1
|
Jin H, Park SY, Lee JE, Park H, Jeong M, Lee H, Cho J, Lee YS. GTSE1-driven ZEB1 stabilization promotes pulmonary fibrosis through the epithelial-to-mesenchymal transition. Mol Ther 2024; 32:4138-4157. [PMID: 39342428 PMCID: PMC11573610 DOI: 10.1016/j.ymthe.2024.09.029] [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: 04/14/2024] [Revised: 08/06/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024] Open
Abstract
G2 and S phase-expressed protein 1 (GTSE1) has been implicated in the development of pulmonary fibrosis (PF); however, its biological function, molecular mechanism, and potential clinical implications remain unknown. Here, we explored the genomic data of patients with idiopathic PF (IPF) and found that GTSE1 expression is elevated in their lung tissues, but rarely expressed in normal lung tissues. Thus, we explored the biological role and downstream events of GTSE1 using IPF patient tissues and PF mouse models. The comprehensive bioinformatics analyses suggested that the increase of GTSE1 in IPF is linked to the enhanced gene signature for the epithelial-to-mesenchymal transition (EMT), leading us to investigate the potential interaction between GTSE1 and EMT transcription factors. GTSE1 preferentially binds to the less stable form of zinc-finger E-box-binding homeobox 1 (ZEB1), the unphosphorylated form at Ser585, inhibiting ZEB1 degradation. Consistently, the ZEB1 protein level in IPF patient and PF mouse tissues correlates with the GTSE1 protein level and the amount of collagen accumulation, representing fibrosis severity. Collectively, our findings highlight the GTSE1-ZEB1 axis as a novel driver of the pathological EMT characteristic during PF development and progression, supporting further investigation into GTSE1-targeting approaches for PF treatment.
Collapse
Affiliation(s)
- Hee Jin
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - So-Yeon Park
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea; Center for Genome Engineering, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Ji Eun Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Hangyeol Park
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Michaela Jeong
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Hyukjin Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University Health System, Seoul 120-749, Republic of Korea
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea.
| |
Collapse
|
2
|
El-Hussieny M, Thabet DM, Tawfik HM, Gayyed MF, Toni ND. The Overexpression of NUSAP1 and GTSE1 Could Predict An Unfavourable Prognosis and Shorter Disease Free Survival in ccRenal Cell Carcinoma. Asian Pac J Cancer Prev 2024; 25:2551-2559. [PMID: 39068590 PMCID: PMC11480612 DOI: 10.31557/apjcp.2024.25.7.2551] [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: 04/16/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Although it has been reported that NUSAP1 and GTSE1 are highly expressed in different types of tumors and associated with malignant progression and poor clinical prognosis, their significances with clinicopathological data and correlations with patients' survival in ccRCC are still poorly understood. Therefore, in our study we attempted to evaluate the link between NUSAP1 and GTSE1 in ccRCC and to correlate their immunoexpression with clinico-pathological parameters and the patients' survival to identify their significance as potential therapeutic targets, indicators for tumor progression, and patients' prognosis. METHOD NUSAP1 and GTSE1 were examined in 100 ccRCC patients by immunohistochemistry. The association between NUSAP1 and GTSE1 immunoreactivity and clinicopathological variables were evaluated. The disease free survival (DFS) was examined by the Kaplan-Meier method. The multivariate Cox regressions was estimated to detect the prognostic role of both proteins. RESULTS We detected high NUSAP1 and GTSE1 expression in 60% and 62% of the cases, respectively. A significant association was detected between NUSAP1 and GTSE1 immunoexpression and size (p=0.007 and p=0.026, respectively), Fuhrman grade (p=0.022 and p=0.004, respectively), tumor stage (p=0.003 and p=0.019, respectively), TILs (p=0.026 and p=0.04 respectively), capsular invasion (p=0.002 and p=0.009, respectively), Distant metastasis (p=0.007 and p=0.009, respectively), and DFS (p=0.007 and 0.009, respectively). Multivariate Cox regression showed that high NUSAP1 and GTSE1 expression levels were independently associated with an unfavourable poor prognosis of ccRCC cases. CONCLUSION We demonstrated that NUSAP1 and GTSE1 overexpression was closely related to the poor prognostic clinicopathological features of ccRCC and predicted an unfavorable prognosis. Therefore, NUSAP1 and GTSE1 might act together as potential futuristic prognostic indicators and therapeutic targets for ccRCC patients. However, further analysis in molecular studies on larger scale are mandatory to highlight the interactive crosstalk regulatory mechanisms between both markers and their combined effect on ccRCC.
Collapse
Affiliation(s)
| | - Dalia M. Thabet
- Department of Pathology, Faculty of Medicine, Minia University 61511, El-Minia, Egypt.
| | | | | | | |
Collapse
|
3
|
Wu J, Yu F, Di Z, Bian L, Yang J, Wang L, Jiang Q, Yin Y, Zhang L. Transcriptome analysis of adipose tissue and muscle of Laiwu and Duroc pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:134-143. [PMID: 38766520 PMCID: PMC11101945 DOI: 10.1016/j.aninu.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/10/2023] [Accepted: 12/15/2023] [Indexed: 05/22/2024]
Abstract
Fat content is an important trait in pig production. Adipose tissue and muscle are important sites for fat deposition and affect production efficiency and quality. To regulate the fat content in these tissues, we need to understand the mechanisms behind fat deposition. Laiwu pigs, a Chinese indigenous breed, have significantly higher fat content in both adipose tissue and muscle than commercial breeds such as Duroc. In this study, we analyzed the transcriptomes in adipose tissue and muscle of 21-d-old Laiwu and Duroc piglets. Results showed that there were 828 and 671 differentially expressed genes (DEG) in subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT), respectively. Functional enrichment analysis showed that these DEG were enriched in metabolic pathways, especially carbohydrate and lipid metabolism. Additionally, in the longissimus muscle (LM) and psoas muscle (PM), 312 and 335 DEG were identified, demonstrating enrichment in the cell cycle and metabolic pathways. The protein-protein interaction (PPI) networks of these DEG were analyzed and potential hub genes were identified, such as FBP1 and SCD in adipose tissues and RRM2 and GADL1 in muscles. Meanwhile, results showed that there were common DEG between adipose tissue and muscle, such as LDHB, THRSP, and DGAT2. These findings showed that there are significant differences in the transcriptomes of the adipose tissue and muscle between Laiwu and Duroc piglets (P < 0.05), especially in metabolic patterns. This insight serves to advance our comprehensive understanding of metabolic regulation in these tissues and provide targets for fat content regulation.
