1
|
Fan Y, Xiao H, Wu Y, Yan R, Li P, Ma H. Targeting TSPAN1 promotes ferroptosis in endometrial cancer cells, reduces energy metabolism and inhibits malignant behavior. Gene 2025; 962:149553. [PMID: 40339767 DOI: 10.1016/j.gene.2025.149553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND This research endeavored to explore the significance of tetraspanin 1 (TSPAN1) in endometrial cancer, with a particular focus on its influence on ferroptosis and glycolytic activity. EXPERIMENTAL PROCEDURE To achieve this, we established cell models that either overexpressed or silenced TSPAN1. We employed the CCK8 assay and EdU to assess the impact of TSPAN1 on cell proliferation. Additionally, scratch and Transwell assays were conducted to evaluate the cells' malignant biological behavior. The expression of ferroptosis-associated marker proteins was monitored. Further studies were conducted to investigate the effects of TSPAN1 on mitochondrial function, energy metabolism, and the cytoskeleton, and elucidated the interaction between TSPAN1 and SLC44A4. RESULTS Our findings indicated that the downregulation of TSPAN1 markedly suppressed cell proliferation and the incidence of malignant biological behaviors. Moreover, the suppression of TSPAN1 was found to enhance ferroptosis and to reduce glycolytic activity. CONCLUSIONS This study provides a comprehensive analysis of TSPAN1's role in endometrial cancer, shedding light on its potential as a key regulator of ferroptosis and glycolysis.
Collapse
Affiliation(s)
- Yang Fan
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Hongyan Xiao
- Pathology Department, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), China
| | - Yang Wu
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Ruihua Yan
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Peipei Li
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Hongyun Ma
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
2
|
Zhu W, Ni Q, Wang Z, Zhang R, Liu F, Chang H. MiR-101-3p targets the PI3K-AKT signaling pathway via Birc5 to inhibit invasion, proliferation, and epithelial-mesenchymal transition in hepatocellular carcinoma. Clin Exp Med 2025; 25:88. [PMID: 40106068 PMCID: PMC11923034 DOI: 10.1007/s10238-025-01622-1] [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: 12/17/2024] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate numerous genes in cells. Abnormal expression of miRNAs can lead to cancer. However, the roles and underlying mechanisms of miRNAs in hepatocellular carcinoma (HCC) are not fully understood. Using molecular biology techniques, we designed eukaryotic expression vectors with enhanced expression of miR-101-3p to transfect human hepatocellular carcinoma cell lines. Subsequent to this, cell cloning experiments, CCK8 assays, and Transwell migration experiments were executed to assess their impact on liver cancer cell proliferation and invasion. Dual-luciferase assays were employed to validate the molecular interaction between miR-101-3p and Birc5. Through rescue experiments aimed at manipulating the expression levels of Birc5, we scrutinized the influence of miR-101-3p on liver cancer cell proliferation and invasion. Furthermore, Western blot analysis was utilized to monitor alterations in the expression levels of E-cadherin, N-cadherin, and vimentin proteins within each cell group. In vivo investigations were conducted using nude mice implanted with hepatocellular carcinoma cells transfected with Birc5. Additionally, further exploration was carried out by combining this model with the PI3K/AKT pathway inhibitor miltefosine to elucidate its effects on tumor proliferation. In vitro functional analysis of miR-101-3p revealed that treatment of HCC cells with its corresponding mimic significantly inhibited cell proliferation, colony formation, invasion, and epithelial-mesenchymal transition. Additionally, miR-101-3p exerts its anti-tumor effects by targeting the shared gene Birc5. Experiments using nude mouse models demonstrate that Birc5 promotes tumor proliferation by phosphorylating the PI3K/AKT signaling pathway. Inhibiting the PI3K/AKT signaling pathway shows suppressive effects on liver cancer proliferation. MiR-101-3p plays crucial roles in inhibiting the proliferation, invasion and epithelial-mesenchymal transition of HCC cells by targeting Birc5 and downregulating the PI3K-AKT signaling pathway. These findings provide new insights for the molecular treatment of HCC.
Collapse
Affiliation(s)
- Wenyuan Zhu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qingqiang Ni
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Zhengjian Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Ruxuan Zhang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Fangfeng Liu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| | - Hong Chang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|
3
|
Abooshahab R, Zarkesh M, Sameni M, Akbarzadeh M, Skandari F, Hedayati M. Expression of TSPAN1 and its link to thyroid nodules: one step forward on the path to thyroid tumorigenesis biomarkers. BMC Cancer 2024; 24:1414. [PMID: 39548464 PMCID: PMC11568580 DOI: 10.1186/s12885-024-13176-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Thyroid cancer is ranked as the most common malignancy within the endocrine system and the seventh most prevalent cancer in women globally. Thyroid malignancies require evaluating biomarkers capable of distinguishing between them for accurate diagnosis. We examined both mRNA and protein levels of TSPAN1 in plasma and tissue samples from individuals with thyroid nodules to aid this endeavour. METHODS In this case-control study, TSPAN1 was assessed at both protein and mRNA levels in 90 subjects, including papillary thyroid cancer (PTC; N = 60), benign (N = 30), and healthy subjects (N = 26) using enzyme-linked immunosorbent assay (ELISA) and SYBR-Green Real-Time PCR, respectively. RESULTS TSPAN1 plasma levels were decreased in PTC and benign compared to healthy subjects (P = 0.002). TSPAN1 mRNA levels were also decremented in the tumoral compared to the paired normal tissues (P = 0.012); this drop was also observed in PTC patients compared to benign patients (P = 0.001). Further, TSPAN1 had an appropriate diagnostic value for detecting PTC patients from healthy plasma samples with a sensitivity of 76.7% and specificity of 65.4% at the cutoff value < 2.7 (ng/ml). CONCLUSION TSPAN1 levels are significantly reduced in patients with benign and PTC, demonstrating its potential value as a diagnostic biomarker. Additionally, the significant reduction in TSPAN1 mRNA expression within PTC tumor tissues may suggest its involvement in tumor progression and development. Further studies, including larger-scale validation studies and mechanistic investigations, are imperative to clarify the molecular mechanisms behind TSPAN1 and, ultimately, its clinical utility for treating thyroid disorders.
Collapse
MESH Headings
- Humans
- Tetraspanins/genetics
- Tetraspanins/metabolism
- Female
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/blood
- Male
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Thyroid Neoplasms/diagnosis
- Thyroid Neoplasms/blood
- Thyroid Nodule/genetics
- Thyroid Nodule/metabolism
- Thyroid Nodule/pathology
- Case-Control Studies
- Middle Aged
- Adult
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- Thyroid Cancer, Papillary/genetics
- Thyroid Cancer, Papillary/metabolism
- Thyroid Cancer, Papillary/pathology
- Thyroid Cancer, Papillary/blood
- Thyroid Cancer, Papillary/diagnosis
- Carcinoma, Papillary/genetics
- Carcinoma, Papillary/metabolism
- Carcinoma, Papillary/pathology
Collapse
Affiliation(s)
- Raziyeh Abooshahab
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box: 19395-4763, Tehran, Iran
- Curtin Medical School, Curtin University, Bentley, 6102, Australia
| | - Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box: 19395-4763, Tehran, Iran.
| | - Marzieh Sameni
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Zhino-Gene Research Services Co, Tehran, Iran
| | - Mahdi Akbarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box: 19395-4763, Tehran, Iran
| | - Fatemeh Skandari
- Department of Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box: 19395-4763, Tehran, Iran.
| |
Collapse
|
4
|
Sa L, Jiang J, Li Y, Huo Y, Zheng W, Zhang H, Zhang L, Wang T, Shan L. Diagnostic and prognostic significance of tetraspanin 6 and its role in facilitating glioma progression. Eur J Med Res 2024; 29:522. [PMID: 39472973 PMCID: PMC11520469 DOI: 10.1186/s40001-024-02119-5] [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: 02/06/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Several tetraspanin (TSPAN) proteins have been implicated in tumorigenesis and disease progression. However, the precise function of tetraspanin 6 (TSPAN6) in glioma remains unclear. METHODS Integrated raw data from the Tumor Genome Atlas (TCGA) and Gene Expression Profiling (GEO) databases were processed using R4.2.1. Bioinformatic methods were used to analyze the gene expression levels of TSPAN6 in both glioma and normal brain tissues, correlating them with clinical characteristics. Additionally, the predictive value of TSPAN6 in relation to the immune checkpoint blockade (ICB) therapy response was assessed. In vitro experiments were conducted to investigate the effects of TSPAN6 knockdown on glioma cell proliferation, migration, cell cycle regulation, and macrophage recruitment. RESULTS TSPAN6 expression was significantly upregulated in glioma tissues compared to normal tissues. Elevated TSPAN6 expression is strongly correlated with unfavorable clinical characteristics in gliomas. Bioinformatic analyses revealed a significant correlation between elevated TSPAN6 expression and reduced overall survival. Additionally, TSPAN6 was co-expressed with several immune checkpoint genes and revealed a prognostic value in the context of ICB therapy. Functional enrichment analysis revealed the involvement of TSPAN6 in cell-cycle regulation. Furthermore, TSPAN6 expression positively correlated with macrophage and neutrophil infiltration. In vitro experiments confirmed that the downregulation of TSPAN6 inhibited U251 cell proliferation, disrupted the cell cycle, diminished migratory capabilities, and reduced the recruitment of macrophages. CONCLUSION Our findings emphasize its potential as both a diagnostic and therapeutic target as well as a predictor of immune therapy response in gliomas.
Collapse
Affiliation(s)
- Longqi Sa
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Junwei Jiang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yifan Li
- The Fifth Cadet Regiment, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Huo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenjing Zheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Han Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Lingling Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Tao Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Lequn Shan
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
5
|
Li W, Yang LJ, Xiong YY, Li ZS, Li X, Wen Y. 4,5-Dimethoxycanthin-6-one Inhibits Glioblastoma Stem Cell and Tumor Growth by Inhibiting TSPAN1 Interaction with TM4SF1. Neurochem Res 2024; 49:2897-2909. [PMID: 39060768 DOI: 10.1007/s11064-024-04211-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
Glioblastoma stem cells (GSCs) have been implicated in the self-renewal and treatment resistance of glioblastoma (GBM). Our previous study found that 4,5-dimethoxycanthin-6-one has the potential to inhibit GBM cell proliferation. This current study aims to elucidate the molecular mechanism underlying the effects of 4,5-dimethoxycanthin-6-one in GBM development. The effect of 4,5-dimethoxycanthin-6-one on GSC formation and differentiation was explored in human GBM cell lines U251 and U87. Subsequently, 4,5-dimethoxycanthin-6-one binding to tetraspanin 1 (TSPAN1) / transmembrane 4 L six family member 1 (TM4SF1) was analyzed by molecular simulation docking. Co-immunoprecipitation (Co-IP) and immunofluorescence (IF) were used to assess the interactions between TSPAN1 and TM4SF1 in GSCs. Cell proliferation was detected by cell counting kit-8 (CCK-8) and colony formation assay. To evaluate cell migration, invasion and apoptosis, we employed wound healing assay, transwell and flow cytometry, respectively. Furthermore, subcutaneous xenograft tumor models in nude mice were constructed to evaluate the impact of 4,5-dimethoxycanthin-6-one on GSCs in vivo by examining tumor growth and histological characteristics. 4,5-Dimethoxycanthin-6-one inhibited GSC formation and promoted stem cell differentiation in a concentration-dependent manner. Molecular docking models of 4,5-dimethoxycanthin-6-one with TM4SF1 and TSPAN1 were constructed. Then, the interaction between TSPAN1 and TM4SF1 in GSC was clarified. Moreover, 4,5-dimethoxycanthin-6-one significantly inhibited the expressions of TM4SF1 and TSPAN1 in vitro and in vivo. Overexpression of TSPAN1 partially reversed the inhibitory effects of 4,5-dimethoxycanthin-6-one on GSC formation, proliferation, migration and invasion. 4,5-Dimethoxycanthin-6-one inhibited GBM progression by inhibiting TSPAN1/TM4SF1 axis. 4,5-Dimethoxycanthin-6-one might be a novel and effective drug for the treatment of GBM.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Li-Jian Yang
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yuan-Yuan Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zeng-Shi Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xi Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yi Wen
- Department of Imaging, Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China.
- Department of Imaging, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
6
|
Liu X, Zhang J, Zhao J, Cheng Y, Jiang D. TSPAN1 overexpression as an indicator of poor prognosis in estrogen receptor-positive breast cancer. Transl Cancer Res 2024; 13:4159-4171. [PMID: 39262478 PMCID: PMC11385530 DOI: 10.21037/tcr-24-409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/25/2024] [Indexed: 09/13/2024]
Abstract
Background Tetraspanin 1 (TSPAN1) is a newly discovered protein of the tetrameric protein family encoded by the TSPAN1 gene localized in the 1p34 chromosome region. TSPAN1 has been shown to be associated with various malignancies. In this study, we aimed to investigate the prognostic significance of TSPAN1 in breast cancer. Estrogen receptor-positive (ER+) breast cancer is the largest breast cancer subgroup, and ER-targeted therapies have significantly prolonged survival and improved symptoms in advanced breast cancer. TSPAN1 overexpression was found to be associated with a poor prognosis in ER+ breast cancer. Methods We analyzed the expression of TSPAN1 in breast cancer tissues and cell lines using western blotting and quantitative reverse transcription polymerase chain reaction (RT-qPCR). Results TSPAN1 expression was higher in breast cancer cells as compared with normal breast tissue. There was a significant association between a high TSPAN1 level and a low survival rate. Inhibition of TSPAN1 significantly reduced the proliferation and invasion of BT474 cells both in vitro and in vivo. The downregulation of TSPAN1 in breast cancer cells significantly reduced the levels of p-mitogen-activated protein kinase 1 (MEK1) (S298) and p-extracellular signal-regulating kinase (ERK) 1/2. Conclusions TSPAN1 modulates downstream extracellular matrix (ECM) receptor signaling cascades and promotes cellular proliferation and invasion in breast cancer. TSPAN1 inhibition may be a potential new treatment strategy for breast cancer.
