1
|
Niu X, Ma F, Li F, Wei C, Zhang J, Gao Z, Wang J, Da M. Integration of bioinformatics and cellular experiments unveils the role of SYT12 in gastric cancer. BMC Cancer 2024; 24:1331. [PMID: 39472897 PMCID: PMC11520883 DOI: 10.1186/s12885-024-13077-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
OBJECTIVE This study employs integrated bioinformatics analysis and in vitro cellular experiments to elucidate the role of Synaptotagmin-12 (SYT12) in the progression of gastric cancer. METHODS We utilized databases and platforms such as Xiantao Academic Tools, UALCAN, Kaplan-Meier plotter analysis, and The Cancer Genome Atlas (TCGA) to extract datasets on SYT12 in gastric cancer. We analyzed the relationship between SYT12 expression and the clinicopathological features, prognosis, diagnosis, and immune infiltration of stomach adenocarcinoma (STAD) patients. Verification was conducted using samples from 31 gastric cancer patients. Additionally, in vitro cellular experiments were performed to determine the role and potential mechanisms of SYT12 in the malignant behavior of gastric cancer cells. RESULTS Comprehensive bioinformatics analysis indicated that SYT12 is highly expressed in most cancers and is associated with promoter DeoxyriboNucleic Acid (DNA) methylation levels. SYT12 expression correlated with clinicopathological features, immune cell infiltration, immune checkpoint gene expression, and poor prognosis in STAD patients. In vitro experiments suggest that SYT12 may promote the proliferation and migration of gastric cancer cells by inducing epithelial-mesenchymal transition (EMT). CONCLUSIONS This study highlights the significant role of SYT12 in gastric cancer, suggesting its potential as a new target for early diagnosis, treatment, immunological, and prognostic evaluation in gastric cancer, offering new insights for precision medicine in this disease.
Collapse
Affiliation(s)
- Xingdong Niu
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Fubin Ma
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Fangying Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cunchun Wei
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Junrui Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhenhua Gao
- Department of General Surgery, The First People's Hospital of Baiyin (Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine), Baiyin, China
| | - Junhong Wang
- The First Clinical Medical College, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- Department of General Surgery, The First People's Hospital of Baiyin (Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine), Baiyin, China.
| | - Mingxu Da
- The First Clinical Medical College, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- Department of Surgical Oncology, Gansu Provincial Hospital, Donggang West Road, 204, lanzhou, Lanzhou, China.
| |
Collapse
|
2
|
Li X, Ding Z, Tong Y. Correlations of m 6A Methylation-Related lncRNAs with the Prognosis of Papillary Thyroid Carcinoma. Int J Gen Med 2024; 17:775-790. [PMID: 38476625 PMCID: PMC10929225 DOI: 10.2147/ijgm.s449827] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Purpose Papillary thyroid carcinoma (PTC) is the most common subtype of thyroid cancer. Recurrence makes the prognosis for some patients with PTC worse. Increasing evidence have suggested that N6-methyladenosine (m6A) RNA methylation plays an important role in tumorigenesis. However, the significance of m6A-related lncRNAs in the malignant progression of PTC remains unknown. In this study, we explored the significance of M6A-related lncrnas in the malignant progression of PTC. Patients and Methods Transcriptome and clinical data of PTC were achieved and integrated from The Cancer Genome Atlas (TCGA). Firstly, a Spearman correlation analysis was performed to obtain m6A RNA methylation-associated lncRNAs. Next, We constructed a prognostic signature and assessed the accuracy of the signature by receiver operating characteristic (ROC) curve and Kaplan Meier survival analyses. Furthermore, functional enrichment analysis was performed on the high- and low-risk groups. Finally, we determined prognostic gene expression in clinical samples using quantitative reverse transcription polymerase chain reaction (RT-qPCR). Results We identified 56 differentially expressed lncRNAs associated with m6A RNA methylation. Univariate Cox and Least Absolute Shrinkage and Selection Operator (LASSO) regression analyses showed that the survival-related lncRNAs associated with m6A RNA methylation were detected, which showed superior calibration and discrimination. Moreover, the biological processes related to energy metabolism were significantly activated in the high-risk group. Finally, the co-expressed genes of lncRNAs in the risk model were significantly enriched in biological processes related to copper ion response. Finally, we validated the expression levels of three prognostic genes in clinical samples using RT-qPCR. Conclusion Our study revealed m6A RNA methylation-associated lncRNAs were significantly associated with disease-free survival in patients with papillary thyroid cancer, which would improve our understanding of the relationship between m6A RNA methylation-associated lncRNAs and PTC.
