1
|
Zhang W, Qin S, Li X, Chang M, Wei T, Huang X, Tong H, Guo X, Lu Y, Zhang J. SMC4 Promotes Prostate Cancer Cell Proliferation and Metastasis via the Rheb/mTOR Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500369. [PMID: 40278414 DOI: 10.1002/advs.202500369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/16/2025] [Indexed: 04/26/2025]
Abstract
Structural maintenance of chromosome protein 4 (SMC4), is a key structural component of mitotic chromosomes. While existing evidence indicates a plausible link between SMC4 and oncogenic manifestations, its precise role in the trajectory of prostate cancer remains ambiguous. The Cancer Genome Atlas (TCGA) database analysis reveals that aberrant expression of SMC4 exhibits a robust prognostic association with metastatic progression. To investigate the function of SMC4, the SMC4 gene is knocked down in RM1-LM cells, a highly metastatic cell clone is developed, using the CRISPR/Cas9 system. The results show that SMC4 knockdown significantly diminished cell proliferation and migration in vitro. Furthermore, in a murine model, RM1-LM cells display higher lung metastasis capabilities than SMC4 knockdown cells. SMC4 knockdown inhibited the activation of mTOR and downregulated the expression of Rheb. KEGG enrichment analyses of the RNA-seq results reveal that cancer signaling pathways and metabolic pathways are enriched. The SMC4 interactome is uncovered through IP-MS and indicates that SMC4 interacts with GLUT1, encoded by Slc2a1. Glycolytic rate assay illustrates that knocking down SMC4 inhibits the cell glycolysis rate and ATP production. Collectively, the data suggests that the interaction between SMC4 and GLUT1, as confirmed by co-IP, promotes prostate cancer cell metastasis through the Rheb/mTOR pathway.
Collapse
Affiliation(s)
- Wei Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518053, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaokang Li
- Reproductive Medicine Center, Wuhan Jinxin Integrated Chinese and Western Medicine Obstetrics and Gynecology Hospital, Wuhan, 430000, China
| | - Ming Chang
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441000, China
| | - Tianzi Wei
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518053, China
| | - Xin Huang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518053, China
| | - Haibo Tong
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Xia Guo
- Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 518101, China
| | - Yi Lu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518053, China
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518101, China
| | - Jian Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518053, China
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518101, China
- SUSTech-KCL School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
2
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
3
|
Shi T, Geng Q, Wang Z, Wen C, Xu J, Jiao Y, Diao W, Gu J, Deng T, Xiao C, Zhong B, Wang J. "Friends or foes": a new perspective of tumour metabolic transcriptional modification. Cell Death Dis 2025; 16:106. [PMID: 39962057 PMCID: PMC11833121 DOI: 10.1038/s41419-025-07429-y] [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: 11/09/2024] [Revised: 01/11/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025]
Abstract
Energy metabolism plays a pivotal role in cancer clinical treatment and has become an important means of clinical diagnosis of tumour progression. However, current research mostly focuses on changes in metabolic products and neglects the deeper mechanisms of transcriptional regulation. This paper proposes a new perspective, establishing a comprehensive network that reveals the interaction between metabolism and transcription, which explores how tumour metabolism affects tumour progression through transcriptional modifications, and provides a novel approach for optimizing tumour treatment strategies. This viewpoint is conducive to overcoming current bottlenecks in treatment and promoting the development of drug combinations and personalized medicine.
Collapse
Affiliation(s)
- Tong Shi
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Qishun Geng
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Zhaoran Wang
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Chaoying Wen
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jiahe Xu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yi Jiao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
- Beijing University of Chinese Medicine, China-Japan Friendship Hospital Clinical Medicine, Beijing, China
| | - Wenya Diao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
- Beijing University of Chinese Medicine, China-Japan Friendship Hospital Clinical Medicine, Beijing, China
| | - Jienan Gu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
- Beijing University of Chinese Medicine, China-Japan Friendship Hospital Clinical Medicine, Beijing, China
| | - Tingting Deng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Cheng Xiao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.
| | - Baoyuan Zhong
- Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
| | - Jianfeng Wang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
4
|
Chi S, Ma J, Ding Y, Lu Z, Zhou Z, Wang M, Li G, Chen Y. Integrated multi-omics analysis identifies a machine learning-derived signature for predicting prognosis and therapeutic vulnerability in clear cell renal cell carcinoma. Life Sci 2025; 363:123396. [PMID: 39809381 DOI: 10.1016/j.lfs.2025.123396] [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: 07/31/2024] [Revised: 01/02/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
AIMS Clear cell renal cell carcinoma (ccRCC) shows considerable variation within and between tumors, presents varying treatment responses among patients, possibly due to molecular distinctions. This study utilized a multi-center and multi-omics analysis to establish and validate a prognosis and treatment vulnerability signature (PTVS) capable of effectively predicting patient prognosis and drug responsiveness. MATERIALS AND METHODS To address this complexity, we constructed an integrative multi-omics analysis using 10 clustering algorithms on ccRCC patient data. Afterwards, we applied bootstrapping in univariate Cox regression and the Boruta algorithm to pinpoint clinically relevant genes. Based on this, we developed a robust PTVS using seven machine learning algorithms. KEY FINDINGS Our analysis revealed two distinct ccRCC subtypes with differential prognostic implications, notably identifying subtype 2 with poorer outcomes. Patients in the low PTVS group exhibited superior prognosis statistics and an augmented sensitivity to immunotherapy, features consistent with a 'hot tumor' phenotype. Conversely, individuals within the high PTVS group exhibited diminished prognosis statistic and restricted advantages from immunotherapy. Importantly, the PTVS holds future potential as a notable biomarker for guiding personalized treatment strategies, with four prospective targets (CTSK, XDH, PKMYT1, and EGLN2) indicating therapeutic promise in patients scoring high on PTVS. SIGNIFICANCE The integrative analysis of multi-omics data profoundly enhances the molecular stratification of ccRCC, underscoring far-reaching impact of such comprehensive profiling on its therapeutic strategies.
Collapse
Affiliation(s)
- Shengqiang Chi
- Research Center for Data Hub and Security, Zhejiang Laboratory, Hangzhou 311121, China; Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Jing Ma
- Research Center for Data Hub and Security, Zhejiang Laboratory, Hangzhou 311121, China
| | - Yiming Ding
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Zeyi Lu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Zhenwei Zhou
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Mingchao Wang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yuanlei Chen
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| |
Collapse
|
5
|
Fan Y, Wu L, Qiu X, Shi H, Wu L, Lin J, Lin J, Teng T. Single-cell RNA-seq analysis reveals microenvironmental infiltration of myeloid cells and pancreatic prognostic markers in PDAC. Discov Oncol 2025; 16:81. [PMID: 39847195 PMCID: PMC11757659 DOI: 10.1007/s12672-025-01830-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a heterogeneous make-up of myeloid cells that influences the therapeutic response and prognosis. However, understanding the myeloid cell at both a genetic and cellular level remains a significant challenge. METHODS Single-cell RNA sequencing (scRNA-seq) data were downloaded from t the Tumor Immune Single-cell Hub and gene expression data were retrieved from The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database. Gene set variation analysis (GSVA) was used to estimate the relative proportions of each cell type based on the signatures identified by scRNA-seq or previous literature. Cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) was performed to evaluate the abundance of immune infiltrating cells. For further analysis, LASSO and Cox analyses were used to construct a risk model using univariate Cox regression. RESULTS Using the scRNA-seq dataset, we identified 7 clusters of myeloid cells, and these clusters were assigned a cell type based on their marker genes. In addition, the results of the CellChat analysis and SCENIC analysis indicate that TAM-spp1 cells may promote the migration of pancreatic tumor cells on different levels. Moreover, the TAM-spp1 cell is most closely related to poor prognoses. An 8-gene risk model was constructed by using univariate Cox and LASSO analyses. In the GEO cohorts, this risk model demonstrated excellent predictive abilities for prognosis. Further, patients with high-risk scores had a lower likelihood of benefiting from immunotherapy. CONCLUSION Using bulk RNA-seq and single-cell RNA-seq, we analyzed myeloid heterogeneity at the single-cell level, and we developed an 8-gene model that predicts survival outcomes and immunotherapy response in PADC.
Collapse
Affiliation(s)
- Yanying Fan
- Fuzhou First General Hospital Affiliated With Fujian Medical University, Fuzhou, China
| | - Lili Wu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
- Department of Surgical Nursing, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Xinyu Qiu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Han Shi
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Longhang Wu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Juan Lin
- Fuzhou First General Hospital Affiliated With Fujian Medical University, Fuzhou, China
| | - Jie Lin
- Fuzhou First General Hospital Affiliated With Fujian Medical University, Fuzhou, China.
| | - Tianhong Teng
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China.
| |
Collapse
|
6
|
Wang H, Sahu A, Chuong MD, Li R. TCF4 as a potential prognostic biomarker and an anticancer target in gastric cancer. Transl Cancer Res 2024; 13:5073-5086. [PMID: 39430834 PMCID: PMC11483414 DOI: 10.21037/tcr-24-1290] [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: 07/25/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024]
Abstract
Background Lymphoid enhancer-binding factor 1 (LEF1)/T cell factor (TCF) family members are key transcription factors in malignant tumors. In this study, the role of T cell factor 4 (TCF4) in the progression of gastric cancer (GC) cell migration and invasion was investigated. Methods Fifty-five pairs of GC tissues and adjacent non-tumor tissues were collected for evaluating the expression of LEF1/TCF family members, which were also evaluated by the Gene Expression Profiling Interactive Analysis (GEPIA) database, an online analysis platform based on The Cancer Genome Atlas and Genotype-Tissue Expression databases. Results Through GEPIA online analysis and our experimental specimens, we found that TCF4 messenger RNA (mRNA) expression was significantly upregulated in GC tissues compared with normal non-tumor tissues. The findings from protein-protein interaction (PPI) analysis suggested that myocyte enhancer factor 2C (MEF2C) may function as a regulatory gene for TCF4 and play a role in the progression of GC. A significant increase in TCF4 mRNA expression was observed in the GC cell lines. Silencing of TCF4 led to significant inhibition of the proliferation, migration, and invasion of the MGC-803 and SGC-7901 cells. TdT-mediated dUTP nick end labeling (TUNEL)-positive staining cells were significantly increased after transfection with TCF4 small interfering (si)-RNA into GC cells. In addition, patients with GC with high TCF4 expression were associated with poor T stage, pathologic stages, histologic grade, overall survival, and recurrence-free survival, indicating that TCF4 may be a potential prognostic marker of GC. Conclusions TCF4 potentially exerts a carcinogenic role in the progression of GC. TCF4 may serve as a prognostic indicator and therapeutic target for GC.
