1
|
Su X, Ding X, Ding C, Wang G, Fu C, Liu F, Shi J, He W. The role of JMJD2A in immune evasion and malignant behavior of esophageal squamous cell carcinoma. Int Immunopharmacol 2024; 137:112401. [PMID: 38878485 DOI: 10.1016/j.intimp.2024.112401] [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: 11/26/2023] [Revised: 06/01/2024] [Accepted: 06/02/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE This study aimed to investigate the role of JMJD2A in radiotherapy tolerance of esophageal squamous cell carcinoma (ESCC). METHODS The levels of H3K9me3 modification were analyzed in anti-PD-1 therapy non-responder or responder patients, and the expression differences of H3K9me3-related modifying enzymes were assessed in TCGA-ESCC and ICGC cohorts. Subsequently, JMJD2A was knocked down in ESCC cells using CRISPR-Cas9 or lentivirus-mediated shRNA, and changes in malignant behavior of ESCC cells were observed. RNA-seq, ATAC-seq, and ChIP-seq analyses were then conducted to investigate the genes and downstream signaling pathways regulated by JMJD2A, and functional validation experiments were performed to analyze the role of downstream regulated genes and pathways in ESCC malignant behavior and immune evasion. RESULTS JMJD2A was significantly overexpressed in ESCC and anti-PD-1 therapy non-responders. Knockdown or deletion of JMJD2A significantly promoted the malignant behavior and immune evasion of ESCC. JMJD2A facilitated the structural changes in chromatin and promoted the binding of SMARCA4 to super-enhancers, thereby inducing the expression of GPX4. This resulted in the inhibition of radiation-induced DNA damage and cell ferroptosis, ultimately promoting the malignant behavior and immune evasion of ESCC cells. CONCLUSION JMJD2A plays an indispensable role in the malignant behavior and immune evasion of ESCC. It regulates the binding of SMARCA4 to super-enhancers and affects the chromatin's epigenetic landscape, thereby promoting the expression of GPX4 and attenuating iron-mediated cell death caused by radiotherapy. Consequently, it triggers the malignant behavior and immune evasion of ESCC cells.
Collapse
Affiliation(s)
- Xiangyu Su
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China; School of Medicine, Southeast University, Nanjing 210009, China
| | - Xu Ding
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China; School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenxi Ding
- Department of Internal Medicine of Traditional Chinese Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Guoqing Wang
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenchun Fu
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fei Liu
- Department of Medical Oncology, Luhe People's Hospital of Nanjing, Nanjing 210000, China
| | - Jinjun Shi
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China.
| | - Wei He
- Department of Thoracic surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China.
| |
Collapse
|
2
|
Pei W, Yin W, Yu T, Zhang X, Zhang Q, Yang X, Shi C, Shen W, Liu G. Dual-Specificity Phosphatase 4 Promotes Malignant Features in Colorectal Cancer Through Cyclic-AMP Response Element Binding Protein/Protein Kinase CAMP-Activated Catalytic Subunit Beta Activation. Dig Dis Sci 2024; 69:2856-2874. [PMID: 38824257 DOI: 10.1007/s10620-024-08481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Previous studies have demonstrated that Dual-specificity phosphatase 4 (DUSP4) plays an important role in the progression of different tumor types. However, the role and mechanism of DUSP4 in colorectal cancer (CRC) remain unclear. AIMS We investigate the role and mechanisms of DUSP4 in CRC. METHODS Immunohistochemistry was used to investigate DUSP4 expression in CRC tissues. Cell proliferation, apoptosis and migration assays were used to validate DUSP4 function in vitro and in vivo. RNA-sequence assay was used to identify the target genes of DUSP4. Human phosphokinase array and inhibitor assays were used to explore the downstream signaling of DUSP4. RESULTS DUSP4 expression was upregulated in CRC tissues relative to normal colorectal tissues, and DUSP4 expression showed a significant positive correlation with CRC stage. Consistently, we found that DUSP4 was highly expressed in colorectal cancer cells compared to normal cells. DUSP4 knockdown inhibits CRC cell proliferation, migration and promotes apoptosis. Furthermore, the ectopic expression of DUSP4 enhanced CRC cell proliferation, migration and diminished apoptosis in vitro and in vivo. Human phosphokinase array data showed that ectopic expression of DUSP4 promotes CREB activation. RNA-sequencing data showed that PRKACB acts as a downstream target gene of DUSP4/CREB and enhances CREB activation through PKA/cAMP signaling. In addition, xenograft model results demonstrated that DUSP4 promotes colorectal tumor progression via PRKACB/CREB activation in vivo. CONCLUSION These findings suggest that DUSP4 promotes CRC progression. Therefore, it may be a promising therapeutic target for CRC.