Collapse
Affiliation(s)
- Xiang Li
- Department of General Surgery, The Affiliated Hospital of Jiujiang University, Jiujiang, People's Republic of China
| | - Zigang Ding
- Department of General Surgery, The Affiliated Hospital of Jiujiang University, Jiujiang, People's Republic of China
| | - Yun Tong
- Department of Pain, The Affiliated Hospital of Jiujiang University, Jiujiang, People's Republic of China
| |
Collapse
|
3
|
Bektas S, Kaptan E. RNA-Seq transcriptome analysis reveals Maackia amurensis leukoagglutinin has antitumor activity in human anaplastic thyroid cancer cells. Mol Biol Rep 2022; 49:9257-9266. [PMID: 36057880 PMCID: PMC9441018 DOI: 10.1007/s11033-022-07759-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/30/2022] [Indexed: 11/23/2022]
Abstract
Background Lectins are carbohydrate-binding molecules that can bind specifically to the sugar residues of glycoconjugates and are found in almost all organisms. Plant lectins subjected to many studies reported exhibiting anti-cancer activity. This study aimed to investigate the possible molecular mechanisms of Maackia amurensis leukoagglutinin II (MAL-II) treated ATCCs. Methods and results We tested the effects of MAL-II, which is isolated from Amur seeds, on cancerous features of 8505C human anaplastic thyroid cancer cells (ATCCs) on a large scale using RNA-Seq. Transcriptome analysis was performed using Illumina next-generation sequencing technology by using cDNA libraries obtained from total RNA isolates of ATCCs treated with 0.25 µM MAL-II for 24 h. Gene ontology and pathway enrichment analysis were performed for the systematic analysis of gene functions. Moreover, we validated RNA-Seq findings using qPCR. Our results showed that many cancer-related genes such as TENM4, STIM2, SYT12, PIEZO2, ABCG1, SPNS2, ARRB1, and IRX5 were downregulated and many anticancer genes such as HSPA6, G0S2, TNFAIP3, GEM, GADD45G, RND1, SERPINB2, and IL24 were upregulated. Also, pathway enrichment analysis showed that differentially expressed genes were found to be associated with Ras, p53, and apoptosis signaling pathways, which are some important signal transduction pathways in development, proliferation, stem cell control, and carcinogenesis. Conclusion Collectively, our results show that MAL-II treatment reveals significant antitumor activity by changing the expression of many cancer-related genes and implies that MAL-II treatment might be a potential candidate molecule to inhibit the malignancy of human anaplastic thyroid cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s11033-022-07759-6.
Collapse
Affiliation(s)
- Suna Bektas
- Department of Biology, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey
| | - Engin Kaptan
- Department of Biology, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey.