Collapse
Affiliation(s)
- Hailong Wang
- Department of Laboratory Medicine, Inner Mongolia Hospital Beijing Hospital of Traditional Chinese Medicine, Bayannaoer, China
| | - Arvind Sahu
- Department of Oncology, Goulburn Valley Health, Shepparton, VIC, Australia
| | - Michael D. Chuong
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Ruiping Li
- Department of Laboratory Medicine, Maternal and Child Health Care Hospital of Linhe District, Bayannaoer City, China
| |
Collapse
|
7
|
Jiang JZ, Qiao YB, Zhu XR, Gu QH, Lu JJ, Ye ZY, Xu L, Liu YY. Identification of Gαi3 as a promising molecular oncotarget of pancreatic cancer. Cell Death Dis 2024; 15:699. [PMID: 39349432 PMCID: PMC11442978 DOI: 10.1038/s41419-024-07079-6] [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: 06/06/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
The increasing mortality rate of pancreatic cancer globally necessitates the urgent identification for novel therapeutic targets. This study investigated the expression, functions, and mechanistic insight of G protein inhibitory subunit 3 (Gαi3) in pancreatic cancer. Bioinformatics analyses reveal that Gαi3 is overexpressed in human pancreatic cancer, correlating with poor prognosis, higher tumor grade, and advanced classification. Elevated Gαi3 levels are also confirmed in human pancreatic cancer tissues and primary/immortalized cancer cells. Gαi3 shRNA or knockout (KO) significantly reduced cell viability, proliferation, cell cycle progression, and mobility in primary/immortalized pancreatic cancer cells. Conversely, Gαi3 overexpression enhanced pancreatic cancer cell growth. RNA-sequencing and bioinformatics analyses of Gαi3-depleted cells indicated Gαi3's role in modulating the Akt-mTOR and PKA-Hippo-YAP pathways. Akt-S6 phosphorylation was decreased in Gαi3-depleted cells, but was increased with Gαi3 overexpression. Additionally, Gαi3 depletion elevated PKA activity and activated the Hippo pathway kinase LATS1/2, leading to YAP/TAZ inactivation, while Gαi3 overexpression exerted the opposite effects. There is an increased binding between Gαi3 promoter and the transcription factor TCF7L2 in pancreatic cancer tissues and cells. Gαi3 expression was significantly decreased following TCF7L2 silencing, but increased with TCF7L2 overexpression. In vivo, intratumoral injection of Gαi3 shRNA-expressing adeno-associated virus significantly inhibited subcutaneous pancreatic cancer xenografts growth in nude mice. A significant growth reduction was also observed in xenografts from Gαi3 knockout pancreatic cancer cells. Akt-mTOR inactivation and increased PKA activity coupled with YAP/TAZ inactivation were also detected in xenograft tumors upon Gαi3 depletion. Furthermore, bioinformatic analysis and multiplex immunohistochemistry (mIHC) staining on pancreatic cancer tissue microarrays showed a reduced proportion of M1-type macrophages and an increase in PD-L1 positive cells in Gαi3-high pancreatic cancer tissues. Collectively, these findings highlight Gαi3's critical role in promoting pancreatic cancer cell growth, potentially through the modulation of the Akt-mTOR and PKA-Hippo-YAP pathways and its influence on the immune landscape.
Collapse
Affiliation(s)
- Jian-Zhuo Jiang
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yin-Biao Qiao
- General Surgery, Cancer Center, Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Qian-Hui Gu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Jing-Jing Lu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Zhen-Yu Ye
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Lu Xu
- Department of general surgery, The first affiliated hospital of Soochow university, Suzhou, China.
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
8
|
Han X, Zhu Y, Ke J, Zhai Y, Huang M, Zhang X, He H, Zhang X, Zhao X, Guo K, Li X, Han Z, Zhang Y. Progression of m 6A in the tumor microenvironment: hypoxia, immune and metabolic reprogramming. Cell Death Discov 2024; 10:331. [PMID: 39033180 PMCID: PMC11271487 DOI: 10.1038/s41420-024-02092-2] [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: 12/24/2023] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
Recently, N6-methyladenosine (m6A) has aroused widespread discussion in the scientific community as a mode of RNA modification. m6A comprises writers, erasers, and readers, which regulates RNA production, nuclear export, and translation and is very important for human health. A large number of studies have found that the regulation of m6A is closely related to the occurrence and invasion of tumors, while the homeostasis and function of the tumor microenvironment (TME) determine the occurrence and development of tumors to some extent. TME is composed of a variety of immune cells (T cells, B cells, etc.) and nonimmune cells (tumor-associated mesenchymal stem cells (TA-MSCs), cancer-associated fibroblasts (CAFs), etc.). Current studies suggest that m6A is involved in regulating the function of various cells in the TME, thereby affecting tumor progression. In this manuscript, we present the composition of m6A and TME, the relationship between m6A methylation and characteristic changes in TME, the role of m6A methylation in TME, and potential therapeutic strategies to provide new perspectives for better treatment of tumors in clinical work.
Collapse
Affiliation(s)
- Xuan Han
- First Clinical College of Changzhi Medical College, Changzhi, China
| | - Yu Zhu
- Linfen Central Hospital, Linfen, China
| | - Juan Ke
- Linfen Central Hospital, Linfen, China
| | | | - Min Huang
- Linfen Central Hospital, Linfen, China
| | - Xin Zhang
- Linfen Central Hospital, Linfen, China
| | | | | | | | | | | | - Zhongyu Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | |
Collapse
|
9
|
Rathee M, Umar SM, Dev AJR, Kashyap A, Mathur SR, Gogia A, Mohapatra P, Prasad CP. Canonical WNT/β-catenin signaling upregulates aerobic glycolysis in diverse cancer types. Mol Biol Rep 2024; 51:788. [PMID: 38970704 DOI: 10.1007/s11033-024-09694-0] [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: 01/23/2024] [Accepted: 05/31/2024] [Indexed: 07/08/2024]
Abstract
Despite many efforts, a comprehensive understanding and clarification of the intricate connections within cancer cell metabolism remain elusive. This might pertain to intracellular dynamics and the complex interplay between cancer cells, and cells with the tumor stroma. Almost a century ago, Otto Warburg found that cancer cells exhibit a glycolytic phenotype, which continues to be a subject of thorough investigation. Past and ongoing investigations have demonstrated intricate mechanisms by which tumors modulate their functionality by utilizing extracellular glucose as a substrate, thereby sustaining the essential proliferation of cancer cells. This concept of "aerobic glycolysis," where cancer cells (even in the presence of enough oxygen) metabolize glucose to produce lactate plays a critical role in cancer progression and is regulated by various signaling pathways. Recent research has revealed that the canonical wingless-related integrated site (WNT) pathway promotes aerobic glycolysis, directly and indirectly, thereby influencing cancer development and progression. The present review seeks to gather knowledge about how the WNT/β-catenin pathway influences aerobic glycolysis, referring to relevant studies in different types of cancer. Furthermore, we propose the concept of impeding the glycolytic phenotype of tumors by employing specific inhibitors that target WNT/β-catenin signaling.
Collapse
Affiliation(s)
- Meetu Rathee
- Department of Medical Oncology Lab, DR BRA IRCH, All India Institute of Medical Sciences (AIIMS), 4thFloor, Ansari Nagar, New Delhi, 110029, India
| | - Sheikh Mohammad Umar
- Department of Medical Oncology Lab, DR BRA IRCH, All India Institute of Medical Sciences (AIIMS), 4thFloor, Ansari Nagar, New Delhi, 110029, India
| | - Arundhathi J R Dev
- Department of Medical Oncology Lab, DR BRA IRCH, All India Institute of Medical Sciences (AIIMS), 4thFloor, Ansari Nagar, New Delhi, 110029, India
| | - Akanksha Kashyap
- Department of Medical Oncology Lab, DR BRA IRCH, All India Institute of Medical Sciences (AIIMS), 4thFloor, Ansari Nagar, New Delhi, 110029, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Ajay Gogia
- Department of Medical Oncology, DR BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | | | - Chandra Prakash Prasad
- Department of Medical Oncology Lab, DR BRA IRCH, All India Institute of Medical Sciences (AIIMS), 4thFloor, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
10
|
Qiao Q, Hu S, Wang X. The regulatory roles and clinical significance of glycolysis in tumor. Cancer Commun (Lond) 2024; 44:761-786. [PMID: 38851859 PMCID: PMC11260772 DOI: 10.1002/cac2.12549] [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: 11/09/2023] [Revised: 05/05/2024] [Accepted: 05/12/2024] [Indexed: 06/10/2024] Open
Abstract
Metabolic reprogramming has been demonstrated to have a significant impact on the biological behaviors of tumor cells, among which glycolysis is an important form. Recent research has revealed that the heightened glycolysis levels, the abnormal expression of glycolytic enzymes, and the accumulation of glycolytic products could regulate the growth, proliferation, invasion, and metastasis of tumor cells and provide a favorable microenvironment for tumor development and progression. Based on the distinctive glycolytic characteristics of tumor cells, novel imaging tests have been developed to evaluate tumor proliferation and metastasis. In addition, glycolytic enzymes have been found to serve as promising biomarkers in tumor, which could provide assistance in the early diagnosis and prognostic assessment of tumor patients. Numerous glycolytic enzymes have been identified as potential therapeutic targets for tumor treatment, and various small molecule inhibitors targeting glycolytic enzymes have been developed to inhibit tumor development and some of them are already applied in the clinic. In this review, we systematically summarized recent advances of the regulatory roles of glycolysis in tumor progression and highlighted the potential clinical significance of glycolytic enzymes and products as novel biomarkers and therapeutic targets in tumor treatment.
Collapse
Affiliation(s)
- Qiqi Qiao
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
| | - Shunfeng Hu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
| | - Xin Wang
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
- Taishan Scholars Program of Shandong ProvinceJinanShandongP. R. China
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongP. R. China
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| |
Collapse
|
11
|
E M, Ren F, Yu Y, Li H, Shen C. The role of lncRNAKCNQ1OT1/miR-301b/Tcf7 axis in cardiac hypertrophy. Cardiol Young 2024; 34:1493-1505. [PMID: 38456301 DOI: 10.1017/s1047951124000155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
OBJECTIVE Cardiac hypertrophy, acting as a pathologic process of chronic hypertension and coronary disease, and its underlying mechanisms still need to be explored. Long non-coding RNA (LncRNA) potassium voltage-gated channel subfamily Q member 1 Transcript 1 (KCNQ1OT1) has been implicated in myocardial infarction. However, its role in cardiac hypertrophy remains reported. METHOD To explore the regulated effect of lncRNAKCNQ1OT1 and miR-301b in cardiac hypertrophy, gain-and-lose function assays were tested. The expression of lncRNAKCNQ1OT1 and miR-301b were tested by quantitative real time polymerase chain reaction (qRT-PCR). The levels of transcription factor 7 (Tcf7), Proto-oncogene c-myc (c-myc), Brainnatriureticpeptide (BNP) and β-myosin heavy chain (β-MHC) were detected by Western blot. Additionally, luciferase analysis revealed interaction between lncRNAKCNQ1OT1, BNPβ-MHCmiR-301b, and Tcf7. RESULT LncRNAKCNQ1OT1 overexpression significantly induced cardiac hypertrophy. Furthermore, lncRNAKCNQ1OT1 acts as a sponge for microRNA-301b, which exhibited lower expression in cardiac hypertrophy model, indicating an anti-hypertrophic role. Furthermore, the BNP and β-MHC expression increased, as well as cardiomyocyte surface area, with Ang II treatment, while the effect was repealed by miR-301b. Moreover, the protein expression of Tcf7 was inversely regulated by miR-301b and Antisense miRNA oligonucleotides (AMO)-301b. CONCLUSION Our study has shown that overexpression of lncRNAKCNQ1OT1 could promote the development of cardiac hypertrophy by regulating miR-301b and Tcf7. Therefore, inhibition of lncRNAKCNQ1OT1 might be a potential therapeutic strategy for cardiac hypertrophy.
