1.Lamin B1 regulates the growth of hepatocellular carcinoma cells by influencing telomerase activity.
Ruiguan WANG ; Si CHEN ; Zhijia SUN ; Shikun WANG ; Jie WANG ; Lingmei QIN ; Jiangbo LI
Chinese Journal of Biotechnology 2023;39(4):1609-1620
Lamin B1 (LMNB1) is highly expressed in liver cancer tissues, and its influence and mechanism on the proliferation of hepatocellular carcinoma cells were explored by knocking down the expression of the protein. In liver cancer cells, siRNAs were used to knock down LMNB1. Knockdown effects were detected by Western blotting. Changes in telomerase activity were detected by telomeric repeat amplification protocol assay (TRAP) experiments. Telomere length changes were detected by quantitative real-time polymerase chain reaction (qPCR). CCK8, cloning formation, transwell and wound healing were performed to detect changes in its growth, invasion and migration capabilities. The lentiviral system was used to construct HepG2 cells that steadily knocked down LMNB1. Then the changes of telomere length and telomerase activity were detected, and the cell aging status was detected by SA-β-gal senescence staining. The effects of tumorigenesis were detected by nude mouse subcutaneous tumorigenesis experiments, subsequent histification staining of tumors, SA-β-gal senescence staining, fluorescence in situ hybridization (FISH) for telomere analysis and other experiments. Finally, the method of biogenesis analysis was used to find the expression of LMNB1 in clinical liver cancer tissues, and its relationship with clinical stages and patient survival. Knockdown of LMNB1 in HepG2 and Hep3B cells significantly reduced telomerase activity, cell proliferation, migration and invasion abilities. Experiments in cells and tumor formation in nude mice had demonstrated that stable knockdown of LMNB1 reduced telomerase activity, shortened telomere length, senesced cells, reduced cell tumorigenicity and KI-67 expression. Bioinformatics analysis showed that LMNB1 was highly expressed in liver cancer tissues and correlated with tumor stage and patient survival. In conclusion, LMNB1 is overexpressed in liver cancer cells, and it is expected to become an indicator for evaluating the clinical prognosis of liver cancer patients and a target for precise treatment.
Animals
;
Mice
;
Telomerase/metabolism*
;
Carcinoma, Hepatocellular/genetics*
;
Liver Neoplasms/genetics*
;
Telomere Shortening
;
In Situ Hybridization, Fluorescence
;
Mice, Nude
;
Telomere/pathology*
;
Carcinogenesis
2.Research Progress of Long Non-Coding RNA in Hematological Tumors --Review.
Feng LI ; Fei-Fei YANG ; Yan-Li XU
Journal of Experimental Hematology 2023;31(1):306-310
Long non-coding RNA (lncRNA) is a hot topic in the field of researching tumor pathogenesis, and the importance in hematologic malignancies has been gradually being elucidated. LncRNA not only regulates hematological tumorigenesis and progression through affecting various biological processes such as cell proliferation, differentiation, pluripotency and apoptosis; moreover, abnormal expression and mutation of lncRNA are closely related to drug resistance and prognosis. Thus lncRNA can be used as novel biomarker and potential therapeutic target for hematological tumors. In this review, we will focus on the latest progress of lncRNA in hematological tumors to provide new ideas for the clinical diagnosis, prognostic evaluation together with research and development of target drugs for hematologic malignancies.
Humans
;
RNA, Long Noncoding/metabolism*
;
Hematologic Neoplasms/genetics*
;
Neoplasms
;
Carcinogenesis/pathology*
;
Cell Transformation, Neoplastic/genetics*
;
Gene Expression Regulation, Neoplastic
3.Effect of long non-coding RNA 114227 on gastric cancer cell proliferation and migration.
Haining GAN ; Huiying XIANG ; Yue XI ; Min YAO ; Chen SHAO ; Shihe SHAO
Journal of Central South University(Medical Sciences) 2023;48(2):157-164
OBJECTIVES:
Gastric cancer is a common cancer of the digestive system. Long non-coding RNA (lncRNA) plays an important role in the formation and development of gastric cancer. This study aims to investigate the effect of long non-coding lncRNA 114227 on biologic behaviors in gastric cancer cells.