Collapse
Affiliation(s)
- Jie Wu
- National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Fangyuan Yu
- National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zhaoyang Di
- National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Liwen Bian
- National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jie Yang
- National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Lina Wang
- National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Qingyan Jiang
- National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Lin Zhang
- National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| |
Collapse
|
4
|
Chen H, Lu Y, Dai Z, Yang Y, Li Q, Rao Y. Comprehensive single-cell RNA-seq analysis using deep interpretable generative modeling guided by biological hierarchy knowledge. Brief Bioinform 2024; 25:bbae314. [PMID: 38960404 PMCID: PMC11221887 DOI: 10.1093/bib/bbae314] [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: 10/13/2023] [Revised: 12/13/2023] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
Recent advances in microfluidics and sequencing technologies allow researchers to explore cellular heterogeneity at single-cell resolution. In recent years, deep learning frameworks, such as generative models, have brought great changes to the analysis of transcriptomic data. Nevertheless, relying on the potential space of these generative models alone is insufficient to generate biological explanations. In addition, most of the previous work based on generative models is limited to shallow neural networks with one to three layers of latent variables, which may limit the capabilities of the models. Here, we propose a deep interpretable generative model called d-scIGM for single-cell data analysis. d-scIGM combines sawtooth connectivity techniques and residual networks, thereby constructing a deep generative framework. In addition, d-scIGM incorporates hierarchical prior knowledge of biological domains to enhance the interpretability of the model. We show that d-scIGM achieves excellent performance in a variety of fundamental tasks, including clustering, visualization, and pseudo-temporal inference. Through topic pathway studies, we found that d-scIGM-learned topics are better enriched for biologically meaningful pathways compared to the baseline models. Furthermore, the analysis of drug response data shows that d-scIGM can capture drug response patterns in large-scale experiments, which provides a promising way to elucidate the underlying biological mechanisms. Lastly, in the melanoma dataset, d-scIGM accurately identified different cell types and revealed multiple melanin-related driver genes and key pathways, which are critical for understanding disease mechanisms and drug development.
Collapse
Affiliation(s)
- Hegang Chen
- School of Computer Science and Engineering, Sun Yat-sen University, 132 Waihuan East Road, Guangzhou University Town, 510006, Guangzhou, China
| | - Yuyin Lu
- School of Computer Science and Engineering, Sun Yat-sen University, 132 Waihuan East Road, Guangzhou University Town, 510006, Guangzhou, China
| | - Zhiming Dai
- School of Computer Science and Engineering, Sun Yat-sen University, 132 Waihuan East Road, Guangzhou University Town, 510006, Guangzhou, China
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, 132 Waihuan East Road, Guangzhou University Town, 510006, Guangzhou, China
| | - Qing Li
- Department of Computing, The Hong Kong Polytechnic University, PQ806, Mong Man Wai Building, 999077, Hong Kong SAR
| | - Yanghui Rao
- School of Computer Science and Engineering, Sun Yat-sen University, 132 Waihuan East Road, Guangzhou University Town, 510006, Guangzhou, China
| |
Collapse
|
5
|
Dong J, Chen J, Wu Y, Yan J. GTSE1 promotes nasopharyngeal carcinoma proliferation and angiogenesis by upregulating STMN1. Cell Div 2024; 19:16. [PMID: 38698443 PMCID: PMC11064356 DOI: 10.1186/s13008-024-00119-9] [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: 10/27/2023] [Accepted: 04/11/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a malignant tumor with poor survival rate. G2 and S phase-expressed-1 (GTSE1) takes part in the progression of diverse tumors as an oncogene, but its role and potential mechanism in NPC remain unknown. METHODS The GTSE1 expression was analyzed by western blot in NPC tissues and cells. Knock-down experiments were conducted to determine the function of GTSE1 in NPC by cell counting kit-8, the 5-ethynyl-2'-deoxyuridine (EdU) incorporation experiment, cell scratch wound-healing experiment, transwell assays, tube forming experiment and western blot. In addition, the in vivo role of GTSE1 was addressed in tumor-bearing mice. RESULTS The expression of was increased in NPC. Silencing of GTSE1 suppressed cell viability, the percent of EdU positive cells, and the number of invasion cells and tubes, but enhanced the scratch ratio in NPC cells. Mechanically, downregulation of GTSE1 decreased the expressions of FOXM1 and STMN1, which were restored with the upregulation of FOXM1. Increased expression of STMN1 reversed the effects of the GTSE1 silencing on proliferation, migration, invasion and angiogenesis of NPC cells. Furthermore, knockdown of GTSE1 repressed the tumor volume and tumor weight of xenografted mice. CONCLUSION GTSE1 was highly expressed in NPC, and silencing of GTSE1 ameliorated the malignant processes of NPC cells by upregulating STMN1, suggesting a possible therapeutical target for NPC.
Collapse
Affiliation(s)
- Jiadi Dong
- Department of Otorhinolaryngology Head and Neck Surger, Ningbo Medical Center Lihuili Hospital, No. 57, Xingning, Yinzhou, 315000, Zhejiang, China
| | - Jingjing Chen
- Department of Otorhinolaryngology Head and Neck Surger, Ningbo Medical Center Lihuili Hospital, No. 57, Xingning, Yinzhou, 315000, Zhejiang, China.
| | - Yidong Wu
- Department of Otorhinolaryngology Head and Neck Surger, Ningbo Medical Center Lihuili Hospital, No. 57, Xingning, Yinzhou, 315000, Zhejiang, China
| | - Jiangyu Yan
- Department of Otorhinolaryngology Head and Neck Surger, Ningbo Medical Center Lihuili Hospital, No. 57, Xingning, Yinzhou, 315000, Zhejiang, China
| |
Collapse
|
6
|
Nie J, Yang H, Liu X, Deng W, Fu B. Identification and validation of shared gene signature of kidney renal clear cell carcinoma and COVID-19. PeerJ 2024; 12:e16927. [PMID: 38464749 PMCID: PMC10921934 DOI: 10.7717/peerj.16927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/22/2024] [Indexed: 03/12/2024] Open
Abstract
Background COVID-19 is a severe infectious disease caused by the SARS-CoV-2 virus, and previous studies have shown that patients with kidney renal clear cell carcinoma (KIRC) are more susceptible to SARS-CoV-2 infection than the general population. Nevertheless, their co-pathogenesis remains incompletely elucidated. Methods We obtained shared genes between these two diseases based on public datasets, constructed a prognostic risk model consisting of hub genes, and validated the accuracy of the model using internal and external validation sets. We further analyzed the immune landscape of the prognostic risk model, investigated the biological functions of the hub genes, and detected their expression in renal cell carcinoma cells using qPCR. Finally, we searched the candidate drugs associated with hub gene-related targets from DSigDB and CellMiner databases. Results We obtained 156 shared genes between KIRC and COVID-19 and constructed a prognostic risk model consisting of four hub genes. Both shared genes and hub genes were highly enriched in immune-related functions and pathways. Hub genes were significantly overexpressed in COVID-19 and KIRC. ROC curves, nomograms, etc., showed the reliability and robustness of the risk model, which was validated in both internal and external datasets. Moreover, patients in the high-risk group showed a higher proportion of immune cells, higher expression of immune checkpoint genes, and more active immune-related functions. Finally, we identified promising drugs for COVID-19 and KIRC, such as etoposide, fulvestrant, and topotecan. Conclusion This study identified and validated four shared genes for KIRC and COVID-19. These genes are associated with immune functions and may serve as potential prognostic biomarkers for KIRC. The shared pathways and genes may provide new insights for further mechanistic research and treatment of comorbidities.
