Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Aging (Albany NY) ; 16(8): 6954-6989, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38613802

ABSTRACT

BACKGROUND: Glioma, a highly invasive and deadly form of human neoplasm, presents a pressing need for the exploration of potential therapeutic targets. While the lysosomal protein transmembrane 4A (LATPM4A) has been identified as a risk factor in pancreatic cancer patients, its role in glioma remains unexplored. METHODS: The analysis of differentially expressed genes (DEG) was conducted from The Cancer Genome Atlas (TCGA) glioma dataset and the Genotype Tissue Expression (GTEx) dataset. Through weighted gene co-expression network analysis (WGCNA), the key glioma-related genes were identified. Among these, by using Kaplan-Meier (KM) analysis and univariate/multivariate COX methods, LAPTM4A emerged as the most influential gene. Moreover, the bioinformatics methods and experimental verification were employed to analyze its relationships with diagnosis, clinical parameters, epithelial-mesenchymal transition (EMT), metastasis, immune cell infiltration, immunotherapy, drug sensitivity, and ceRNA network. RESULTS: Our findings revealed that LAPTM4A was up-regulated in gliomas and was associated with clinicopathological features, leading to poor prognosis. Furthermore, functional enrichment analysis demonstrated that LATPM4A played a role in the immune system and cancer progression. In vitro experiments indicated that LAPTM4A may influence metastasis through the EMT pathway in glioma. Additionally, we found that LAPTM4A was associated with the tumor microenvironment (TME) and immunotherapy. Notably, drug sensitivity analysis revealed that patients with high LAPTM4A expression were sensitive to doxorubicin, which contributed to a reduction in LAPTM4A expression. Finally, we uncovered the FGD5-AS1-hsa-miR-103a-3p-LAPTM4A axis as a facilitator of glioma progression. CONCLUSIONS: In conclusion, our study identifies LATPM4A as a promising biomarker for prognosis and immune characteristics in glioma.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Computational Biology , Glioma , Membrane Proteins , Female , Humans , Male , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Prognosis
2.
Am J Transl Res ; 15(8): 5110-5119, 2023.
Article in English | MEDLINE | ID: mdl-37692964

ABSTRACT

PURPOSE: To analyze the clinical efficacy of gasless submental-transoral endoscopic thyroidectomy (ETE) with Kirschner wire suspension in patients with papillary thyroid carcinoma (PTC). METHODS: Retrospectively, we enrolled 112 patients with PTC who received treatment in The Second Affiliated Hospital of Nanchang University between December 2020 and December 2021. Among them, 60 cases (laparoscopic group) received gasless submental-transoral ETE with Kirschner wire suspension, and the other 52 cases (open group) were treated by traditional thyroidectomy. Surgical indicators (operative time (OT), intraoperative blood loss (IBL), and postoperative drainage volume (DV)), number of central lymph node (CLN) dissected, length of hospital stay (LOS), Visual Analogue Scale (VAS) score, aesthetic satisfaction score, and complications were observed and compared between the two groups. RESULTS: There was no significant difference between the two groups in OT (55.73±5.49 min vs. 55.00±7.79 min), IBL (20.67±7.75 mL vs. 23.08±6.24 mL), postoperative DV (33.17±15.09 mL vs. 39.52±19.22 mL), number of CLN dissected (5.54±2.75 vs. 5.43±3.15), LOS (3.63±0.69 d vs. 3.68±0.57 d), postoperative VAS score (3.19±1.07 points vs. 3.38±1.09 points), and total complication rate (3.85% vs. 8.33%; all P>0.05). However, the laparoscopic group exhibited a significantly higher aesthetic satisfaction score than the open group (7.10±1.46 points vs. 6.42±1.46 points; P<0.05). In addition, patients in both groups were followed up for at least 3 months, and no recurrence or metastasis was observed. CONCLUSIONS: Gasless submental-transoral ETE with Kirschner wire suspension offers comparable curative effect as traditional thyroidectomy and safety, but it provides superior esthetic results, making it a viable treatment option for patients with PTC.