Collapse
Affiliation(s)
- Xiangjuan Liu
- Department of Breast Surgery, Yantaishan Hospital, Yantai, China
| | - Jiahong Zhang
- Department of First General Surgery, Yantai Penglai People's Hospital, Yantai, China
| | - Jun Zhao
- Department of Oncology, Yantai Penglai People's Hospital, Yantai, China
| | - Yan Cheng
- Department of Breast Surgery, Yantaishan Hospital, Yantai, China
| | - Dandan Jiang
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Yang Y, Wang J, Wan J, Cheng Q, Cheng Z, Zhou X, Wang O, Shi K, Wang L, Wang B, Zhu X, Chen J, Feng D, Liu Y, Jahan-Mihan Y, Haddock AN, Edenfield BH, Peng G, Hohenstein JD, McCabe CE, O'Brien DR, Wang C, Ilyas SI, Jiang L, Torbenson MS, Wang H, Nakhleh RE, Shi X, Wang Y, Bi Y, Gores GJ, Patel T, Ji B. PTEN deficiency induces an extrahepatic cholangitis-cholangiocarcinoma continuum via aurora kinase A in mice. J Hepatol 2024; 81:120-134. [PMID: 38428643 PMCID: PMC11259013 DOI: 10.1016/j.jhep.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/09/2024] [Accepted: 02/18/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND & AIMS The PTEN-AKT pathway is frequently altered in extrahepatic cholangiocarcinoma (eCCA). We aimed to evaluate the role of PTEN in the pathogenesis of eCCA and identify novel therapeutic targets for this disease. METHODS The Pten gene was genetically deleted using the Cre-loxp system in biliary epithelial cells. The pathologies were evaluated both macroscopically and histologically. The characteristics were further analyzed by immunohistochemistry, reverse-transcription PCR, cell culture, and RNA sequencing. Some features were compared to those in human eCCA samples. Further mechanistic studies utilized the conditional knockout of Trp53 and Aurora kinase A (Aurka) genes. We also tested the effectiveness of an Aurka inhibitor. RESULTS We observed that genetic deletion of the Pten gene in the extrahepatic biliary epithelium and peri-ductal glands initiated sclerosing cholangitis-like lesions in mice, resulting in enlarged and distorted extrahepatic bile ducts in mice as early as 1 month after birth. Histologically, these lesions exhibited increased epithelial proliferation, inflammatory cell infiltration, and fibrosis. With aging, the lesions progressed from low-grade dysplasia to invasive carcinoma. Trp53 inactivation further accelerated disease progression, potentially by downregulating senescence. Further mechanistic studies showed that both human and mouse eCCA showed high expression of AURKA. Notably, the genetic deletion of Aurka completely eliminated Pten deficiency-induced extrahepatic bile duct lesions. Furthermore, pharmacological inhibition of Aurka alleviated disease progression. CONCLUSIONS Pten deficiency in extrahepatic cholangiocytes and peribiliary glands led to a cholangitis-to-cholangiocarcinoma continuum that was dependent on Aurka. These findings offer new insights into preventive and therapeutic interventions for extrahepatic CCA. IMPACT AND IMPLICATIONS The aberrant PTEN-PI3K-AKT signaling pathway is commonly observed in human extrahepatic cholangiocarcinoma (eCCA), a disease with a poor prognosis. In our study, we developed a mouse model mimicking cholangitis to eCCA progression by conditionally deleting the Pten gene via Pdx1-Cre in epithelial cells and peribiliary glands of the extrahepatic biliary duct. The conditional Pten deletion in these cells led to cholangitis, which gradually advanced to dysplasia, ultimately resulting in eCCA. The loss of Pten heightened Akt signaling, cell proliferation, inflammation, fibrosis, DNA damage, epigenetic signaling, epithelial-mesenchymal transition, cell dysplasia, and cellular senescence. Genetic deletion or pharmacological inhibition of Aurka successfully halted disease progression. This model will be valuable for testing novel therapies and unraveling the mechanisms of eCCA tumorigenesis.
Collapse
Affiliation(s)
- Yan Yang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA; Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jiale Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jianhua Wan
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Qianqian Cheng
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zenong Cheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xueli Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Oliver Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Kelvin Shi
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Lingxiang Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Bin Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Xiaohui Zhu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jiaxiang Chen
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Dongfeng Feng
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Yang Liu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Ashley N Haddock
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Guang Peng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Chantal E McCabe
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel R O'Brien
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Liuyan Jiang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Michael S Torbenson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Raouf E Nakhleh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Xuemei Shi
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Ying Wang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Tushar Patel
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, USA
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA.
| |
Collapse
|
8
|
Zhang H, Song Q, Shang K, Li Y, Jiang L, Yang L. Tspan protein family: focusing on the occurrence, progression, and treatment of cancer. Cell Death Discov 2024; 10:187. [PMID: 38649381 PMCID: PMC11035590 DOI: 10.1038/s41420-024-01961-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
The Tetraspanins (Tspan) protein family, also known as the tetraspanin family, contains 33 family members that interact with other protein molecules such as integrins, adhesion molecules, and T cell receptors by forming dimers or heterodimers. The Tspan protein family regulates cell proliferation, cell cycle, invasion, migration, apoptosis, autophagy, tissue differentiation, and immune response. More and more studies have shown that Tspan proteins are involved in tumorigenesis, epithelial-mesenchymal transition, thrombosis, tumor stem cell, and exosome signaling. Some drugs and microRNAs can inhibit Tspan proteins, thus providing new strategies for tumor therapy. An in-depth understanding of the functions and regulatory mechanisms of the Tspan protein family, which can promote or inhibit tumor development, will provide new strategies for targeted interventions in the future.
Collapse
Affiliation(s)
- Huhu Zhang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, 266071, China
| | - Qinghang Song
- Health Science Center, Qingdao University, Qingdao, 266071, China
| | - Kaiwen Shang
- Health Science Center, Qingdao University, Qingdao, 266071, China
| | - Ya Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, 266071, China
| | - Liangqian Jiang
- Department of Medical Genetics, Linyi People's Hospital, Linyi, China
| | - Lina Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
9
|
Wang MD, Li HT, Peng LX, Mei Y, Zheng LS, Li CZ, Meng DF, Lang YH, Xu L, Peng XS, Liu ZJ, Xie DH, Guo LL, Ma MG, Ding LY, Huang BJ, Cao Y, Qian CN. TSPAN1 inhibits metastasis of nasopharyngeal carcinoma via suppressing NF-kB signaling. Cancer Gene Ther 2024; 31:454-463. [PMID: 38135697 DOI: 10.1038/s41417-023-00716-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
Nasopharyngeal carcinoma (NPC) originates in the epithelial cells of the nasopharynx and is a common malignant tumor in southern China and Southeast Asia. Metastasis of NPC remains the main cause of death for NPC patients even though the tumor is sensitive to radiotherapy and chemotherapy. Here, we found that the transmembrane protein tetraspanin1 (TSPAN1) potently inhibited the in vitro migration and invasion, as well as, the in vivo metastasis of NPC cells via interacting with the IKBB protein. In addition, TSPAN1 was essential in preventing the overactivation of the NF-kB pathway in TSPAN1 overexpressing NPC cells. Furthermore, reduced TSPAN1 expression was associated with NPC metastasis and the poor prognosis of NPC patients. These results uncovered the suppressive role of TSPAN1 against NF-kB signaling in NPC cells for preventing NPC metastasis. Its therapeutic value warrants further investigation.
Collapse
Affiliation(s)
- Ming-Dian Wang
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Hui-Ting Li
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Li-Xia Peng
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yan Mei
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, P. R. China
| | - Li-Sheng Zheng
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, P. R. China
| | - Chang-Zhi Li
- Medical School, Pingdingshan University, Pingdingshan, Henan Province, 467021, P. R. China
| | - Dong-Fang Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, P. R. China
| | - Yan-Hong Lang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Liang Xu
- Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, P. R. China
| | - Xing-Si Peng
- Department of radiation oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, P. R. China
| | - Zhi-Jie Liu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Radiotherapy, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China
| | - De-Huan Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ling-Ling Guo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Mao-Guang Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
| | - Liu-Yan Ding
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Bi-Jun Huang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yun Cao
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China.
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- Guangzhou Concord Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
10
|
Shao S, Bu Z, Xiang J, Liu J, Tan R, Sun H, Hu Y, Wang Y. The role of Tetraspanins in digestive system tumor development: update and emerging evidence. Front Cell Dev Biol 2024; 12:1343894. [PMID: 38389703 PMCID: PMC10882080 DOI: 10.3389/fcell.2024.1343894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Digestive system malignancies, including cancers of the esophagus, pancreas, stomach, liver, and colorectum, are the leading causes of cancer-related deaths worldwide due to their high morbidity and poor prognosis. The lack of effective early diagnosis methods is a significant factor contributing to the poor prognosis for these malignancies. Tetraspanins (Tspans) are a superfamily of 4-transmembrane proteins (TM4SF), classified as low-molecular-weight glycoproteins, with 33 Tspan family members identified in humans to date. They interact with other membrane proteins or TM4SF members to form a functional platform on the cytoplasmic membrane called Tspan-enriched microdomain and serve multiple functions including cell adhesion, migration, propagation and signal transduction. In this review, we summarize the various roles of Tspans in the progression of digestive system tumors and the underlying molecular mechanisms in recent years. Generally, the expression of CD9, CD151, Tspan1, Tspan5, Tspan8, Tspan12, Tspan15, and Tspan31 are upregulated, facilitating the migration and invasion of digestive system cancer cells. Conversely, Tspan7, CD82, CD63, Tspan7, and Tspan9 are downregulated, suppressing digestive system tumor cell metastasis. Furthermore, the connection between Tspans and the metastasis of malignant bone tumors is reviewed. We also summarize the potential role of Tspans as novel immunotherapy targets and as an approach to overcome drug resistance. Finally, we discuss the potential clinical value and therapeutic targets of Tspans in the treatments of digestive system malignancies and provide some guidance for future research.
Collapse
Affiliation(s)
- Shijie Shao
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhen Bu
- Department of General Surgery, Xinyi People's Hospital, Xinyi, China
| | - Jinghua Xiang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiachen Liu
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Rui Tan
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Han Sun
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yuanwen Hu
- Department of Gastroenterology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yimin Wang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
11
|
Zhang Z, Zhang T, Zhang R, Zhang Z, Tan S. Migrasomes and tetraspanins in hepatocellular carcinoma: current status and future prospects. Future Sci OA 2023; 9:FSO890. [PMID: 37752917 PMCID: PMC10518826 DOI: 10.2144/fsoa-2023-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/24/2023] [Indexed: 09/28/2023] Open
Abstract
In recent years, many studies have attempted to clarify the formation, structure and biological function of migrasomes, which are defined as specialized organelles formed by the tips and intersections of Retraction Fibrils during cell migration. It has confirmed that migrasomes were involved in various critical biological processes and diseases, and has became a new research hotspot. In this paper, we reviewed the formation and biological functions of migrasomes, explored the relationship between migrasomes, tetraspanins and hepatocellular carcinoma and discussed the potential applications of migrasomes in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhongqi Zhang
- Guangxi Key Laboratory of Environmental Exposomics & Entire Lifecycle Health, Guilin Medical University, Guilin, 541004, Guangxi, China
- Department of Epidemiology & Health Statistics, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Tianmiao Zhang
- Guangxi Key Laboratory of Environmental Exposomics & Entire Lifecycle Health, Guilin Medical University, Guilin, 541004, Guangxi, China
- Department of Epidemiology & Health Statistics, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Rongcheng Zhang
- Guangxi Key Laboratory of Environmental Exposomics & Entire Lifecycle Health, Guilin Medical University, Guilin, 541004, Guangxi, China
- Department of Epidemiology & Health Statistics, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Zhengbao Zhang
- Guangxi Key Laboratory of Environmental Exposomics & Entire Lifecycle Health, Guilin Medical University, Guilin, 541004, Guangxi, China
- Department of Epidemiology & Health Statistics, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Shengkui Tan
- Guangxi Key Laboratory of Environmental Exposomics & Entire Lifecycle Health, Guilin Medical University, Guilin, 541004, Guangxi, China
- Department of Epidemiology & Health Statistics, Guilin Medical University, Guilin, 541004, Guangxi, China
| |
Collapse
|
12
|
Tang HY, Lin M, Liang YQ, Wang JH, Yi HG, Yang M. Tspan5 promotes the EMT process to regulate the syncytialization of trophoblast cells by activating Notch signalling. ZYGOTE 2023; 31:498-506. [PMID: 37485669 DOI: 10.1017/s0967199423000369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Placental trophoblastic cells play important roles in placental development and fetal health. However, the mechanism of trophoblastic cell fusion is still not entirely clear. The level of Tspan5 in the embryo culture medium was detected using enzyme-linked immunosorbent assay (ELISA). Fusion of BeWo cells was observed by immunofluorescence. Cell fusion-related factors and EMT-related factors were identified by qRT-PCR and western blotting. Notch protein repressor DAPT was used to verify the role of Tspan5 in BeWo cells. The expression of Tspan5 was significantly increased in embryo culture medium. The fusion of BeWo cells was observed after treatment with forskolin (FSK). Cell fusion-related factors (i.e. β-hCG and syncytin 1/2) and Tspan5 were significantly increased after FSK treatment. In addition, FSK treatment promoted EMT-related protein expression in BeWo cells. Knockdown of Tspan5 inhibited cell fusion and EMT-related protein levels. Notch-1 and Jagged-1 protein levels were significantly upregulated, and the EMT process was activated by overexpression of Tspan5 in FSK-treated BeWo cells. Interestingly, blocking the Notch pathway by the repressor DAPT had the opposite results. These results indicated that Tspan5 could promote the EMT process by activating the Notch pathway, thereby causing cell fusion. These findings contribute to a better understanding of trophoblast cell syncytialization and embryonic development. Tspan5 may be used as a therapeutic target for normal placental development.