Collapse
Affiliation(s)
- Jianqiang Nie
- First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hailang Yang
- First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqiang Liu
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| | - Wen Deng
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| | - Bin Fu
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| |
Collapse
|
7
|
Geng R, Kang SG, Huang K, Tong T. Dietary supplementation with α-ionone alleviates chronic UVB exposure-induced skin photoaging in mice. Food Funct 2024; 15:1884-1898. [PMID: 38328833 DOI: 10.1039/d3fo04379g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Photoaging is widely regarded as the most significant contributor to skin aging damage. It is triggered by prolonged exposure to ultraviolet (UV) light and typically manifests as dryness and the formation of wrinkles. Nutritional intervention is a viable strategy for preventing and treating skin photoaging. In previous studies, we demonstrated that α-ionone had ameliorating effects on photoaging in both epidermal keratinocytes and dermal fibroblasts. Here, we investigated the potential anti-photoaging effects of dietary α-ionone using a UVB-irradiated male C57BL/6N mouse model. Our findings provided compelling evidence that dietary α-ionone alleviates wrinkle formation, skin dryness, and epidermal thickening in chronic UVB-exposed mice. α-Ionone accumulated in mouse skin after 14 weeks of dietary intake of α-ionone. α-Ionone increased collagen density and boosted the expression of collagen genes, while attenuating the UVB-induced increase of matrix metalloproteinase genes in the skin tissues. Furthermore, α-ionone suppressed the expression of senescence-associated secretory phenotypes and reduced the expression of the senescence marker p21 and DNA damage marker p53 in the skin of UVB-irradiated mice. Transcriptome sequencing results showed that α-ionone modifies gene expression profiles of skin. Multiple pathway enrichment analyses on both the differential genes and the entire genes revealed that α-ionone significantly affects multiple physiological processes and signaling pathways associated with skin health and diseases, of which the p53 signaling pathway may be the key signaling pathway. Taken together, our findings reveal that dietary α-ionone intervention holds promise in reducing the risks of skin photoaging, offering a potential strategy to address skin aging concerns.
Collapse
Affiliation(s)
- Ruixuan Geng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, PR China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, PR China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun 58554, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, PR China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, PR China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, PR China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, PR China
| |
Collapse
|
8
|
Jin L, Duan Y, Li X, Li Z, Hu J, Shi H, Su Z, Li Z, Du B, Chen Y, Tan Y. High expression ITGA2 affects the expression of MET, PD-L1, CD4 and CD8 with the immune microenvironment in pancreatic cancer patients. Front Immunol 2023; 14:1209367. [PMID: 37881431 PMCID: PMC10594995 DOI: 10.3389/fimmu.2023.1209367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/22/2023] [Indexed: 10/27/2023] Open
Abstract
Purpose Pancreatic cancer is characterized by a grim prognosis and is regarded as one of the most formidable malignancies. Among the genes exhibiting high expression in different tumor tissues, ITGA2 stands out as a promising candidate for cancer therapy. The promotion of cancer in pancreatic cancer is not effective. The objective of this study is to assess the presence of ITGA2, EMT and PD-L1 in pancreatic cancer. Experimental design We examined the expression of ITGA2, MET, E-cadherin, PD-L1, CD4, and CD8 proteins in 62 pancreatic cancer tissue samples using multi-tissue immunofluorescence and immunohistochemistry techniques. Functional assays, such as the cell migration assay and transwell assay, were used to determine the biological role of ITGA2 in pancreatic cancer. The relationship of ITGA2,EMT and PD-L1 were examined using Western blot analysis and RT-qPCR assay. Results In our study, we observed the expression of ITGA2, E-cadherin, and PD-L1 in both tumor and stroma tissues of pancreatic cancer. Additionally, a positive correlation between ITGA2, E-cadherin, and PD-L1 in the tumor region (r=0.559, P<0.001 and r=0.511, P<0.001), and PD-L1 in the stroma region (r=0.512, P<0.001).The expression levels of ITGA2, CD4, and CD8 were found to be higher in pancreatic cancer tissues compared to adjacent tissues (P < 0.05). Additionally, ITGA2 was negatively correlated with CD4 and CD8 (r = -0.344, P < 0.005 and r = -0.398, P < 0.005).Furthermore, ITGA2, CD4, and CD8 were found to be correlated with the survival time of patients (P < 0.05). Blocking ITGA2 inhibited the proliferation and invasion ability of pancreatic cancer cells significantly, Additionally, sh-ITGA2 can down-regulate the expression of EMT and PD-L1. Conclusions We identified a novel mechanism in which ITGA2 plays a crucial role in the regulation of pancreatic cancer growth and invasion. This mechanism involves the upregulation of MET and PD-L1 expression in pancreatic cancer cells. Additionally, we found that increased expression of ITGA2 is associated with a poor prognosis in pancreatic cancer patients. Furthermore, ITGA2 also affects immune regulation in these patients. Therefore, targeting ITGA2 is an effective method to enhance the efficacy of checkpoint immunotherapy and prohibiting tumor growth against pancreatic cancer.