Collapse
Affiliation(s)
- Mingyao E
- Department of Pharmacology, Baicheng Medical College, Baicheng, China
| | - Feifei Ren
- Nursing Department of Baicheng Hospital, Baicheng Medical College, Baicheng, China
| | - Yanhua Yu
- Department of Pharmacology, Baicheng Medical College, Baicheng, China
| | - Haiyan Li
- Department of Pharmacology, Baicheng Medical College, Baicheng, China
| | - Chao Shen
- Department of Pharmacology, Baicheng Medical College, Baicheng, China
| |
Collapse
|
12
|
Huang Y, Zhang R, Lyu H, Xiao S, Guo D, Chen XZ, Zhou C, Tang J. LncRNAs as nodes for the cross-talk between autophagy and Wnt signaling in pancreatic cancer drug resistance. Int J Biol Sci 2024; 20:2698-2726. [PMID: 38725864 PMCID: PMC11077374 DOI: 10.7150/ijbs.91832] [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: 11/02/2023] [Accepted: 02/06/2024] [Indexed: 05/12/2024] Open
Abstract
Pancreatic cancer is a malignancy with high mortality. In addition to the few symptoms until the disease reaches an advanced stage, the high fatality rate is attributed to its rapid development, drug resistance and lack of appropriate treatment. In the selection and research of therapeutic drugs, gemcitabine is the first-line drug for pancreatic cancer. Solving the problem of gemcitabine resistance in pancreatic cancer will contribute to the progress of pancreatic cancer treatment. Long non coding RNAs (lncRNAs), which are RNA transcripts longer than 200 nucleotides, play vital roles in cellular physiological metabolic activities. Currently, our group and others have found that some lncRNAs are aberrantly expressed in pancreatic cancer cells, which can regulate the process of cancer through autophagy and Wnt/β-catenin pathways simultaneously and affect the sensitivity of cancer cells to therapeutic drugs. This review presents an overview of the recent evidence concerning the node of lncRNA for the cross-talk between autophagy and Wnt/β-catenin signaling in pancreatic cancer, together with the practicability of lncRNAs and the core regulatory factors as targets in therapeutic resistance.
Collapse
Affiliation(s)
- Yuhan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada, T6G2R3
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| |
Collapse
|
13
|
Nishikiori N, Watanabe M, Sato T, Furuhashi M, Okura M, Hida T, Uhara H, Ohguro H. Significant and Various Effects of ML329-Induced MITF Suppression in the Melanoma Cell Line. Cancers (Basel) 2024; 16:263. [PMID: 38254754 PMCID: PMC10814414 DOI: 10.3390/cancers16020263] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
To study the inhibitory effects on microphthalmia-associated transcription factor (MITF)-related biological aspects in malignant melanomas (MMs) in the presence or absence of the low-molecular MITF specific inhibitor ML329, cell viability, cellular metabolic functions, and three-dimensional (3D) spheroid formation efficacy were compared among MM cell lines including SK-mel-24, A375, dabrafenib- and trametinib-resistant A375 (A375DT), and WM266-4. Upon exposure to 2 or 10 μM of ML329, cell viability was significantly decreased in WM266-4, SK-mel-24, and A375DT cells, but not A375 cells, in a dose-dependent manner, and these toxic effects of ML329 were most evident in WM266-4 cells. Extracellular flux assays conducted using a Seahorse bioanalyzer revealed that treatment with ML329 increased basal respiration, ATP-linked respiration, proton leakage, and non-mitochondrial respiration in WM266-4 cells and decreased glycolytic function in SK-mel-24 cells, whereas there were no marked effects of ML329 on A375 and A375DT cells. A glycolytic stress assay under conditions of high glucose concentrations also demonstrated that the inhibitory effect of ML329 on the glycolytic function of WM266-4 cells was dose-dependent. In addition, ML329 significantly decreased 3D-spheroid-forming ability, though the effects of ML329 were variable among the MM cell lines. Furthermore, the mRNA expression levels of selected genes, including STAT3 as a possible regulator of 3D spheroid formation, KRAS and SOX2 as oncogenic-signaling-related factors, PCG1a as the main regulator of mitochondrial biogenesis, and HIF1a as a major hypoxia transcriptional regulator, fluctuated among the MM cell lines, possibly supporting the diverse ML329 effects mentioned above. The findings of diverse ML329 effects on various MM cell lines suggest that MITF-associated biological activities are different among various types of MM.
Collapse
Affiliation(s)
- Nami Nishikiori
- Department of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan; (N.N.); (M.W.)
| | - Megumi Watanabe
- Department of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan; (N.N.); (M.W.)
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan; (T.S.); (M.F.)
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan; (T.S.); (M.F.)
| | - Masae Okura
- Department of Dermatology, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan; (M.O.); (T.H.); (H.U.)
| | - Tokimasa Hida
- Department of Dermatology, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan; (M.O.); (T.H.); (H.U.)
| | - Hisashi Uhara
- Department of Dermatology, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan; (M.O.); (T.H.); (H.U.)
| | - Hiroshi Ohguro
- Department of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Spporo 060-8556, Japan; (N.N.); (M.W.)
| |
Collapse
|
14
|
Kuroda Y, Oda T, Shimomura O, Louphrasitthiphol P, Mathis BJ, Tateno H, Hatano K. Novel positron emission tomography imaging targeting cell surface glycans for pancreatic cancer: 18 F-labeled rBC2LCN lectin. Cancer Sci 2023; 114:3364-3373. [PMID: 37203465 PMCID: PMC10394132 DOI: 10.1111/cas.15846] [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: 01/09/2023] [Revised: 04/11/2023] [Accepted: 04/26/2023] [Indexed: 05/20/2023] Open
Abstract
Advancement in early detection modalities will greatly improve the overall prognoses of pancreatic ductal adenocarcinoma (PDAC). For this purpose, we report a novel class of tumor-specific probes for positron emission tomography (PET) based on targeting cell surface glycans. The PDAC-targeting ability of rBC2LCN lectin, combined with fluorine-18 (18 F) ([18 F]FB-rBC2LCN), resulted in reproducible, high-contrast PET imaging of tumors in a PDAC xenograft mouse model. [18 F]N-succinimidyl-4-fluorobenzoate ([18 F]SFB) was conjugated to rBC2LCN, and [18 F]FB-rBC2LCN was successfully prepared with a radiochemical purity >95%. Cell binding and uptake revealed that [18 F]FB-rBC2LCN binds to H-type-3-positive Capan-1 pancreatic cancer cells. As early as 60 min after [18 F]FB-rBC2LCN (0.34 ± 0.15 MBq) injection into the tail vein of nude mice subcutaneously bearing Capan-1 tumors, tumor uptake was high (6.6 ± 1.8 %ID/g), and the uptake increased over time (8.8 ± 1.9 %ID/g and 11 ± 3.2 %ID/g at 150 and 240 min after injection, respectively). Tumor-to-muscle ratios increased over time, up to 19 ± 1.8 at 360 min. High-contrast PET imaging of tumors relative to background muscle was also achieved as early as 60 min after injection of [18 F]FB-rBC2LCN (0.66 ± 0.12 MBq) and continued to increase up to 240 min. Our 18 F-labeled rBC2LCN lectin warrants further clinical development to improve the accuracy and sensitivity of early-stage pancreatic cancer detection.
Collapse
Affiliation(s)
- Yukihito Kuroda
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Osamu Shimomura
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Pakavarin Louphrasitthiphol
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Bryan J. Mathis
- International Medical CenterUniversity of Tsukuba HospitalTsukubaJapan
| | - Hiroaki Tateno
- Cellular and Molecular Biotechnology Research InstituteNational Institute of Advanced Industrial Science and TechnologyTsukubaJapan
| | - Kentaro Hatano
- Department of Applied Molecular Imaging, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| |
Collapse
|
15
|
Ge W, Meng L, Cao S, Hou C, Zhu X, Huang D, Li Q, Peng Y, Jiang K. The SIX1/LDHA Axis Promotes Lactate Accumulation and Leads to NK Cell Dysfunction in Pancreatic Cancer. J Immunol Res 2023; 2023:6891636. [PMID: 36937004 PMCID: PMC10022590 DOI: 10.1155/2023/6891636] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/18/2022] [Accepted: 02/08/2023] [Indexed: 03/10/2023] Open
Abstract
Background Pancreatic cancer (PC) is a malignant cancer with poor prognosis and high mortality rate. Sine oculis homeobox homolog 1 (SIX1) participates in the development of many cancers. However, the function of SIX1 in PC is not fully understood. Methods SIX1 expression was determined using immunohistochemistry in PC tissues and cell lines. Glucose consumption, lactate production, and ATP assays were used to detect the function of SIX1. PC cells and NK cells were cocultured to study the effect of SIX1 overexpression in PC cells on NK cell function. Chromatin immunoprecipitation (ChIP) assays were used to study the relationship between SIX1 and lactate dehydrogenase A (LDHA). A series of in vitro and in vivo assays were further applied to elucidate the important role of the SIX1/LDHA axis in metabolism and NK cell dysfunction in PC. Results SIX1 was significantly upregulated in PC tissue; SIX1 overexpression promoted the glycolysis capacity of PANC-1 and CFPAC-1 cells and resulted in NK cell dysfunction after the NK cells had been cultured with PC cells. LDHA inhibitor partially restored the promotion of PC caused by SIX1 overexpression. According to ChIP assays, SIX1 directly binds to the LDHA promoter region. Moreover, LDHA inhibitor and lactate transporter blocker treatment promoted the function of NK cells cocultured with PC cells. In vivo experiments yielded the same results. Conclusion The SIX1/LDHA axis promotes lactate accumulation and leads to NK cell dysfunction in PC.
Collapse
Affiliation(s)
- Wanli Ge
- 1Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- 2Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Lingdong Meng
- 1Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- 2Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Shouji Cao
- 3Nanjing Medical University, Nanjing, China
- 4Thyroid Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Chaoqun Hou
- 1Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- 2Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Xiaole Zhu
- 1Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- 2Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Dongya Huang
- 1Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- 2Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Qiang Li
- 1Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- 2Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yunpeng Peng
- 1Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- 2Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- 1Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- 2Pancreas Institute, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Dong S, Li W, Li X, Wang Z, Chen Z, Shi H, He R, Chen C, Zhou W. Glucose metabolism and tumour microenvironment in pancreatic cancer: A key link in cancer progression. Front Immunol 2022; 13:1038650. [PMID: 36578477 PMCID: PMC9792100 DOI: 10.3389/fimmu.2022.1038650] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Early and accurate diagnosis and treatment of pancreatic cancer (PC) remain challenging endeavors globally. Late diagnosis lag, high invasiveness, chemical resistance, and poor prognosis are unresolved issues of PC. The concept of metabolic reprogramming is a hallmark of cancer cells. Increasing evidence shows that PC cells alter metabolic processes such as glucose, amino acids, and lipids metabolism and require continuous nutrition for survival, proliferation, and invasion. Glucose metabolism, in particular, regulates the tumour microenvironment (TME). Furthermore, the link between glucose metabolism and TME also plays an important role in the targeted therapy, chemoresistance, radiotherapy ineffectiveness, and immunosuppression of PC. Altered metabolism with the TME has emerged as a key mechanism regulating PC progression. This review shed light on the relationship between TME, glucose metabolism, and various aspects of PC. The findings of this study provide a new direction in the development of PC therapy targeting the metabolism of cancer cells.
