1.Role of miRNA in prostate cancer and research progress of traditional Chinese medicine intervention.
Sheng-Long LI ; Yong-Lin LIANG ; Xiu-Juan YANG ; Yong-Qiang ZHAO ; Hui LI ; Gang-Gang LU ; Xu MA ; Da-Cheng TIAN
China Journal of Chinese Materia Medica 2025;50(10):2619-2630
Prostate cancer(PCa) is a common malignant tumor among elderly men, with high incidence and mortality rates worldwide, posing a serious threat to human health. Traditional treatments face limitations, highlighting the urgent need for novel therapeutic strategies. Recent studies on the regulatory mechanisms of micro ribonucleic acid(microRNA, miRNA) in tumor development has identified miRNA as new targets for PCa diagnosis and treatment. Traditional Chinese medicine(TCM), with its multi-mechanism, multi-target, and multi-pathway regulatory properties, shows promising potential in miRNA-based PCa therapy. This review summarized recent findings on miRNA' roles in PCa and research progress of TCM intervention and found that a variety of miRNA played important regulatory roles in cell differentiation, proliferation, apoptosis, invasion, metastasis, immune microenvironment, and drug resistance, and their potential as biomarkers for PCa diagnosis, prognosis, and therapy, indicating the potential to be a biomarker for the diagnosis, prognosis evaluation, and treatment of PCa. The review concluded that the active components of TCM(terpenoids, flavonoids, alkaloids, and others) and compounds(Yishen Tonglong Decoction, Shenhu Decoction, Zhoushi Qiling Decoction, Fuzheng Yiliu Decoction, and Qilan Formula) could regulate the expression of their downstream target genes by acting on specific miRNA and affect the above biological behaviors of PCa cells, thus playing a role in the treatment of PCa. This review aims to provide a theoretical basis for miRNA as potential biomarkers and therapeutic targets for PCa and suggest new avenues for further development of targeted therapy strategies against miRNA.
Humans
;
MicroRNAs/metabolism*
;
Prostatic Neoplasms/metabolism*
;
Male
;
Drugs, Chinese Herbal/therapeutic use*
;
Medicine, Chinese Traditional
;
Animals
;
Gene Expression Regulation, Neoplastic/drug effects*
2.CCDC97 influences the immune microenvironment and biological functions in HCC.
Lingling MO ; Xinyue WU ; Xiaohua PENG ; Chuang CHEN
Chinese Journal of Cellular and Molecular Immunology 2025;41(1):23-30
Objective To explore the clinical and immunological significance of CCDC97 in hepatocellular carcinoma (HCC). Methods Clinical data and RNA sequencing results from HCC patients were retrieved from TCGA and ICGC databases. Bioinformatics analysis and in vitro experiments were performed to investigate the role of CCDC97 in HCC. Results The expression level of CCDC97 was elevated in HCC patients and HCC cells, closely associated with pathological features and prognosis. CCDC97 was identified as a novel prognostic biomarker. It is linked to the spliceosome pathway, which is significantly active in tumors and potentially promotes carcinogenesis. CCDC97 is also highly expressed in various immune cells and is associated with microenvironment. Furthermore, knocking down CCDC97 in vitro suppressed cell migration, invasion, and proliferation. Conclusion CCDC97 plays a critical role in HCC progression and the immune microenvironment, making it a potential target for prognosis and therapeutic intervention.
Humans
;
Carcinoma, Hepatocellular/metabolism*
;
Liver Neoplasms/metabolism*
;
Tumor Microenvironment/genetics*
;
Cell Movement/genetics*
;
Cell Proliferation
;
Prognosis
;
Cell Line, Tumor
;
Gene Expression Regulation, Neoplastic
;
Biomarkers, Tumor/genetics*
;
Male
3.A novel glycolysis-related prognostic risk model for colorectal cancer patients based on single-cell and bulk transcriptomic data.