Collapse
Affiliation(s)
- Liquan Jin
- 1St Department of General Surgery, The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Yaoqiang Duan
- Clinical Medical College of Dali University, Dali, Yunnan, China
| | - Xiaoxi Li
- Clinical Medical College of Dali University, Dali, Yunnan, China
| | - Zhenqi Li
- Clinical Medical College of Dali University, Dali, Yunnan, China
| | - Jifu Hu
- Clinical Medical College of Dali University, Dali, Yunnan, China
| | - Hongbo Shi
- 1St Department of General Surgery, The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Ziting Su
- 1St Department of General Surgery, The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Zhe Li
- Clinical Medical College of Dali University, Dali, Yunnan, China
| | - Bilian Du
- Clinical Medical College of Dali University, Dali, Yunnan, China
| | - Yiming Chen
- 1St Department of General Surgery, The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Yunbo Tan
- 1St Department of General Surgery, The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| |
Collapse
|
9
|
Wang C, Wen M, Xu J, Gao P, Liu S, Liu J, Chen Y, Zhou L. GTSE1 promotes the growth of NSCLC by regulating microtubule-associated proteins through the ERK/MAPK pathway. Thorac Cancer 2023; 14:1624-1634. [PMID: 37079439 PMCID: PMC10260487 DOI: 10.1111/1759-7714.14908] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/21/2023] Open
Abstract
The role of G2 and S phase-expressed-1 (GTSE1), a microtubule-localized protein, in non-small-cell lung cancer (NSCLC) remains unknown. We explored its role in NSCLC growth. GTSE1 was detected in NSCLC tissues and cell lines using quantitative real-time polymerase chain reaction. The clinical significance of GTSE1 levels was evaluated. Biological and apoptotic effects of GTSE1 were evaluated using transwell, cell-scratch, and MTT assays, and flow cytometry and western blotting, respectively. Its association with cellular microtubules was shown by western blotting and immunofluorescence. GTSE1 expression was upregulated in NSCLC tissues and cell lines. GTSE1 levels correlated with lymph node metastasis. Higher GTSE1 mRNA expression correlated with shorter progression-free survival. GTSE1-knockdown decreased proliferation, colony formation, invasion, and migration of NSCLC cells, and inhibited tau and stathmin-1 microtubule-associated protein expression, via the extracellular-regulated protein kinase/mitogen-activated protein kinase (ERK/MAPK) signaling pathway, and microtubule disruption. GTSE1 may promote NSCLC growth by regulating tau and stathmin-1 through the ERK/MAPK signaling pathway.
Collapse
Affiliation(s)
- Chuanlin Wang
- Department of Clinical NutritionYunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Department of Clinical NutritionYunnan Cancer CenterKunmingChina
| | - Meiyan Wen
- Department of Clinical NutritionYunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Department of Clinical NutritionYunnan Cancer CenterKunmingChina
| | - Jiali Xu
- Department of Clinical NutritionYunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Department of Clinical NutritionYunnan Cancer CenterKunmingChina
| | - Pengning Gao
- Department of Clinical NutritionYunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Department of Clinical NutritionYunnan Cancer CenterKunmingChina
| | - Shanling Liu
- Department of Clinical NutritionYunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Department of Clinical NutritionYunnan Cancer CenterKunmingChina
| | - Jiayu Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, School of Public HealthAnhui Medical UniversityHefeiChina
| | - Ying Chen
- Department of Clinical NutritionYunnan Cancer CenterKunmingChina
- Department of Thoracic SurgeryYunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Lan Zhou
- Department of Clinical NutritionYunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Department of Clinical NutritionYunnan Cancer CenterKunmingChina
| |
Collapse
|
10
|
Chen W, Wang H, Lu Y, Huang Y, Xuan Y, Li X, Guo T, Wang C, Lai D, Wu S, Zhao W, Mai H, Li H, Wang B, Ma X, Zhang X. GTSE1 promotes tumor growth and metastasis by attenuating of KLF4 expression in clear cell renal cell carcinoma. J Transl Med 2022; 102:1011-1022. [PMID: 36775416 DOI: 10.1038/s41374-022-00797-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 11/09/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most common malignant tumors and is characterized by a poor prognosis. Although G2- and S -phase expressed-1 (GTSE1) is known to be involved in the progression and metastasis of various cancers, its significance and mechanism in ccRCC remain unknown. In the present study, we found that GTSE1 was overexpressed in ccRCC tissues, especially in metastatic samples. Moreover, high GTSE1 expression was positively correlated with higher pT stage, tumor size, clinical stage, and WHO/ISUP grade and worse prognosis. And GTSE1 expression served as an independent prognostic factor for overall survival (OS). In addition, GTSE1 knockdown inhibited ccRCC cell proliferation, migration, and invasion, and enhanced cell apoptosis in vitro and in vivo. GTSE1 was crucial for epithelial-mesenchymal transition (EMT) in ccRCC. Mechanistically, GTSE1 depletion could upregulate the expression of Krüppel-like factor 4 (KLF4), which acts as a tumor suppressor in ccRCC. Downregulation of KLF4 effectively rescued the inhibitory effect induced by GTSE1 knockdown and reversed the EMT process. Overall, our results revealed that GTSE1 served as an oncogene regulating EMT through KLF4 in ccRCC, and that GTSE1 could also serve as a novel prognostic biomarker and may represent a promising therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Weihao Chen
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Hanfeng Wang
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yongliang Lu
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Yan Huang
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yundong Xuan
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Xiubin Li
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Tao Guo
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Paediatrics, the Seventh Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
| | - Chenfeng Wang
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Dong Lai
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Shengpan Wu
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Wenlei Zhao
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Haixing Mai
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Hongzhao Li
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Baojun Wang
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xin Ma
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xu Zhang
- Department of Urology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
11
|
Xie C, Xiang W, Shen H, Shen J. GTSE1 is possibly involved in the DNA damage repair and cisplatin resistance in osteosarcoma. J Orthop Surg Res 2021; 16:713. [PMID: 34876170 PMCID: PMC8650252 DOI: 10.1186/s13018-021-02859-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/24/2021] [Indexed: 11/18/2022] Open
Abstract
Background G2 and S phase-expressed-1 (GTSE1) negatively regulates the tumor-suppressive protein p53 and is potentially correlated with chemoresistance of cancer cells. This study aims to explore the effect of GTSE1 on the DNA damage repair and cisplatin (CDDP) resistance in osteosarcoma (OS). Materials and methods Expression of GTSE1 in OS was predicted in bioinformatics system GEPIA and then validated in clinically obtained tissues and acquired cell lines using RT-qPCR, immunohistochemical staining, and western blot assays. Gain- and loss-of-function studies of GTSE1 were performed in MG-63 and 143B cells to examine its function in cell cycle progression, DNA replication, and CDDP resistance. Stably transfected MG-63 cells were administrated into mice, followed by CDDP treatment to detect the role of GTSE1 in CDDP resistance in vivo. Results GTSE1 was highly expressed in patients with OS and correlated with poor survival according to the bioinformatics predictions. Elevated GTSE1 expression was detected in OS tissues and cell lines. GTSE1 silencing reduced S/G2 transition and DNA replication, and it increased the CDDP sensitivity and decreased the expression of DNA repair-related biomarkers in MG-63 cells. GTSE1 overexpression in 143B cells led to inverse trends. In vivo, downregulation of GTSE1 strengthened the treating effect of CDDP and significantly repressed growth of xenograft tumors in nude mice. However, overexpression of GTSE1 blocked the anti-tumor effect of CDDP. Conclusion This study demonstrates that GTSE1 is possibly involved in the DNA damage repair and cisplatin resistance in OS.