Collapse
Affiliation(s)
- Shi Dong
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Wancheng Li
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Xin Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhengfeng Wang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhou Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Huaqing Shi
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Ru He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Chen Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Wence Zhou
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
17
|
Zhou B, Lei JH, Wang Q, Qu TF, Cha LC, Zhan HX, Liu SL, Hu X, Sun CD, Guo WD, Qiu FB, Cao JY. Cancer-associated fibroblast-secreted miR-421 promotes pancreatic cancer by regulating the SIRT3/H3K9Ac/HIF-1α axis. Kaohsiung J Med Sci 2022; 38:1080-1092. [PMID: 36200682 DOI: 10.1002/kjm2.12590] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/18/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
This study was designed to explore the effects of exosomal miR-421 secreted by cancer-associated fibroblasts (CAFs) on pancreatic cancer (PC) progression and the mechanisms involved. CAFs and exosomes (exos) were isolated and identified. PC cells were treated with CAF-derived exos (CAF-exos). Western blotting and quantitative polymerase chain reaction (qPCR) were used to measure miR-421, sirtuin-3 (SIRT3), and hypoxia duciblefactors-1 alpha (HIF-1α) levels. Cell counting kit-8 (CCK-8), wound-healing, and transwell migration assays were used to measure proliferation, migration, and invasion abilities of the cells. Dual-luciferase assay and RNA immunoprecipitation (RIP) experiment analyzed the relationship between miR-421 and SIRT3. Chromatin immunoprecipitation (f)-verified H3K9Ac enrichment in the HIF-1α promoter region. In vivo tumorigenesis experiments were performed to further explore the effects of exosomal miR-421 from CAFs on PC. CAFs and exos were successfully isolated. CAF-exo-treated PC cells highly expressed miR-421 and had increased cell proliferation, migration, and invasion abilities. Knocking down miR-421 increased the expression of SIRT3. SIRT3 is a target of miR-421, and inhibiting the expression of SIRT3 reversed the negative effects of miR-421 knockdown on PC cell. Knocking down miR-421 in CAF-exo inhibited the expression of HIF-1α in PC cells. Moreover, SIRT3-mediated HIF-1α expression by regulating H3K9Ac. HIF-1α overexpression reversed the inhibiting effects of SIRT3 overexpression on PC progression and counteracted the inhibiting effects of miR-421 knockdown on glycolysis. Moreover, in vivo tumorigenesis experiments showed that knocking down miR-421 attenuated CAF-exo induced tumor growth. Exosomal miR-421 from CAFs promoted PC progression by regulating the SIRT3/H3K9Ac/HIF-1α axis. This study provided insights into the molecular mechanism of PC.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Jing-Hao Lei
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Qiang Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Teng-Fei Qu
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Li-Chao Cha
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Han-Xiang Zhan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Shang-Long Liu
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Xiao Hu
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Chuan-Dong Sun
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Wei-Dong Guo
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Fa-Bo Qiu
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Jing-Yu Cao
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| |
Collapse
|
18
|
MITF-Mediated lncRNA CCDC183-As1 Promotes the Tumorigenic Properties and Aerobic Glycolysis of Bladder Cancer via Upregulating TCF7L2. JOURNAL OF ONCOLOGY 2022; 2022:6785956. [PMID: 35957803 PMCID: PMC9357683 DOI: 10.1155/2022/6785956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/04/2022] [Indexed: 12/02/2022]
Abstract
As a primary malignancy tumor of the urology system, bladder cancer (BC) is characterized by its high recurrence and metastasis characteristics. Despite the great improvement in clinical interventions over the past decades, the outcomes of BC patients are still unsatisfactory. Novel molecular mechanisms for developing effective diagnostic and therapeutic strategies are urgently needed; therefore, we screened the lncRNA expression profile in four pairs of BC tissues, showing that CCDC183-AS1 was the most upregulated lncRNA. Subsequently, results of CCK-8, EdU, Transwell, and aerobic glycolysis detection showed that CCDC183-AS1 plays an oncogene role in BC progression. Furthermore, an investigation of the downstream and upstream factors of CCDC183-AS1 identified a novel MITF/CCDC183-AS1/miR-4731-5p/TCF7L2 axis in BC progression, which might furnish novel insights for developing effective diagnostic and therapeutic strategies for BC.
Collapse
|
19
|
Zhang T, Wang B, Su F, Gu B, Xiang L, Gao L, Zheng P, Li XM, Chen H. TCF7L2 promotes anoikis resistance and metastasis of gastric cancer by transcriptionally activating PLAUR. Int J Biol Sci 2022; 18:4560-4577. [PMID: 35864968 PMCID: PMC9295057 DOI: 10.7150/ijbs.69933] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/14/2022] [Indexed: 12/04/2022] Open
Abstract
Gastric cancer (GC) is the most common gastrointestinal malignant tumor, and distant metastasis is a critical factor in the prognosis of patients with GC. Understanding the mechanism of GC metastasis will help improve patient prognosis. Studies have confirmed that urokinase-type plasminogen activator receptor (PLAUR) promotes GC metastasis; however, its relationship with anoikis resistance and associated mechanisms remains unclear. In this study, we demonstrated that PLAUR promotes the anoikis resistance and metastasis of GC cells and identified transcription Factor 7 Like 2 (TCF7L2) as an important transcriptional regulator of PLAUR. We also revealed that TCF7L2 is highly expressed in GC and promotes the anoikis resistance and metastasis of GC cells. Moreover, we found that TCF7L2 transcription activates PLAUR. Finally, we confirmed that TCF7L2 is an independent risk factor for poor prognosis of patients with GC. Our results show that TCF7L2 and PLAUR are candidate targets for developing therapeutic strategies for GC metastasis.
Collapse
Affiliation(s)
- Tao Zhang
- Department of oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Bofang Wang
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Fei Su
- Department of oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Baohong Gu
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lin Xiang
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lei Gao
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Peng Zheng
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xue-Mei Li
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hao Chen
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Cancer center, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
20
|
The multifaceted role of EGLN family prolyl hydroxylases in cancer: going beyond HIF regulation. Oncogene 2022; 41:3665-3679. [PMID: 35705735 DOI: 10.1038/s41388-022-02378-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 12/22/2022]
Abstract
EGLN1, EGLN2 and EGLN3 are proline hydroxylase whose main function is the regulation of the HIF factors. They work as oxygen sensors and are the main responsible of HIFα subunits degradation in normoxia. Being their activity strictly oxygen-dependent, when oxygen tension lowers, their control on HIFα is released, leading to activation of systemic and cellular response to hypoxia. However, EGLN family members activity is not limited to HIF modulation, but it includes the regulation of essential mechanisms for cell survival, cell cycle metabolism, proliferation and transcription. This is due to their reported hydroxylase activity on a number of non-HIF targets and sometimes to hydroxylase-independent functions. For these reasons, EGLN enzymes appear fundamental for development and progression of different cancer types, playing either a tumor-suppressive or a tumor-promoting role, according to EGLN isoform and to tumor context. Notably, EGLN1, the most studied isoform, has been shown to have also a central role in tumor micro-environment modulation, mediating CAF activation and impairing HIF1α -related angiogenesis, thus covering an important function in cancer metastasis promotion. Considering the recent knowledge acquired on EGLNs, the possibility to target these enzymes for cancer treatment is emerging. However, due to their multifaceted and controversial roles in different cancer types, the use of EGLN inhibitors as anti-cancer drugs should be carefully evaluated in each context.
Collapse
|
21
|
Zhang J, Liu Z, Zhang Z, Tang R, Zeng Y, Chen P. Identification of a glycolysis-related gene signature for predicting pancreatic cancer survival. J Gastrointest Oncol 2022; 13:380-399. [PMID: 35284107 PMCID: PMC8899750 DOI: 10.21037/jgo-22-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/21/2022] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is one of the most common malignant tumors of the digestive tract. Its clinical symptoms are obscure and atypical. It is difficult to diagnose and treat. Tumor cells mainly obtain energy through glycolysis to promote their growth. Inhibiting glycolysis can inhibit proliferation and kill tumor cells. METHODS Using bioinformatics method, we investigate the relationships between glycolysis-related genes and PC tumor samples' epidemiologic information comprehensively. RESULTS Different expression levels of 27 genes were identified. Using bioinformatics methods, we plotted two subgroup curves based on glycolysis-related gene expression level. Potential predictive genes were screened and their prognostic values were analyzed. Survival among high-risk group and low risk group had significant difference. Receiver operating characteristic (ROC) curve analysis indicated that area under curve (AUC) of 10 genes was greater than 0.8. These genes could be used for clinical diagnosis and prediction for PC. Two potential predictors [Kinesin Family Member 20A (KIF20A) and MET Proto-Oncogene, Receptor Tyrosine Kinase (MET)] that met the independent predictive value were selected. In univariate analysis, we screened out 3 regulators MET, protein kinase CAMP-activated catalytic subunit alpha (PRKACA) and KIF20A. According to the 3 regulatory factors, the prognostic signals of PC were constructed, by which the samples with good prognosis and poor prognosis can be clearly distinguished independently of potential confounding factors. CONCLUSIONS Our results indicate that for PC, glycolysis -related genes could be promising therapeutic targets or prognostic indicators.
Collapse
Affiliation(s)
- Jiachao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhehao Liu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhensheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Rong Tang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yongchao Zeng
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Pingping Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
22
|
Watts D, Jaykar MT, Bechmann N, Wielockx B. Hypoxia signaling pathway: A central mediator in endocrine tumors. Front Endocrinol (Lausanne) 2022; 13:1103075. [PMID: 36699028 PMCID: PMC9868855 DOI: 10.3389/fendo.2022.1103075] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Adequate oxygen levels are essential for the functioning and maintenance of biological processes in virtually every cell, albeit based on specific need. Thus, any change in oxygen pressure leads to modulated activation of the hypoxia pathway, which affects numerous physiological and pathological processes, including hematopoiesis, inflammation, and tumor development. The Hypoxia Inducible Factors (HIFs) are essential transcription factors and the driving force of the hypoxia pathway; whereas, their inhibitors, HIF prolyl hydroxylase domain (PHDs) proteins are the true oxygen sensors that critically regulate this response. Recently, we and others have described the central role of the PHD/HIF axis in various compartments of the adrenal gland and its potential influence in associated tumors, including pheochromocytomas and paragangliomas. Here, we provide an overview of the most recent findings on the hypoxia signaling pathway in vivo, including its role in the endocrine system, especially in adrenal tumors.
Collapse
|
23
|
Zhang Z, Zhang HJ. Glycometabolic rearrangements-aerobic glycolysis in pancreatic ductal adenocarcinoma (PDAC): roles, regulatory networks, and therapeutic potential. Expert Opin Ther Targets 2021; 25:1077-1093. [PMID: 34874212 DOI: 10.1080/14728222.2021.2015321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Glycometabolic rearrangements (aerobic glycolysis) is a hallmark of pancreatic ductal adenocarcinoma (PDAC) and contributes to tumorigenesis and progression through numerous mechanisms. The targeting of aerobic glycolysis is recognized as a potential therapeutic strategy which offers the possibility of improving treatment outcomes for PDAC patients. AREAS COVERED In this review, the role of aerobic glycolysis and its regulatory networks in PDAC are discussed. The targeting of aerobic glycolysis in PDAC is examined, and its therapeutic potential is evaluated. The relevant literature published from 2001 to 2021 was searched in databases including PubMed, Scopus, and Embase. EXPERT OPINION Regulatory networks of aerobic glycolysis in PDAC are based on key factors such as c-Myc, hypoxia-inducible factor 1α, the mammalian target of rapamycin pathway, and non-coding RNAs. Experimental evidence suggests that modulators or inhibitors of aerobic glycolysis promote therapeutic effects in preclinical tumor models. Nevertheless, successful clinical translation of drugs that target aerobic glycolysis in PDAC is an obstacle. Moreover, it is necessary to identify the potential targets for future interventions from regulatory networks to design efficacious and safer agents.