| |
Collapse
|
4
|
Suo H, Xiao N, Wang K. Potential roles of synaptotagmin family members in cancers: Recent advances and prospects. Front Med (Lausanne) 2022; 9:968081. [PMID: 36004367 PMCID: PMC9393329 DOI: 10.3389/fmed.2022.968081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
With the continuous development of bioinformatics and public database, more and more genes that play a role in cancers have been discovered. Synaptotagmins (SYTs) are abundant, evolutionarily conserved integral membrane proteins composed of a short N-terminus, a variable linker domain, a single transmembrane domain, and two C2 domains, and they constitute a family of 17 isoforms. The synaptotagmin family members are known to regulate calcium-dependent membrane fusion events. Some SYTs play roles in hormone secretion or neurotransmitter release or both, and much evidence supports SYTs as Ca2+ sensors of exocytosis. Since 5 years ago, an increasing number of studies have found that SYTs also played important roles in the occurrence and development of lung cancer, gastric cancer, colon cancer, and other cancers. Down-regulation of SYTs inhibited cell proliferation, migration, and invasion of cancer cells, but promoted cell apoptosis. Growth of peritoneal nodules is inhibited and survival is prolonged in mice administrated with siSYTs intraperitoneally. Therefore, most studies have found SYTs serve as an oncogene after overexpression and may become potential prognostic biomarkers for multiple cancers. This article provides an overview of recent studies that focus on SYT family members’ roles in cancers and highlights the advances that have been achieved.
Collapse
Affiliation(s)
- Huandan Suo
- Department of Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Nan Xiao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Kewei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Kewei Wang,
| |
Collapse
|
5
|
Jin L, Zheng D, Chen D, Xia E, Guan Y, Wen J, Bhandari A, Wang O. SYT12 is a novel oncogene that promotes thyroid carcinoma progression and metastasis. J Cancer 2021; 12:6851-6860. [PMID: 34659573 PMCID: PMC8518017 DOI: 10.7150/jca.62555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/01/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Thyroid malignancy is the most frequent endocrine malignant tumor whose incidence is still increasing. Mechanisms genomic variations play a major part in the pathogenesis of many types of malignancy. Synaptotagmin 12 (SYT12) is a member gene of the synaptotagmins family and SYT12's variants were shown to be associated with some malignancies. Nevertheless, SYT12's specific function and probable clinical value in papillary cancer were still unknown. Methods: We conducted complete genome sequence of 39 pairs PTC malignant neoplasm and matched non-neoplastic tissues. We found that SYT12 was significantly overexpressed in thyroid malignancy. Next, we investigated the expression level of SYT12 and the relation between clinical information and SYT12 expression in thyroid cancer in the Cancer Genome Atlas (TCGA). QRt-PCR of else 40 pairs local verified cohort was performed to confirm the sequencing data and TCGA cohort. Then, we used small interfering RNA (si-RNA) to knock down the expression of SYT12 in PTC cells. Finally, proliferation, cell colony formation, migration, invasion, and apoptosis assays were done to demonstrate the function of SYT12. Results: SYT12 is significantly overexpressed and higher expression of SYT12 upsurges the risk of lymph node metastatic and incidence rate of primary neoplasm multivariate focus type and classical histological type for PTC patients in TCGA cohort. In vitro experiments, the results of functional assays presented that knock-down of SYT12 inhibited the cell proliferation, cell colony formation, trans-well migration, and trans-well invasion and promoted cell apoptotic in PTC cell lines. Conclusion: SYT12 was a novel oncogene that promotes thyroid carcinoma progression and metastasis potential and a potential biomarker for diagnosis and treatment in PTC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Adheesh Bhandari