Kai YAO ; Jingyi XIA ; Shuo ZHANG ; Yun SUN ; Junjie MA ; Bo ZHU ; Li REN ; Congli ZHANG
Chinese Journal of Cellular and Molecular Immunology 2025;41(2):105-115
Objective To explore the prognostic value of glycolysis-related genes in colorectal cancer (CRC) patients and formulate a novel glycolysis-related prognostic risk model. Methods Single-cell and bulk transcriptomic data of CRC patients, along with clinical information, were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Glycolysis scores for each sample were calculated using single-sample Gene Set Enrichment Analysis (ssGSEA). Kaplan-Meier survival curves were generated to analyze the relationship between glycolysis scores and overall survival. Novel glycolysis-related subgroups were defined among the cell type with the highest glycolysis scores. Gene enrichment analysis, metabolic activity assessment, and univariate Cox regression were performed to explore the biological functions and prognostic impact of these subgroups. A prognostic risk model was built and validated based on genes significantly affecting the prognosis. Gene Set Enrichment Analysis (GSEA) was conducted to explore differences in biological processes between high- and low-risk groups. Differences in immune microenvironment and drug sensitivity between these groups were assessed using R packages. Potential targeted agents for prognostic risk genes were predicted using the Enrichr database. Results Tumor tissues showed significantly higher glycolysis scores than normal tissues, which was associated with a poor prognosis in CRC patients. The highest glycolysis score was observed in epithelial cells, within which we defined eight novel glycolysis-related cell subpopulations. Specifically, the P4HA1+ epithelial cell subpopulation was associated with a poor prognosis. Based on signature genes of this subpopulation, a six-gene prognostic risk model was formulated. GSEA revealed significant biological differences between high- and low-risk groups. Immune microenvironment analysis demonstrated that the high-risk group had increased infiltration of macrophages and tumor-associated fibroblasts, along with evident immune exclusion and suppression, while the low-risk group exhibited higher levels of B cell and T cell infiltration. Drug sensitivity analysis indicated that high-risk patients were more sensitive to Abiraterone, while low-risk patients responded to Cisplatin. Additionally, Valproic acid was predicted as a potential targeted agent. Conclusion High glycolytic activity is associated with a poor prognosis in CRC patients. The novel glycolysis-related prognostic risk model formulated in this study offers significant potential for enhancing the diagnosis and treatment of CRC.
Humans
;
Colorectal Neoplasms/pathology*
;
Glycolysis/genetics*
;
Prognosis
;
Transcriptome
;
Tumor Microenvironment/genetics*
;
Gene Expression Profiling
;
Single-Cell Analysis
;
Gene Expression Regulation, Neoplastic
;
Male
;
Female
;
Kaplan-Meier Estimate
4.Potential molecular mechanism of lncRNAs HOTAIR in malignant metastasis of esophageal cancer.
Kaijin LU ; Jiangfeng SHEN ; Guang HAN ; Quan CHEN
Chinese Journal of Cellular and Molecular Immunology 2025;41(3):236-244
Objective To elucidate the molecular mechanism by which exosomes (Exo) derived from cancer-associated fibroblasts (CAF) carrying HOX transcript antisense intergenic RNA (lncRNA HOTAIR) promote the metastasis of esophageal squamous cell carcinoma (ESCC). Methods CAFs were collected from tumor tissues, and non-cancer associated fibroblasts (NFs) were obtained from adjacent normal tissues at least 5 cm away from the tumor. Exosomes (CAFs-Exo and NFs-Exo) were isolated from conditioned media collected from CAFs or NFs. CAFs-Exo and NFs-Exo were incubated with human ESCC cell line TE-1 for 24 hours, and CCK-8 was used to determine the cell proliferation ability. Scratch test and Transwell test were performed to determine the cell migration and invasion ability. TE-1 cells were divided into the following two groups: NC group and KD group. The NC group and KD group were transfected with control siRNAs or siRNAs targeting HOTAIR respectively. The effects of HOTAIR knock-down on cell proliferation, migration, invasion and glycolysis were determined. Results CAFs-Exo promoted the proliferation of TE-1 cells more significantly than NFs-Exo. Compared with NFs-Exo group, the migration and invasion ability of TE-1 cells treated with CAFs-Exo were improved significantly. In addition, CAFs-Exo treatment inhibited the expression of E-cadherin and enhanced the expression of N-cadherin. The expression of HOTAIR in CAFs was significantly higher than that in NFs. Compared with NFs-Exo, the expression level of HOTAIR in CAFs-Exo increased significantly. Compared with NC group, the proliferation, migration and invasion of TE-1 cells in KD group decreased significantly. Compared with NC group, hexokinase 2 (HK2), extracellular acidification rate (ECAR) and ATP/ADP ratio of TE-1 cells in KD group decreased significantly. Conclusion HOTAIR, an exosome derived from CAFs, may be involved in metastasis and EMT by regulating glycolysis in ESCC cells.