Collapse
Affiliation(s)
- Hai-Yu Tang
- Department of Reproductive Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou514000, Guangdong Province, China
| | - Mei Lin
- Department of Reproductive Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou514000, Guangdong Province, China
| | - Yong-Qian Liang
- Department of Reproductive Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou514000, Guangdong Province, China
| | - Jin-Hua Wang
- Department of Reproductive Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou514000, Guangdong Province, China
| | - Hong-Gan Yi
- Department of Reproductive Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou514000, Guangdong Province, China
| | - Man Yang
- Department of Reproductive Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou514000, Guangdong Province, China
| |
Collapse
|
13
|
Qian Y, Feng D, Wang J, Wei W, Wei Q, Han P, Yang L. Establishment of cancer-associated fibroblasts-related subtypes and prognostic index for prostate cancer through single-cell and bulk RNA transcriptome. Sci Rep 2023; 13:9016. [PMID: 37270661 DOI: 10.1038/s41598-023-36125-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/30/2023] [Indexed: 06/05/2023] Open
Abstract
Current evidence indicate that cancer-associated fibroblasts (CAFs) play an important role in prostate cancer (PCa) development and progression. In this study, we identified CAF-related molecular subtypes and prognostic index for PCa patients undergoing radical prostatectomy through integrating single-cell and bulk RNA sequencing data. We completed analyses using software R 3.6.3 and its suitable packages. Through single-cell and bulk RNA sequencing analysis, NDRG2, TSPAN1, PTN, APOE, OR51E2, P4HB, STEAP1 and ABCC4 were used to construct molecular subtypes and CAF-related gene prognostic index (CRGPI). These genes could clearly divide the PCa patients into two subtypes in TCGA database and the BCR risk of subtype 1 was 13.27 times higher than that of subtype 2 with statistical significance. Similar results were observed in MSKCC2010 and GSE46602 cohorts. In addtion, the molucular subtypes were the independent risk factor of PCa patients. We orchestrated CRGPI based on the above genes and divided 430 PCa patients in TCGA database into high- and low- risk groups according to the median value of this score. We found that high-risk group had significant higher risk of BCR than low-risk group (HR: 5.45). For functional analysis, protein secretion was highly enriched in subtype 2 while snare interactions in vesicular transport was highly enriched in subtype 1. In terms of tumor heterogeneity and stemness, subtype 1 showd higher levels of TMB than subtype 2. In addition, subtype 1 had significant higher activated dendritic cell score than subtype 2. Based on eight CAF-related genes, we developed two prognostic subtypes and constructed a gene prognostic index, which could predict the prognosis of PCa patients very well.
Collapse
Affiliation(s)
- Youliang Qian
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, People's Republic of China
- Department of Urology, Chengdu Second People's Hospital, Chengdu, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, People's Republic of China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, People's Republic of China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, People's Republic of China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, People's Republic of China
| | - Ping Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, People's Republic of China.
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
14
|
Zhou Z, Yang Z, Zhou L, Yang M, He S. The versatile roles of testrapanins in cancer from intracellular signaling to cell-cell communication: cell membrane proteins without ligands. Cell Biosci 2023; 13:59. [PMID: 36941633 PMCID: PMC10025802 DOI: 10.1186/s13578-023-00995-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
The tetraspanins (TSPANs) are a family of four-transmembrane proteins with 33 members in mammals. They are variably expressed on the cell surface, various intracellular organelles and vesicles in nearly all cell types. Different from the majority of cell membrane proteins, TSPANs do not have natural ligands. TSPANs typically organize laterally with other membrane proteins to form tetraspanin-enriched microdomains (TEMs) to influence cell adhesion, migration, invasion, survival and induce downstream signaling. Emerging evidence shows that TSPANs can regulate not only cancer cell growth, metastasis, stemness, drug resistance, but also biogenesis of extracellular vesicles (exosomes and migrasomes), and immunomicroenvironment. This review summarizes recent studies that have shown the versatile function of TSPANs in cancer development and progression, or the molecular mechanism of TSPANs. These findings support the potential of TSPANs as novel therapeutic targets against cancer.
Collapse
Affiliation(s)
- Zhihang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, SAR, People's Republic of China.
| | - Zihan Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, SAR, People's Republic of China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, China
| | - Li Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, SAR, People's Republic of China
| | - Mengsu Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, SAR, People's Republic of China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, China
| | - Song He
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
15
|
Liu Y, Wang CL, Pang ZQ, Gao K, Shen LK, Xu WH, Ren MH. Endostatin 33 Peptide Is a Deintegrin α6β1 Agent That Exerts Antitumor Activity by Inhibiting the PI3K-Akt Signaling Pathway in Prostate Cancer. J Clin Med 2023; 12:1861. [PMID: 36902648 PMCID: PMC10003382 DOI: 10.3390/jcm12051861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the leading cause of death in men and has poor therapeutic outcomes. METHODS A novel endostatin 33 peptide was synthesized by adding a specific QRD sequence on the basis of the endostatin 30 peptide (PEP06) with antitumor activity. Then, bioinformatic analysis and subsequent experiments were performed to validate the antitumor function of this endostatin 33 peptide. RESULTS We found that the 33 polypeptides significantly inhibited growth, invasion and metastasis and promoted the apoptosis of PCa in vivo or vitro, which is more significant than PEP06 under the same conditions. According to 489 cases from the TCGA data portal, the α6β1 high expression group was closely associated with the poor prognosis (Gleason score, pathological N stage, etc.) of PCa and was mainly enriched in the PI3K-Akt pathway. Subsequently, we demonstrated that endostatin 33 peptide can down-regulate the PI3K-Akt pathway via the targeted inhibition of α6β1, thereby inhibiting the epithelial-mesenchymal transition and matrix metalloproteinase in C42 cell lines. CONCLUSION The endostatin 33 peptide can exert antitumor effects by inhibiting the PI3K-Akt pathway, especially in tumors with a high expression of the integrin α6β1 subtype, such as prostate cancer. Therefore, our study will provide a new method and theoretical basis for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Yang Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Chang-Lin Wang
- Department of Urology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Zhong-Qi Pang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ke Gao
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lin-Kun Shen
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wan-Hai Xu
- Department of Urology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ming-Hua Ren
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
16
|
Ou Y, Chen R, Qian Q, Cui N, Miao Q, Tang R, You Z, Ma X, Wang Q. The immunological characteristics of TSPAN1 expressing B cells in autoimmune hepatitis. Front Immunol 2022; 13:1076594. [PMID: 36591302 PMCID: PMC9797502 DOI: 10.3389/fimmu.2022.1076594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Background and aims Tetraspanin proteins are closely related to the functional changes of B cells, including antigen presentation, production of cytokines, and transduction. We aim to explore the potential role of Tetraspanin 1 (TSPAN1) in the biological activities of B cells in AIH. Methods and results Herein, this study found that numbers of cells expressing TSPAN1 were significantly increased in AIH patients compared to PBC, chronic hepatitis B, and healthy control (P < 0.0001). Moreover, there was a positive correlation between numbers of TSPAN1+ cells and AIH disease severity (P < 0.0001). Immunofluorescence staining further confirmed that TSPAN1 was primarily expressed on CD19+ B cells. Flow-cytometric analysis showed that TSPAN1+ B cells secreted more inflammatory cytokines and expressed higher level of CD86 than TSPAN1- B cells. Furthermore, compared with TSAPN1- cells, the expression of CXCR3 on TSPAN1+ cells was also higher. Meanwhile, CXCL10, the ligand of CXCR3, was significantly elevated in the liver of AIH (P < 0.01) and had positive correlation with the quantities of TSPAN1 (P < 0.05). Interestingly, the numbers of TSPAN1+ B cells were decreased in AIH patients after immunosuppressive therapy. Conclusions TSPAN1+ B cells in the liver may promote the progression of AIH via secreting cytokines and presenting antigens. The chemotactic movement of TSPAN1+ B cells toward the liver of AIH was possibly due to CXCR3 - CXCL10 interaction.
Collapse
Affiliation(s)
- Yiyan Ou
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Ruiling Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Qiwei Qian
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Nana Cui
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Miao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Ruqi Tang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengrui You
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Qixia Wang, ; Xiong Ma, ; Zhengrui You,
| | - Xiong Ma
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China,Division of Infectious Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Qixia Wang, ; Xiong Ma, ; Zhengrui You,
| | - Qixia Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China,Division of Infectious Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Qixia Wang, ; Xiong Ma, ; Zhengrui You,
| |
Collapse
|
17
|
Li J, Gao H, Chen B, Li L, Wang Q, Gao Z. lncRNA DARS-AS1 Modulates TSPAN1-Mediated ITGA2 Hypomethylation by Interaction with miR-194-5p Thus Promoting Ovarian Cancer Progression. Stem Cells Int 2022; 2022:4041550. [PMID: 36187230 PMCID: PMC9522497 DOI: 10.1155/2022/4041550] [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: 07/28/2022] [Revised: 08/27/2022] [Accepted: 09/01/2022] [Indexed: 11/18/2022] Open
Abstract
Objective Ovarian cancer (OC) is usually called the "silent killer" due to its asymptomatic characteristics until advanced stages, thus being a significant threat to female health worldwide. In this work, we characterized an oncogenic DARS-AS1 role in OC. Methods The aggressiveness behaviors of the OC cell model were examined by CCK-8 assay, transwell invasion assay, flow cytometry, and immunoblotting analysis of apoptosis-related proteins. Interactions of miR-194-5p with lncRNA DARS-AS1 or TSPAN1 and of TSPAN1 with ITGA2 were validated by using a luciferase activity assay and chromatin immunoprecipitation (ChIP) assay. Results The OC cell model exhibited overexpressed lncRNA DARS-AS1 compared to normal cells. lncRNA DARS-AS1 knockdown led to reduced OC cell growth and metastasis while inducing the apoptosis in the OC cell model. lncRNA DARS-AS1 positively regulated TSPAN1 expression by binding with miR-194-5p and TSPAN1-mediated ITGA2 hypomethylation in OC cells. Further rescue function studies demonstrated that lncRNA DARS-AS1 affected OC cell viability, migration, invasion, and apoptosis ability by modulating miR-194-5p and TSPAN1 expressions. Conclusion Our work demonstrates that lncRNA DARS-AS1 promotes OC progression by modulating TSPAN1 and ITGA2 hypomethylation by binding with miR-194-5p.
Collapse
Affiliation(s)
- Jun Li
- Gynecologic Oncology Department, Xinxiang Central Hospital, China
- Xinxiang Medical University, The Fourth Clinical University, China
| | - Haoyu Gao
- Xinxiang Medical University, School of Basic Medical Sciences, China
| | - Beibei Chen
- Gynecologic Oncology Department, Xinxiang Central Hospital, China
| | - Li Li
- Gynecologic Oncology Department, Xinxiang Central Hospital, China
| | - Qianqing Wang
- Gynecologic Oncology Department, Xinxiang Central Hospital, China
| | - Zhihui Gao
- Gynecologic Oncology Department, Xinxiang Central Hospital, China
| |
Collapse
|
18
|
Takashima Y, Komatsu S, Ohashi T, Kiuchi J, Kamiya H, Shimizu H, Arita T, Konishi H, Shiozaki A, Kubota T, Okamoto K, Fujiwara H, Tsuda H, Otsuji E. Overexpression of Tetraspanin31 contributes to malignant potential and poor outcomes in gastric cancer. Cancer Sci 2022; 113:1984-1998. [PMID: 35307915 PMCID: PMC9207375 DOI: 10.1111/cas.15342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 12/24/2022] Open
Abstract
Tetraspanin has important functions in many cancers by aggregating with various proteins that interact with intracellular signaling proteins. The molecular function of Tetraspanin31 (TSPAN31), located in the 12q14 amplified region in various cancers, remains unclear in gastric cancer (GC). We tested whether TSPAN31 acts as a cancer‐promoting gene through its activation or overexpression in GC. We analyzed seven GC cell lines and 189 primary tumors, which were curatively resected in our hospital between 2011 and 2013. Overexpression of the TSPAN31 protein was frequently detected in three GC cell lines (42.9%) and 62 primary GC specimens (32.8%). Overexpression of TSPAN31 was significantly correlated with lymphatic invasion, venous invasion, more advanced pT and pN stages, and a higher recurrence rate. Moreover, TSPAN31 positivity was an independent factor predicting worse patient outcomes (p = 0.0283, hazard ratio 3.97). Ectopic overexpression of TSPAN31 facilitated cell proliferation of GC cells, and knockdown of TSPAN31 inhibited cell proliferation, migration, invasion, and epithelial–mesenchymal transition of GC cells through the PI3K‐Akt pathway and increased cell apoptosis in a TP53 mutation‐independent manner. In vivo analysis also revealed knockdown of TSPAN31 suppressed tumor progression. In addition, knockdown of TSPAN31 improved chemosensitivity to cisplatin through the suppression of ABCC2. These findings suggest that TSPAN31 plays a crucial role in tumor‐malignant potential through overexpression, highlighting its utility as a prognostic factor and a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Yusuke Takashima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hajime Kamiya
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitoshi Tsuda
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan.,Department of Basic Pathology, National Defense Medical College, Tokorozawa, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
19
|
Shao S, Piao L, Wang J, Guo L, Wang J, Wang L, Tong L, Yuan X, Han X, Fang S, Zhu J, Wang Y. Tspan9 Induces EMT and Promotes Osteosarcoma Metastasis via Activating FAK-Ras-ERK1/2 Pathway. Front Oncol 2022; 12:774988. [PMID: 35280793 PMCID: PMC8906905 DOI: 10.3389/fonc.2022.774988] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/21/2022] [Indexed: 01/14/2023] Open
Abstract
Object At present, there are few effective treatment options available to patients suffering from osteosarcoma (OS). Clarifying the signaling pathways that govern OS oncogenesis may highlight novel approaches to treating this deadly form of cancer. Recent experimental evidence suggests that the transmembrane protein tetraspanin-9 (Tspan9) plays a role in tumor development. This study was thus formulated to assess the molecular role of Tspan9 as a regulator of OS cell metastasis. Methods Gene expression in OS cell lines was evaluated via qRT-PCR, while CCK-8, colony formation, Transwell, and wound healing assays were used to explore the in vitro proliferative, invasive, and migratory activities of OS cells. The relationship between Tspan9 and in vivo OS cell metastasis was assessed by injecting these cells into the tail vein of nude mice. Interactions between the Tspan9 and integrin β1 proteins were explored through mass spectrometric and co-immunoprecipitation, and Western blotting to assess the functional mechanisms whereby Tspan9 shapes OS pathogenesis. Results Both primary OS tumors and OS cell lines commonly exhibited Tspan9 upregulation, and the knockdown of this tetraspanin suppressed the migration, invasion, and epithelial-mesenchymal transition (EMT) activity in OS cells, whereas Tspan9 overexpression resulted in opposite phenotypes. Tumor lung metastasis were significantly impaired in mice implanted with HOS cells in which Tspan9 was downregulated as compared to mice implanted with control HOS cells. Tspan9 was also found to interact with β1 integrin and to contribute to OS metastasis via the amplification of integrin-mediated downstream FAK/Ras/ERK1/2 signaling pathway. Conclusion These data suggest that Tspan9 can serve as a promising therapeutic target in OS.