3.
Adv Healthc Mater ; 12(20): e2300012, 2023 08.
Article in English | MEDLINE | ID: mdl-36929147

ABSTRACT

The development of nanomedicines that combine photothermal therapy (PTT) with photodynamic therapy (PDT) is considered promising for cancer treatment, but still faces the challenge of enhancing tumoricidal efficiency. Fortunately, apart from the well-acknowledged effect on direct tumor cell-killing, nitric oxide (NO) is also considered to be effective for the enhancement of both PTT and PDT. However, both the low loading efficiency of NO precursor and the short half-life time and diffusion distance of NO hamper the synergistic therapeutic efficacy of NO. Taking the aforementioned factors into account, a mitochondria-targeted nitric oxide nanogenerator, EArgFe@Ce6, is constructed to achieve high loading of the NO donor l-Arginine (l-Arg) for synergistic photodynamic/gas/photothermal therapy upon single 660 nm light irradiation. The coordination of epigallocatechin gallate (EGCG) and ferric ions (Fe3+ ) provides EArgFe@Ce6 supreme photothermal capability to perform low-temperature PTT (mPTT). EGCG endows EArgFe@Ce6 with mitochondria-targeting capability and meanwhile favors hypoxia alleviation for enhanced PDT. The PDT-produced massive reactive oxygen species (ROS) further catalyzes l-Arg to generate a considerable amount of NO to perform gas therapy and sensitize both mPTT and PDT. In vitro and in vivo studies demonstrate that the synergistic photodynamic/gas/photothermal therapy triggered by single 660 nm light irradiation is highly effective for tumor treatments.


Subject(s)
Nanoparticles , Photochemotherapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Nitric Oxide , Photothermal Therapy , Phototherapy , Cell Line, Tumor
4.
Adv Healthc Mater ; 12(18): e2300054, 2023 07.
Article in English | MEDLINE | ID: mdl-36977362

ABSTRACT

To overcome the limitations of doxorubicin (DOX) chemotherapy, nanomedicines that integrate additional photothermal therapy (PTT) and chemodynamic therapy (CDT) strategies are highlighted as promising alternatives for the treatment of malignant tumors. However, time-consuming preparation processes, biosafety concerns, and the bottlenecks of individual therapeutic modalities often limit the practical applications of this strategy. To address these issues, this work designs an oxygen economizer that additionally serves as a Fenton reaction amplifier through the simple assembly of epigallocatechin gallate (EGCG), pluronic F-127 (PF127), iron (III) ions, and doxorubicin (DOX) for the enhancement of synergistic PTT/CDT/chemotherapy. The resulting nanoformulation, EFPD, can target mitochondria and inhibit cell respiration to reduce O2 consumption, thus boosting DOX-mediated H2 O2 generation for enhanced CDT and simultaneously improving hypoxia-limited DOX chemotherapy efficacy. Moreover, the coordination between EGCG and Fe3+ provides EFPD with excellent photothermal conversion efficiencies (η = 34.7%) for PTT and photothermal-accelerated drug release. Experimental results indicate that EFPD-mediated synergistic enhancement of PTT/CDT/chemotherapy can achieve excellent therapeutic outcomes, including enhanced ablation of solid tumors, reduced metastasis and cardiotoxicity, and extended life spans.


Subject(s)
Doxorubicin , Nanoparticles , Neoplasms , Humans , Cell Line, Tumor , Doxorubicin/pharmacology , Hydrogen Peroxide , Hypoxia , Iron , Metals , Neoplasms/therapy , Oxygen , Photothermal Therapy , Drug Synergism
5.
J Formos Med Assoc ; 122(8): 738-746, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36739231

ABSTRACT

PURPOSE: The purpose of this study was to clarify the effect of ZC3H13 on the growth of papillary thyroid carcinoma (PTC). METHODS: Firstly, we used qRT-PCR and Western blot to compare the difference in the expression of ZC3H13 between normal thyroid epithelial cells and PTC cell lines. Then, ZC3H13 overexpression/knockout thyroid cancer cells were constructed by lentivirus transfection, and the effects of overexpression of ZC3H13 on the proliferation, migration and invasion of PTC cells were detected by CCK8 and transwell experiments. Lastly, MeRIP-qPCR, RIP and o Actinomycin D were used to verify that ZC3H13 regulated the expression of downstream target gene IQGAP1 through m6A modification. RESULTS: ZC3H13 expression was decreased in PTC cell lines BCPAP, KTC-1, k1, HTH83, and TPC-1. Proliferation, invasion, and migration of PTC cells were inhibited by overexpressed ZC3H13 but increased by knockdown of ZC3H13. IQGAP1 expression was suppressed by ZC3H13 overexpression but enhanced by ZC3H13 knockdown. In ZC3H13-overexpressed PTC cells, the m6A level of IQGAP1 mRNA was increased, and the IQGAP1 mRNA expression was decreased with the increasing time of Actinomycin D treatment. YTHDF2 enriched more IQGAP1 mRNA than IgG and knockdown of YTHDF2 reversed the effect of ZC3H13 overexpression on IQGAP1 mRNA stability. The xenograft tumor experiment in nude mice confirmed that the overexpression of ZC3H13 inhibited tumor growth, while overexpression of IQGAP1 could reverse the inhibitory effect of ZC3H13 overexpression on tumor growth. CONCLUSION: ZC3H13 mediates IQGAP1 mRNA degradation by promoting m6A modification of IQGAP1 mRNA, this provides a prospective therapeutic target for PTC.