Collapse
Affiliation(s)
- Chaofan Xie
- Department of Orthopaedic, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, Guangdong, People's Republic of China.,Department of Orthopaedic, The Eighth Affiliated Hospital of Sun Yat-Sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Wei Xiang
- Department of Orthopaedic, The Eighth Affiliated Hospital of Sun Yat-Sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Huiyong Shen
- Department of Orthopaedic, The Eighth Affiliated Hospital of Sun Yat-Sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518033, Guangdong, People's Republic of China.
| | - Jingnan Shen
- Department of Muscularskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510000, Guangdong, People's Republic of China.
| |
Collapse
|
12
|
Bernardes SS, Ferreira I, Elder DE, Nobre AB, Martínez‐Said H, Adams DJ, Robles‐Espinoza CD, Possik PA. More than just acral melanoma: the controversies of defining the disease. J Pathol Clin Res 2021; 7:531-541. [PMID: 34213090 PMCID: PMC8503895 DOI: 10.1002/cjp2.233] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/13/2021] [Accepted: 06/09/2021] [Indexed: 01/17/2023]
Abstract
Acral melanoma (AM) is a malignant cutaneous melanocytic tumour specifically located on the palms, soles, and nail apparatus, which are areas of glabrous (hairless) skin. Acral lentiginous melanoma, a subtype of AM, represents a histopathological subtype diagnosis of cutaneous melanoma with unique morphological and structural features. Despite clear definitions, the misuse of these terms and the inconsistency in reporting the histopathological features of AM cases have become a major obstacle to the study of the disease. In this review, we discuss the epidemiology, histopathological features, prognosis, and genetic profile of AM, highlighting the differences observed when histopathological subtypes are considered. The increasing global effort to characterise AM cases from ethnically diverse populations would benefit greatly from a more consistent classification of the disease.
Collapse
Affiliation(s)
- Sara S Bernardes
- Program of Immunology and Tumour BiologyBrazilian National Cancer InstituteRio de JaneiroBrazil
- Tissue Microenvironment Laboratory, Department of General PathologyFederal University of Minas GeraisBelo HorizonteBrazil
| | - Ingrid Ferreira
- Experimental Cancer GeneticsWellcome Sanger InstituteHinxtonUK
- Université Libre de BruxellesBrusselsBelgium
| | - David E Elder
- Division of Anatomic PathologyHospital of the University of PennsylvaniaPhiladelphiaPAUSA
| | - Aretha B Nobre
- Division of PathologyBrazilian National Cancer InstituteRio de JaneiroBrazil
- Serviço de Patologia, Maternidade EscolaUniversidade Federal do Rio de JaneiroRio de JaneiroBrazil
| | - Héctor Martínez‐Said
- Servicio de Piel y Partes BlandasInstituto Nacional de CancerologíaCiudad de MéxicoMexico
| | - David J Adams
- Experimental Cancer GeneticsWellcome Sanger InstituteHinxtonUK
| | - Carla Daniela Robles‐Espinoza
- Experimental Cancer GeneticsWellcome Sanger InstituteHinxtonUK
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de MéxicoSantiago de QuerétaroMexico
| | - Patricia A Possik
- Program of Immunology and Tumour BiologyBrazilian National Cancer InstituteRio de JaneiroBrazil
| |
Collapse
|
13
|
Nurzat Y, Su W, Min P, Li K, Xu H, Zhang Y. Identification of Therapeutic Targets and Prognostic Biomarkers Among Integrin Subunits in the Skin Cutaneous Melanoma Microenvironment. Front Oncol 2021; 11:751875. [PMID: 34660316 PMCID: PMC8514842 DOI: 10.3389/fonc.2021.751875] [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: 08/02/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
The roles of different integrin alpha/beta (ITGA/ITGB) subunits in skin cutaneous melanoma (SKCM) and their underlying mechanisms of action remain unclear. Oncomine, UALCAN, GEPIA, STRING, GeneMANIA, cBioPortal, TIMER, TRRUST, and Webgestalt analysis tools were used. The expression levels of ITGA3, ITGA4, ITGA6, ITGA10, ITGB1, ITGB2, ITGB3, ITGB4, and ITGB7 were significantly increased in SKCM tissues. The expression levels of ITGA1, ITGA4, ITGA5, ITGA8, ITGA9, ITGA10, ITGB1, ITGB2, ITGB3, ITGB5, ITGB6 and ITGB7 were closely associated with SKCM metastasis. The expression levels of ITGA1, ITGA4, ITGB1, ITGB2, ITGB6, and ITGB7 were closely associated with the pathological stage of SKCM. The expression levels of ITGA6 and ITGB7 were closely associated with disease-free survival time in SKCM, and the expression levels of ITGA6, ITGA10, ITGB2, ITGB3, ITGB6, ITGB7, and ITGB8 were markedly associated with overall survival in SKCM. We also found significant correlations between the expression of integrin subunits and the infiltration of six types of immune cells (B cells, CD8+ T cells, CD4+T cells, macrophages, neutrophils, and dendritic cells). Finally, Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed, and protein-protein interaction (PPI) networks were constructed. We have identified abnormally-expressed genes and gene regulatory networks associated with SKCM, improving understanding of the underlying pathogenesis of SKCM.
Collapse
Affiliation(s)
- Yeltai Nurzat
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Weijie Su
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Peiru Min
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ke Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Heng Xu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Chen L, Zhong Y, Yang X, Zhang Q, Wu X. Downregulation of GTSE1 leads to the inhibition of proliferation, migration, and Warburg effect in cervical cancer by blocking LHDA expression. J Obstet Gynaecol Res 2021; 47:3913-3922. [PMID: 34482592 DOI: 10.1111/jog.15000] [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: 10/29/2020] [Revised: 06/15/2021] [Accepted: 08/18/2021] [Indexed: 01/11/2023]
Abstract
AIM G2 and S phase-expressed-1 (GTSE1) has been identified to play a vital role in several kinds of cancers, but its role in cervical cancer development remains unknown. Herein, we aimed to reveal the role and underlying mechanism of GTSE1 in cervical cancer cell growth, migration, and aerobic glycolysis. METHODS GTSE1 expression levels in cervical cancer tissues and normal cervical tissues were determined by real time PCR and immunohistochemistry. Human short hairpin RNA was used to downregulate GTSE1 level in cervical cancer cells SiHa and HeLa cells. Colony formation, cell counting kit-8, and wound-healing assays were used for cell function evaluation. Lactate production, lactate dehydrogenase activity, and glucose concentration were tested to assess the Warburg effect. RESULTS GTSE1 expressions at both mRNA and protein levels were significantly elevated in cervical cancer tissues compared with normal tissues. Downregulation of GTSE1 induced significant repressions in cell colony formation, viability and migration, and Warburg effect, as well as reduced expression of lactate dehydrogenase isoform A (LDHA) at mRNA and protein levels. Additionally, downregulation of GTSE1 weakened the tumorigenesis of HeLa and SiHa cells in vivo. CONCLUSION This study demonstrated that downregulation of GTSE1 led to significant inhibitions in cell proliferation, migration, tumorigenesis, and Warburg effect in cervical cancer by blocking the expression of LHDA.