Collapse
Affiliation(s)
- Zhong Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People's Republic of China
| | - Hai-Jun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
24
|
Wang C, Huang B, Sun L, Wang X, Zhou B, Tang H, Geng W. MK8722, an AMPK activator, inhibiting carcinoma proliferation, invasion and migration in human pancreatic cancer cells. Biomed Pharmacother 2021; 144:112325. [PMID: 34656065 DOI: 10.1016/j.biopha.2021.112325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND MK8722 is a potent and systemic pan-AMPK activator. It is an effective, direct, allosteric activator of AMPK complex in many mammals. This study tried to explore the underlying anti-cancer molecular mechanism of MK8722 in human pancreatic cancer cells (PCCs). METHODS The anti-proliferation, invasion and migration functions of MK8722 in human pancreatic cancer analyzed by real time cellular analysis, colony formation assay, cell migration assay, transwell assay and flow cytometery analysis. Moreover, the potential targeted signaling pathway was tested via RNA-seq and pathway enrichment analysis. RESULTS In the present study, we investigated the anti-PCCs effects of MK8722 on two different human pancreatic cancer cell lines (PANC-1 and Patu8988). The results showed that MK8722 significantly inhibited human tumor cells proliferation and migration/invasion in a dose-dependent manner. Additionally, the influence of MK8722 was examined by analyzing the expression of potential key genes and pathways, which may provide novel insights to the mechanism of MK8722. CONCLUSION The inhibition of pancreatic cancer by MK8722 through a number of pathways that inhibit carcinoma proliferation, invasion and migration. The potential effect of MK8722 might be determined by regulating the expression of AL162151, IER2, REPIN1, KRT80 to inhibit cycle arrest and migration.
Collapse
Affiliation(s)
- Cheng Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Baojun Huang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Linxiao Sun
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Xi Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Baofeng Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Hongli Tang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Wujun Geng
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; Wenzhou Key Laboratory of perioperative medicine (NO. 2021HZSY0037).
| |
Collapse
|
25
|
Zhang D, Li Y, Yang S, Wang M, Yao J, Zheng Y, Deng Y, Li N, Wei B, Wu Y, Zhai Z, Dai Z, Kang H. Identification of a glycolysis-related gene signature for survival prediction of ovarian cancer patients. Cancer Med 2021; 10:8222-8237. [PMID: 34609082 PMCID: PMC8607265 DOI: 10.1002/cam4.4317] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 08/22/2021] [Accepted: 08/31/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Ovarian cancer (OV) is deemed the most lethal gynecological cancer in women. The aim of this study was to construct an effective gene prognostic model for predicting overall survival (OS) in patients with OV. METHODS The expression profiles of glycolysis-related genes (GRGs) and clinical data of patients with OV were extracted from The Cancer Genome Atlas (TCGA) database. Univariate, multivariate, and least absolute shrinkage and selection operator Cox regression analyses were conducted, and a prognostic signature based on GRGs was constructed. The predictive ability of the signature was analyzed using training and test sets. RESULTS A gene risk signature based on nine GRGs (ISG20, CITED2, PYGB, IRS2, ANGPTL4, TGFBI, LHX9, PC, and DDIT4) was identified to predict the survival outcome of patients with OV. The signature showed a good prognostic ability for OV, particularly high-grade OV, in the TCGA dataset, with areas under the curve (AUC) of 0.709 and 0.762 for 3- and 5-year survival, respectively. Similar results were found in the test sets, and the AUCs of 3-, 5-year OS were 0.714 and 0.772 in the combined test set. And our signature was an independent prognostic factor. Moreover, a nomogram combining the prediction model and clinical factors was developed. CONCLUSION Our study established a nine-GRG risk model and nomogram to better predict OS in patients with OV. The risk model represents a promising and independent prognostic predictor for patients with OV. Moreover, our study on GRGs could offer guidance for the elucidation of underlying mechanisms in future studies.
Collapse
Affiliation(s)
- Dai Zhang
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Department of Thyroid, Breast and Vascular SurgeryXijing HospitalThe Air Force Medical UniversityXi'anChina
| | - Yiche Li
- Department of Tumor SurgeryShaanxi Provincial People's HospitalXi'anChina
| | - Si Yang
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Meng Wang
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Jia Yao
- Department of Breast SurgeryThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Yi Zheng
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yujiao Deng
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Na Li
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Bajin Wei
- Department of Breast SurgeryThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Ying Wu
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Department of Breast SurgeryThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Zhen Zhai
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Zhijun Dai
- Department of Breast SurgeryThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Huafeng Kang
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
26
|
Bao M, Shang F, Liu F, Hu Z, Wang S, Yang X, Yu Y, Zhang H, Jiang C, Jiang J, Liu Y, Wang X. Comparative transcriptomic analysis of the brain in Takifugu rubripes shows its tolerance to acute hypoxia. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:1669-1685. [PMID: 34460041 DOI: 10.1007/s10695-021-01008-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
Hypoxia in water that caused by reduced levels of oxygen occurred frequently, due to the complex aquatic environment. Hypoxia tolerance for fish depends on a complete set of coping mechanisms such as oxygen perception and gene-protein interaction regulation. The present study examined the short-term effects of hypoxia on the brain in Takifugu rubripes. We sequenced the transcriptomes of the brain in T. rubripes to study their response mechanism to acute hypoxia. A total of 167 genes were differentially expressed in the brain of T. rubripes after exposed to acute hypoxia. Gene ontology and KEGG enrichment analysis indicated that hypoxia could cause metabolic and neurological changes, showing the clues of their adaptation to acute hypoxia. As the most complex and important organ, the brain of T. rubripes might be able to create a self-protection mechanism to resist or reduce damage caused by acute hypoxia stress.
Collapse
Affiliation(s)
- Mingxiu Bao
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Fengqin Shang
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
- College of Marine Technology and Environment, Dalian Ocean University, Dalian, 116023, China
| | - Fujun Liu
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Ziwen Hu
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Shengnan Wang
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Xiao Yang
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Yundeng Yu
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Hongbin Zhang
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Chihang Jiang
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Jielan Jiang
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China
| | - Yang Liu
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China.
| | - Xiuli Wang
- College of Fisheries and Life Science, Dalian Ocean University, 52 Heishijiao Street, DalianLiaoning, 116023, China.
| |
Collapse
|
27
|
Tao Y, Shao F, Cai M, Liu Z, Peng Y, Huang Q, Meng F. Activated Pancreatic Stellate Cells Enhance the Warburg Effect to Cause the Malignant Development in Chronic Pancreatitis. Front Oncol 2021; 11:714598. [PMID: 34540683 PMCID: PMC8446660 DOI: 10.3389/fonc.2021.714598] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/16/2021] [Indexed: 12/03/2022] Open
Abstract
Chronic pancreatitis (CP) is a precancerous condition associated with pancreatic ductal adenocarcinoma (PDAC), but its evolutionary mechanism is unclear. Pancreatic stellate cells (PSCs) are closely related to the occurrence and development of CP and PDAC, but it is not clear whether PSCs play a key role in this “inflammation-cancer transition”. Our research found that co-culture with activated PSCs promoted the proliferation, migration and invasion of normal pancreatic duct epithelial cells and pancreatic cancer cells. At the same time, activated PSCs had a significant effect on the expression of the glycolysis markers (pyruvate kinase M2, lactate dehydrogenase A, glucose transporter 1, hexokinase-II and monocarboxylate transporter 4; PKM2, LDHA, GLUT1, HK2 and MCT4) in normal pancreatic duct epithelial cells and pancreatic cancer cells and increased lactic acid production and glucose consumption in these two cells. In vivo experiments showed that the expression of the glycolysis markers in pancreatic duct epithelial cells and the marker protein (α-SMA) of activated PSCs in the pancreatic duct peripancreatic interstitium were higher in pancreatic cancer tissues and chronic pancreatitis tissues than in normal pancreatic tissues in both animals and humans. In addition, analysis of human tissue specimens showed that there is a correlation between the expression of glycolysis markers and α-SMA. These findings indicate that activated PSCs play an important role in the development and progression of chronic pancreatitis into pancreatic cancer by regulating and promoting aerobic glycolysis. Our research provides a new theoretical basis for further understanding the mechanism of CP malignancy and the selection of targets for reversing CP malignancy.
Collapse
Affiliation(s)
- Ye Tao
- Department of General Surgery, Anhui Provincial Hospital Affiliated of Anhui Medical University, Hefei, China
| | - Feng Shao
- Department of General Surgery, The First Affiliated Hospital of University of Science & Technology of China, Anhui Provincial Hospital, Hefei, China
| | - Ming Cai
- Department of General Surgery, Anhui Provincial Hospital Affiliated of Anhui Medical University, Hefei, China
| | - Zhen Liu
- Department of General Surgery, The First Affiliated Hospital of University of Science & Technology of China, Anhui Provincial Hospital, Hefei, China
| | - Yao Peng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiang Huang
- Department of General Surgery, Anhui Provincial Hospital Affiliated of Anhui Medical University, Hefei, China
| | - Futao Meng
- Department of General Surgery, The First Affiliated Hospital of University of Science & Technology of China, Anhui Provincial Hospital, Hefei, China.,Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
28
|
Abou Khouzam R, Rao SP, Venkatesh GH, Zeinelabdin NA, Buart S, Meylan M, Nimmakayalu M, Terry S, Chouaib S. An Eight-Gene Hypoxia Signature Predicts Survival in Pancreatic Cancer and Is Associated With an Immunosuppressed Tumor Microenvironment. Front Immunol 2021; 12:680435. [PMID: 34093582 PMCID: PMC8173254 DOI: 10.3389/fimmu.2021.680435] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022] Open
Abstract
Intratumoral hypoxia is a widely established element of the pancreatic tumor microenvironment (TME) promoting immune escape, tumor invasion, and progression, while contributing to treatment resistance and poor survival. Despite this critical role, hypoxia is underrepresented in molecular signatures of pancreatic ductal adenocarcinoma (PDA) and concurrent investigations into the hypoxia-immune status are lacking. In this work a literature-based approach was applied to derive an eight-gene hypoxia signature that was validated in fourteen cancer cell lines and in a cohort of PDA. The eight-gene hypoxia signature was significantly associated with overall survival in two distinct PDA datasets and showed independent prognostic value in multivariate analysis. Comparative analysis of tumors according to their hypoxia score (high versus low) determined that tumors with high hypoxia were significantly less enriched in cytotoxic T-cells, and cytolytic activity. In addition, they had lower expression of cytokines and tumor inflammatory markers, pointing to the signature’s ability to discern an immune “cold”, hypoxic TME. Combining the signature with an immune metric highlighted a worse survival probability in patients with high hypoxia and low immune reactivity, indicating that this approach could further refine survival estimates. Hypoxia as determined by our signature, was significantly associated with certain immune checkpoint inhibitors (ICI) biomarkers, suggesting that the signature reflects an aspect of the TME that is worth pursuing in future clinical trials. This is the first work of its kind in PDA, and our findings on the hypoxia-immune tumor contexture are not only relevant for ICI but could also guide combinatorial hypoxia-mediated therapeutic strategies in this cancer type.