METHODS:
The experiment was divided into 4 groups: a negative control (NC) group, a lncRNA 114227 small interference (si-lncRNA 114227) group, an empty vector (Vector) group, and an overexpression vector (OE-lncRNA 114227) group. The expressions of lncRNA 114227 in gastric mucosa and gastric cancer tissues, gastric mucosal epithelial cells and different gastric cancer strains were determined by real-time reverse transcription PCR (real-time RT-PCR).The proliferation were detected by CCK-8 assay in gastric cancer cells. The epithelial-mesenchymal transformation (EMT) was utilized by Transwell assay, scratch healing assay, and Western blotting in gastric cancer cells. The effect of lncRNA 114227 on proliferation of gastric cancer cells was detected by tumor bearing experiment in nude mice in vivo.
RESULTS:
The expression level of lncRNA 114227 in the gastric cancer tissues was significantly lower than that in the gastric mucosa tissues, and in 4 kinds of gastric cancer strains was all significantly lower than that in gastric mucosal epithelial cells (all P<0.01). In vitro, the proliferation and migration abilities of gastric cells were significantly reduced after overexpressing lncRNA 114227, and cell proliferation and migration were enhanced after silencing lncRNA 114227 (all P<0.05). The results of in vivo subcutaneous tumorigenesis in nude mice showed that the tumorigenic volume of the tumor-bearing mice in the OE-lncRNA 114227 group was significantly smaller than that of the Vector group, and the tumorigenic quality was lower than that of the Vector group (P<0.05), indicating that lncRNA 114227 inhibited tumorigenesis.
CONCLUSIONS
The expression of lncRNA 114227 is downregulated in gastric cancer gastric cancer tissues and cell lines. LncRNA 114227 may inhibit the proliferation and migration of gastric cancer cells through EMT process.
Animals
;
Mice
;
RNA, Long Noncoding/metabolism*
;
Stomach Neoplasms/pathology*
;
Mice, Nude
;
Cell Line, Tumor
;
Cell Proliferation/genetics*
;
Carcinogenesis/genetics*
;
Cell Movement/genetics*
;
Epithelial-Mesenchymal Transition/genetics*
;
Gene Expression Regulation, Neoplastic
;
Apoptosis/genetics*
4.Organoids: approaches and utility in cancer research.
Bingrui ZHOU ; Zhiwei FENG ; Jun XU ; Jun XIE
Chinese Medical Journal 2023;136(15):1783-1793
Organoids are three-dimensional cellular structures with self-organizing and self-differentiation capacities. They faithfully recapitulate structures and functions of in vivo organs as represented by functionality and microstructural definitions. Heterogeneity in in vitro disease modeling is one of the main reasons for anti-cancer therapy failures. Establishing a powerful model to represent tumor heterogeneity is crucial for elucidating tumor biology and developing effective therapeutic strategies. Tumor organoids can retain the original tumor heterogeneity and are commonly used to mimic the cancer microenvironment when co-cultured with fibroblasts and immune cells; therefore, considerable effort has been made recently to promote the use of this new technology from basic research to clinical studies in tumors. In combination with gene editing technology and microfluidic chip systems, engineered tumor organoids show promising abilities to recapitulate tumorigenesis and metastasis. In many studies, the responses of tumor organoids to various drugs have shown a positive correlation with patient responses. Owing to these consistent responses and personalized characteristics with patient data, tumor organoids show excellent potential for preclinical research. Here, we summarize the properties of different tumor models and review their current state and progress in tumor organoids. We further discuss the substantial challenges and prospects in the rapidly developing tumor organoid field.
Humans
;
Neoplasms/genetics*
;
Organoids/pathology*
;
Carcinogenesis
;
Models, Biological
;
Precision Medicine/methods*
;
Tumor Microenvironment
5.MiR-203a-5p Inhibits Multiple Myeloma Cell Proliferation and Cell Cycle Progression via Targeting JAG1.
Yue ZHANG ; Ting-Ting CHEN ; He-Bing ZHOU ; Wen-Ming CHEN
Journal of Experimental Hematology 2023;31(3):801-809
OBJECTIVE:
To investigate the biological function of miR-203a-5p and the underlying mechanism in multiple myeloma (MM).