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Ouchen Wang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| |
Collapse
|
6
|
Jin L, Zheng D, Bhandari A, Chen D, Xia E, Guan Y, Wen J, Wang O. PSD3 is an oncogene that promotes proliferation, migration, invasion, and G1/S transition while inhibits apoptotic in papillary thyroid cancer. J Cancer 2021; 12:5413-5422. [PMID: 34405004 PMCID: PMC8364633 DOI: 10.7150/jca.60885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Background: The morbidity of thyroid cancer is gradually increasing, meanwhile, the average age of the morbidity population also becomes younger. Mechanisms genomic variations serve an important function for the pathogenesis of many cancer types. Pleckstrin and sec7 domain-containing 3 (PSD3), also known as EFA6R, was shown to be associated with some cancers such as acute myeloid leukemia, breast cancer metastasis, and astrocytoma. But it was unknown that whether PSD3 took effect and how did it work in thyroid cancer. Methods: We guessed that PSD3 might play an important role in thyroid cancer by consulting previous literature. Then, we analyzed the level of PSD3 expression in thyroid malignancy and the connection with clinical manifestation in The Cancer Genome Atlas (TCGA). And RNA extraction, reverse transcription, and real-time quantitative polymerase chain reaction (qRt-PCR) of 40 pairs of local samples were done to verify the result of TCGA. Then, PSD3 was knocked down by small interfering RNA (siRNA) for flowing functional experiments. Results: Bioinformatics and qRt-PCR analysis shown PSD3 was overexpressed in papillary thyroid cancer (PTC) and connected with the histological type (P=0.009) and risk of lymph node metastasis (P=0.016). In vitro assays, we confirmed that down-regulation PSD3 could not only suppress the cell proliferation, colony formation, cell migration, cell invasion, and G1/S cell cycle transition but also promote apoptosis in PTC cells. Conclusion: PSD3 promotes proliferation, migration, invasion, and G1/S transition while inhibits apoptotic in PTC and a possible biomarker in PTC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ouchen Wang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| |
Collapse
|
7
|
Han R, Sun W, Zhang H. Identification of a Signature Comprising 5 Soluble Carrier Family Genes to Predict the Recurrence of Papillary Thyroid Carcinoma. Technol Cancer Res Treat 2021; 20:15330338211036314. [PMID: 34590520 PMCID: PMC8489750 DOI: 10.1177/15330338211036314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/23/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
RNA-sequencing data and relevant clinical data in The Cancer Genome Atlas for 502 samples of papillary thyroid cancer (PTC) were analyzed to determine the prognostic value of soluble carrier family genes in PTC. We analyzed soluble carrier family gene expression and function in the samples. Clustering identified 2 clusters in the data. Risk characteristics were identified using LASSO and Univariate Cox regression analysis, which divided the patients into low and high-risk groups. The expression levels of 88 soluble carrier genes were significantly different between tumors and normal tissue. The 2 PTC clusters had different clinical outcomes and distributions of gene expression. The expression levels of SFXN1, SLC12A4, SLC35A1, SLC35E1, and SLCO1C1 were markedly different between the 2 groups. The high risk and low risk groups had significant different prognoses (P < 0.05). Significant differences were identified for disease free survival (DFS), sex and T stage between the 2 subgroups. The risk score was identified as an independent prognostic variable (P < 0.05) and as a predictor of clinicopathological variables. In patients with PTC, solute carrier gene expression showed differential associations with clinicopathological variables. The 5 genes could be used as prognostic factors for PTC, particularly to predict PTC recurrence.