Collapse
Affiliation(s)
- Raefa Abou Khouzam
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Shyama Prasad Rao
- Bioinformatics Division, Yenepoya Research Center, Yenepoya University, Mangalore, India
| | - Goutham Hassan Venkatesh
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Nagwa Ahmed Zeinelabdin
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Stephanie Buart
- INSERM UMR 1186, Integrative Tumor Immunology and Cancer Immunotherapy, Gustave Roussy, EPHE, Faculty De médecine Univ. Paris-Sud, University Paris-Saclay, Villejuif, France
| | - Maxime Meylan
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, F-75006, Paris, France
| | - Manjunath Nimmakayalu
- Graduate Program in Diagnostic Genetics and Genomics, School of Health Professions, MD Anderson Cancer Center, The University of Texas, Houston, TX, United States
| | - Stéphane Terry
- INSERM UMR 1186, Integrative Tumor Immunology and Cancer Immunotherapy, Gustave Roussy, EPHE, Faculty De médecine Univ. Paris-Sud, University Paris-Saclay, Villejuif, France
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates.,INSERM UMR 1186, Integrative Tumor Immunology and Cancer Immunotherapy, Gustave Roussy, EPHE, Faculty De médecine Univ. Paris-Sud, University Paris-Saclay, Villejuif, France
| |
Collapse
|
29
|
Zhang Z, Xu L, Xu X. The role of transcription factor 7-like 2 in metabolic disorders. Obes Rev 2021; 22:e13166. [PMID: 33615650 DOI: 10.1111/obr.13166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/08/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022]
Abstract
Transcription factor 7-like 2 (TCF7L2), a member of the T cell factor/lymphoid enhancer factor family, generally forms a complex with β-catenin to regulate the downstream target genes as an effector of the canonical Wnt signalling pathway. TCF7L2 plays a vital role in various biological processes and functions in many organs and tissues, including the liver, islet and adipose tissues. Further, TCF7L2 down-regulates hepatic gluconeogenesis and promotes lipid accumulation. In islets, TCF7L2 not only affects the insulin secretion of the β-cells but also has an impact on other cells. In addition, TCF7L2 influences adipogenesis in adipose tissues. Thus, an out-of-control TCF7L2 expression can result in metabolic disorders. The TCF7L2 gene is composed of 17 exons, generating 13 different transcripts, and has many single-nucleotide polymorphisms (SNPs). The discovery that these SNPs have an impact on the risk of type 2 diabetes (T2D) has attracted thorough investigations in the study of TCF7L2. Apart from T2D, TCF7L2 SNPs are also associated with type 1, posttransplant and other types of diabetes. Furthermore, TCF7L2 variants affect the progression of other disorders, such as obesity, cancers, metabolic syndrome and heart diseases. Finally, the interaction between TCF7L2 variants and diet also needs to be investigated.
Collapse
Affiliation(s)
- Zhensheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Zhejiang University School of Medicine, Hangzhou, China
| | - Li Xu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
30
|
DUOX2 As a Potential Prognostic Marker which Promotes Cell Motility and Proliferation in Pancreatic Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6530298. [PMID: 33748270 PMCID: PMC7943273 DOI: 10.1155/2021/6530298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/04/2020] [Accepted: 01/18/2021] [Indexed: 12/25/2022]
Abstract
DUOX2 has been reported to highly express in several types of cancers. However, the prognostic significance and the biological function of DUOX2 expression with pancreatic cancer (PC) still remain unclear. The present study is aimed at investigating whether DUOX2 could act as a novel biomarker of prognosis and evaluating its effect on PC cell progression. The mRNA and protein expression of DUOX2 in PC cells and tissues were assessed by quantitative real-time PCR (RT-qPCR) and immunohistochemistry. The effect of DUOX2 expression on PC cell motility and proliferation was evaluated in vitro. The correlation between DUOX2 mRNA expression and clinicopathological features and its prognostic significance were analyzed according to the Gene Expression Profiling Interactive Analysis (GEPIA) website based on The Cancer Genome Atlas (TCGA) and the GTEx databases combined with our clinical information. According to bioinformatics analysis, we forecasted the upstream transcription factors (TFs) and microRNA (miRNA) regulatory mechanism of DUOX2 in PC. The expression of DUOX2 at transcriptional and protein level was dramatically increased in PC specimens when compared to adjacent nontumor specimens. Functionally, DUOX2 knockdown inhibited cell motility and proliferation activities. Our clinical data revealed that the patients had better postoperative overall survival (OS) with lower expression of DUOX2, which is consistent with GEPIA data. Multivariate analysis revealed that high DUOX2 expression was considered as an independent prognostic indicator for OS (P = 0.031). Based on Cistrome database, the top 5 TFs of each positively and negatively association with DUOX2 were predicted. hsa-miR-5193 and hsa-miR-1343-3p targeting DUOX2 were forecasted from TargetScan, miRDB, and DIANA-TarBase databases, which were negatively correlated with OS (P = 0.043 and P = 0.0088, respectively) and DUOX2 expression (P = 0.0093 and P = 0.0032, respectively) in PC from TCGA data. These findings suggest that DUOX2 acts as a promising predictive biomarker and an oncogene in PC, which could be a therapeutic target for PC.
Collapse
|
31
|
Tao J, Yang G, Zhou W, Qiu J, Chen G, Luo W, Zhao F, You L, Zheng L, Zhang T, Zhao Y. Targeting hypoxic tumor microenvironment in pancreatic cancer. J Hematol Oncol 2021; 14:14. [PMID: 33436044 PMCID: PMC7805044 DOI: 10.1186/s13045-020-01030-w] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/25/2020] [Indexed: 12/13/2022] Open
Abstract
Attributable to its late diagnosis, early metastasis, and poor prognosis, pancreatic cancer remains one of the most lethal diseases worldwide. Unlike other solid tumors, pancreatic cancer harbors ample stromal cells and abundant extracellular matrix but lacks vascularization, resulting in persistent and severe hypoxia within the tumor. Hypoxic microenvironment has extensive effects on biological behaviors or malignant phenotypes of pancreatic cancer, including metabolic reprogramming, cancer stemness, invasion and metastasis, and pathological angiogenesis, which synergistically contribute to development and therapeutic resistance of pancreatic cancer. Through various mechanisms including but not confined to maintenance of redox homeostasis, activation of autophagy, epigenetic regulation, and those induced by hypoxia-inducible factors, intratumoral hypoxia drives the above biological processes in pancreatic cancer. Recognizing the pivotal roles of hypoxia in pancreatic cancer progression and therapies, hypoxia-based antitumoral strategies have been continuously developed over the recent years, some of which have been applied in clinical trials to evaluate their efficacy and safety in combinatory therapies for patients with pancreatic cancer. In this review, we discuss the molecular mechanisms underlying hypoxia-induced aggressive and therapeutically resistant phenotypes in both pancreatic cancerous and stromal cells. Additionally, we focus more on innovative therapies targeting the tumor hypoxic microenvironment itself, which hold great potential to overcome the resistance to chemotherapy and radiotherapy and to enhance antitumor efficacy and reduce toxicity to normal tissues.
Collapse
Affiliation(s)
- Jinxin Tao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Wenchuan Zhou
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, 200092, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Guangyu Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China. .,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
| |
Collapse
|
32
|
Zhang D, Zheng Y, Yang S, Li Y, Wang M, Yao J, Deng Y, Li N, Wei B, Wu Y, Zhu Y, Li H, Dai Z. Identification of a Novel Glycolysis-Related Gene Signature for Predicting Breast Cancer Survival. Front Oncol 2021; 10:596087. [PMID: 33489894 PMCID: PMC7821871 DOI: 10.3389/fonc.2020.596087] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
To identify a glycolysis-related gene signature for the evaluation of prognosis in patients with breast cancer, we analyzed the data of a training set from TCGA database and four validation cohorts from the GEO and ICGC databases which included 1,632 patients with breast cancer. We conducted GSEA, univariate Cox regression, LASSO, and multiple Cox regression analysis. Finally, an 11-gene signature related to glycolysis for predicting survival in patients with breast cancer was developed. And Kaplan-Meier analysis and ROC analyses suggested that the signature showed a good prognostic ability for BC in the TCGA, ICGC, and GEO datasets. The analyses of univariate Cox regression and multivariate Cox regression revealed that it's an important prognostic factor independent of multiple clinical features. Moreover, a prognostic nomogram, combining the gene signature and clinical characteristics of patients, was constructed. These findings provide insights into the identification of breast cancer patients with a poor prognosis.
Collapse
Affiliation(s)
- Dai Zhang
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yi Zheng
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Si Yang
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yiche Li
- Breast Center Department, The Fourth Hospital of Hebei Medical University, Hebei Medical University, Shijiazhuang, China
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jia Yao
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yujiao Deng
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Na Li
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bajin Wei
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Wu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yuyao Zhu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hongtao Li
- Department of Breast Head and Neck surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Tumor Hospital), Urumqi, China
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
33
|
Abstract
Pancreatic cancer (PC) is one of the deadliest malignancies. The high mortality rate of PC largely results from delayed diagnosis and early metastasis. Therefore, identifying novel treatment targets for patients with PC is urgently required to improve survival rates. A major barrier to successful treatment of PC is the presence of a hypoxic tumor microenvironment, which is associated with poor prognosis, treatment resistance, increased invasion and metastasis. Recent studies have identified a number of novel molecules and pathways in PC cells that promote cancer cells progression under hypoxic conditions, which may provide new therapy strategies to inhibit the development and metastasis of PC. This review summarizes the latest research of hypoxia in PC and provides an overview of how the current therapies have the capacity to overcome hypoxia and improve PC patient treatment. These findings will eventually provide guidance for future PC management and clinical trials and hopefully improve the survival of patients with PC.
Collapse
Affiliation(s)
- Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
34
|
Wu D, Jia H, Zhang Z, Li S. Circ-PRMT5 promotes breast cancer by the miR-509-3p/TCF7L2 axis activating the PI3K/AKT pathway. J Gene Med 2020; 23:e3300. [PMID: 33277756 DOI: 10.1002/jgm.3300] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 11/22/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Breast cancer is the most prevalent malignancy occurring in females. In recent years, emerging evidence has suggested that circular RNAs are involved in the development of multiple cancers. Circ-PRMT5 has recently attracted attention as a tumor-promoting circular RNA. In the present study, we focused on exploring the biological effects of circ-PRMT5 in breast cancer. METHODS A quantitative real-time polymerase chain reaction was used to determine the expression of circ-PRMT5 in breast cancer. In vitro experiments, including cell-counting kit-8, 5-ethynyl-2'-deoxyuridine, flow cytometry and tube formation assays, were performed to test the effects of circ-PRMT5 on the cellular progression of breast cancer. Bioinformatic analysis, luciferase reporter, radioimmunoprecipitation and RNA-pull down assays were performed to predict the potential microRNAs interacting with circ-PRMT5 and mRNAs that can be targeted by miR-509-3p. RESULTS Circ-PRMT5 is up-regulated in breast cancer tissues and cells. Importantly, an elevation of circ-PRMT5 indicates a poor prognosis in patients with breast cancer. Functionally, knockdown of circ-PRMT5 suppresses cell proliferation and angiogenesis and increases cell apoptosis in breast cancer. Mechanistically, we identified that circ-PRMT5 up-regulates TCF7L2 expression by acting as a miR-509-3p sponge. The negative expression correlation between miR-509-3p and circ-PRMT5 or TCF7L2 in clinical tissues was further demonstrated. Rescue assays showed that TCF7L2 overexpression reverses the antitumoral effects of circ-PRMT5 knockdown on breast cancer cell processes. Additionally, we demonstrated that circ-PRMT5 activates the phosphoinositide 3-kinase (PI3K)/AKT pathway by up-regulation of TCF7L2. CONCLUSIONS Overall, our data indicate that the circ-PRMT5/miR-509-3p/TCF7L2 axis can aggravate the malignant character of breast cancer cells by the regulation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Di Wu
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongyao Jia
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhiru Zhang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
35
|
Wei M, Tan C, Tang Z, Lian Y, Huang Y, Chen Y, Chen C, Zhou W, Cai T, Hu J. Proteome-Wide Alterations of Asymmetric Arginine Dimethylation Associated With Pancreatic Ductal Adenocarcinoma Pathogenesis. Front Cell Dev Biol 2020; 8:545934. [PMID: 33344439 PMCID: PMC7744470 DOI: 10.3389/fcell.2020.545934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 11/06/2020] [Indexed: 02/04/2023] Open
Abstract
Arginine methylation catalyzed by protein arginine methyltransferases (PRMTs) performs essential roles in regulating cancer initiation and progression, but its implication in pancreatic ductal adenocarcinoma (PDAC) requires further elucidation. In this study, asymmetric dimethylarginine (ADMA)-containing peptides in PDAC cell line PANC-1 were identified by label-free quantitative proteomics combined with affinity purification, using human non-cancerous pancreatic ductal epithelium cell line HPDE6c7 as the control. In total, 289 ADMA sites in 201 proteins were identified in HPDE6c7 and PANC-1 cells, including 82 sites with lower dimethylation and 37 sites with higher dimethylation in PANC-1 cells compared with HPDE6c7 cells. These ADMA-containing peptides demonstrated significant enrichment of glycine and proline residues in both cell lines. Importantly, leucine residues were significantly enriched in ADMA-containing peptides identified only in HPDE6c7 cells or showing lower dimethylation in PANC-1 cells. ADMA-containing proteins were significantly enriched in multiple biological processes and signaling cascades associated with cancer development, such as spliceosome machinery, the Wnt/β-catenin, Hedgehog, tumor growth factor beta (TGF-β), and mitogen-activated protein kinase (MAPK) signaling pathways. Moreover, PDAC cell lines with enhanced cell viability showed lower PRMT4 protein abundance and global ADMA-containing protein levels compared with HPDE6c7. PRMT4 overexpression partially recovered ADMA-containing protein levels and repressed viability in PANC-1 cells. These results revealed significantly altered ADMA-containing protein profiles in human pancreatic carcinoma cells, which provided a basis for elucidating the pathogenic roles of PRMT-mediated protein methylation in pancreatic cancer.