Humans
;
RNA, Long Noncoding/metabolism*
;
Esophageal Neoplasms/metabolism*
;
Cell Movement/genetics*
;
Cell Proliferation/genetics*
;
Cell Line, Tumor
;
Esophageal Squamous Cell Carcinoma
;
Exosomes/genetics*
;
Neoplasm Metastasis
;
Neoplasm Invasiveness
;
Gene Expression Regulation, Neoplastic
;
Glycolysis/genetics*
;
Cancer-Associated Fibroblasts/metabolism*
;
Carcinoma, Squamous Cell/metabolism*
;
Cadherins/genetics*
5.Correlation analysis of low expression of LY86-AS1 and KHDRBS2 with immune cell invasion and prognosis in glioblastoma.
Shasha WANG ; Wenhao ZHAO ; Xining HE ; Yangyang ZHANG ; Wenli CHANG
Chinese Journal of Cellular and Molecular Immunology 2025;41(3):245-253
Objective To investigate the expression and correlation of LY86-AS1 and KHDRBS2 in glioblastoma (GBM), and their impacts on the prognosis of patients and immune cell infiltration. Methods Based on the GSE50161 dataset from the Gene Expression Omnibus (GEO) database, LY86-AS1 and KHDRBS2, which are closely related to the development of GBM, were identified by WGCNA and differential expression analysis. The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) databases were used to analyze the relationship between the expression of LY86-AS1 and KHDRBS2 and the prognosis of GBM patients. Multiple datasets were employed to analyze the correlation between the expression levels of LY86-AS1 and KHDRBS2 and its relationship with immune cell infiltration. Real-time quantitative PCR was used to verify the expression of LY86-AS1 and KHDRBS2 in GBM and normal brain tissues. The Human Protein Atlas (HPA) database was accessed to obtain the protein expression of KHDRBS2, and immunohistochemical staining was conducted to verify the protein expression of KHDRBS2. Results LY86-AS1 and KHDRBS2 were lowly expressed in GBM tissues and were closely related to the development of GBM, showing a significant positive correlation. Patients with low expression levels of LY86-AS1 and KHDRBS2 had a lower overall survival rate than those with high expression levels. LY86-AS1 was positively correlated with naive B cells, plasma cells, activated NK cells, M1 macrophages, activated mast cells and monocytes. KHDRBS2 was positively correlated with naive B cells, plasma cells, helper T cells, activated NK cells and monocytes. Conclusion The low expression levels of LY86-AS1 and KHDRBS2 in GBM, which is associated with poor prognosis, affect the tumor immune microenvironment and may serve as potential new biomarkers for the diagnosis of GBM and the prognosis assessment of patients.
Humans
;
Glioblastoma/metabolism*
;
Prognosis
;
Brain Neoplasms/pathology*
;
Gene Expression Regulation, Neoplastic
;
RNA-Binding Proteins/metabolism*
6.Sialyltransferase ST3GAL1 promotes malignant progression in glioma.
Zihao ZHAO ; Wenjing ZHENG ; Lingling ZHANG ; Wenjie SONG ; Tao WANG
Chinese Journal of Cellular and Molecular Immunology 2025;41(4):308-317
Objective To investigate the clinical relevance and diagnostic or prognostic value of ST3β-galactoside α-2, 3-sialyltransferase 1 (ST3GAL1) in glioma and to confirm its role in promoting malignant phenotypes. Methods Using data from The Cancer Genome Atlas (TCGA) database, we analyzed the correlation between ST3GAL1 expression levels in glioma and clinical parameters to evaluate its diagnostic and prognostic value. The impact of ST3GAL1 on malignant phenotypes of glioma cells-including proliferation, cell cycle progression, apoptosis, and invasion was further validated through ST3GAL1 knockdown experiments. Results The expression level of ST3GAL1 was significantly higher in glioma tissues compared to healthy brain tissues and showed a strong correlation with clinical characteristics of glioma patients. Survival analysis and receiver operating characteristic (ROC) curve demonstrated that ST3GAL1 could serve as a potential diagnostic and prognostic biomarker for glioma. Knockdown of ST3GAL1 suppressed proliferation, invasion, and migration capabilities of glioma cell lines, and induced G1-phase cell cycle arrest. Conclusion ST3GAL1 promotes malignant phenotypes in glioma and plays a critical role in its malignant progression, suggesting its potential as a biomarker for glioma diagnosis and prognosis.