Collapse
Affiliation(s)
- Rui Han
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, People’s Republic of China
- Rui Han and Wei Sun contributed equally to this article
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, People’s Republic of China
- Rui Han and Wei Sun contributed equally to this article
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
8
|
Liu K, Luo J, Shao C, Ren Z, Sun S, Zhu Y, Zhou H, Jiang Z, Li X, Gu W, Xu Y, Qiang Y, Ren B, Xu L, Wu H, Shen Y. Synaptotagmin 12 (SYT12) Gene Expression Promotes Cell Proliferation and Progression of Lung Adenocarcinoma and Involves the Phosphoinositide 3-Kinase (PI3K)/AKT/Mammalian Target of Rapamycin (mTOR) Pathway. Med Sci Monit 2020; 26:e920351. [PMID: 32108133 PMCID: PMC7063850 DOI: 10.12659/msm.920351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background This study aimed to use bioinformatics analysis to compare data from tissue microarrays from patients with lung adenocarcinoma (LUAD) and normal lung tissue, and human lung adenocarcinoma cells with normal lung epithelial cells in vitro to investigate the role of synaptotagmin 12 (SYT12) gene expression in LUAD. Material/Methods Human lung adenocarcinoma cell lines (A549, SPC-A-1, H1299, H1975, and PC9) and the normal HBE cell line were compared, and tumor xenografts were developed in mice. The Cancer Genome Atlas (TCGA) tissue microarray data were used to compare SYT12 expression and overall survival (OS). The in vivo and in vitro effects of down-regulation and upregulation of SYT12 were studied using short-interfering RNA (si-RNA) and overexpression plasmids, respectively. The Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) pathway analysis, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and Western blot investigated the molecular mechanisms of SYT12 expression in LUAD. Results SYT12 expression was increased in tissues from patients with LUAD from TCGA and was associated with advanced tumor stage and reduced prognosis. Knockdown of SYT12 suppressed the proliferation and migration of LUAD cells, and upregulation of SYT12 increased the proliferation and migration of LUAD cells in vitro. Phosphorylation of PIK3R3 activated the PI3K/AKT/mTOR pathway. In the mouse xenograft model, expression of SYT12 increased the volume and weight of the xenograft tumors. Conclusions Bioinformatics analysis, human LUAD cells, and mouse xenograft studies showed that SYT12 acted as a possible oncogene by phosphorylation of PIK3R3 to activate the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Kaichao Liu
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China (mainland).,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, China (mainland)
| | - Jing Luo
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China (mainland).,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, China (mainland)
| | - Chenye Shao
- Department of Cardiothoracic Surgery, Jingling Hospital, Jingling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Zhijian Ren
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, China (mainland)
| | - Sai Sun
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, China (mainland)
| | - Yihao Zhu
- The Fourth Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Hai Zhou
- Department of Cardiothoracic Surgery, Jingling Hospital, Jingling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China (mainland).,Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing, Jiangsu, China (mainland)
| | - Zhisheng Jiang
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing, Jiangsu, China (mainland)
| | - Xiaokun Li
- Department of Cardiothoracic Surgery, Jinling Hospital, Jinling Hospital, China (mainland)
| | - Wenfeng Gu
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China (mainland).,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, China (mainland)
| | - Youtao Xu
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, China (mainland).,Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, China (mainland)
| | - Yong Qiang
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing, Jiangsu, China (mainland)
| | - Binhui Ren
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, China (mainland).,Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, China (mainland)
| | - Lin Xu
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, China (mainland).,Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, China (mainland)
| | - Haiwei Wu
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China (mainland).,Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing, Jiangsu, China (mainland)
| | - Yi Shen
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China (mainland).,Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
9
|
Eizuka K, Nakashima D, Oka N, Wagai S, Takahara T, Saito T, Koike K, Kasamatsu A, Shiiba M, Tanzawa H, Uzawa K. SYT12 plays a critical role in oral cancer and may be a novel therapeutic target. J Cancer 2019; 10:4913-4920. [PMID: 31598163 PMCID: PMC6775516 DOI: 10.7150/jca.32582] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 06/27/2019] [Indexed: 02/06/2023] Open
Abstract
Synaptotagmin12 (SYT12) has been well characterized as the regulator of transmitter release in the nervous system, however the relevance and molecular mechanisms of SYT12 in oral squamous cell carcinoma (OSCC) are not understood. In the current study, we investigated the expression of SYT12 and its molecular biological functions in OSCC by quantitative reverse transcriptase polymerase chain reaction, immunoblot analysis, and immunohistochemistry. SYT12 were up-regulated significantly in OSCC-derived cell lines and primary OSCC tissue compared with the normal counterparts (P<0.05) and the SYT12 expression levels were correlated significantly with clinical indicators, such as the primary tumoral size, lymph node metastasis, and TNM stage (P<0.05). SYT12 knockdown OSCC cells showed depressed cellular proliferation, migration, and invasion with cell cycle arrest at G1 phase. Surprisingly, we found increased calcium/calmodulin-dependent protein kinase 2 (CAMK2) inhibitor 1 (CAMK2N1) and decreased CAMK2-phosphorylation in the knockdown cells. Furthermore, treatment with L-3, 4-dihydroxyphenylalanine (L-dopa), a drug approved for Parkinson's disease, led to down-regulation of SYT12 and similar phenotypes to SYT12 knockdown cells. Taken together, we concluded that SYT12 plays a significant role in OSCC progression via CAMK2N1 and CAMK2, and that L-dopa would be a new drug for OSCC treatment through the SYT12 expression.