Collapse
Affiliation(s)
- Meijin Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chaochao Tan
- Department of Clinical Laboratory, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China.,Translational Medicine Research Institute, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zhouqin Tang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingying Lian
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Huang
- Department of Emergency, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yi Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Congwei Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wen Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tao Cai
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiliang Hu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
36
|
Chiba N, Sunamura M, Nakagawa M, Koganezawa I, Yokozuka K, Kobayashi T, Hikita K, Ozawa Y, Okihara M, Sano T, Tomita K, Tsutsui R, Sugimoto M, Kawachi S. Overexpression of hydroxyproline via EGLN/HIF1A is associated with distant metastasis in pancreatic cancer. Am J Cancer Res 2020; 10:2570-2581. [PMID: 32905516 PMCID: PMC7471362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/10/2020] [Indexed: 06/11/2023] Open
Abstract
For pancreatic cancer, the probability of distant metastasis can help choose the best course of treatment. The aim of this study is to establish the efficacy of hydroxyproline as a biomarker for distant metastasis for pancreatic cancer and to clarify the mechanism of EGLN/HIF1A axis that controls the invasion and metastasis. Metabolites (hydroxyproline) and genes (EGLN2 and EGLN3) were identified by metabolome analysis of the serum with pancreatic cancers with and without distant metastasis. The mechanism of EGLN/HIF1A axis including angiogenesis was examined in pancreatic cancer cells. Hydroxyproline associated with these mechanisms was evaluated to suggest the association with overall survival in pancreatic cancer. Decreased expression of EGLN2 and EGLN3 in pancreatic cancer, via the HIF1A and TGF ß1 pathway, was associated with the induction of angiogenic factors, increased vascular invasion, and poor overall patient survival. Hydroxyproline concentrations were regulated via the HIF1A pathway by EGLN2 and EGLN3, and that increased concentrations of hydroxyproline promote the invasion and metastasis of pancreatic cancer cells. These results suggested that the expression of hydroxyproline through the HIF1A pathway induced by EGLN2 and EGLN3 could be a surrogate marker for treatment and might predict distant metastasis in pancreatic cancer.
Collapse
Affiliation(s)
- Naokazu Chiba
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Makoto Sunamura
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Masashi Nakagawa
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Itsuki Koganezawa
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Kei Yokozuka
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Toshimichi Kobayashi
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Kosuke Hikita
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Yosuke Ozawa
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Masaaki Okihara
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Toru Sano
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Koichi Tomita
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Rina Tsutsui
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| | - Masahiro Sugimoto
- Research and Development Center for Minimally Invasive Therapies Health Promotion and Preemptive Medicine, Tokyo Medical UniversityTokyo, Japan
| | - Shigeyuki Kawachi
- Department of Digestive and Transplantation Surgery, Tokyo Medical University Hachioji Medical CenterTokyo, Japan
| |
Collapse
|
37
|
Feng J, Li J, Wu L, Yu Q, Ji J, Wu J, Dai W, Guo C. Emerging roles and the regulation of aerobic glycolysis in hepatocellular carcinoma. J Exp Clin Cancer Res 2020; 39:126. [PMID: 32631382 PMCID: PMC7336654 DOI: 10.1186/s13046-020-01629-4] [Citation(s) in RCA: 398] [Impact Index Per Article: 79.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Liver cancer has become the sixth most diagnosed cancer and the fourth leading cause of cancer death worldwide. Hepatocellular carcinoma (HCC) is responsible for up to 75-85% of primary liver cancers, and sorafenib is the first targeted drug for advanced HCC treatment. However, sorafenib resistance is common because of the resultant enhancement of aerobic glycolysis and other molecular mechanisms. Aerobic glycolysis was firstly found in HCC, acts as a hallmark of liver cancer and is responsible for the regulation of proliferation, immune evasion, invasion, metastasis, angiogenesis, and drug resistance in HCC. The three rate-limiting enzymes in the glycolytic pathway, including hexokinase 2 (HK2), phosphofructokinase 1 (PFK1), and pyruvate kinases type M2 (PKM2) play an important role in the regulation of aerobic glycolysis in HCC and can be regulated by many mechanisms, such as the AMPK, PI3K/Akt pathway, HIF-1α, c-Myc and noncoding RNAs. Because of the importance of aerobic glycolysis in the progression of HCC, targeting key factors in its pathway such as the inhibition of HK2, PFK or PKM2, represent potential new therapeutic approaches for the treatment of HCC.
Collapse
Affiliation(s)
- Jiao Feng
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
| |
Collapse
|
38
|
Jin X, Dai L, Ma Y, Wang J, Liu Z. Implications of HIF-1α in the tumorigenesis and progression of pancreatic cancer. Cancer Cell Int 2020; 20:273. [PMID: 32587480 PMCID: PMC7313137 DOI: 10.1186/s12935-020-01370-0] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer-related deaths worldwide and is characterized by highly hypoxic tumor microenvironment. Hypoxia-inducible factor-1 alpha (HIF-1α) is a major regulator of cellular response to changes in oxygen concentration, supporting the adaptation of tumor cells to hypoxia in an oxygen-deficient tumor microenvironment. Numerous studies revealed the central role of HIF-1α in the carcinogenesis and progression of pancreatic cancer. This article reviewed the molecular mechanisms of how HIF-1α regulated tumorigenesis and progression of pancreatic cancer and suggested that targeting HIF-1α and its signaling pathways could be promising therapeutics for pancreatic cancer.
Collapse
Affiliation(s)
- Xiao Jin
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| | - Lu Dai
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| | - Yilan Ma
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| | - Jiayan Wang
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| | - Zheng Liu
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| |
Collapse
|
39
|
Wenzel J, Rose K, Haghighi EB, Lamprecht C, Rauen G, Freihen V, Kesselring R, Boerries M, Hecht A. Loss of the nuclear Wnt pathway effector TCF7L2 promotes migration and invasion of human colorectal cancer cells. Oncogene 2020; 39:3893-3909. [PMID: 32203164 PMCID: PMC7203011 DOI: 10.1038/s41388-020-1259-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/25/2022]
Abstract
The transcription factor TCF7L2 is indispensable for intestinal tissue homeostasis where it transmits mitogenic Wnt/β-Catenin signals in stem and progenitor cells, from which intestinal tumors arise. Yet, TCF7L2 belongs to the most frequently mutated genes in colorectal cancer (CRC), and tumor-suppressive functions of TCF7L2 were proposed. This apparent paradox warrants to clarify the role of TCF7L2 in colorectal carcinogenesis. Here, we investigated TCF7L2 dependence/independence of CRC cells and the cellular and molecular consequences of TCF7L2 loss-of-function. By genome editing we achieved complete TCF7L2 inactivation in several CRC cell lines without loss of viability, showing that CRC cells have widely lost the strict requirement for TCF7L2. TCF7L2 deficiency impaired G1/S progression, reminiscent of the physiological role of TCF7L2. In addition, TCF7L2-negative cells exhibited morphological changes, enhanced migration, invasion, and collagen adhesion, albeit the severity of the phenotypic alterations manifested in a cell-line-specific fashion. To provide a molecular framework for the observed cellular changes, we performed global transcriptome profiling and identified gene-regulatory networks in which TCF7L2 positively regulates the proto-oncogene MYC, while repressing the cell cycle inhibitors CDKN2C/CDKN2D. Consistent with its function in curbing cell motility and invasion, TCF7L2 directly suppresses the pro-metastatic transcription factor RUNX2 and impinges on the expression of cell adhesion molecules. Altogether, we conclude that the proliferation-stimulating activity of TCF7L2 persists in CRC cells. In addition, TCF7L2 acts as invasion suppressor. Despite its negative impact on cell cycle progression, TCF7L2 loss-of-function may thereby increase malignancy, which could explain why TCF7L2 is mutated in a sizeable fraction of colorectal tumors.
Collapse
Affiliation(s)
- Janna Wenzel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
| | - Katja Rose
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, 79104, Freiburg, Germany
| | - Elham Bavafaye Haghighi
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79104, Freiburg, Germany
| | - Constanze Lamprecht
- Institute of Physics, University of Freiburg, Hermann-Herder-Str. 3a, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Freiburg Center for Interactive Materials and Bioinspired Technology (FIT), University of Freiburg, Georges-Köhler-Allee 105, 79110, Freiburg, Germany
| | - Gilles Rauen
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Vivien Freihen
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, 79104, Freiburg, Germany
| | - Rebecca Kesselring
- Department of General and Visceral Surgery, Center for Surgery, Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79104, Freiburg, Germany
- German Cancer Consortium (DKTK), Hugstetter Straße 55, 79106, Freiburg, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Andreas Hecht
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, 79104, Freiburg, Germany.
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
| |
Collapse
|
40
|
Carreira-Barbosa F, Nunes SC. Wnt Signaling: Paths for Cancer Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:189-202. [PMID: 32130700 DOI: 10.1007/978-3-030-34025-4_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Wnt signaling pathways are well known for having several pivotal roles during embryonic development. However, the same developmental signaling pathways also present key roles in cancer initiation and progression. In this chapter, several issues regarding the roles of both canonical and non-canonical Wnt signaling pathways in cancer will be explored, mainly concerning their role in the maintenance of cancer stemness, in the metabolism reprograming of cancer cells and in the modulation of the tumor microenvironment. The role of Wnt signaling cascades in the response of cancer cells to anti-cancer treatments will be also discussed, as well as its potential therapeutic targeting during cancer treatment. Collectively, increasing evidence has been supporting pivotal roles of Wnt signaling in several features of cancer biology, however; a lot is still to be elucidated.