Collapse
Affiliation(s)
- Shijie Shao
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lianhua Piao
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou, China
| | - Jiangsong Wang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liwei Guo
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiawen Wang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Luhui Wang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lei Tong
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaofeng Yuan
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xu Han
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Sheng Fang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Junke Zhu
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yimin Wang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
20
|
Deng S, Zhang L, Li J, Jin Y, Wang J. Activation of the PI3K-AKT signaling pathway by SPARC contributes to the malignant phenotype of cholangiocarcinoma cells. Tissue Cell 2022; 76:101756. [PMID: 35217388 DOI: 10.1016/j.tice.2022.101756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/22/2022]
Abstract
Cholangiocarcinoma (CCA) is a primary biliary epithelium malignancy with limited therapies, poor prognosis and high mortality rate. Nowadays, the molecular mechanisms of CCA remain elusive. SPARC has been proposed to be highly expressed in clinical CCA tissues, but few studies has been elucidated its functions in CCA. In the current study, we aimed to investigate the functional role of SPARC in the progression of CCA. In this study, a significantly increased expression of SPARC was observed in CCA tissues and cells. Knockdown of SPARC by RNA interference significantly impeded the proliferation of CCA cells. Moreover, SPARC silencing hampered the migration and invasion of CCA cells by inhibiting EMT. In parallel, overexpression of SPARC in RBE cells had the opposite effects. Mechanically, SPARC promoted proliferation, migration, invasion, and EMT of CCA cells in vitro via activating the PI3K-AKT signaling. Overall, our integrated analysis revealed that SPARC plays a crucial role in CCA progression via the PI3K-AKT signaling pathway, which suggests that targeting SPARC might represent a promising approach for improving CCA patient's clinical outcome.
Collapse
Affiliation(s)
- Shikang Deng
- Medical School, Kunming University of Science and Technology, Kumming, Yunnan, China; Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China
| | - Li Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China
| | - Jiao Li
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China
| | - Yan Jin
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China
| | - Junfeng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China.
| |
Collapse
|
21
|
Garcia-Mayea Y, Mir C, Carballo L, Sánchez-García A, Bataller M, LLeonart ME. TSPAN1, a novel tetraspanin member highly involved in carcinogenesis and chemoresistance. Biochim Biophys Acta Rev Cancer 2021; 1877:188674. [PMID: 34979155 DOI: 10.1016/j.bbcan.2021.188674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022]
Abstract
The tetraspanin (TSPAN) family constitutes a poorly explored family of membrane receptors involved in various physiological processes, with relevant roles in anchoring multiple proteins, acting as scaffolding proteins, and cell signaling. Recent studies have increasingly demonstrated the involvement of TSPANs in cancer. In particular, tetraspanin 1 (also known as TSPAN1, NET-1, TM4C, C4.8 or GEF) has been implicated in cell survival, proliferation and invasion. Recently, our laboratory revealed a key role of TSPAN1 in the acquired resistance of tumor cells to conventional chemotherapy (e.g., cisplatin). In this review, we summarize and discuss the latest research on the physiological mechanisms of TSPANs in cancer and, in particular, on TSPAN1 regulating resistance to chemotherapy. A model of TSPAN1 action is proposed, and the potential of targeting TSPAN1 in anticancer therapeutic strategies is discussed.
Collapse
Affiliation(s)
- Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Laia Carballo
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Almudena Sánchez-García
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marina Bataller
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Matilde E LLeonart
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Spanish Biomedical Research Network Center in Oncology, CIBERONC, Spain.
| |
Collapse
|
22
|
Zhou S, Qu KL, Li JA, Chen SL, Zhang YG, Zhu C, Jin H, Wang Y, Pang Q, Liu HC. YY1 activates EMI2 and promotes the progression of cholangiocarcinoma through the PI3K/Akt signaling axis. Cancer Cell Int 2021; 21:699. [PMID: 34933678 PMCID: PMC8693494 DOI: 10.1186/s12935-021-02328-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is one of the deadliest cancers of the digestive tract. The prognosis of CCA is poor and the 5-year survival rate is low. Bioinformatic analysis showed that early mitotic inhibitor 2 (EMI2) was overexpressed in CCA but the underlying mechanism is not known. METHODS The data on bile duct carcinoma from TCGA and GEO databases were used to detect the expression of EMI2. The transcription factors of EMI2 were predicted using JASPAR and PROMO databases. Among the predicted transcription factors, YY1 has been rarely reported in cholangiocarcinoma, and was verified using the luciferase reporter gene assay. RT-PCR was performed to predict the downstream pathway of EMI2, and PI3K/Akt was suspected to be associated with it. Subsequently, in vivo and in vitro experiments were conducted to verify the effects of silencing and overexpressing EMI2 and YY1 on the proliferation, invasion, and metastasis of the bile duct cancer cells. RESULTS EMI2 was highly expressed in CCA. Silencing EMI2 inhibited the proliferation, invasion, and migration of CCA cells, arrested cell cycle in the G1 phase, and promoted of apoptosis. The luciferase reporter gene assay showed that YY1 bound to the promoter region of EMI2, and after silencing YY1, the expression of EMI2 decreased and the progression of CCA was inhibited. Moreover, key proteins in the PI3K/Akt signaling pathway decreased after silencing EMI2. CONCLUSION EMI2 may be one of the direct targets of YY1 and promotes the progression of CCA through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Shuai Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Kang Lin Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Jin Ang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Shi Lei Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Yi Gang Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Chao Zhu
- The Fourth Department of General Surgery, Second People's Hospital of Anhui Province, No. 1868 Dangshan Road, North Second Ring, Hefei, 230041, Anhui, China
| | - Hao Jin
- The Fourth Department of General Surgery, Second People's Hospital of Anhui Province, No. 1868 Dangshan Road, North Second Ring, Hefei, 230041, Anhui, China
| | - Yong Wang
- The Fourth Department of General Surgery, Second People's Hospital of Anhui Province, No. 1868 Dangshan Road, North Second Ring, Hefei, 230041, Anhui, China
| | - Qing Pang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China.
| | - Hui Chun Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China.
- The Fourth Department of General Surgery, Second People's Hospital of Anhui Province, No. 1868 Dangshan Road, North Second Ring, Hefei, 230041, Anhui, China.
| |
Collapse
|
23
|
Wu Y, Chen W, Gong Y, Liu H, Zhang B. Tetraspanin 1 (TSPAN1) promotes growth and transferation of breast cancer cells via mediating PI3K/Akt pathway. Bioengineered 2021; 12:10761-10770. [PMID: 34852709 PMCID: PMC8809960 DOI: 10.1080/21655979.2021.2003130] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The incidence and mortality of breast cancer rank first among all types of female tumors. To improve patients’ prognosis with advanced breast cancer, new and more effective targets still need to be explored and identified. Tetraspanin 1 (TSPAN1) is highly expressed in several cancers and affects the progression of these tumors. However, there are few studies focused on its role in breast cancer. Previous study showed that TSPAN1 promoted epithelial-mesenchymal transition (EMT) and metastasis, and whether TSPAN1 could promote breast cancer via regulating EMT needs further study. In this study, we found high TSPAN1 expression in breast cancer tumor samples and cell lines which was confirmed by bioinformation analysis. The ablation of TSPAN1 suppressed the growth, and motility of breast cancer cells. We further found that TSPAN1 affected the EMT and mediated the PI3K/Akt pathway in breast cancer cells. In addition, TSPAN1 depletion suppressed tumor growth of breast cancer in mice. In summary, we thought TSPAN1 suppressed growth and motility of breast cancer via mediating EMT and PI3K/AKT pathway, and could serve as a potential target for treatment of breast cancer.
Collapse
Affiliation(s)
- Yange Wu
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, Guangdong Province, China
| | - Wenxiu Chen
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, Guangdong Province, China
| | - Yufeng Gong
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, Guangdong Province, China
| | - Hongxia Liu
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, Guangdong Province, China
| | - Bo Zhang
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, Guangdong Province, China
| |
Collapse
|
24
|
STK39 enhances the progression of Cholangiocarcinoma via PI3K/AKT pathway. iScience 2021; 24:103223. [PMID: 34746696 PMCID: PMC8551078 DOI: 10.1016/j.isci.2021.103223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/10/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023] Open
Abstract
Serine/threonine kinase 39 (STK39) is overexpressed in various tumor tissues and plays an essential role in tumor progression. In this study, we investigated the clinical value, as well as the potential functions and mechanisms of STK39 in cholangiocarcinoma (CCA). The results showed that STK39 was overexpressed in CCA and negatively associated with the prognosis of patients with CCA. Functionally, STK39 knockdown suppressed cell proliferation, migration, and invasion, while STK39 overexpression facilitated tumor aggressiveness. The tumor-promoting effects of STK39 in CCA were also validated by in vivo experiments. Mechanistically, RNA-seq analysis identified that STK39 enhanced the progression of CCA by activating PI3K/AKT signaling pathway. Furthermore, overexpression of STK39 could induce gemcitabine resistance in CCA cells. Moreover, the increased expression of STK39 may be mediated by the dysregulation of miR-26a-5p. In summary, STK39 could be served as a valuable prognostic candidate and a potential therapeutic target of CCA. STK39 was overexpressed in CCA, negatively associated with the prognosis of patients with CCA STK39 knockdown suppressed cell proliferation and invasion. STK39 overexpression facilitated tumor aggressiveness STK39 mediates oncogenic effects on CCA cells by activating the PI3K/AKT signaling pathway STK39 reduces CCA sensitivity to gemcitabine. Increased expression of STK39 may be mediated by dysregulation of miR-26a-5p
Collapse
|
25
|
Zhou C, Liang Y, Zhou L, Yan Y, Liu N, Zhang R, Huang Y, Wang M, Tang Y, Ali DW, Wang Y, Michalak M, Chen XZ, Tang J. TSPAN1 promotes autophagy flux and mediates cooperation between WNT-CTNNB1 signaling and autophagy via the MIR454-FAM83A-TSPAN1 axis in pancreatic cancer. Autophagy 2021; 17:3175-3195. [PMID: 32972302 PMCID: PMC8525961 DOI: 10.1080/15548627.2020.1826689] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 09/12/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is one of the most aggressive tumors associated with a poor clinical prognosis, weakly effective therapeutic options. Therefore, there is a strong impetus to discover new therapeutic targets in pancreatic cancer. In the present study, we first demonstrated that TSPAN1 is upregulated in pancreatic cancer and that TSPAN1 depletion decreases pancreatic cancer cell proliferation in vitro and in vivo. TSPAN1 expression was correlated with poor overall survival of pancreatic cancer patients. Moreover, we demonstrated that TSPAN1 is a novel positive regulator of macroautophagy/autophagy characterized by decreased LC3-II and SQSTM1/p62 expressions, inhibited puncta formation of GFP-LC3 and autophagic vacuoles. We also demonstrated that tspan1 mutation impaired autophagy in the zebrafish model. Furthermore, we showed that TSPAN1 promoted autophagy maturation via direct binding to LC3 by two conserved LIR motifs. Mutations in the LIR motifs of TSPAN1 resulted in a loss of the ability to induce autophagy and promote pancreatic cancer proliferation. Second, we discovered two conservative TCF/LEF binding elements present in the promoter region of the TSPAN1 gene, which was further verified through luciferase activity and ChIP assays. Furthermore, TSPAN1 was upregulated by FAM83A through the canonical WNT-CTNNB1 signaling pathway. We further demonstrated that both TSPAN1 and FAM83A are both direct targets of MIR454 (microRNA 454). Additionally, we revealed the role of MIR454-FAM83A-TSPAN1 in the proliferation of pancreatic cancer cells in vitro and in vivo. Our findings suggest that components of the MIR454-FAM83A-TSPAN1 axis may be valuable prognosis markers or therapeutic targets for pancreatic cancer.Abbreviations: AMPK: adenosine 5'-monophosphate (AMP)-activated protein kinase; APC: APC regulator of WNT signaling pathway; ATG: autophagy related; AXIN2: axin 2; BECN1: beclin 1; CCND1: cyclin D1; CSNK1A1/CK1α: casein kinase 1 alpha 1; CTNNB1/β-catenin: catenin beta 1; DAPI: 4'6-diamino-2-phenylindole; EBSS: Earle's balanced salt solution; EdU: 5-ethynyl-20-deoxyuridine; FAM83A: family with sequence similarity 83 member A; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GSEA: gene set enrichment analysis; GSK3B: glycogen synthase kinase 3 beta; IHC: immunohistochemical; LAMP1: lysosomal associated membrane protein 1; LIR: LC3-interacting region; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; MIR454: microRNA 454; miRNA: microRNA; MKI67: antigen identified by monoclonal antibody Ki 67; MTOR: mechanistic target of rapamycin kinase; MTT: 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide; MYC: MYC proto-oncogene, bHLH transcription factor; OS: overall survival; PDAC: pancreatic ductal adenocarcinoma; RAB7A: RAB7A, member RAS oncogene family; shRNA: short hairpin RNA; SQSTM1: sequestosome 1; TBE: TCF/LEF binding element; TCGA: The Cancer Genome Atlas; TCF/LEF: transcription factor/lymphoid enhancer binding factor; TCF4: transcription factor 4; TSPAN1: tetraspanin 1; TUNEL: terminal deoxynucleotidyl transferase mediated dUTP nick end labeling; UTR: untranslated region; WT: wild type.