Humans
;
Sialyltransferases/metabolism*
;
Glioma/diagnosis*
;
Cell Proliferation/genetics*
;
Cell Line, Tumor
;
Brain Neoplasms/enzymology*
;
beta-Galactoside alpha-2,3-Sialyltransferase
;
Disease Progression
;
Prognosis
;
Cell Movement/genetics*
;
Apoptosis/genetics*
;
Male
;
Female
;
Gene Expression Regulation, Neoplastic
;
Biomarkers, Tumor/metabolism*
;
Middle Aged
7.Molecular mechanisms of TPT1-AS1 in regulating epithelial ovarian cancer cell invasion, migration, and angiogenesis by targeting the miR-324/TWIST1 axis.
Chinese Journal of Cellular and Molecular Immunology 2025;41(6):536-543
Objective To explore the mechanism of TPT1-AS1 targeting miR-324/TWIST1 axis to regulate the proliferation, invasion, migration and angiogenesis of epithelial ovarian cancer (EOC) cells, thereby affecting ovarian cancer (OC) progression. Methods RT-qPCR was used to detect the expression of TPT1-AS1 and miR-324 in 29 OC lesions and adjacent tissue samples. The two OC cell models of TPT1-AS1 overexpression and miRNA324 knockdown were constructed, and the cell proliferation, invasion and migration abilities were detected by CCK-8, TranswellTM and scratch test. Western blot analysis was used to detect the protein expression levels of TWIST1, epithelial cadherin (E-cadherin), Vimentin, and vascular endothelial growth factor A (VEGF-A) in OC cells. Fluorescence in situ hybridization (FISH) and RNA pull-down experiments were used to verify the interaction between TPT1-AS1 and miR-324. Immunohistochemistry and Targetscan bioinformatics analysis were used to verify the negative regulatory role of miR-324 in the epithelial-mesenchymal transition (EMT) process. Results The TPT1-AS1 expression was significantly higher in OC tissues than that in para-cancerous tissues, while the miR-324 expression was significantly lower. In SKOV3 cells with TPT1-AS1 overexpression, the miR-324 expression decreased significantly, and TPT1-AS1 was negatively correlated with miR-324. It was also found that TPT1-AS1 and miR-324 were co-expressed in OC cells, and there was a direct binding relationship between them. Down-regulation of miR-324 significantly promoted the proliferation, invasion and migration of SKOV3 cells. Further studies revealed that miR-324 had a binding site at the 3'-UTR end of the TWIST1, a key transcription factor for EMT. Inhibiting miR-324 expression increased the transcription level of TWIST1, leading to a decrease in E-cadherin protein expression and an increase in Vimentin protein expression. Additionally, the downregulation of miR-324 resulted in an increased expression level of VEGF-A protein, which in turn enhanced angiogenesis of OC. Conclusion TPT1-AS1 promotes EOC cell proliferation, invasion, migration and angiogenesis by negatively regulating the miR-324/TWIST1 axis, thus promoting the development of OC. These findings provide new potential targets for the diagnosis and treatment of OC.
Humans
;
MicroRNAs/metabolism*
;
Female
;
Cell Movement/genetics*
;
Ovarian Neoplasms/blood supply*
;
Twist-Related Protein 1/metabolism*
;
Cell Line, Tumor
;
Neovascularization, Pathologic/genetics*
;
Neoplasm Invasiveness
;
Carcinoma, Ovarian Epithelial/metabolism*
;
Nuclear Proteins/metabolism*
;
Cell Proliferation/genetics*
;
Epithelial-Mesenchymal Transition/genetics*
;
Gene Expression Regulation, Neoplastic
;
RNA, Long Noncoding/metabolism*
;
Cadherins/genetics*
;
Vascular Endothelial Growth Factor A/genetics*
;
Vimentin/genetics*
;
Angiogenesis
8.The expression characteristics of TXN in pan cancer and its impact on tumor immunity and prognosis.