Collapse
Affiliation(s)
- Keitaro Eizuka
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Dai Nakashima
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Noritoshi Oka
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sho Wagai
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshikazu Takahara
- Department of Dentistry and Oral-Maxillofacial Surgery, Chiba University Hospital, Chiba, Japan
| | - Tomoaki Saito
- Department of Dentistry and Oral-Maxillofacial Surgery, Chiba University Hospital, Chiba, Japan
| | - Kazuyuki Koike
- Department of Dentistry and Oral-Maxillofacial Surgery, Chiba University Hospital, Chiba, Japan
| | - Atsushi Kasamatsu
- Department of Dentistry and Oral-Maxillofacial Surgery, Chiba University Hospital, Chiba, Japan
| | - Masashi Shiiba
- Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideki Tanzawa
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Dentistry and Oral-Maxillofacial Surgery, Chiba University Hospital, Chiba, Japan
| | - Katsuhiro Uzawa
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Dentistry and Oral-Maxillofacial Surgery, Chiba University Hospital, Chiba, Japan
| |
Collapse
|
10
|
Gao Y, Elamin E, Zhou R, Yan H, Liu S, Hu S, Dong J, Wei M, Sun L, Zhao Y. FKBP51 promotes migration and invasion of papillary thyroid carcinoma through NF-κB-dependent epithelial-to-mesenchymal transition. Oncol Lett 2018; 16:7020-7028. [PMID: 30546435 PMCID: PMC6256738 DOI: 10.3892/ol.2018.9517] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 08/03/2018] [Indexed: 12/19/2022] Open
Abstract
FK506-binding protein 51 (FKBP51) is a member of the immunophilin family, with relevant roles in multiple signaling pathways, tumorigenesis and chemoresistance. However, the function of FKBP51 in papillary thyroid carcinoma (PTC) remains largely unknown. In the present study, increased FKBP51 expression was detected in PTC tissues as compared with adjacent normal tissues, and the expression level was associated with clinical tumor, node and metastasis stage. Using FKBP51-overexpressing K1 cells and FKBP51-knockdown TPC-1 cells, both human PTC cell lines, it was identified that FKBP51 promoted the migration and invasion of PTC, without affecting cell proliferation. Further investigation revealed that FKBP51 activated the NF-κB pathway and epithelial-to-mesenchymal transition (EMT) genes, and EMT was suppressed when NF-κB was inhibited. It was also assessed whether FKBP51 promoted the formation of cytoskeleton to promote migration and invasion of PTC using a tubulin tracker; however, no evidence of such an effect was observed. These results suggested that FKBP51 promotes migration and invasion through NF-κB-dependent EMT.
Collapse
Affiliation(s)
- Ying Gao
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China.,Department of Laboratory Medicine, Shandong Qianfoshan Hospital, Shandong University, Shandong, Jinan 250014, P.R. China
| | - Elham Elamin
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China
| | - Rongfang Zhou
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China
| | - Huili Yan
- Department of Medicine and Life Science, University of Jinan-Shandong Academy of Medical Sciences, Shandong, Jinan 250062, P.R. China
| | - Shuang Liu
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China
| | - Shengnan Hu
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China
| | - Jing Dong
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China
| | - Muyun Wei
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China
| | - Linying Sun
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China
| | - Yueran Zhao
- Department of Central Lab, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan 250021, P.R. China
| |
Collapse
|