Collapse
Affiliation(s)
- Cefan Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Yanyan Liang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Li Zhou
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, China
| | - Yanan Yan
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Nanxi Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Yuan Huang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Ming Wang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yongfei Tang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Yefu Wang
- The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| |
Collapse
|
26
|
Dai L, Lu Y, Jiang L, Zhu L, Zhang J, Wang F, Gao Y, Xin W. SRY-Box Transcription Factor 9 (SOX9) Affects the Proliferation, Invasion and Epithelial to Mesenchymal Transition (EMT) of Intrahepatic Cholangiocarcinoma by Regulating Transforming Growth Factor β (TGF β)/Smad Signaling. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Intrahepatic cholangiocarcinoma (ICC) develops rapidly with a high malignancy. SOX9 expression is increased in several tumors. However, its expression and role in intrahepatic cholangiocarcinoma have not yet been elucidated. Real time PCR and Western blot were done to assess SOX9 expression
in tumor tissues and adjacent tissues of ICC. ICC cell line QBC939 cells were separated into control group, SOX9 overexpression group and SOX9 siRNA group followed by analysis of cell survival by MTT assay, cell migration by cell scratch assay, cell invasion by transwell chamber, E-cadherin
and Vimentin level by western blot, TGFβ/Smad signaling protein level by real time PCR. SOX9 level in tumor tissues was significantly increased compared to adjacent tissues (P < 0.05) and it was associated with TNM stage, tissue type and metastasis, and survival time
(P < 0.05). Transfection of pcDNA3.1-SOX9 upregulated SOX9, promoted cell proliferation, migration and invasion, downregulated E-cadherin, upregulated Vimentin, TGF-β1 and Smad4 (P < 0.05). SOX9 siRNA transfection into QBC939 cells could significantly reverse
the above mentioned changes (P < 0.05). SOX9 level is increased in intrahepatic cholangiocarcinoma and targeting SOX9 can inhibit cell migration and invasion, and EMT via regulating TGFβ/Smad signaling.
Collapse
Affiliation(s)
- Ling Dai
- Intensive Care Second Unit (Second Ward), Wuhan No. 1 Hospital, Wuhan, Hubei, 430000, China
| | - Yuqing Lu
- Department of Hepatobiliary Surgery, Wuhan No. 1 Hospital, Wuhan, Hubei, 430000, China
| | - Lu Jiang
- Intensive Care Second Unit (Second Ward), Wuhan No. 1 Hospital, Wuhan, Hubei, 430000, China
| | - Liping Zhu
- Department of Hepatobiliary Surgery, Wuhan No. 1 Hospital, Wuhan, Hubei, 430000, China
| | - Jing Zhang
- Taizhou Vocational and Technical College, Taizhou, Zhejiang, 318000, China
| | - Fang Wang
- Department of Hepatobiliary Surgery, Wuhan No. 1 Hospital, Wuhan, Hubei, 430000, China
| | - Yuanyuan Gao
- Intensive Care Second Unit (Second Ward), Wuhan No. 1 Hospital, Wuhan, Hubei, 430000, China
| | - Wenwei Xin
- Department Emergency, Taizhou First Peoples Hospital, Taizhou, Zhejiang, 318000, China
| |
Collapse
|
27
|
Hao J, Deng H, Yang Y, Chen L, Wu Q, Yao P, Li J, Li B, Jin X, Wang H, Duan H. Downregulation of MCM8 expression restrains the malignant progression of cholangiocarcinoma. Oncol Rep 2021; 46:235. [PMID: 34523691 PMCID: PMC8453687 DOI: 10.3892/or.2021.8186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/10/2021] [Indexed: 12/26/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive malignant tumor with an extremely poor prognosis. Minichromosome maintenance 8 homologous recombination repair factor (MCM8) is a helicase involved in the elongation step of DNA replication and tumorigenesis. In the present study, the clinical significance and biological function of MCM8 in CCA were investigated. The expression levels of MCM8 in CCA and paracancerous tissues were analyzed using immunohistochemical staining. The potential mechanisms underlying MCM8 and the biological effects of MCM8 in CCA cells were explored using in vitro assays and in vivo mouse xenograft models. The high expression levels of MCM8 in CCA has important clinical significance in predicting disease progression. Knockdown of MCM8 decreased proliferation, promoted apoptosis and suppressed migration of CCA cells. MCM8 knockdown also suppressed tumor growth in vivo. Mechanistically, MCM8 knockdown led to the abnormal downregulation of survivin, XIAP, HSP27, IGF‑1sR, sTNF‑R1, sTNF‑R2, TNF‑α and TNF‑β. Furthermore, downregulation of MCM8 expression inhibited the PI3K/Akt signaling pathway and induced the MAPK9 signaling pathway. MCM8 promoted the malignant progression of CCA, indicating that inhibition of MCM8 may have the potential to serve as a novel molecular targeted therapy.
Collapse
Affiliation(s)
- Jingcheng Hao
- Department of Hepatobiliary and Vascular Surgery, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Haimin Deng
- Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Yuan Yang
- Department of Rheumatology and Immunology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Lidan Chen
- Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Qiang Wu
- Department of Hepatobiliary and Vascular Surgery, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Pei Yao
- Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Junen Li
- Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Bowen Li
- Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Xueli Jin
- Department of Hepatobiliary and Vascular Surgery, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Haiqing Wang
- Department of Hepatopancreatobiliary Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Huaxin Duan
- Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
28
|
Liu S, Cai Y, Changyong E, Sheng J, Zhang X. Screening and Validation of Independent Predictors of Poor Survival in Pancreatic Cancer. Pathol Oncol Res 2021; 27:1609868. [PMID: 34321959 PMCID: PMC8310909 DOI: 10.3389/pore.2021.1609868] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is a digestive system malignant tumor with high mortality and poor prognosis, but the mechanisms of progression remain unclear in pancreatic cancer. It's necessary to identify the hub genes in pancreatic cancer and explore the novel potential predictors in the prognosis of pancreatic cancer. We downloaded two mRNA expression profiles from Gene Expression Omnibus and The Cancer Genome Atlas Pancreatic Cancer (TCGA-PAAD) datasets to screen the commonly differentially expressed genes in pancreatic cancer by limma package in R. Subsequently, measurement of the functional similarity among the 38 DEGs in common was performed to identify the hub genes using GOSemSim package. Then, survival analysis and Cox regression were applied to explore prognosis-related hub genes using the survival package. Statistics analysis by two-tailed Student's t-test or one-way based on TCGA-PAAD datasets and qPCR detection in clinical samples were performed to explore the correlations between expression of hub genes in pancreatic cancer tissues and clinical parameters. Based on integrated analysis of TCGA and GEO datasets, we screened 38 DEGs in common, which were all up-regulated. The functional similarity results showed that 10 DEGs including TSPAN1, MSLN, C1orf116, PKP3, CEACAM6, BAIAP2L1, PPL, RAB25, ERBB3, and AP1M2 in the DEGs in common, which had the higher average functional similarity, were considered as the hub genes. Survival analysis results and Cox regression analysis showed that TSPAN1, CEACAM6, as well as ERBB3 were all associated with poor overall survival of PC. qPCR results showed that the expression levels of TSPAN1 and ERBB3 were significantly upregulated in the PC tissues. The statistical analysis results revealed that TSPAN1 expression correlated significantly with histologic grade, T stage, clinical stage, and vital status by two-tailed Student's t-test or one-way ANOVA; ERBB3 expression correlated significantly with T stage, clinical stage, and vital status by two-tailed Student's t-test or one-way ANOVA. We found that TSPAN1 and ERBB3 could be independent predictors of poor survival in pancreatic cancer.
Collapse
Affiliation(s)
- Shui Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, China
| | - Yan Cai
- Hospital of Stomatology, Jilin University, Changchun, China
| | - E. Changyong
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, China
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
29
|
Horiguchi K, Fujiwara K, Tsukada T, Nakakura T, Yoshida S, Hasegawa R, Takigami S, Ohsako S. CD9-positive cells in the intermediate lobe migrate into the anterior lobe to supply endocrine cells. Histochem Cell Biol 2021; 156:301-313. [PMID: 34185148 DOI: 10.1007/s00418-021-02009-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 12/20/2022]
Abstract
The adenohypophysis is composed of the anterior and intermediate lobes (AL and IL), and secretes important hormones for growth, sexual development, metabolism, and reproduction. In the marginal cell layer (MCL) facing Rathke's cleft between the IL and AL, cluster of differentiation (CD) 9-, CD81-, S100β-, and SOX2-quadruple positive (CD9/CD81/S100β/SOX2-positive) cells in the adult IL are settled as tissue-resident stem/progenitor cells supplying hormone-producing cells to the AL. However, it is unclear how CD9/CD81/S100β/SOX2-positive cells in the IL-side MCL migrate into the AL across Rathke's cleft. In the present study, we performed chimeric pituitary tissue culture using S100β/GFP-transgenic rats and Wistar rats, and traced the footprint of S100β/GFP-expressing cells. We detected IL-side S100β/GFP-expressing cells in the AL tissue, demonstrating that these cells migrate from the IL to the AL. However, the cells failed to migrate in the opposite direction. Consistently, scanning electron microscopic analysis revealed well-developed cytoplasmic protrusions in the IL-side MCL, but not in the AL-side MCL, suggesting that IL-side CD9/CD81/S100β/SOX2-positive cells had higher migratory activity. We also searched for a specific marker for IL-side CD9/CD81/S100β/SOX2-positive cells and identified tetraspanin 1 (TSPAN1) from microarray analysis. Downregulation of Tspan1 by specific siRNA impaired cell migration and significantly reduced expression of snail family transcriptional repressor 2 (Slug), a marker of epithelial-mesenchymal transition (EMT). Therefore, CD9/CD81/S100β/SOX2-positive cells in the IL-side MCL can be stem/progenitor cells that provide stem/progenitor cells to the AL-side MCL via SLUG-mediated EMT and cell migration.
Collapse
Affiliation(s)
- K Horiguchi
- Laboratory of Anatomy and Cell Biology, Department of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo, 181-8612, Japan.
| | - K Fujiwara
- Department of Biological Science, Faculty of Science, Kanagawa University, 2946 Tsuchiya, Hiratsuka, Kanagawa, 259-1293, Japan
| | - T Tsukada
- Department of Biomolecular Science, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - T Nakakura
- Department of Anatomy, Graduate School of Medicine, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo, 173-8605, Japan
| | - S Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - R Hasegawa
- Laboratory of Anatomy and Cell Biology, Department of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo, 181-8612, Japan
| | - S Takigami
- Laboratory of Anatomy and Cell Biology, Department of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo, 181-8612, Japan
| | - S Ohsako
- Laboratory of Anatomy and Cell Biology, Department of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo, 181-8612, Japan
| |
Collapse
|
30
|
Weidle UH, Birzele F, Brinkmann U, Auslaender S. Gastric Cancer: Identification of microRNAs Inhibiting Druggable Targets and Mediating Efficacy in Preclinical In Vivo Models. Cancer Genomics Proteomics 2021; 18:497-514. [PMID: 34183383 DOI: 10.21873/cgp.20275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 01/06/2023] Open
Abstract
In addition to chemotherapy, targeted therapies have been approved for treatment of locally advanced and metastatic gastric cancer. The therapeutic benefit is significant but more durable responses and improvement of survival should be achieved. Therefore, the identification of new targets and new approaches for clinical treatment are of paramount importance. In this review, we searched the literature for down-regulated microRNAs which interfere with druggable targets and exhibit efficacy in preclinical in vivo efficacy models. As druggable targets, we selected transmembrane receptors, secreted factors and enzymes. We identified 38 microRNAs corresponding to the criteria as outlined. A total of 13 miRs target transmembrane receptors, nine inhibit secreted proteins and 16 attenuate enzymes. These microRNAs are targets for reconstitution therapy of gastric cancer. Further target validation experiments are mandatory for all of the identified microRNAs.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany;
| | - Fabian Birzele
- Pharmaceutical Sciences, Roche Pharma Research and Early Development (pRed), Roche Innovation Center Basel, Basel, Switzerland
| | - Ulrich Brinkmann
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany;
| | - Simon Auslaender
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
31
|
Deng X, Zuo M, Pei Z, Xie Y, Yang Z, Zhang Z, Jiang M, Kuang D. MicroRNA-455-5p Contributes to Cholangiocarcinoma Growth and Mediates Galangin's Anti-Tumor Effects. J Cancer 2021; 12:4710-4721. [PMID: 34149934 PMCID: PMC8210562 DOI: 10.7150/jca.58873] [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: 01/31/2021] [Accepted: 05/22/2021] [Indexed: 01/06/2023] Open
Abstract
Fully understanding the mechanism of how Cholangiocarcinoma (CCA) development and discovering promising therapeutic drugs are important to improve patients' survival time. This study identifies that microRNA-455-5p (miR-455-5p) targets protein phosphatase 1 regulatory subunit 12A (PPP1R12A), an effect that represses mitogen-activated protein kinase (MAPK) and PI3K/AKT pathway activation, thereby controlling CCA cells survival and metastasis. Moreover, miR-455-5p expression is reduced in CCA tissues and negative correlation with PPP1R12A and PPP1R12A knockdown phenotypic mimics miR-455-5p' effects on CCA cells. Furthermore, we demonstrate that galangin inhibits CCA growth both in vitro and in vivo, which is associated with increased miR-455-5p and repressed PPP1R12A expression. In support, overexpression of miR-455-5p abrogates those galangin-mediated anti-CCA effects. These findings establish an essential role of miR-455-5p in CCA development and galangin may provide a potential therapeutic adjuvant agent for anti-CCA treatment.