Subject(s)
MicroRNAs , Thyroid Neoplasms , Mice , Animals , Humans , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , MicroRNAs/genetics , Mice, Nude , Dactinomycin/metabolism , Cell Line, Tumor , Cell Proliferation , Neoplasm Invasiveness , Cell Movement , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , RNA, Messenger , Gene Expression Regulation, Neoplastic , Nuclear Proteins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
6.
Adv Healthc Mater ; 12(3): e2202045, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36239177

ABSTRACT

Doxorubicin (Dox)-mediated generation of reactive oxygen radicals (ROS) for mitochondrial apoptosis is identified as a new cytotoxic mechanism in addition to the well-established one via nuclear DNA replication interference. However, this mechanism contributes far less than the latter to Dox therapy. This newly identified pathway to make Dox therapy function like the combination of chemodynamic therapy (CDT) and chemotherapy-mediated by Dox alone would be amplified. One-pot nanoconstruction (HEBD) is fabricated based on the chemical reactions driven assemblies among epigallocatechin gallate (EGCG), buthionine sulfoximine (BSO) and formaldehyde in aqueous mediums followed by Dox adsorption. Acid tumor microenvironments allow the liberation of EGCG, BSO, and Dox due to the breakage of Schiff base bonds. EGCG component in HEBD is responsible for targeting mitochondria and disrupting mitochondrial electron transport chain (mETC) to compel electrons leakage in favor of their capture by Dox to produce more ROS. EGCG-induced mETC disruption results in mitochondrial respiration inhibition with alleviated hypoxia in tumor cells while BSO inhibits glutathione biosynthesis to protect ROS from redox depletion, further boosting Dox-induced CDT. This strategy of amplifying CDT pathway for the Dox-mediated combined therapy could largely improve antitumor effect, extend lifespan of tumor-bearing mice, reduce risks of cardiotoxicity and metastasis.


Subject(s)
Apoptosis , Doxorubicin , Mice , Animals , Reactive Oxygen Species/metabolism , Doxorubicin/pharmacology , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Mitochondria
7.
Cell Signal ; 88: 110165, 2021 12.
Article in English | MEDLINE | ID: mdl-34606908

ABSTRACT

Radioiodine (131I) is one of the most well-known and widely used targeted therapies. In thyroid carcinoma (THCA), it has been applied for more than eight decades and is still being utilized to eliminate remnants after resection and to reduce tumor metastases. Here, we aimed to investigate if lysine methyltransferase 2B (KMT2B) silencing could confer 131I resistance to THCA cells and the epigenetic mechanism behind. RT-qPCR, immunohistochemistry and western blot revealed that KMT2B was poorly expressed in THCA cells, and 131I resistance of cells led to a further decrease in KMT2B expression. EdU, colony formation, TUNEL, and tumor growth and metastasis assays showed that overexpression of KMT2B sensitized THCA cell to 131I and inhibited cell growth and metastasis. Further bioinformatics prediction and functional assay validation revealed that KMT2B elevated SHPRH expression via H3K4me3 modification in the SHPRH promoter, and that SHPRH modulated FYN ubiquitination, thereby promoting its protein degradation. We finally proved that the 131I-resistant cells regained resistance to 131I by FYN overexpression in the presence of KMT2B overexpression in vitro and in vivo. Therefore, we conclude that the overexpression of KMT2B represents a potential target for THCA therapy.


Subject(s)
Iodine Radioisotopes , Thyroid Neoplasms , DNA Helicases/metabolism , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Iodine Radioisotopes/metabolism , Protein Stability , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism
8.
Oncol Rep ; 45(4)2021 04.
Article in English | MEDLINE | ID: mdl-33649840