Collapse
Affiliation(s)
- Longyi Chen
- Department of Gynecology, First People's Hospital of Kashi, Kashi Prefecture, Xinjiang Uygur Autonomous Region, China
| | - Youwen Zhong
- School of Economics and Finance, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Xiuwei Yang
- Department of Gynecology, First People's Hospital of Kashi, Kashi Prefecture, Xinjiang Uygur Autonomous Region, China
| | - Qingyue Zhang
- Department of Gynecology, First People's Hospital of Kashi, Kashi Prefecture, Xinjiang Uygur Autonomous Region, China
| | - Xiaoling Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
15
|
Guo W, Zhu J, Zhu Y, Wang K. G2 and S phase-expressed-1 acts as a putative tumor promoter in cervical cancer by enhancing Wnt/β-catenin signaling via modulation of GSK-3β. ENVIRONMENTAL TOXICOLOGY 2021; 36:1628-1639. [PMID: 33974332 DOI: 10.1002/tox.23158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/23/2021] [Accepted: 04/25/2021] [Indexed: 06/12/2023]
Abstract
G2 and S phase-expressed-1 (GTSE1) is currently identified as a key regulator of carcinogenesis. However, the involvement of GTSE1 in cervical cancer is unclear. The aims of this work were to explore the relationship between GTSE1 and cervical cancer. Our data elucidated high GTSE1 expression in cervical cancer tissue, which predicted a poor prognosis in cervical cancer patients. GTSE1 knockdown had tumor-suppressive effects in cervical cancer cells by inhibiting cell proliferative and invasive abilities. GTSE1 knockdown decreased the level of phosphorylated glycogen synthase kinase-3β (GSK-3β) and active β-catenin, resulted in inactivation of Wnt/β-catenin signaling. Suppression of GSK-3β remarkably abolished the GTSE1-knockdown-induced inhibitory effects on Wnt/β-catenin signaling. Suppression of Wnt/β-catenin signaling abolished the GTSE1-overexpression-induced oncogenic effects. Notably, GTSE1 knockdown impeded the in vivo tumorigenicity of cervical cancer cells. In short, this work demonstrates that GTSE1 is overexpressed in cervical cancer and GTSE1 suppression exerts a tumor-inhibiting role in cervical cancer by down-regulating Wnt/β-catenin signaling. Our work underlines a crucial relevance between GTSE1 and cervical cancer progression and suggests GTSE1 as a promising therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Wenting Guo
- Obstetrics and Gynecology Department, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Jing Zhu
- Obstetrics and Gynecology Department, Yulin NO.2 Hospital, Yulin, China
| | - Yuan Zhu
- Gynecology Department, Maternal and Child Health Care Hospital of Zibo, Zibo City, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, China
- Oncology Department, Daxing Hospital, Xi'an, China
| |
Collapse
|
16
|
Xiong J, Zhang J, Li H. Identification of G2 and S Phase-Expressed-1 as a Potential Biomarker in Patients with Prostate Cancer. Cancer Manag Res 2020; 12:9259-9269. [PMID: 33061616 PMCID: PMC7532308 DOI: 10.2147/cmar.s272795] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 08/28/2020] [Indexed: 11/23/2022] Open
Abstract
Background This study aimed to predict and explore the possible clinical value and mechanism of genetic markers in prostate cancer (PCa) using a bioinformatics analysis method. Materials and Methods The RNA-seq data were downloaded from The Cancer Genome Atlas (TCGA) database to identify the differentially expressed genes (DEGs). The hub genes were screened by building protein–protein interaction (PPI) subnetworks with four topological analysis methods. The overall survival analysis of hub genes was conducted using Kaplan–Meier curves. Furthermore, the bioinformatics results were confirmed in 102 PCa samples collected in our hospital. Gene Set Enrichment Analysis (GSEA) was performed to provide information about the molecular mechanisms underlying PCa. Results Among 13 hub genes, the high expression of GTSE1 or KIF18B was associated with worse overall survival according to the TCGA samples. Immunoreactive scores for GTSE1 staining were significantly higher in PCa tissues than in paracancerous tissues (P<0.01). The overall survival time of patients with high GTSE1 expression was shorter than that of patients with low GTSE1 expression (P=0.015). GSEA demonstrated that high GTSE1 expression was mainly enriched in the cell cycle (P<0.001), DNA replication (P<0.001), mismatch repair (P<0.001), and p53 signaling pathway (P<0.001). Conclusion GTSE1 expression was significantly high in PCa and associated with poor prognosis. GTSE1 may serve as a potential biomarker and therapeutic target in PCa patients.
Collapse
Affiliation(s)
- Jian Xiong
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China
| | - Jianzhong Zhang
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Hongjun Li
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China
| |
Collapse
|
17
|
Liu H, Li H, Luo K, Sharma A, Sun X. Prognostic gene expression signature revealed the involvement of mutational pathways in cancer genome. J Cancer 2020; 11:4510-4520. [PMID: 32489468 PMCID: PMC7255374 DOI: 10.7150/jca.40237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/18/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Over the years, many efforts have been made to use the gene expression profiles of cancer types/subtypes to identify the prognostic genes with their potential clinical applications. However, one major challenge remains is to predict the common prognostic genes using simultaneously the dataset of multiple cancers, especially by considering the differences in survival, expression and the associated mutated pathways. Methods: Herein, we carried out a comprehensive examination for the prognostic genes and linked them to the mutational status of 29 cancers, so as to find independent prognostic genes and mechanisms. Additionally, their diagnostic value of them was also assessed. Results: our extensive analysis revealed: 1) the number of prognostic and diagnostic genes differs greatly across the cancers, 2) the potentially implicated 22 genes harbor the diagnostic as well as prognostic capacity, 3) the universal prognostic genes (CDC20, CDCA8, ASPM, ERCC6L, and GTSE1) were found to be involved in the spindle assembly checkpoint, 4) the prognostic genes were found to be statistically linked to the frequently mutated TP53-, MAPK-, PI3K- and AKT- related pathways. We also manually mined possible biological mechanisms for some of the statistical links in literatures. Conclusions: Taken together, we identified the prognostic genes and in addition we assessed their diagnostic capacity. Our analysis provides an important insight about the considerable overlapping between gene expression variation and the further associated altered mutational pathways across the cancer genome. We thus hypothesized that cancer related (mutated) genes are tightly connected and are capable to reshape the genome in multiple cancer types.