METHODS:
Three miRNA expression profiles (GSE16558, GSE24371 and GSE17498) were downloaded from the GEO database. The three miRNA expression profiles contained 131 MM samples and 17 normal plasmacyte samples. The robust rank aggregation (RRA) method was used to identify the differentially expressed miRNAs between MM and normal plasmacytes. In order to carry out cytological experiments, MM cell line with stable over-expression of miR-203a-5p was constructed with lentivirus. Expression levels of miR-203a-5p in MM cells were quantified by qRT-PCR. The effects of miR-203a-5p on MM cells were investigated using assays of cell viability and cell cycle. Cell proliferation was measured using the Cell Counting kit (CCK)8 assay. The percentage of cells in each cell cycle was measured with a FACSCalibur system. Xenograft tumor models were established to evaluate the role of miR-203a-5p in tumorigenesis in vivo . To elucidate the underlying molecular mechanisms of miR-203a-5p in mediating cell proliferation inhibition and cell cycle arrest in MM, we used TargetScan and miRanda to predict the candidate targets of miR-203a-5p. The potential target of miR-203a-5p in MM cells was explored using the luciferase reporter assay, qRT-PCR, and Western blot.
RESULTS:
An integrated analysis of three MM miRNA expression datasets showed that the levels of miR-203a-5p in MM were notably downregulated compared with those in normal plasmacytes. Accordingly, the relative expression levels of miR-203a-5p were decreased in MM cell lines. In addition, overexpression of miR-203a-5p inhibited the proliferation and cell cycle progression of RPMI8226 and U266 cells. In vivo experiments demonstrated that upregulation of miR-203a-5p expression could significantly inhibit the tumorigenesis of subcutaneous myeloma xenografts in nude mice. Mechanistic investigation led to the identification of Jagged 1 (JAG1) as a novel and direct downstream target of miR-203a-5p. Interestingly, the reintroduction of JAG1 abrogated miR-203a-5p-induced MM cell growth inhibition and cell cycle arrest.
CONCLUSION
Our data demonstrate that miR-203a-5p inhibits cell proliferation and cell cycle progression in MM cells by targeting JAG1, supporting the utility of miR-203a-5p as a novel and potential therapeutic agent for miRNA-based MM therapy.
Animals
;
Mice
;
Humans
;
Multiple Myeloma/pathology*
;
Cell Line, Tumor
;
Mice, Nude
;
MicroRNAs/metabolism*
;
Cell Division
;
Cell Proliferation
;
Disease Models, Animal
;
Carcinogenesis/genetics*
;
Gene Expression Regulation, Neoplastic
;
Jagged-1 Protein/metabolism*
6.Identification of BRAF V600E mutation in odontogenic tumors by high-performance MALDI-TOF analysis.
Lucrezia TOGNI ; Antonio ZIZZI ; Roberta MAZZUCCHELLI ; Andrea SANTARELLI ; Corrado RUBINI ; Marco MASCITTI
International Journal of Oral Science 2022;14(1):22-22
Odontogenic tumors are rare lesions with unknown etiopathogenesis. Most of them are benign, but local aggressiveness, infiltrative potential, and high recurrence rate characterize some entities. The MAP-kinase pathway activation can represent a primary critical event in odontogenic tumorigenesis. Especially, the BRAF V600E mutation has been involved in 80-90% of ameloblastic lesions, offering a biological rationale for developing new targeted therapies. The study aims to evaluate the BRAF V600E mutation in odontogenic lesions, comparing three different detection methods and focusing on the Sequenom MassARRAY System. 81 surgical samples of odontogenic lesions were subjected to immunohistochemical analysis, Sanger Sequencing, and Matrix-Assisted Laser Desorption/Ionization-Time of Flight mass spectrometry (Sequenom). The BRAF V600E mutation was revealed only in ameloblastoma samples. Moreover, the presence of BRAF V600E was significantly associated with the mandibular site (ρ = 0.627; P value <0.001) and the unicystic histotype (ρ = 0.299, P value <0.001). However, any significant difference of 10-years disease-free survival time was not revealed. Finally, Sequenom showed to be a 100% sensitive and 98.1% specific, suggesting its high-performance diagnostic accuracy. These results suggest the MAP-kinase pathway could contribute to ameloblastic tumorigenesis. Moreover, they could indicate the anatomical specificity of the driving mutations of mandibular ameloblastomas, providing a biological rational for developing new targeted therapies. Finally, the high diagnostic accuracy of Sequenom was confirmed.