ABSTRACT

Radioactive iodine (RAI, 131I) therapy is the main treatment for thyroid carcinoma (TC). Long noncoding RNA (lncRNA)/microRNA (miR) competing endogenous RNA (ceRNA) networks have aroused great interest for their roles in gene expression. The present study aimed to investigate the effect of lncRNA SNHG7 on the growth and 131I resistance of TC. Differentially expressed lncRNAs in TC and paracancerous tissues were analyzed. The binding of miR­9­5p with small nucleolar RNA host gene 7 (SNHG7) and dipeptidyl­peptidase 4 (DPP4) was identified. Gain­ and loss­of­function analyses of SNHG7 and miR­9­5p were performed to determine their effects on the growth and 131I resistance of TC cells. The activity of the PI3K/Akt pathway was evaluated. Consequently, upregulated SNHG7 was revealed in TC tissues and correlated with 131I resistance. Silencing of SNHG7 or overexpressing miR­9­5p inhibited the growth and 131I resistance of TC cells. SNHG7 acted as a ceRNA of miR­9­5p to enhance DPP4 expression. Overexpressed SNHG7 increased DPP4 expression and activated the PI3K/Akt signaling pathway by sponging miR­9­5p. The in vitro results were reproduced in vivo. In summary, the present study provided evidence that the SNHG7/miR­9­5p/DPP4 ceRNA network could promote the growth and 131I resistance of TC cells via PI3K/Akt activation. The present study may offer novel options for TC treatment.


Subject(s)
Dipeptidyl Peptidase 4/genetics , Iodine Radioisotopes/pharmacology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/radiotherapy , Animals , Cell Growth Processes/radiation effects , Dipeptidyl Peptidase 4/metabolism , Enzyme Activation , Female , Gene Regulatory Networks , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , Radiation Tolerance , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
10.
Mol Cell Biochem ; 476(2): 727-739, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33201383

ABSTRACT

Kin17 DNA and RNA binding protein (Kin17) is an extremely conserved nuclear protein that is almost expressed in every type of mammal cells. Recently, Kin17 has been implicated into the regulation of tumorigenesis of diverse human cancers. However, its functions in thyroid cancer (TC) are still largely unexplored. Kin17 mRNA and protein level were tested by qRT-PCR and western blot, respectively. Effects of Kin17 on TC cell proliferation were estimated by colony formation assay and flow cytometry analysis in vitro as well as by in vivo tumor growth experiment. TC cell migratory and invasive capacities were assessed via wound-healing and transwell experiments. Epithelial-mesenchymal transition (EMT)-related proteins (E-cadherin and N-cadherin) and p38 MAPAK signaling pathway-related proteins (p-p38, p38, Cyclin D1, and p27) were examined via western blot. Kin17 was remarkably increased in TC tissue samples and cell lines at both mRNA and protein levels compared to normal tissue and control cell line. Knockdown of Kin17 obviously repressed TC cell proliferation, arrested cell cycle, and inhibited TC cell migration and invasion in vitro, while overexpression of Kin17 produced opposite effects. Kin17 knockdown suppressed p38 MAPK signaling pathway, while Kin17 overexpression activated this pathway. Treatment of p38 agonist (p79350) abolished the repressive effects of sh-Kin17 on TC cell proliferation, migration, and invasion, as well as on p38 pathway. Kin17 knockdown was also found to enhance the sensitivity of Doxorubicin of TC cells. In addition, Kin17 knockdown in vivo also markedly repressed TC tumor growth and p38 pathway. Kin17 functioned as an oncogene of TC by activating p38 MAPK signaling pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , RNA-Binding Proteins/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , Humans , Male , Mice, Nude , Neoplasm Invasiveness , RNA-Binding Proteins/genetics , Signal Transduction , Thyroid Neoplasms/genetics , Topoisomerase II Inhibitors/pharmacology , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/genetics
11.
Cancer Cell Int ; 20: 203, 2020.
Article in English | MEDLINE | ID: mdl-32514248

ABSTRACT

BACKGROUND: Papillary thyroid cancer (PTC) is a type of malignant tumor with excellent prognosis, accounting for more than 80% of thyroid cancer. Recently, numerous studies illustrated the importance of N 6-methyladenosine (m6A) RNA modification to tumorigenesis, but it has never been reported in PTC. METHODS: We downloaded data from The Cancer Genome Atlas (TCGA) and analyzed RNA expression, single nucleotide polymorphisms (SNPs) and copy number variations (CNVs) of 19 m6A RNA methylation regulators in PTC. Then we used nonnegative matrix factorization (NMF) to cluster patients into two m6A subtypes and compared them in overall survival (OS) and disease-free survival (DFS). The Weighted correlation network analysis (WGCNA) and univariate Cox proportional hazard model (CoxPH) were used to select genes for the construction of a m6A-related signature. The accuracy and prognostic value of this signature were validated by using receiver operating characteristic (ROC) curves, K-M (Kaplan-Meier) survival analysis, univariant and multivariant analyses. RESULTS: CNVs and differential expression of m6A regulators were observed in PTC patients. Especially IGF2BP2 (Insulin-like growth factor 2 mRNA binding protein 2), which was most significantly overexpressed in tumor tissue. We chose 4 genes in the m6A-related module from WGCNA: IGF2BP2, STT3A, MTHFD1 and GSTM4, and used them to construct a m6A-related signature. The prognostic value of this signature was validated, and risk scores provided by the signature was the independent prognostic factor for PTC. A nomogram was also provided for clinical usage. CONCLUSIONS: We performed a comprehensive evaluation of the m6A RNA modification landscape of PTC and explored its underlying mechanisms. Our m6A-related signature was of great significance in predicting the DFS of patients with PTC. And IGF2BP2 was a gene worthy for further analysis as its strong correlation with DFS and clinical phenotypes of PTC.