Collapse
Affiliation(s)
- Xu Deng
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Meiling Zuo
- Department of Pharmacy, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zhifang Pei
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuanlin Xie
- Department of Pharmacy, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zhongbao Yang
- Department of Pharmacy, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zhihui Zhang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Minna Jiang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dabin Kuang
- Department of Pharmacy, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
32
|
Zhou J, Nie W, Yuan J, Zhang Z, Mi L, Wang C, Huang R. GSG2 knockdown suppresses cholangiocarcinoma progression by regulating cell proliferation, apoptosis and migration. Oncol Rep 2021; 45:91. [PMID: 33846801 PMCID: PMC8042665 DOI: 10.3892/or.2021.8042] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 03/18/2021] [Indexed: 01/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is the second most common type of hepatocellular carcinoma characterized by high aggressiveness and extremely poor patient prognosis. The germ cell-specific gene 2 protein (GSG2) is a histone H3 threonine-3 kinase required for normal mitosis. Nevertheless, the role and mechanism of GSG2 in the progression and development of CCA remain elusive. In the present study, the association between GSG2 and CCA was elucidated. Firstly, we demonstrated that GSG2 was overexpressed in CCA specimens and HCCC-9810 and QBC939 cells by immunohistochemical (IHC) staining. It was further revealed that high expression of GSG2 in CCA had significant clinical significance in predicting disease deterioration. Subsequently, cell proliferation, apoptosis, cell cycle distribution and migration were measured by MTT, flow cytometry, and wound healing assays, respectively in vitro. The results demonstrated that downregulation of GSG2 decreased proliferation, promoted apoptosis, arrested the cell cycle and weakened migration in the G2 phase of CCA cells. Additionally, GSG2 knockdown inhibited CCA cell migration by suppressing epithelial-mesenchymal transition (EMT)-related proteins, such as N-cadherin and vimentin. Mechanistically, GSG2 exerted effects on CCA cells by modulating the PI3K/Akt, CCND1/CDK6 and MAPK9 signaling pathways. In vivo experiments further demonstrated that GSG2 knockdown suppressed tumor growth. In summary, GSG2 was involved in the progression of CCA, suggesting that GSG2 may be a potential therapeutic target for CCA patients.
Collapse
Affiliation(s)
- Jun Zhou
- Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Wanpin Nie
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of The Central South University, Changsha, Hunan 410013, P.R. China
| | - Jiajia Yuan
- Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Zeyu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of The Central South University, Changsha, Hunan 410013, P.R. China
| | - Liangliang Mi
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of The Central South University, Changsha, Hunan 410013, P.R. China
| | - Changfa Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of The Central South University, Changsha, Hunan 410013, P.R. China
| | - Ranglang Huang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of The Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
33
|
Li C, Hou X, Yuan S, Zhang Y, Yuan W, Liu X, Li J, Wang Y, Guan Q, Zhou Y. High expression of TREM2 promotes EMT via the PI3K/AKT pathway in gastric cancer: bioinformatics analysis and experimental verification. J Cancer 2021; 12:3277-3290. [PMID: 33976737 PMCID: PMC8100818 DOI: 10.7150/jca.55077] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/15/2021] [Indexed: 01/17/2023] Open
Abstract
Background: To date, the pathogenesis of gastric cancer (GC) remains unclear. We combined public database resources and bioinformatics analysis methods, explored some novel genes and verified the experiments to further understand the pathogenesis of GC and to provide a promising target for anti-tumor therapy. Methods: We downloaded the chip data related to GC from the Gene Expression Omnibus (GEO) database, extracted differentially expressed genes (DEGs), and then determined the key genes in the development of GC via PPI networks and model analysis. Functional annotation via GO and KEGG enrichment of DEGs was used to understand the latent roles of DEGs. The expression of the triggering receptor expressed on myeloid cells 2 (TREM2) gene in GC cell lines was verified via RT-PCR and western blotting. Moreover, the CCK-8, wound healing assay, and transwell migration and invasion assays were used to understand the changes in the proliferation, migration, and invasion abilities of GC cells after silencing TREM2. Western blotting verified the interaction between TREM2 and PI3K predict of the string website, as well as the effect of TREM2 on EMT. Finally, a lung metastasis model was used to explore the relationship between TREM2 and metastasis. Results: Our study identified 16 key genes, namely BGN, COL1A1, COL4A1, COL5A2, NOX4, SPARC, HEYL, SPP1, TIMP1, CTHRC1, TREM2, SFRP4, FBXO32, GPX3, KIF4A, and MMP9 genes associated with GC. The EMT-related pathway was the most significantly altered pathway. TREM2 expression was higher in GC cell lines and was remarkably associated with tumor invasion depth, TNM stage, histological grade, histological type, anatomic subdivision, and Helicobacter pylori state. Knockdown of TREM2 expression inhibited the proliferation, migration, and invasion of GC cells as well as the progression of EMT by PI3K/AKT signaling in vitro. In addition, lung metastasis were decreased in vivo. Conclusions: We identified some important genes associated with the progression of GC via public database analysis, explored and verified the effects of proto-oncogene TREM2 on EMT via the PI3K/AKT pathway. TREM2 may be a novel target in the GC therapy.
Collapse
Affiliation(s)
- Chunmei Li
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoming Hou
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Shuqiao Yuan
- Department of medical laboratory, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yigan Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Wenzhen Yuan
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoguang Liu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,Department of Rheumatology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Juan Li
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,Department of Gastroenterology, Gansu Provincial Hospital, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Quanlin Guan
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
34
|
Liang J, Liu T, Liao J, Zhang L, Zhou M, Xu W, He Y, Cai G, Jin G, Song J, Li G, Liang H, Ding Z, Zhang B. Development and validation of a CpG island methylator phenotype-related prognostic signature for cholangiocarcinoma. J Cell Physiol 2021; 236:3143-3156. [PMID: 32996133 DOI: 10.1002/jcp.30082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022]
Abstract
Cholangiocarcinoma (CCA) still has a very unfavorable prognosis with a very high mortality, which is complicated by a lack of prognostic biomarkers. In this study, CCA patients in the Gene Expression Omnibus (GEO) cohort were categorized into two subtypes. Differentially expressed and methylated genes were identified, and the impact of DNA methylation in the trans-regulation of gene expression was investigated. Finally, a CIMP-related methylation signature specific for CCA (CMSC) was trained in GEO and validated in the Tongji cohort. A subset of patients with CIMP-H was identified, which was correlated with an unfavorable prognosis. Gene enrichment analysis implied the potential mechanism of CIMP as a promoter of carcinogenesis by regulating proliferation. The trans-regulation among differentially methylated CpG sites and genes with the same change trends was positively correlated, while the converse situation showed a negative correlation. Notably, CMSC based on four genes could significantly classify CCA patients into low- and high-risk groups in the GEO cohort, and the robustness of CMSC was validated in the Tongji cohort. The results of receiver operating characteristic analysis further indicated that CMSC was capable of highly sensitive and specific prediction of the patient outcomes in CCA. In conclusion, our work highlights the clinical significance of CMSC in the prognosis of CCA.
Collapse
Affiliation(s)
- Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tongtong Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingyu Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lu Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mi Zhou
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weiqi Xu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi He
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guangzhen Cai
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guannan Jin
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jia Song
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ganxun Li
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zeyang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
35
|
Liu K, Gao R, Wu H, Wang Z, Han G. Single-cell analysis reveals metastatic cell heterogeneity in clear cell renal cell carcinoma. J Cell Mol Med 2021; 25:4260-4274. [PMID: 33759378 PMCID: PMC8093989 DOI: 10.1111/jcmm.16479] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) is one of the leading causes of cancer-related death worldwide. Tumour metastasis and heterogeneity lead to poor survival outcomes and drug resistance in patients with metastatic RCC (mRCC). In this study, we aimed to assess intratumoural heterogeneity (ITH) in mRCC cells by performing a combined analysis of bulk data and single-cell RNA-sequencing data, and develop novel biomarkers for prognosis prediction on the basis of the potential molecular mechanisms underlying tumorigenesis. Eligible single-cell cohorts related to mRCC were acquired using the Gene Expression Omnibus (GEO) dataset to identify potential mRCC subpopulations. We then performed gene set variation analysis to understand the differential function in primary RCC and mRCC samples. Subsequently, we applied weighted correlation network analysis to identify coexpressing gene modules that were related to the external trait of metastasis. Protein-protein interactions were used to screen hub subpopulation-difference (sub-dif) markers (ACTG1, IL6, CASP3, ACTB and RAP1B) that might be involved in the regulation of RCC metastasis and progression. Cox regression analysis revealed that ACTG1 was a protective factor (HR < 1), whereas the other four genes (IL6, CASP3, ACTB and RAP1B) were risk factors (HR > 1). Kaplan-Meier survival analysis suggested the potential prognostic value of these sub-dif markers. The expression of sub-dif markers in mRCC was further evaluated in clinical samples by immunohistochemistry (IHC). Additionally, the genetic features of sub-dif marker expression patterns, such as genetic variation profiles, correlations with tumour-infiltrating lymphocytes (TILs), and targeted signalling pathway activities, were assessed in bulk RNA-seq datasets. In conclusion, we established novel subpopulation markers as key prognostic factors affecting EMT-related signalling pathway activation in mRCC, which could facilitate the implementation of a treatment for mRCC patients.
Collapse
Affiliation(s)
- Kun Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Gao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hao Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhe Wang
- Department of Gastrointestinal Oncology, Cancer Hospital of China Medical University Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Guang Han
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
36
|
Shin HY, Yang W, Chay DB, Lee EJ, Chung JY, Kim HS, Kim JH. Tetraspanin 1 promotes endometriosis leading to ovarian clear cell carcinoma. Mol Oncol 2021; 15:987-1004. [PMID: 33331115 PMCID: PMC8024726 DOI: 10.1002/1878-0261.12884] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/13/2020] [Accepted: 12/14/2020] [Indexed: 11/29/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) reportedly develops from endometriosis. However, the molecular mechanism underlying its malignant progression to OCCC remains elusive. This study aimed to identify an essential gene in the malignant transformation of endometriosis to OCCC. We performed RNA sequencing in formalin‐fixed, paraffin‐embedded (FFPE) tissues of endometriosis (n = 9), atypical endometriosis (AtyEm) (n = 18), adjacent endometriosis to OCCC (AdjEm) (n = 7), and OCCC (n = 17). We found that tetraspanin 1 (TSPAN1) mRNA level was significantly increased by 2.4‐ (DESeq2) and 3.4‐fold (edgeR) in AtyEm and by 80.7‐ (DESeq2) and 101‐fold (edgeR) in OCCC relative to endometriosis. We confirmed that TSPAN1 protein level was similarly overexpressed in OCCC tissues and cell lines. In immortalized endometriosis cell lines, TSPAN1 overexpression enhanced cell growth and invasion. Mechanistically, TSPAN1 triggered AMP‐activated protein kinase (AMPK) activity, promoting endometriosis and cell growth. Upregulated levels of TSPAN1 are considered an early event in the development of high‐risk endometriosis that could progress to ovarian cancer. Our study suggests the potential of TSPAN1 as a screening candidate for high‐risk endometriosis.
Collapse
Affiliation(s)
- Ha-Yeon Shin
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Wookyeom Yang
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Doo Byung Chay
- Department of Obstetrics and Gynecology, Sahmyook Medical Center, Seoul, Korea
| | - Eun-Ju Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Joon-Yong Chung
- Experimental Pathology Lab., Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hyun-Soo Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
miR-573 suppresses pancreatic cancer cell proliferation, migration, and invasion through targeting TSPAN1. Strahlenther Onkol 2020; 197:438-448. [PMID: 33320287 DOI: 10.1007/s00066-020-01728-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/30/2020] [Indexed: 01/22/2023]
Abstract
PURPOSE To explore whether miR-573 can suppress pancreatic cancer cell proliferation, migration, and invasion by targeting TSPAN1. METHODS The expression of miR-573 and TSPAN1 in pancreatic cancer tissues and cells lines was analyzed using RT-qPCR. The human pancreatic cancer cell line PANC‑1 was transfected with miR-573 mimic, pcDNA3.1-TSPAN1, or genOFFTM st-h-TSPAN1. The effects of miR-573 and TSPAN1 on cell proliferation, colony formation, migration, and invasion were analyzed by CCK‑8, colony formation, transwell migration, and invasion assay, respectively. Target genes of miR-573 were screened using bioinformatics tools and confirmed by dual-luciferase reporter assay and real-time PCR. The effects of miR-573 in vivo were observed using tumor xenografts. RESULTS We found that miR-573 is downregulated and TSPAN1 is upregulated in pancreatic cancer tissues and cells lines. Function assays demonstrated that overexpression of miR-573 inhibited cell proliferation, colony formation, migration, and invasion of pancreatic cancer cells, as well as suppressing tumor growth in vivo. Target genes of miR-573 were predicted using bioinformatics tools and confirmed by dual-luciferase reporter assay and RT-qPCR or western blotting. Downregulation of TSPAN1 also inhibited cell proliferation, colony formation, migration, and invasion of pancreatic cancer cells. Furthermore, overexpression of TSPAN1 attenuated miR-573-induced inhibition of pancreatic cancer cell proliferation and migration. CONCLUSION Our findings indicated that miR-573 suppresses pancreatic cancer cell proliferation, migration, and invasion through targeting TSPAN1. TSPAN1 targeted by miR-573 might be a potential therapeutic target for clinical treatment of pancreatic cancer.