Annan SUN ; Luna SUN ; Hao WU ; Pu LI
Chinese Journal of Cellular and Molecular Immunology 2025;41(8):706-716
Objective TXN is a thioredoxin (TXN) that participates in many redox reactions and plays a crucial role in various signaling pathways. However, the role of TXN in many cancers is still unclear. The objective of this study is to investigate and visualize the diagnostic, prognostic, and immunological implications of TXN expression across various cancer types. Methods The clinical data were downloaded from the cancer genome mapping project(TCGA) database to analyze the expression level of TXN in pan cancer, and the expression level was preliminarily verified by human protein mapping (HPA)(https://www.proteinatlas.org/)database. The ESTIMATE algorithm and CIBERSORT algorithm were applied to calculate the correlation between TXN expression and immune cell infiltration. The correlation between TXN and microsatellite instability (MSI) and tumor mutation burden (TMB) was analyzed using Spearman method. Gene Set Enrichment Analysis (GSEA) is used for gene biology functional analysis and sensitivity analysis of genes to pan cancer therapeutic drugs. Results TXN is highly expressed in most malignant tumors. The high expression of TXN is associated with overall survival (OS), disease-specific survival (DSS), disease-free interval (DFI), and progression free interval (PFI) in various cancers. Moreover, TXN expression is associated with TMB, MSI, tumor microenvironment, chemotherapy sensitivity and so on. Conclusion TXN may become a potential prognostic biomarker in pan cancer, providing strong theoretical basis for future tumor diagnosis and prognosis judgment. The retinoic acid-inducible gene-I (RIG-I)-like receptor signaling pathway, Toll-like receptor (TLR) signaling pathway, and nucleotide binding oligomerization domain (NOD)-like receptor signaling pathway may be crucial pathways through which TXN influences tumor immunity.
Humans
;
Prognosis
;
Neoplasms/diagnosis*
;
Thioredoxins/metabolism*
;
Microsatellite Instability
;
Gene Expression Regulation, Neoplastic
;
Biomarkers, Tumor/genetics*
;
Mutation
;
Tumor Microenvironment
9.Effects of lncRNA DHRS4-AS1 on proliferation, invasion, migration, and apoptosis of thyroid cancer cells by regulating the miR-221-3p/SOCS3 signaling axis.
Hui WANG ; Yu GUO ; Peipei ZHANG ; Haoyu YANG ; Chuntao TIAN ; Mingming JIN
Chinese Journal of Cellular and Molecular Immunology 2025;41(9):798-805
Objective To explore the influences of long-chain noncoding RNA DHRS4-AS1 (lncRNA DHRS4-AS1) on the proliferation, invasion, migration, and apoptosis of thyroid cancer (TC) cells by regulating the microRNA-221-3p (miR-221-3p)/suppressor of cytokine signaling 3 (SOCS3) signaling axis. Methods Quantitative real-time PCR (qRT-PCR) was applied to detect the expression of lncRNA DHRS4-AS1, miR-221-3p, and SOCS3 mRNA in TC cell lines, and the optimal cell line was selected for subsequent experiments. FTC-133 cells were divided into five groups: control group, pcDNA-NC group, DHRS4-AS1 group, DHRS4-AS1 combined with agomir NC group, and DHRS4-AS1 combined with miR-221-3p-agomir group. Transfection efficiency was assessed using qRT-PCR. Dual luciferase reporter assays were applied to verify the targeting interaction between lncRNA DHRS4-AS1, SOCS3, and miR-221-3p. Western blot analysis was used to detect the expression of SOCS3 in FTC-133 cells. EdU method was used to measure cell proliferation. Flow cytometry was applied to measure the apoptosis of FTC-133 cells. Scratch experiment was applied to measure the migration of FTC-133 cells. Transwell chamber was applied to detect the invasion of FTC-133 cells. Nude mouse transplantation tumor experiment was used to observe the effect of lncRNA DHRS4-AS1 on the growth of TC transplantation tumors. Results Dual luciferase reporter assays showed a targeting relationship between lncRNA DHRS4-AS1, miR-221-3p, and SOCS3. LncRNA DHRS4-AS1 and SOCS3 were downregulated and miR-221-3p was upregulated in FTC-133 cells. Overexpression of lncRNA DHRS4-AS1 inhibited proliferation, migration, and invasion of FTC-133 cells, while inducing apoptosis. Conversely, miR-221-3p overexpression reversed these inhibitory effects, and suppressed the apoptosis. Nude mouse transplantation experiment observed that overexpression of lncRNA DHRS4-AS1 resulted in a decrease in tumor tissue quality and volume, and a decrease in miR-221-3p expression and an increase in SOCS3 expression. Conclusion LncRNA DHRS4-AS1 is downregulated in FTC-133 cells. Overexpression of lncRNA DHRS4-AS1 can inhibit the proliferation, invasion, and migration of TC cells and induce apoptosis by regulating the miR-221-3p/SOCS3 signaling axis.
