phosphorylation
;
Real-Time Polymerase Chain Reaction
;
Serine
;
Tyrosine
6.Suppressor of Variegation 3–9 Homolog 2, a Novel Binding Protein of Translationally Controlled Tumor Protein, Regulates Cancer Cell Proliferation
A Reum KIM ; Jee Young SUNG ; Seung Bae RHO ; Yong Nyun KIM ; Kyungsil YOON
Biomolecules & Therapeutics 2019;27(2):231-239
Suppressor of Variegation 3–9 Homolog 2 (SUV39H2) methylates the lysine 9 residue of histone H3 and induces heterochromatin formation, resulting in transcriptional repression or silencing of target genes. SUV39H1 and SUV39H2 have a role in embryonic development, and SUV39H1 was shown to suppress cell cycle progression associated with Rb. However, the function of human SUV39H2 has not been extensively studied. We observed that forced expression of SUV39H2 decreased cell proliferation by inducing G1 cell cycle arrest. In addition, SUV39H2 was degraded through the ubiquitin-proteasomal pathway. Using yeast two-hybrid screening to address the degradation mechanism and function of SUV39H2, we identified translationally controlled tumor protein (TCTP) as an SUV39H2-interacting molecule. Mapping of the interacting regions indicated that the N-terminal 60 amino acids (aa) of full-length SUV39H2 and the C-terminus of TCTP (120–172 aa) were critical for binding. The interaction of SUV39H2 and TCTP was further confirmed by co-immunoprecipitation and immunofluorescence staining for colocalization. Moreover, depletion of TCTP by RNAi led to up-regulation of SUV39H2 protein, while TCTP overexpression reduced SUV39H2 protein level. The half-life of SUV39H2 protein was significantly extended upon TCTP depletion. These results clearly indicate that TCTP negatively regulates the expression of SUV39H2 post-translationally. Furthermore, SUV39H2 induced apoptotic cell death in TCTP-knockdown cells. Taken together, we identified SUV39H2, as a novel target protein of TCTP and demonstrated that SUV39H2 regulates cell proliferation of lung cancer cells.
Amino Acids
;
Apoptosis
;
Carrier Proteins
;
Cell Cycle
;
Cell Death
;
Cell Proliferation
;
Embryonic Development
;
Female
;
Fluorescent Antibody Technique
;
G1 Phase Cell Cycle Checkpoints
;
Half-Life
;
Heterochromatin
;
Histones
;
Humans
;
Immunoprecipitation
;
Lung Neoplasms
;
Lysine
;
Mass Screening
;
Pregnancy
;
Repression, Psychology
;
RNA Interference
;
Up-Regulation
;
Yeasts
7.MS-5, a Naphthalene Derivative, Induces the Apoptosis of an Ovarian Cancer Cell CAOV-3 by Interfering with the Reactive Oxygen Species Generation.
Eunsook MA ; Seon Ju JEONG ; Joon Seok CHOI ; Thi Ha NGUYEN ; Chul Ho JEONG ; Sang Hoon JOO
Biomolecules & Therapeutics 2019;27(1):48-53
Reactive oxygen species (ROS) are widely generated in biological processes such as normal metabolism and response to xenobiotic exposure. While ROS can be beneficial or harmful to cells and tissues, generation of ROS by diverse anti-cancer drugs or phytochemicals plays an important role in the induction of apoptosis. We recently identified a derivative of naphthalene, MS-5, that induces apoptosis of an ovarian cell, CAOV-3. Interestingly, MS-5 induced apoptosis by down-regulating the ROS. Cell viability was evaluated by water-soluble tetrazolium salt (WST-1) assay. Apoptosis was evaluated by flow cytometry analysis. Intracellular ROS (H₂O₂), mitochondrial superoxide, mitochondrial membrane potential (MMP) and effect on cycle were determined by flow cytometry. Protein expression was assessed by western blotting. The level of ATP was measured using ATP Colorimetric/Fluorometric Assay kit. MS-5 inhibited growth of ovarian cancer cell lines, CAOV-3, in a concentration- and time-dependent manner. MS-5 also induced G1 cell cycle arrest in CAOV-3 cells, while MS-5 decreased intracellular ROS generation. In addition, cells treated with MS-5 showed the decrease in MMP and ATP production. In this study, we found that treatment with MS-5 in CAOV-3 cells induced apoptosis but decreased ROS level. We suspect that MS-5 might interfere with the minimum requirements of ROS for survival. These perturbations appear to be concentration-dependent, suggesting that MS-5 may induce apoptosis by interfering with ROS generation. We propose that MS-5 may be a potent therapeutic agent for inducing apoptosis in ovarian cancer cell through regulation of ROS.
Adenosine Triphosphate
;
Apoptosis*
;
Biological Processes
;
Blotting, Western
;
Cell Line
;
Cell Survival
;
Flow Cytometry
;
G1 Phase Cell Cycle Checkpoints
;
Membrane Potential, Mitochondrial
;
Metabolism
;
Ovarian Neoplasms*
;
Phytochemicals
;
Reactive Oxygen Species*
;
Superoxides
8.Novel insights into cell cycle regulation of cell fate determination.