Ameloblastoma/pathology*
;
Carcinogenesis
;
Humans
;
Mitogen-Activated Protein Kinases/genetics*
;
Mutation
;
Odontogenic Tumors/pathology*
;
Proto-Oncogene Proteins B-raf/metabolism*
;
Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.Effects of MnSOD silence on in vitro tumorigenicity in NCI-H446 cells.
Qing YUAN ; Min WEN ; Xiang LI ; Ling SHU ; Jianguo CAO ; Jiansong ZHANG
Journal of Central South University(Medical Sciences) 2018;43(6):583-588
To investigate the effect of manganese superoxide dismutase (MnSOD) silence on the in vitro tumorigenicity in human small cell lung cancer NCI-H446 cells and the underlying mechanisms.
Methods: Sphere formation cells from NCI-H446 cells were obtained by suspension culture, while the expression of MnSOD and urokinase type plasminogen activator (uPAR) was analyzed by Western blot. Silence of MnSOD was performed by adenovirus infection in the second passage formation cells, and the effect of MnSOD silence on tumorigenicity in NCI-H446 cells was evaluated by sphere formation assay and soft-agar colony formation assay, while the expression of uPAR was analyzed by Western blot.
Results: Compared with NCI-H446 cells, the sphere formation rate, colony formation rate, and the expression of MnSOD and uPAR were significantly increased in the second passage sphere formation cells in NCI-H446 cells (P<0.05). Silence of MnSOD inhibited the sphere formation rate, colony formation rate, and the expression level of uPAR in the second passage sphere formation cells in NCI-H446 cells.
Conclusion: MnSOD may promote tumorigenicity in NCI-H446 cells by up-regulation of uPAR expression in vitro.
Adenoviridae
;
Carcinogenesis
;
Cell Line, Tumor
;
Humans
;
In Vitro Techniques
;
Lung Neoplasms
;
etiology
;
metabolism
;
RNA Interference
;
Receptors, Urokinase Plasminogen Activator
;
genetics
;
metabolism
;
Small Cell Lung Carcinoma
;
etiology
;
metabolism
;
Spheroids, Cellular
;
pathology
;
Superoxide Dismutase
;
genetics
;
metabolism
;
Tumor Stem Cell Assay
;
Up-Regulation
8.Progress of Long Non-coding RNA in Non-small Cell Lung Cancer.
Yachen ZHANG ; Di LIANG ; Jing JIN ; Congmin LIU ; Yutong HE
Chinese Journal of Lung Cancer 2018;21(1):43-49
Lung cancer is one of the most important malignant tumors in the world. The morbidity and mortality rank the first in all kinds of cancer. Long non-coding RNA (lncRNA) is at least 200 nt long and has no protein coding capacity. It plays an important role in the epigenetic regulation, cell cycle regulation, the regulation of cell differentiation, and many other life activities. The studies indicate that dysregulation of lncRNAs in non-small cell lung cancer (NSCLC) tissue and blood circulation is associated with the occurrence and development of cancer. The lncRNAs play an significant role in proliferation, differentiation, migration and apoptosis of the tumor cells. Explore the potential mechanism between lncRNAs and NSCLC is beneficial for the early diagnosis, target therpy and improve prognosis. Therefore, the study aims to demonstrate the latest studies on the lncRNAs related to occurence, diagnosis, therpy and prognosis of NSCLC. It can help to deeply understanding of lncRNA, and provide new ideas for the prevention of NSCLC.
Carcinogenesis
;
genetics
;
Carcinoma, Non-Small-Cell Lung
;
diagnosis
;
drug therapy
;
genetics
;
pathology
;
Humans
;
Molecular Targeted Therapy
;
RNA, Long Noncoding
;
genetics
9.Multiple Functions of Ten-eleven Translocation 1 during Tumorigenesis.
Yi-Ping TIAN ; Yi-Min ZHU ; Xiao-Hui SUN ; Mao-De LAI ;
Chinese Medical Journal 2016;129(14):1744-1751
OBJECTIVEAberrant expression of ten-eleven translocation 1 (TET1) plays a critical role in tumor development and progression. We systematically summarized the latest research progress on the role and mechanisms of TET1 in cancer biology.