MicroRNAs/metabolism*
;
Suppressor of Cytokine Signaling 3 Protein/metabolism*
;
Humans
;
RNA, Long Noncoding/metabolism*
;
Apoptosis/genetics*
;
Cell Proliferation/genetics*
;
Cell Movement/genetics*
;
Thyroid Neoplasms/physiopathology*
;
Animals
;
Signal Transduction/genetics*
;
Cell Line, Tumor
;
Mice, Nude
;
Neoplasm Invasiveness
;
Gene Expression Regulation, Neoplastic
;
Mice
;
Mice, Inbred BALB C
10.NIP7 upregulates the expression of ubiquitin-conjugating enzyme E2 C to promote tumor growth in anaplastic thyroid cancer.
Yingying GONG ; Ziwen FANG ; Yixuan WANG ; Minghua GE ; Zongfu PAN
Journal of Zhejiang University. Medical sciences 2025;54(3):372-381
OBJECTIVES:
To investigate the role of nucleolar pre-rRNA processing protein NIP7 (NIP7) in maintaining the malignant phenotype of anaplastic thyroid cancer (ATC) and its molecular mechanisms.
METHODS:
NIP7 expression in ATC tissues and its gene knock-out effects in ATC cells were analyzed using gene expression microarray (GSE33630), proteome database (IPX0008941000) and the Dependency Map database, respectively. Expression and localization of NIP7 in normal thyroid cells, papillary thyroid cancer cells, and ATC cells were detected by Western blotting. Small interfering RNA (siRNA) was transfected into ATC cells, and the knockdown efficiency of NIP7 was detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting. Cell proliferation was assessed by CCK-8 assay, colony formation was evaluated by colony formation assay, and tumor growth was assessed by xenograft tumor model in nude mice. SUnSET (surface sensing of translation) assay combined with co-immunoprecipitation were employed to evaluate the effect of NIP7 silencing on ubiquitin-conjugating enzyme E2 C (UBE2C) translation. Finally, gene set enrichment analysis was used to identify shared pathways of NIP7 and UBE2C, which were validated by qRT-PCR.
RESULTS:
Compared with normal tissues and papillary thyroid cancer, NIP7 was significantly upregulated in ATC tissues, and had a gene knock-out fitness effect on different ATC cell lines. The relative protein levels of NIP7 in ATC cells were significantly higher than those in normal thyroid follicular cells, and the protein was mainly expressed in the nucleus. NIP7 silencing significantly inhibited cell proliferation and reduced colony formation. Xenograft tumor model showed that NIP7 knockdown significantly slowed down the growth of ATC xenograft, and the tumor volume and weight were significantly lower than those in the control group (all P<0.05). NIP7 silencing downregulated the protein level of UBE2C, but did not affect the expression of UBE2C mRNA. Compared to the control group, UBE2C silencing significantly inhibited ATC cells proliferation (P<0.01) and colony formation (P<0.05). UBE2C overexpression reversed the proliferation-inhibitory effect induced by NIP7 silencing (P<0.01). Gene set enrichment analysis indicated that NIP7 and UBE2C were both involved in DNA replication. NIP7 or UBE2C silencing could significantly downregulate the expression levels of DNA polymerase epsilon, catalytic subunit 2 and replication factor C4 in DNA replication pathway.
CONCLUSIONS
NIP7 promotes ATC tumor growth by upregulating UBE2C to mediate DNA replication.
Humans
;
Ubiquitin-Conjugating Enzymes/genetics*
;
Thyroid Neoplasms/genetics*
;
Thyroid Carcinoma, Anaplastic/genetics*
;
Animals
;
Mice, Nude
;
Mice
;
Cell Line, Tumor
;
Cell Proliferation
;
Up-Regulation
;
RNA, Small Interfering/genetics*
;
Nuclear Proteins/metabolism*
;
Gene Expression Regulation, Neoplastic

Result Analysis
Print
Save
E-mail