Collapse
Affiliation(s)
- Hongde Liu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Huamei Li
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Kun Luo
- Department of Neurosurgery, Xinjiang Evidence-Based Medicine Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Amit Sharma
- Department of Ophthalmology, University Hospital Bonn, Germany
| | - Xiao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
18
|
Ruan X, Zheng J, Liu X, Liu Y, Liu L, Ma J, He Q, Yang C, Wang D, Cai H, Li Z, Liu J, Xue Y. lncRNA LINC00665 Stabilized by TAF15 Impeded the Malignant Biological Behaviors of Glioma Cells via STAU1-Mediated mRNA Degradation. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:823-840. [PMID: 32464546 PMCID: PMC7256440 DOI: 10.1016/j.omtn.2020.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/23/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022]
Abstract
Glioma is a brain cancer characterized by strong invasiveness with limited treatment options and poor prognosis. Recently, dysregulation of long non-coding RNAs (lncRNAs) has emerged as an important component in cellular processes and tumorigenesis. In this study, we demonstrated that TATA-box binding protein associated factor 15 (TAF15) and long intergenic non-protein coding RNA 665 (LINC00665) were both downregulated in glioma tissues and cells. TAF15 overexpression enhanced the stability of LINC00665, inhibiting malignant biological behaviors of glioma cells. Both metal regulatory transcription factor 1 (MTF1) and YY2 transcription factor (YY2) showed high expression levels in glioma tissues and cells, and their knockdown inhibited malignant progression. Mechanistically, overexpression of LINC00665 was confirmed to destabilize MTF1 and YY2 mRNA by interacting with STAU1, and knockdown of STAU1 could rescue the MTF1 and YY2 mRNA degradation caused by LINC00665 overexpression. G2 and S-phase expressed 1 (GTSE1) was identified as an oncogene in glioma, and knockdown of MTF1 or YY2 decreased the mRNA and protein expression levels of GTSE1 through direct binding to the GTSE1 promoter region. Our study highlights a key role of the TAF15/LINC00665/MTF1(YY2)/GTSE1 axis in modulating the malignant biological behaviors of glioma cells, suggesting novel mechanisms by which lncRNAs affect STAU1-mediated mRNA stability, which can inform new molecular therapies for glioma.
Collapse
Affiliation(s)
- Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Jun Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Qianru He
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Jing Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| |
Collapse
|
19
|
Kaushik AC, Mehmood A, Wei DQ, Dai X. Systems Biology Integration and Screening of Reliable Prognostic Markers to Create Synergies in the Control of Lung Cancer Patients. Front Mol Biosci 2020; 7:47. [PMID: 32318583 PMCID: PMC7154114 DOI: 10.3389/fmolb.2020.00047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 03/05/2020] [Indexed: 12/16/2022] Open
Abstract
This study aims to achieve a clearer and stronger understanding of all the mechanisms involved in the occurrence as well as in the progression of lung cancer along with discovering trustworthy prognostic markers. We combined four gene expression profiles (GSE19188, GSE19804, GSE101929, and GSE18842) from the GEO database and screened the commonly differentially expressed genes (CDEGs). We performed differentially expressed group analysis on CDEGs, alteration and mutational analysis, and expression level verification of core differential genes. Systems biology discoveries in our examination are predictable with past reports. Curiously, our examination revealed that screened biomarker adjustments, for the most part, coexist in lung cancer. After screening 952 CDEGs, we found that the up-regulation of neuromedin U (NMU) and GTSE1 in the case of lung cancer is related to poor prognosis. On the other hand, FOS CDKN1C expression is associated with poor prognosis and is responsible for the down-regulation of CDKN1C and FOS. Changes in these qualities are on free pathways to lung cancer and are not usually of combined quality variety. Even though biomarkers were related to both survival occasions in our examination, it gives us another point of view while playing out the investigation of hereditary changes and clinical highlights employing information mining. Based on our results, we found potential and prospective clinical applications in GTSE1, NMU, FOS, and CDKN1C to act as prognostic markers in case of lung cancer.
Collapse
Affiliation(s)
- Aman Chandra Kaushik
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Aamir Mehmood
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Dong-Qing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
20
|
Lei X, Du L, Zhang P, Ma N, Liang Y, Han Y, Qu B. Knockdown GTSE1 enhances radiosensitivity in non-small-cell lung cancer through DNA damage repair pathway. J Cell Mol Med 2020; 24:5162-5167. [PMID: 32202046 PMCID: PMC7205821 DOI: 10.1111/jcmm.15165] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an important strategy for NSCLC. However, although a variety of comprehensive radiotherapy-based treatments have dominated the treatment of NSCLC, it cannot be avoided to overcome the growing radioresistance during radiotherapy. The purpose of this study was to elucidate the radiosensitizing effects of NSCLC via knockdown GTSE1 expression and its mechanism. Experiments were performed by using multiple NSCLC cells such as A549, H460 and H1299. Firstly, we found GTSE1 conferred to radioresistance via clonogenic assay and apoptosis assay. Then, we detected the level of DNA damage through comet assay and γH2AX foci, which we could clearly observe knockdown GTSE1 enhance DNA damage after IR. Furthermore, through using laser assay and detecting DNA damage repair early protein expression, we found radiation could induce GTSE1 recruited to DSB site and initiate DNA damage response. Our finding demonstrated that knockdown GTSE1 enhances radiosensitivity in NSCLC through DNA damage repair pathway. This novel observation may have therapeutic implications to improve therapeutic efficacy of radiation.