Collapse
Affiliation(s)
| | - Sofia C Nunes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Lisbon, Portugal
| |
Collapse
|
41
|
Wensheng L, Bo Z, Qiangsheng H, Wenyan X, Shunrong J, Jin X, Quanxing N, Xianjun Y, Xiaowu X. MBD1 promotes the malignant behavior of gallbladder cancer cells and induces chemotherapeutic resistance to gemcitabine. Cancer Cell Int 2019; 19:232. [PMID: 31516389 PMCID: PMC6734348 DOI: 10.1186/s12935-019-0948-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 08/27/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Methyl-CpG binding domain protein 1 (MBD1), which couples DNA methylation to transcriptional repression, has been implicated in transcriptional regulation, heterochromatin formation, genomic stability, cell cycle progression and development. It has also been proven that MBD1 is involved in tumor development and progression. However, whether MBD1 is involved in tumorigenesis, especially in gallbladder cancer, is totally unknown. METHODS Human GBC-SD and SGC996 cells were used to perform experiments. Invasion, wound healing and colony formation assays were performed to evaluate cell viability. A CCK-8 assay was performed to assess gallbladder cancer cell viability after gemcitabine treatment. Western blot analysis was used to evaluate changes in protein expression. Human gallbladder cancer tissues and adjacent nontumor tissues were subjected to immunohistochemical staining to detect protein expression. RESULTS We found that MBD1 expression was significantly upregulated in gallbladder cancer tissues compared with that in surrounding normal tissues according to immunohistochemical analysis of 84 surgically resected gallbladder cancer specimens. These data also indicated that higher MBD1 expression was correlated with lymph node metastasis and poor survival in gallbladder cancer patients. Overexpression and deletion in vitro validated MBD1 as a potent oncogene promoting malignant behaviors in gallbladder cancer cells, including invasion, proliferation and migration, as well as epithelial-mesenchymal transition. Studies have demonstrated that epithelial-mesenchymal transition is common in gallbladder cancer, and it is well known that drug resistance and epithelial-mesenchymal transition are very closely correlated. Herein, our data show that targeting MBD1 restored gallbladder cancer cell sensitivity to gemcitabine chemotherapy. CONCLUSIONS Taken together, the results of our study revealed a novel function of MBD1 in gallbladder cancer tumor development and progression through participation in the gallbladder cancer epithelial-mesenchymal transition program, which is involved in resistance to gemcitabine chemotherapy. Thus, MBD1 may be a potential therapeutic target for gallbladder cancer.
Collapse
Affiliation(s)
- Liu Wensheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| | - Zhang Bo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| | - Hu Qiangsheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| | - Xu Wenyan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| | - Ji Shunrong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| | - Xu Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| | - Ni Quanxing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| | - Yu Xianjun
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| | - Xu Xiaowu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032 Shanghai, People’s Republic of China
- Shanghai Pancreatic Cancer Institute, 200032 Shanghai, China
| |
Collapse
|
42
|
Chen X, Zhou W, Liang C, Shi S, Yu X, Chen Q, Sun T, Lu Y, Zhang Y, Guo Q, Li C, Zhang Y, Jiang C. Codelivery Nanosystem Targeting the Deep Microenvironment of Pancreatic Cancer. NANO LETTERS 2019; 19:3527-3534. [PMID: 31058513 DOI: 10.1021/acs.nanolett.9b00374] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered as one of the most aggressive malignancies due to its unique microenvironment of which the cardinal histopathological feature is the remarkable desmoplasia of the stroma, taking up about 80% of the tumor mass. The desmoplastic stroma negatively affects drug diffusion and the infiltration of T cells, leading to an immunosuppressive microenvironment. However, this unique microenvironment can limit the physical spread of pancreatic cancer via a neighbor suppression effect. Here, a tumor central stroma targeting and microenvironment responsive strategy was applied to generate a nanoparticle coloading paclitaxel and phosphorylated gemcitabine. The designed nanoparticle disrupted the central stroma while preserving the external stroma, thereby promoting the antitumor effectiveness of chemotherapeutics. Additionally, the resulting nanoparticle can modulate the tumor immunosuppressive microenvironment by augmenting the number of cytotoxic T cells and restraining the percentage of T regulatory cells. The relatively intact external stroma can effectively maintain the neighbor suppression effect and prevent tumor metastasis. Combining stroma targeting with the delivery of stimuli-responsive polymeric nanoparticles embodies an effective tumor-tailored drug delivery system.
Collapse
Affiliation(s)
- Xinli Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Wenxi Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Chen Liang
- Department of Pancreatic and Hepatobiliary Surgery , Fudan University Shanghai Cancer Center , 270 Dongan Road , Shanghai 200032 , China
- Department of Oncology, Shanghai Medical College , Fudan University , Shanghai 200032 , China
| | - Si Shi
- Department of Pancreatic and Hepatobiliary Surgery , Fudan University Shanghai Cancer Center , 270 Dongan Road , Shanghai 200032 , China
- Department of Oncology, Shanghai Medical College , Fudan University , Shanghai 200032 , China
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery , Fudan University Shanghai Cancer Center , 270 Dongan Road , Shanghai 200032 , China
- Department of Oncology, Shanghai Medical College , Fudan University , Shanghai 200032 , China
| | - Qinjun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Yifei Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Yujie Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Qin Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Chao Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Yu Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Research Center on Aging and Medicine, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| |
Collapse
|
43
|
Liu C, Li Y, Wei M, Zhao L, Yu Y, Li G. Identification of a novel glycolysis-related gene signature that can predict the survival of patients with lung adenocarcinoma. Cell Cycle 2019; 18:568-579. [PMID: 30727821 DOI: 10.1080/15384101.2019.1578146] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lung cancer is one of the most malignant cancers worldwide, and lung adenocarcinoma (LUAD) is the most common histologic subtype. Thousands of biomarkers related to the survival and prognosis of patients with this cancer type have been investigated through database mining; however, the prediction effect of a single gene biomarker is not satisfactorily specific or sensitive. Thus, the present study aimed to develop a novel gene signature of prognostic values for patients with LUAD. Using a data-mining method, we performed expression profiling of 1145 mRNAs in large cohorts with LUAD (n = 511) from The Cancer Genome Atlas database. Using the Gene Set Enrichment Analysis, we selected 198 genes related to GLYCOLYSIS, which is the most important enrichment gene set. Moreover, these genes were identified using Cox proportional regression modeling. We established a risk score staging system to predict the outcome of patients with LUAD and subsequently identified four genes (AGRN, AKR1A1, DDIT4, and HMMR) that were closely related to the prognosis of patients with LUAD. The identified genes allowed us to classify patients into the high-risk group (with poor outcome) and low-risk group (with better outcome). Compared with other clinical factors, the risk score has a better performance in predicting the outcome of patients with LUAD, particularly in the early stage of LUAD. In conclusion, we developed a four-gene signature related to glycolysis by utilizing the Cox regression model and a risk staging model for LUAD, which might prove valuable for the clinical management of patients with LUAD.
Collapse
Affiliation(s)
- Chang Liu
- a Department of Radiation Oncology , The First Affiliated Hospital of China Medical University , Shenyang , China
| | - Yinyan Li
- b Department of Ultrasound , The First Affiliated Hospital of China Medical University , Shenyang , China
| | - Minjie Wei
- c Department of Pharmacology, School of Pharmacy , China Medical University , Shenyang , China
| | - Lin Zhao
- c Department of Pharmacology, School of Pharmacy , China Medical University , Shenyang , China
| | - Yangyang Yu
- a Department of Radiation Oncology , The First Affiliated Hospital of China Medical University , Shenyang , China
| | - Guang Li
- a Department of Radiation Oncology , The First Affiliated Hospital of China Medical University , Shenyang , China
| |
Collapse
|
44
|
Wu HH, Li YL, Liu NJ, Yang Z, Tao XM, Du YP, Wang XC, Lu B, Zhang ZY, Hu RM, Wen J. TCF7L2 regulates pancreatic β-cell function through PI3K/AKT signal pathway. Diabetol Metab Syndr 2019; 11:55. [PMID: 31312258 PMCID: PMC6612183 DOI: 10.1186/s13098-019-0449-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/24/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Transcription factor 7-like 2 (TCF7L2), which previously known as TCF-4, is a major form of transcription factor involved in the downstream WNT signaling and exhibits the strongest association to diabetes susceptibility. Although we still do not know mechanistically how TCF7L2 exerts its physiological functions on pancreatic endocrine cells, it had been suggested that TCF7L2 may directly affect β-cell function by regulating the activation of PI3K/AKT signaling pathway. METHODS MIN6 cells were transfected with TCF7L2 knockdown virus or lenti-TCF7L2 virus for 48 h to evaluate the contribution of TCF7L2 to the PI3K/AKT signaling pathway and pancreatic β-cell function. This was confirmed by measuring the expression of PI3K p85 and p-Akt by western blotting and insulin secretion by enzyme-linked immunosorbent assay (ELISA), respectively. Chromatin immunoprecipitation (ChIP) and polymerase chain reaction (PCR) experiments were performed to explore the genomic distribution of TCF7L2-binding sites in the promoter of PIK3R1, the affinity between which was analyzed by the luciferase reporter assay. RESULTS In the present study, we strikingly identified that TCF7L2 could profoundly inhibit the expression of PIK3R1 gene and its encoding protein PI3K p85, which then could lead to the activation of PI3K/AKT signaling and stimulate insulin secretion in pancreatic β-cells. However, the integrity and stability of evolutionarily conserved TCF7L2-binding motif plays a very crucial role in the binding events between transcription factor TCF7L2 and its candidate target genes. We also found that the affinity of TCF7L2 to the promoter region of PIK3R1 alters upon the specific binding sites, which further provides statistical validation to the necessity of TCF7L2-binding motif. CONCLUSIONS This study demonstrated that TCF7L2 is closely bound to the specific binding regions of PIK3R1 promoter and prominently controls the transcription of its encoding protein p85, which further affects the activation of PI3K/AKT signaling pathway and insulin secretion.
Collapse
Affiliation(s)
- Hui-Hui Wu
- Department of Endocrinology and Metabolism, Jing’an District Center Hospital of Shanghai, Shanghai, 200040 China
| | - Yan-Liang Li
- Department of Endocrinology and Metabolism, Huashan Hospital of Fudan University, NO. 12 Wulumuqi Mid Road, Building 0#, Jing’an District, Shanghai, 200040 China
| | - Nai-Jia Liu
- Department of Endocrinology and Metabolism, Huashan Hospital of Fudan University, NO. 12 Wulumuqi Mid Road, Building 0#, Jing’an District, Shanghai, 200040 China
| | - Zhen Yang
- Department of Endocrinology and Metabolism, Xin Hua Hospital, Shanghai Jiao Tong University, Shanghai, 200020 China
| | - Xiao-Ming Tao
- Department of Endocrinology and Metabolism, Hua Dong Hospital, Fudan University, Shanghai, 200040 China
| | - Yan-Ping Du
- Department of Endocrinology and Metabolism, Hua Dong Hospital, Fudan University, Shanghai, 200040 China
| | - Xuan-Chun Wang
- Department of Endocrinology and Metabolism, Huashan Hospital of Fudan University, NO. 12 Wulumuqi Mid Road, Building 0#, Jing’an District, Shanghai, 200040 China
| | - Bin Lu
- Department of Endocrinology and Metabolism, Huashan Hospital of Fudan University, NO. 12 Wulumuqi Mid Road, Building 0#, Jing’an District, Shanghai, 200040 China
| | - Zhao-Yun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital of Fudan University, NO. 12 Wulumuqi Mid Road, Building 0#, Jing’an District, Shanghai, 200040 China
| | - Ren-Ming Hu
- Department of Endocrinology and Metabolism, Huashan Hospital of Fudan University, NO. 12 Wulumuqi Mid Road, Building 0#, Jing’an District, Shanghai, 200040 China
| | - Jie Wen
- Department of Endocrinology and Metabolism, Jing’an District Center Hospital of Shanghai, Shanghai, 200040 China
- Department of Endocrinology and Metabolism, Huashan Hospital of Fudan University, NO. 12 Wulumuqi Mid Road, Building 0#, Jing’an District, Shanghai, 200040 China
| |
Collapse
|