12.
Oncol Lett ; 19(3): 2295-2305, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32194729

ABSTRACT

The prognosis of most patients with papillary thyroid carcinoma (PTC) is excellent despite some cases of tumor progression or relapse. The present study was designed to reveal possible prognostic risk indicators for PTC. Differentially expressed genes (DEGs) extracted from 4 Gene Expression Omnibus (GEO) cohorts were subjected to functional enrichment analyses by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) pathway analysis. A dataset from The Cancer Genome Atlas (TCGA) was obtained to filter and validate significant genes using cytoHubba, followed by analysis of their association with clinicopathological characteristics and prognosis. In total, 240 DEGs were identified after data preprocessing. These DEGs were enriched in 'intracellular redox equilibrium', 'release of exosome', 'cell adhesion', 'regulation of extracellular matrix', 'collagen binding' and 'energy metabolism' based on GO analysis which including cellular component, molecular function and biological process. KEGG pathway analysis revealed that the DEGs were enriched in thyroid hormone synthesis, pathways in cancer, focal adhesion, metabolic pathways, apoptosis, PPAR signaling pathway and PI3K/AKT signaling pathway. Using cytoHubba, the following hub genes were identified: Apolipoprotein E (APOE); hemoglobin subunit α1 (HBA1); angiotensin II receptor 1 (AGTR1); collagen I α1 (COL1A1); galectin 3 (LGALS3) and TIMP metallopeptidase inhibitor 1 (TIMP1). The expression of these genes was found to be consistent in TCGA datasets. Kaplan-Meier analysis revealed that APOE was significantly associated with overall survival (P=0.00067) and disease free survival (P=0.00220). Additionally, low expression of APOE was significantly associated with older age (P<0.001) and higher TNM stage (P<0.001) compared with the high expression group. Therefore, APOE may function as a predictive risk indicator for progression as well as prognosis of PTC.

13.
Cancer Biomark ; 23(2): 185-191, 2018.
Article in English | MEDLINE | ID: mdl-30175973

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) were recently identified as crucial regulators of papillary thyroid cancer (PTC). However, the clinical role and regulatory functions of lncRNA cancer susceptibility candidate 2 (CASC2) in PTC remain unknown. METHODS: LncRNA CASC2 expression was examined in plasma samples from 68 PTC patients and 39 patients with nodular goiter (NG). Cell proliferation, migration and invasion abilities were evaluated using CCK8 assay and transwell migration and invasion assay. QRT-PCR and western blot analysis were performed to detect the expression of epithelial-to-mesenchymal (EMT) markers ZEB1, E-cadherin and vimentin in PTC cells. RESULTS: We demonstrated that lncRNA CASC2 expression was significantly downregulated in tumor tissues and plasma samples in patients with PTC compared with those in nodular goiters (P< 0.05). Decreased plasma lncRNA CASC2 expression associated with lymph node metastasis (LNM) of PTC patients and was identified as an independent risk for patients with LNM (P< 0.05). Furthermore, functional assays demonstrated that overexpression of lncRNA CASC2 inhibited cell proliferation, migration and invasion of PTC. Moreover, we demonstrated that overexpression of lncRNA CASC2 suppressed cell epithelial-mesenchymal transition (EMT) process of PTC by increasing the E-cadherin expression, but downregulating ZEB1 and N-cadherin expression. CONCLUSIONS: Thus, these results indicated that lncRNA CASC2 was a predictor for LNM of PTC patients and may serve as a potential target of PTC treatment.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Tumor Suppressor Proteins/genetics , Adult , Aged , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Thyroid Cancer, Papillary/metabolism , Tumor Burden , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...