Journal of Zhejiang University. Science. B 2019;20(6):467-475
The stem/progenitor cell has long been regarded as a central cell type in development, homeostasis, and regeneration, largely owing to its robust self-renewal and multilineage differentiation abilities. The balance between self-renewal and stem/progenitor cell differentiation requires the coordinated regulation of cell cycle progression and cell fate determination. Extensive studies have demonstrated that cell cycle states determine cell fates, because cells in different cell cycle states are characterized by distinct molecular features and functional outputs. Recent advances in high-resolution epigenome profiling, single-cell transcriptomics, and cell cycle reporter systems have provided novel insights into the cell cycle regulation of cell fate determination. Here, we review recent advances in cell cycle-dependent cell fate determination and functional heterogeneity, and the application of cell cycle manipulation for cell fate conversion. These findings will provide insight into our understanding of cell cycle regulation of cell fate determination in this field, and may facilitate its potential application in translational medicine.
Animals
;
Cell Cycle
;
Cell Physiological Phenomena
;
Epigenomics
;
G1 Phase
;
G2 Phase
;
Humans
;
Translational Medical Research
9.C-Type Natriuretic Peptide/Natriuretic Peptide Receptor 2 Is Involved in Cell Proliferation and Testosterone Production in Mouse Leydig Cells
Lei YANG ; Lanjie LEI ; Qihan ZHAO ; Ying GONG ; Gaopeng GUAN ; Shaoxin HUANG
The World Journal of Men's Health 2019;37(2):186-198
PURPOSE: This study investigated the role of natriuretic peptide receptor 2 (NPR2) on cell proliferation and testosterone secretion in mouse Leydig cells. MATERIALS AND METHODS: Mouse testis of different postnatal stages was isolated to detect the expression C-type natriuretic peptide (CNP) and its receptor NPR2 by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Leydig cells isolated from mouse testis were cultured and treated with shNPR2 lentiviruses or CNP. And then the cyclic guanosine monophosphate production, testosterone secretion, cell proliferation, cell cycle and cell apoptosis in mouse Leydig cells were analyzed by ELISA, RT-qPCR, Cell Counting Kit-8, and flow cytometry. Moreover, the expression of NPR2, cell cycle, apoptosis proliferation and cell cycle related gene were detected by RT-qPCR and Western blot. RESULTS: Knockdown of NPR2 by RNAi resulted in S phase cell cycle arrest, cell apoptosis, and decreased testosterone secretion in mouse Leydig cells. CONCLUSIONS: Our study provides more evidences to better understand the function of CNP/NPR2 pathway in male reproduction, which may help us to treat male infertility.
Animals
;
Apoptosis
;
Blotting, Western
;
Cell Count
;
Cell Cycle
;
Cell Cycle Checkpoints
;
Cell Proliferation
;
Enzyme-Linked Immunosorbent Assay
;
Flow Cytometry
;
Germ Cells
;
Guanosine Monophosphate
;
Humans
;
Infertility, Male
;
Lentivirus
;
Leydig Cells
;
Male
;
Mice
;
Natriuretic Peptide, C-Type
;
Polymerase Chain Reaction
;
Receptors, Peptide
;
Reproduction
;
Reverse Transcription
;
RNA Interference
;
S Phase
;
Testicular Diseases
;
Testis
;
Testosterone
10.Jab1 Silencing Inhibits Proliferation and Sensitizes to Cisplatin in Biliary Tract Cancer
Ah Rong NAM ; Ji Won KIM ; Ji Eun PARK ; Ju Hee BANG ; Mei Hua JIN ; Do Youn OH ; Yung Jue BANG
Cancer Research and Treatment 2019;51(3):886-900
PURPOSE: Jab1 is a coactivator of c-Jun that enhances the transcriptional function of c-Jun. Jab1 is frequently overexpressed in various cancers and is associatedwith poor prognosis of cancer patients. Thus, Jab1 could be a potential therapeutic target in cancer. However, the role of Jab1 in biliary tract cancer (BTC) has not been studied. MATERIALS AND METHODS: We performed in vitro and in vivo experiments to evaluate the therapeutic potential ofJab1 inhibition in BTC. RESULTS: Among 8 BTC cell lines, many showed higher Jab1 expression levels. In addition, Jab1 silencing by siRNA increased p27 expression levels. SNU478 and HuCCT-1 cells exhibited profound Jab1 knockdown and increased p27 expression by Jab1-specific siRNA transfection. Jab1 silencing induced anti-proliferative and anti-migratory effects and resulted in G1 cell cycle arrest in SNU478 and HuCCT-1 cells. In addition, Jab1 silencing potentiated the anti-proliferative and anti-migratory effects of cisplatin by increasing DNA damage. Interestingly,Jab1 knockdown increased PTEN protein half-life, resulting in increased PTEN expression. In the HuCCT-1 mouse xenograft model, stable knockdown of Jab1 by shRNA also showed anti-proliferative effects in vivo, with decreased Ki-67 expression and AKT phosphorylation and increased Terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling and p27 expression. CONCLUSION: Jab1 knockdown demonstrated anti-proliferative and anti-migratory effects in BTC cells by increasing DNA damage and stabilizing PTEN, resulting in G1 cell cycle arrest. In addition, Jab1 silencing potentiated the anti-proliferative effects of cisplatin. Our data suggest that Jab1 may be a potential therapeutic target in BTC that is worthy of further investigations.
Animals
;
Biliary Tract Neoplasms
;
Biliary Tract
;
Cell Line
;
Cisplatin
;
DNA Damage
;
G1 Phase Cell Cycle Checkpoints
;
Half-Life
;
Heterografts
;
Humans
;
In Vitro Techniques
;
Mice
;
Phosphorylation
;
Prognosis
;
PTEN Phosphohydrolase
;
RNA, Small Interfering
;
Transfection

Result Analysis
Print
Save
E-mail