DATA SOURCESRelevant articles published in English from 1980 to April 2016 were selected from the PubMed database. The terms "ten-eleven translocation 1," "5mC," "5hmC," "microRNA," "hypoxia," and "embryonic stem cell" were used for the search.
STUDY SELECTIONArticles focusing on the role and mechanism of TET1 in tumor were reviewed, including clinical and basic research articles.
RESULTSTET proteins, the key enzymes converting 5-methylcytosine to 5-hydroxymethylcytosine, play vital roles in DNA demethylation regulation. Recent studies have shown that loss of TET1 is associated with tumorigenesis and can be used as a potential biomarker for cancer therapy, which indicates that TET1 serves as tumor suppressor gene. Moreover, besides its dioxygenase activity, TET1 could induce epithelial-mesenchymal transition and act as a coactivator to regulate gene transcription, such as developmental regulator in embryonic stem cells (ESCs) and hypoxia-responsive gene in cancer. The regulation of TET1 is also correlated with microRNA in a posttranscriptional modification process. Hence, it is complex but critical to comprehend the mechanisms of TET1 in the biology of ESCs and cancer.
CONCLUSIONSTET1 not only serves as a demethylation enzyme but also plays multiple roles during tumorigenesis and progression. More studies should be carried out to elucidate the exact mechanisms of TET1 and its associations with cancer before considering it as a therapeutic tool.
Animals ; Biomarkers ; metabolism ; Carcinogenesis ; genetics ; metabolism ; pathology ; Humans ; MicroRNAs ; genetics ; Mixed Function Oxygenases ; genetics ; metabolism ; Proto-Oncogene Proteins ; genetics ; metabolism
10.Anti-rheumatic drug iguratimod (T-614) alleviates cancer-induced bone destruction via down-regulating interleukin-6 production in a nuclear factor-κB-dependent manner.
Yue SUN ; Da-Wei YE ; Peng ZHANG ; Ying-Xing WU ; Bang-Yan WANG ; Guang PENG ; Shi-Ying YU
Journal of Huazhong University of Science and Technology (Medical Sciences) 2016;36(5):691-699
Cytokines are believed to be involved in a "vicious circle" of progressive interactions in bone metastasis. Iguratimod is a novel anti-rheumatic drug which is reported to have the capability of anti-cytokines. In this study, a rat model was constructed to investigate the effect of iguratimod on bone metastasis and it was found that iguratimod alleviated cancer-induced bone destruction. To further explore whether an anti-tumor activity of iguratimod contributes to the effect of bone resorption suppression, two human breast cancer cell lines MDA-MB-231 and MCF-7 were studied. The effect of iguratimod on tumor proliferation was detected by CCK-8 assay and flow cytometry. The effects of iguratimod on migration and invasion of cancer cells were determined by wound-healing and Transwell assays. Results showed that high dose (30 μg/mL) iguratimod slightly suppressed the proliferation of cancer cells but failed to inhibit their migration and invasion capacity. Interestingly, iguratimod decreased the transcription level of IL-6 in MDA-MB-231 cells in a concentration-dependent manner. Moreover, iguratimod partially impaired NF-κB signaling by suppressing the phosphorylation of NF-κB p65 subunit. Our findings indicated that iguratimod may alleviate bone destruction by partially decreasing the expression of IL-6 in an NF-κB-dependent manner, while it has little effect on the tumor proliferation and invasion.
Animals
;
Apoptosis
;
drug effects
;
Bone Neoplasms
;
complications
;
drug therapy
;
pathology
;
secondary
;
Bone Resorption
;
complications
;
drug therapy
;
pathology
;
Breast Neoplasms
;
complications
;
drug therapy
;
genetics
;
pathology
;
Carcinogenesis
;
drug effects
;
Cell Movement
;
drug effects
;
Cell Proliferation
;
drug effects
;
Chromones
;
administration & dosage
;
Female
;
Humans
;
Interleukin-6
;
biosynthesis
;
genetics
;
MCF-7 Cells
;
Neoplasm Invasiveness
;
genetics
;
pathology
;
Rats
;
Sulfonamides
;
administration & dosage
;
Transcription Factor RelA
;
biosynthesis
;
genetics

Result Analysis
Print
Save
E-mail