Collapse
Affiliation(s)
- Xiao Lei
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lehui Du
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Pei Zhang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Na Ma
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanjie Liang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanan Han
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Baolin Qu
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
21
|
Lin F, Xie YJ, Zhang XK, Huang TJ, Xu HF, Mei Y, Liang H, Hu H, Lin ST, Luo FF, Lang YH, Peng LX, Qian CN, Huang BJ. GTSE1 is involved in breast cancer progression in p53 mutation-dependent manner. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:152. [PMID: 30961661 PMCID: PMC6454633 DOI: 10.1186/s13046-019-1157-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 03/29/2019] [Indexed: 12/24/2022]
Abstract
Background With the rapid development of the high throughput detection techniques, tumor-related Omics data has become an important source for studying the mechanism of tumor progression including breast cancer, one of the major malignancies worldwide. A previous study has shown that the G2 and S phase-expressed-1 (GTSE1) can act as an oncogene in several human cancers. However, its functional roles in breast cancer remain elusive. Method In this study, we analyzed breast cancer data downloaded from The Cancer Genome Atlas (TCGA) databases and other online database including the Oncomine, bc-GenExMiner and PROGgeneV2 database to identify the molecules contributing to the progression of breast cancer. The GTSE1 expression levels were investigated using qRT-PCR, immunoblotting and IHC. The biological function of GTSE1 in the growth, migration and invasion of breast cancer was examined in MDA-MB-231, MDA-MB-468 and MCF7 cell lines. The in vitro cell proliferative, migratory and invasive abilities were evaluated by MTS, colony formation and transwell assay, respectively. The role of GTSE1 in the growth and metastasis of breast cancer were revealed by in vivo investigation using BALB/c nude mice. Results We showed that the expression level of GTSE1 was upregulated in breast cancer specimens and cell lines, especially in triple negative breast cancer (TNBC) and p53 mutated breast cancer cell lines. Importantly, high GTSE1 expression was positively correlated with histological grade and poor survival. We demonstrated that GTSE1 could promote breast cancer cell growth by activating the AKT pathway and enhance metastasis by regulating the Epithelial-Mesenchymal transition (EMT) pathway. Furthermore, it could cause multidrug resistance in breast cancer cells. Interestingly, we found that GTSE1 could regulate the p53 function to alter the cell cycle distribution dependent on the mutation state of p53. Conclusion Our results reveal that GTSE1 played a key role in the progression of breast cancer, indicating that GTSE1 could serve as a novel biomarker to aid in the assessment of the prognosis of breast cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1157-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fen Lin
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Yu-Jie Xie
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Xin-Ke Zhang
- Department of pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Tie-Jun Huang
- Department of Nuclear Medicine, The Second People's Hospital of Shenzhen, Shenzhen, People's Republic of China
| | - Hong-Fa Xu
- Zhuhai Precision Medicine Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Yan Mei
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hu Liang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hao Hu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Si-Ting Lin
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Fei-Fei Luo
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Yan-Hong Lang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Li-Xia Peng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Bi-Jun Huang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
22
|
Liu A, Zeng S, Lu X, Xiong Q, Xue Y, Tong L, Xu W, Sun Y, Zhang Z, Xu C. Overexpression of G2 and S phase-expressed-1 contributes to cell proliferation, migration, and invasion via regulating p53/FoxM1/CCNB1 pathway and predicts poor prognosis in bladder cancer. Int J Biol Macromol 2018; 123:322-334. [PMID: 30414902 DOI: 10.1016/j.ijbiomac.2018.11.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022]
Abstract
Bladder cancer is one of the most common urogenital tumors worldwide. The specific function and molecular mechanism of GTSE1 in bladder cancer remain unknown. In the present study, real-time quantitative polymerase chain reaction and Western blotting were used to identify GTSE1 expression in bladder cancer tissues and cells, and immunohistochemical assays were conducted to investigate GTSE1 expression in tissue microarray. Regression analyses explored the relationship between GTSE1 expression and pathological characteristics. A series of functional tests were performed to observe the effects of GTSE1 knockdown or overexpression, and the related mechanism was also performed. GTSE1 expression was significantly higher in bladder cancer tissues; overexpression of GTSE1 was positively associated with disease recurrence history, lymph node invasion, and progression. Patients with higher GTSE1 expression were more likely to experience shorter survival time, and GTSE1 expression served as a prognostic factor for the disease progression. Knockdown of GTSE1 obviously suppressed the proliferation, migration, and invasion capacity whereas increasing GTSE1 led to the opposite trend, which suggested that GTSE1 could serve as a potential therapeutic target for bladder cancer. GTSE1 overexpression in bladder cancer might participate in the regulation of FoxM1/CCNB1 expression via the induction of the transfer of p53 to cytoplasm.
Collapse
Affiliation(s)
- Anwei Liu
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Shuxiong Zeng
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Xin Lu
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Qiao Xiong
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Yongping Xue
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Liping Tong
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Weidong Xu
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Yinghao Sun
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Zhensheng Zhang
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China.
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China.
| |
Collapse
|
23
|
Xu T, Ma M, Chi Z, Si L, Sheng X, Cui C, Dai J, Yu S, Yan J, Yu H, Wu X, Tang H, Yu J, Kong Y, Guo J. High G2 and S-phase expressed 1 expression promotes acral melanoma progression and correlates with poor clinical prognosis. Cancer Sci 2018; 109:1787-1798. [PMID: 29660787 PMCID: PMC5989838 DOI: 10.1111/cas.13607] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/23/2018] [Accepted: 04/08/2018] [Indexed: 02/07/2023] Open
Abstract
G2 and S‐phase expressed 1 (GTSE1) regulates cell cycle progression in human cancers. However, its significance and mechanism of action in acral melanoma (AM) remain unknown. In the present study, we found that GTSE1 expression was upregulated in advanced stage/metastatic AM tissues and metastatic cell lines, and correlated with higher stage (P = .028) and poor disease‐free survival (DFS) in patients with AM (P = .003). Cox regression assays validated GTSE1 expression to be an independent prognostic factor of DFS for patients with AM (P = .004). Ectopic expression of GTSE1 enhanced primary AM cell proliferation, invasion, and migration. Loss‐of‐function in GTSE1 attenuated metastatic AM cell proliferation and metastatic ability in vitro and in vivo. We additionally observed that inhibition of migration and invasion occurred concomitantly with a GTSE1 knockdown‐mediated increase in E‐cadherin and decreases in N‐cadherin and Slug. We further showed that integrin subunit alpha 2 (ITGA2) interacts with GTSE1 and is a downstream effector of GTSE1. Further, ITGA2 levels were positively correlated with GTSE1 expression in human AM tissues. Ectopic ITGA2 expression rescued siGTSE1‐mediated inhibition of migration and invasion, thereby restoring epithelial‐to‐mesenchymal transition (EMT). In conclusion, GTSE1 expression promotes AM progression and correlates with clinical outcomes of patients with AM, and may represent a promising therapeutic target to suppress AM progression.
Collapse
Affiliation(s)
- Tianxiao Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Junya Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaowen Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiayi Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|