Collapse
|
38
|
Garcia-Mayea Y, Mir C, Carballo L, Castellvi J, Temprana-Salvador J, Lorente J, Benavente S, García-Pedrero JM, Allonca E, Rodrigo JP, LLeonart ME. TSPAN1: A Novel Protein Involved in Head and Neck Squamous Cell Carcinoma Chemoresistance. Cancers (Basel) 2020; 12:cancers12113269. [PMID: 33167355 PMCID: PMC7694336 DOI: 10.3390/cancers12113269] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/24/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Therapy resistance in head and neck squamous cell carcinoma (HNSCC) patients is the main obstacle to achieve more effective treatments that improve survival and quality of life of these patients. Therefore, it is of vital importance to unravel the molecular and cellular mechanisms by which tumor cells acquire resistance to chemotherapy. We conducted a comparative proteomic study involving cisplatin-resistant cells and cancer stem cells with the aim of identifying proteins potentially implicated in the acquisition of cisplatin resistance. Through this study, we identified for the first time tetraspanin-1 (TSPAN1) as an important protein involved in the development, progression and chemoresistance of HNSCC tumors. Abstract Sensitization of resistant cells and cancer stem cells (CSCs) represents a major challenge in cancer therapy. A proteomic study revealed tetraspanin-1 (TSPAN1) as a protein involved in acquisition of cisplatin (CDDP) resistance (Data are available via ProteomeXchange with identifier PXD020159). TSPAN1 was found to increase in CDDP-resistant cells, CSCs and biopsies from head and neck squamous cell carcinoma (HNSCC) patients. TSPAN1 depletion in parental and CDDP-resistant HNSCC cells reduced cell proliferation, induced apoptosis, decreased autophagy, sensitized to chemotherapeutic agents and inhibited several signaling cascades, with phospho-SRC inhibition being a major common target. Moreover, TSPAN1 depletion in vivo decreased the size and proliferation of parental and CDDP-resistant tumors and reduced metastatic spreading. Notably, CDDP-resistant tumors showed epithelial–mesenchymal transition (EMT) features that disappeared upon TSPAN1 inhibition, suggesting a link of TSPAN1 with EMT and metastasis. Immunohistochemical analysis of HNSCC specimens further revealed that TSPAN1 expression was correlated with phospho-SRC (pSRC), and inversely with E-cadherin, thus reinforcing TSPAN1 association with EMT. Overall, TSPAN1 emerges as a novel oncogenic protein and a promising target for HNSCC therapy.
Collapse
Affiliation(s)
- Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells, Vall d’Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (Y.G.-M.); (C.M.); (L.C.); (J.C.); (J.T.-S.)
- Genetic, Microbiology and Statistics Department, Faculty of Biology, University of Barcelona, Avenida Diagonal 643, 08014 Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells, Vall d’Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (Y.G.-M.); (C.M.); (L.C.); (J.C.); (J.T.-S.)
| | - Laia Carballo
- Biomedical Research in Cancer Stem Cells, Vall d’Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (Y.G.-M.); (C.M.); (L.C.); (J.C.); (J.T.-S.)
| | - Josep Castellvi
- Biomedical Research in Cancer Stem Cells, Vall d’Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (Y.G.-M.); (C.M.); (L.C.); (J.C.); (J.T.-S.)
| | - Jordi Temprana-Salvador
- Biomedical Research in Cancer Stem Cells, Vall d’Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (Y.G.-M.); (C.M.); (L.C.); (J.C.); (J.T.-S.)
| | - Juan Lorente
- Otorhinolaryngology Department, Hospital Vall d’Hebron (HUVH), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain;
| | - Sergi Benavente
- Radiotherapy Unit, Vall d’Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain;
| | - Juana M. García-Pedrero
- Department of Otolaryngology-Head and Neck Surgery, Central University Hospital of Asturias, University of Oviedo, ISPA, IUOPA, 33011 Oviedo, Spain; (J.M.G.-P.); (E.A.); (J.P.R.)
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Av. Roma SN, 33011 Oviedo, Spain
| | - Eva Allonca
- Department of Otolaryngology-Head and Neck Surgery, Central University Hospital of Asturias, University of Oviedo, ISPA, IUOPA, 33011 Oviedo, Spain; (J.M.G.-P.); (E.A.); (J.P.R.)
| | - Juan P. Rodrigo
- Department of Otolaryngology-Head and Neck Surgery, Central University Hospital of Asturias, University of Oviedo, ISPA, IUOPA, 33011 Oviedo, Spain; (J.M.G.-P.); (E.A.); (J.P.R.)
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Av. Roma SN, 33011 Oviedo, Spain
| | - Matilde E. LLeonart
- Biomedical Research in Cancer Stem Cells, Vall d’Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (Y.G.-M.); (C.M.); (L.C.); (J.C.); (J.T.-S.)
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Vall d’Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119–129, 08035 Barcelona, Spain
- Correspondence: ; Tel.: +34-934894169; Fax: +34-932746708
| |
Collapse
|
39
|
Qi Y, Li H, Lv J, Qi W, Shen L, Liu S, Ding A, Wang G, Sun L, Qiu W. Expression and function of transmembrane 4 superfamily proteins in digestive system cancers. Cancer Cell Int 2020; 20:314. [PMID: 32694936 PMCID: PMC7364658 DOI: 10.1186/s12935-020-01353-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/15/2020] [Indexed: 02/08/2023] Open
Abstract
Background Although the medical level is constantly improving, cancer is still a major disease that threatens human health, and very effective treatments have not been found. In recent years, studies have found that four-transmembrane superfamily proteins are involved in multiple stages of tumorigenesis and development, but their expression and function in tumors have not been systematically studied. Methods We used the Oncomine database to analyze the mRNA expression levels of TSPAN family in various cancers. Then differentially expressed genes were screened out and verified by liver cancer, colorectal cancer, and gastric cancer cells by q-PCR and Western blot analysis. CCK8 and EDU analysis are used to detect cell proliferation, Cell wound scrape assay and Cell invasion assay are used to analyze cell invasion and metastasis. Nude tumor formation test used to verify the tumor suppressive effect of TSPAN7 in vivo. Results Differential analysis of 33 TSPAN proteins revealed that a total of 11 proteins showed differential expression in 10% of independent analyses, namely TSPAN1, TSPAN3, TSPAN5, TSPAN6, TSPAN7, TSPAN8, TSPAN13, TSPAN25, TSPAN26, TSPAN29, TSPAN30. TSPAN7 is the only four-transmembrane protein with reduced expression in three types of digestive tract tumors, so we chose TSPAN7 to be selected for cellular and molecular level verification. We found that compared with normal cells, the expression of TSPAN7 in liver cancer cells was significantly reduced, while the expression of gastric and colon cancer was not significantly different from that of normal cells. In addition, we also found that the high expression of Tspan7 not only inhibited the proliferation of HCC-LM3 cells, but also inhibited its invasion and metastasis. Conclusions Our study evaluated the expression and function of the TSPANs family in digestive cancers and explored TSPAN7 in hepatoma cells in detail. We found some members of the TSPAN family show significant expression differences between cancer and normal tissues, of which TSPAN7 may be a potential biomarker for liver cancer.
Collapse
Affiliation(s)
- Yaoyue Qi
- Qingdao University, Qingdao, Shandong China
| | - Hui Li
- Qingdao University, Qingdao, Shandong China
| | - Jing Lv
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China
| | - Weiwei Qi
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China
| | - Liwei Shen
- Department of Oncology, Qingdao Women and Children's Hospital, Qingdao, Shandong China
| | - Shihai Liu
- Central Laboratory, Affiliated Hospital of Qingdao University, Qingdao, Shandong China
| | - Aiping Ding
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China
| | | | - Libin Sun
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China
| | - Wensheng Qiu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China
| |
Collapse
|
40
|
Chu KJ, Ma YS, Jiang XH, Wu TM, Wu ZJ, Li ZZ, Wang JH, Gao QX, Yi B, Shi Y, Wang HM, Gu LP, Zhang SQ, Wang GR, Liu JB, Fu D, Jiang XQ. Whole-Transcriptome Sequencing Identifies Key Differentially Expressed mRNAs, miRNAs, lncRNAs, and circRNAs Associated with CHOL. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:592-603. [PMID: 32721879 PMCID: PMC7390861 DOI: 10.1016/j.omtn.2020.06.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022]
Abstract
To systematically evaluate the whole-transcriptome sequencing data of cholangiocarcinoma (CHOL) to gain more insights into the transcriptomic landscape and molecular mechanism of this cancer, we performed whole-transcriptome sequencing based on the tumorous (C) and their corresponding non-tumorous adjacent to the tumors (CP) from eight CHOL patients. Subsequently, differential expression analysis was performed on the C and CP groups, followed by functional interaction prediction analysis to investigate gene-regulatory circuits in CHOL. In addition, The Cancer Genome Atlas (TCGA) for CHOL data was used to validate the results. In total, 2,895 differentially expressed messenger RNAs (dif-mRNAs), 56 differentially expressed microRNAs (dif-miRNAs), 151 differentially expressed long non-coding RNAs (dif-lncRNAs), and 110 differentially expressed circular RNAs (dif-circRNAs) were found in CHOL samples compared with controls. Enrichment analysis on those differentially expressed genes (DEGs) related to miRNA, lncRNA, and circRNA also identified the function of spliceosome. The downregulated hsa-miR-144-3p were significantly enriched in the competing endogenous RNA (ceRNA) complex network, which also included 7 upregulated and 13 downregulated circRNAs, 7 upregulated lncRNAs, and 90 upregulated and 40 downregulated mRNAs. Moreover, most of the DEGs and a few of the miRNAs (such as hsa-miR-144-3p) were successfully validated by TCGA data. The genes involved in RNA splicing and protein degradation processes and miR-144-3p may play fundamental roles in the pathogenesis of CHOL.
Collapse
Affiliation(s)
- Kai-Jian Chu
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai 200438, China
| | - Yu-Shui Ma
- Cancer Institute, Nantong Tumor Hospital, Nantong 226631, China; Department of Radiology, The Fourth Affiliated Hospital of Anhui Medical University, Hefei 230012, China; Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiao-Hui Jiang
- General Surgery, Nantong Tumor Hospital, Nantong 226631, China
| | - Ting-Miao Wu
- Department of Radiology, The Fourth Affiliated Hospital of Anhui Medical University, Hefei 230012, China
| | - Zhi-Jun Wu
- Department of Oncology, Nantong Second People's Hospital, Nantong 226002, China
| | - Zhi-Zhen Li
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai 200438, China
| | - Jing-Han Wang
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai 200438, China
| | - Qing-Xiang Gao
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai 200438, China
| | - Bin Yi
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai 200438, China
| | - Yi Shi
- Cancer Institute, Nantong Tumor Hospital, Nantong 226631, China
| | - Hui-Min Wang
- Cancer Institute, Nantong Tumor Hospital, Nantong 226631, China
| | - Li-Peng Gu
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Su-Qing Zhang
- Department of Hepatobiliary Surgery, Nantong Tumor Hospital, Nantong 226631, China
| | - Gao-Ren Wang
- Cancer Institute, Nantong Tumor Hospital, Nantong 226631, China
| | - Ji-Bin Liu
- Cancer Institute, Nantong Tumor Hospital, Nantong 226631, China.
| | - Da Fu
- Department of Radiology, The Fourth Affiliated Hospital of Anhui Medical University, Hefei 230012, China; Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Xiao-Qing Jiang
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical University, Shanghai 200438, China.
| |
Collapse
|
41
|
Liu JB, Chu KJ, Ling CC, Wu TM, Wang HM, Shi Y, Li ZZ, Wang JH, Wu ZJ, Jiang XQ, Wang GR, Ma YS, Fu D. Prognosis for intrahepatic cholangiocarcinoma patients treated with postoperative adjuvant transcatheter hepatic artery chemoembolization. Curr Probl Cancer 2020; 44:100612. [PMID: 32517878 DOI: 10.1016/j.currproblcancer.2020.100612] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/20/2020] [Accepted: 05/07/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVE We used meta-analysis to evaluate the efficacy of transcatheter hepatic arterial chemoembolization (TACE) for the treatment of intrahepatic cholangiocarcinoma (ICC). METHODS We performed the meta-analysis using the R 3.12 software and the quality evaluation of data using the Newcastle-Ottawa Scale. The main outcomes were recorded as 1-year overall survival (OS), 3-year OS, 5-year OS, and hazard ratio (HR) of TACE treatment or non-TACE treatment. The heterogeneity test was performed using the Q-test based on chi-square and I2 statistics. Egger's test was used to test the publication bias. The odds ratio or HR and 95% confidence interval (CI) were used to represent the effect index. RESULTS Nine controlled clinical trials involving 1724 participants were included in this study; patients came mainly from China, Italy, South Korea, and Germany. In the OS meta-analysis, the 1-year and 3-year OS showed significant heterogeneity, but not the 5-year OS. TACE increased the 1-year OS (odds ratio = 2.66, 95% CI: 1.10-6.46) of the patients with ICC, but the 3- and 5-year OS rates were not significantly increased. The results had no publication bias, but the stability was weak. The HR had significant heterogeneity (I2 = 0%, P= 0.54). TACE significantly decreased the HR of ICC patients (HR = 0.59, 95% CI: 0.48-0.73). The results had no publication bias, and the stability was good. CONCLUSIONS Treatment with TACE is effective for patients with ICC. Regular updating and further research and analysis still need to be carried out.
Collapse
Affiliation(s)
- Ji-Bin Liu
- Cancer Institute, Nantong Tumor Hospital, Nantong, China
| | - Kai-Jian Chu
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Chang-Chun Ling
- Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Ting-Miao Wu
- Department of Radiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui-Min Wang
- Cancer Institute, Nantong Tumor Hospital, Nantong, China; Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Shi
- Cancer Institute, Nantong Tumor Hospital, Nantong, China; Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhi-Zhen Li
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Jing-Han Wang
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Zhi-Jun Wu
- Department of Radiotherapy, Nantong Tumor Hospital, Nantong, China
| | - Xiao-Qing Jiang
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Gao-Ren Wang
- Department of Radiotherapy, Nantong Tumor Hospital, Nantong, China.
| | - Yu-Shui Ma
- Cancer Institute, Nantong Tumor Hospital, Nantong, China; Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Da Fu
- Department of Radiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China; Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
42
|
Cui Y, Liang S, Zhang S, Zhang C, Zhao Y, Wu D, Wang J, Song R, Wang J, Yin D, Liu Y, Pan S, Liu X, Wang Y, Han J, Meng F, Zhang B, Guo H, Lu Z, Liu L. ABCA8 is regulated by miR-374b-5p and inhibits proliferation and metastasis of hepatocellular carcinoma through the ERK/ZEB1 pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:90. [PMID: 32430024 PMCID: PMC7236190 DOI: 10.1186/s13046-020-01591-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/06/2020] [Indexed: 02/07/2023]
Abstract
Background ATP binding cassette subfamily A member 8 (ABCA8) belongs to the ATP binding cassette (ABC) transporter superfamily. ABCA8 is a transmembrane transporter responsible for the transport of organics, such as cholesterol, and drug efflux. Some members of the ABC subfamily, such as ABCA1, may inhibit cancer development. However, the mechanism of ABCA8 in the process of cancer activation is still ambiguous. Methods The expression of ABCA8 in human hepatocellular carcinoma (HCC) tissues and cell lines was examined using qPCR, immunoblotting, and immunohistochemical staining. The effects of ABCA8 on the proliferation and metastasis of HCC were examined using in vitro and in vivo functional tests. A luciferase reporter assay was performed to explore the binding between microRNA-374b-5p (miR-374b-5p) and the ABCA8 3′-untranslated region (UTR). Results ABCA8 was frequently down-regulated in HCC and this down-regulation was negatively correlated with prognosis. The overexpression of ABCA8 inhibited growth and metastasis in HCC, whereas the knockdown of ABCA8 exerted the antithetical effects both in vivo and in vitro. ABCA8 was down-regulated by miR-374b-5p; this down-regulation can induce epithelial transformation to mesenchyme via the ERK/ZEB1 signaling pathway and promote HCC progression. Conclusion We exposed the prognostic value of ABCA8 in HCC, and illuminated a novel pathway in ABCA8-regulated inhibition of HCC tumorigenesis and metastasis. These findings may lead to a new targeted therapy for HCC through the regulation of ABCA8, and miR-374b-5p.
Collapse
Affiliation(s)
- Yifeng Cui
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Shuhang Liang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Shugeng Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Congyi Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Yunzheng Zhao
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Dehai Wu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Ruipeng Song
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Jizhou Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Dalong Yin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Shangha Pan
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Xirui Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Yan Wang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jihua Han
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Fanzheng Meng
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Bo Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Hongrui Guo
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China
| | - Zhaoyang Lu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China. .,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China.
| | - Lianxin Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China. .,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, China. .,Department of Hepatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
43
|
Wang S, Liu X, Khan AA, Li H, Tahir M, Yan X, Wang J, Huang H. miR-216a-mediated upregulation of TSPAN1 contributes to pancreatic cancer progression via transcriptional regulation of ITGA2. Am J Cancer Res 2020; 10:1115-1129. [PMID: 32368389 PMCID: PMC7191091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/05/2020] [Indexed: 06/11/2023] Open
Abstract
Pancreatic cancer (PC) is recognized as the most aggressive and deadliest malignancy because it has the highest mortality of all cancers in humans. Mutations in multiple tumor suppressors and oncogenes have been documented to be involved in pancreatic cancer progression and metastasis. The upregulation of tetraspanin 1 (TSPAN1), a transmembrane protein, has been reportedly observed in many human cancers. However, the role of TSPAN1 and its underlying molecular mechanisms in PC progression have not been fully elucidated. In this study, we validated the oncogenic role of TSPAN1 in PC, showing that TSPAN1 reinforces cell proliferation, migration, invasion and tumorigenesis. To investigate the upregulation of TSPAN1 in PC, we showed that miR-216a is the upstream negative regulator of TSPAN1 via direct binding to the TSPAN1 3'-untranslated region. Through RNA-Seq analysis, we for the first time revealed that TSPAN1 expression transcriptionally regulates ITGA2, which is involved in the actin cytoskeleton pathway. The stimulated cell proliferation and invasion initiated by TSPAN1 overexpression could be abolished by knockdown of ITGA2 in PC cells. Furthermore, TSPAN1 epigenetically regulates the expression of ITGA2 by modulating the levels of TET2 DNMT3B and DNMT1, resulting in hypomethylation of the CpG island of the ITGA2 promoter. In conclusion, the newly identified miR-216a/TSPAN1/ITGA2 axis is involved in the modulation of PC progression and represents a novel therapeutic strategy for future pancreatic cancer treatment.
Collapse
Affiliation(s)
- Shensen Wang
- College of Life Science and Bioengineering, Beijing University of Technology Beijing 100124, China
| | - Xinhui Liu
- College of Life Science and Bioengineering, Beijing University of Technology Beijing 100124, China
| | - Aamir Ali Khan
- College of Life Science and Bioengineering, Beijing University of Technology Beijing 100124, China
| | - Huan Li
- College of Life Science and Bioengineering, Beijing University of Technology Beijing 100124, China
| | - Muhammad Tahir
- College of Life Science and Bioengineering, Beijing University of Technology Beijing 100124, China
| | - Xinlong Yan
- College of Life Science and Bioengineering, Beijing University of Technology Beijing 100124, China
| | - Juan Wang
- College of Life Science and Bioengineering, Beijing University of Technology Beijing 100124, China
| | - Hua Huang
- College of Life Science and Bioengineering, Beijing University of Technology Beijing 100124, China
| |
Collapse
|
44
|
Miwa T, Kanda M, Umeda S, Tanaka H, Shimizu D, Tanaka C, Kobayashi D, Hayashi M, Yamada S, Nakayama G, Koike M, Kodera Y. Establishment of Peritoneal and Hepatic Metastasis Mouse Xenograft Models Using Gastric Cancer Cell Lines. In Vivo 2020; 33:1785-1792. [PMID: 31662503 DOI: 10.21873/invivo.11669] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIM Establishment of mouse xenograft models is necessary for oncological research and depends on the characteristics of the cell lines and the immune system of the host. In this study, we describe the development of mouse xenograft models using human gastric cancer (GC) cell lines. MATERIALS AND METHODS MKN1 stably-expressing luciferase (MKN1-Luc), N87, KATO III, MKN45 stably-expressing luciferase (MKN45-Luc), NUGC4, and OCUM-1 human GC cell lines were injected intraperitoneally into mice to establish peritoneal metastasis models. MKN45-Luc were injected into subcutaneously implanted spleen, and MKN1-Luc and MKN45-Luc were injected directly into the portal veins of mice for the establishment of hepatic metastasis models. RESULTS Peritoneal metastasis was formed after implantation of MKN1-Luc, N87, KATO III, MKN45-Luc, and NUGC4 in nude mice, but not formed in OCUM-1 even in NOD/SCID mice. After intrasplenic injection of MKN45-Luc, we found no hepatic metastasis formation. We identified hepatic metastasis formation after direct injection of MKN45-Luc and MKN1-Luc into the portal veins of NOD/SCID mice. CONCLUSION Peritoneal and hepatic metastasis mouse xenograft models were successfully established using several human GC cell lines.
Collapse
Affiliation(s)
- Takashi Miwa
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Haruyoshi Tanaka
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Kobayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Suguru Yamada
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Goro Nakayama
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiko Koike
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
45
|
Wang P, Xu LL, Zheng XB, Hu YT, Zhang JF, Ren SS, Hao XY, Li L, Zhang M, Xu MQ. Correlation between the expressions of circular RNAs in peripheral venous blood and clinicopathological features in hepatocellular carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:338. [PMID: 32355782 PMCID: PMC7186655 DOI: 10.21037/atm.2020.02.134] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background Recent studies have reported that circular RNAs (circRNAs) are involved in the development of hepatocellular carcinoma (HCC). This study evaluated the expression of preoperative peripheral venous blood circRNAs in HCC patients and their predictive ability for microvascular invasion (MVI). Methods Seven circRNAs (circMTO1, circ-10720, circZKSCAN1, cSMARCA5, circHIPK3, circSETD3 and ciRS-7) were screened from the literature as circRNAs with reported biological functions in HCC. The expression levels of seven circRNAs in preoperative blood samples and HCC tissues were detected by quantitative reverse transcription polymerase chain reaction. The correlations between the circRNA expressions in blood and the clinicopathological factors of HCC patients were analyzed. The risk factors of MVI were analyzed by univariate and multivariate logistic regression. The functional role of circSETD3 in cell migration and invasion was evaluated by wound healing and Transwell assays in vitro. Results The expressions of all seven circRNAs were measured in peripheral venous blood samples. The venous expression levels of circHIPK3 and circMTO1 were significantly associated with gender, while circ-10720 and circMTO1 levels were significantly correlated with gross vascular invasion. Furthermore, circMTO1 and cSMARCA5 levels were significantly associated with alpha-fetoprotein level and ciRS-7 was significantly associated with satellite nodules. Importantly, low venous circSETD3 expression was significantly associated with prothrombin induced by vitamin K absence or antagonist-II (PIVKA-II) level, MVI, gross vascular invasion, and liver capsule. Furthermore, venous circSETD3 expression had predictive ability for MVI. Knockdown of circSETD3 promoted cell invasion and metastasis in vitro. Conclusions CircRNAs were stably present in peripheral venous blood and associated with multiple clinicopathological characteristics of HCC patients. Venous circSETD3 was an independent risk factor of MVI and shows ability to predict MVI in HCC patients before surgery.
Collapse
Affiliation(s)
- Peng Wang
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang-Liang Xu
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiao-Bo Zheng
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi-Tao Hu
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin-Fu Zhang
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Sheng-Sheng Ren
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiang-Yong Hao
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Lian Li
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ming Zhang
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of General Surgery, Mianzhu Hospital of West China Hospital, Sichuan University, Mianzhu 618200, China
| | - Ming-Qing Xu
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
46
|
Lan X, Lin W, Xu Y, Xu Y, Lv Z, Chen W. The detection and analysis of differential regulatory communities in lung cancer. Genomics 2020; 112:2535-2540. [PMID: 32045668 DOI: 10.1016/j.ygeno.2020.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/06/2020] [Accepted: 02/07/2020] [Indexed: 02/07/2023]
Abstract
The tumorgenesis process of lung cancer involves the regulatory dysfunctions of multiple pathways. Although many signaling pathways have been identified to be associated with lung cancer, there are little quantitative models of how inactions between genes change during the process from normal to cancer. These changes belong to different dynamic co-expressions patterns. We quantitatively analyzed differential co-expression of gene pairs in four datasets. Each dataset included a large number of lung cancer and normal samples. By overlapping their results, we got 14 highly confident gene pairs with consistent co-expression change patterns. Some of they, such as ARHGAP30 and GIMAP4, had been recorded in STRING network database while some of them were novel discoveries, such as C9orf135 and MORN5, TEKT1 and TSPAN1 were positively correlated in both normal and cancer but more correlated in normal than cancer. These gene pairs revealed the underlying mechanisms of lung cancer occurrence.
Collapse
Affiliation(s)
- Xiu Lan
- Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Weilong Lin
- Department of Orthopedics, Lishui Traditional Chinese Medicine Hospital, Lishui, China
| | - Yufen Xu
- Department of Oncology, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing, China; Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Yanyan Xu
- Department of Pharmacy, Lishui Central Hospital, Lishui, China
| | - Zhuqing Lv
- Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Wenyu Chen
- Department of Respiration, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing, China.
| |
Collapse
|
47
|
Chen C, Li H, Wang X, Wang L, Zeng Q. Lnc-LFAR1 affects intrahepatic cholangiocarcinoma proliferation, invasion, and EMT by regulating the TGFβ/Smad signaling pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2455-2461. [PMID: 31934072 PMCID: PMC6949567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/25/2019] [Indexed: 06/10/2023]
Abstract
Intrahepatic cholangiocarcinoma (ICC) has had an increasing incidence in recent years. It exhibits high recurrence and metastasis rates, so it is a global health problem. LncRNAs have long been thought to have a role in regulating tumorigenesis and tumor progression and in playing a carcinogenic or tumor suppressor role in malignant cells. Lnc-LFAR1 is enriched in liver tissue, but its role and mechanism in ICC have not been elucidated. ICC cell line QBC939 cells were randomly divided into 3 groups, including a control group, an lnc-LFAR1 overexpression group, and an lnc-LFAR1 siRNA group, the cells of which were transfected with pcDNA3.1-LFAR1 plasmid and lnc-LFAR1 siRNA, respectively. Real-time PCR was used to quantify lnc-LFAR1 expression. A tetrazolium salt (MTT) colorimetric assay was adopted to assess cell viability. A cell scratch assay was selected to assess cell migration. A transwell chamber assay was used to test cell invasion. E-cadherin and vimentin expressions were detected using Western blot. The TGFβ/Smad signaling pathway was measured using real-time PCR. The transfection of pcDNA3.1-LFAR1 significantly increased the expression of lnc-LFAR1, promoted cell proliferation, facilitated cell migration and invasion, reduced E-cadherin levels, enhanced vimentin expression, upregulated TGF-β1 and Smad2 and Smad4 expressions (P < 0.05). Lnc-LFAR1 siRNA transfection clearly downregulated lnc-LFAR1 expression, inhibited cell proliferation, suppressed cell migration and invasion, increased E-cadherin expression, and decreased vimentin, TGF-β1, Smad2, and Smad4 levels (P < 0.05). Targeting lnc-LFAR1 can inhibit cell proliferation, migration, invasion, and EMT by regulating the TGFβ/Smad pathway to affect the occurrence and development of cholangiocarcinoma.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal UniversityChangsha, Hunan, China
| | - Hao Li
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal UniversityChangsha, Hunan, China
| | - Xintian Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal UniversityChangsha, Hunan, China
| | - Lei Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal UniversityChangsha, Hunan, China
| | - Qinghua Zeng
- Department of Nephrology and Immunology Center, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal UniversityChangsha, Hunan, China
| |
Collapse
|