Collapse
Affiliation(s)
- Wenju Pei
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of General Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272067, China
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Wanbin Yin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of General Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272067, China
| | - Tao Yu
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaoyuan Zhang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Qi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaowen Yang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Chunlei Shi
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Wenzhi Shen
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China.
| | - Gang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| |
Collapse
|
3
|
Liu X, Ye Z, Rao D, Chen Q, Zhang Z. DUSP4 maintains the survival and LSD1 protein stability in esophageal squamous cell carcinoma cells by inhibiting JNK signaling-dependent autophagy. In Vitro Cell Dev Biol Anim 2024; 60:115-122. [PMID: 38286920 DOI: 10.1007/s11626-023-00845-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 12/13/2023] [Indexed: 01/31/2024]
Abstract
DUSP4 is a biomarker of esophageal squamous cell carcinoma (ESCC), which is responsible for the prognosis in ESCC. However, the underlying mechanism of DUSP4-regulated ESCC carcinogenesis is unknown. As a negative regulator of JNK, DUSP4 can inhibit autophagy, which contributes to tumorigenesis. This study aimed to explore the role of autophagy in DUSP4-regulated ESCC carcinogenesis. Our results showed that DUSP4 overexpression inhibited autophagy and promoted LSD1 protein expression in ESCC cells, while DUSP4 silencing showed the opposite effects. However, DUSP4 overexpression and silencing did not affect LSD1 mRNA expression. But the regulatory ability of DUSP4 overexpression on autophagy, death level, and LSD1 protein was reversed by rapamycin. In addition, DUSP4 overexpression inhibited JNK and Bcl2 phosphorylation and the dissociation of Bcl2-Beclin1 complex, while DUSP4 silencing promoted JNK and Bcl2 phosphorylation. Moreover, the regulatory ability of DUSP4 overexpression on autophagy, death, and LSD1 protein was reversed by JNK activator anisomycin. The xenograft assays also showed that DUSP4 overexpression-promoted ESCC tumor growth in vivo and LC3II and LSD1 protein expression in tumor tissues were reversed by rapamycin or anisomycin. Overall, DUSP4 inhibits Bcl2-Beclin1-autophagy signal transduction through the negative regulation of JNK, thus suppressing autophagic death and the autophagic degradation of LSD1 in ESCC, by which DUSP4 promotes ESCC carcinogenesis.
Collapse
Affiliation(s)
- Xinxin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Gannan Branch of National Clinical Research Center for Geriatrics Ganzhou 341000, Jiangxi, China
| | - Zhou Ye
- Department of Digestive, The 900Th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350001, Fujian, China
| | - Dingyu Rao
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, No. 3, Outangli, Xingannan Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Qianshun Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian, China.
| | - Zuxiong Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, No. 3, Outangli, Xingannan Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China.
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
4
|
Hou Z, Lin J, Ma Y, Fang H, Wu Y, Chen Z, Lin X, Lu F, Wen S, Yu X, Huang H, Pan Y. Single-cell RNA sequencing revealed subclonal heterogeneity and gene signatures of gemcitabine sensitivity in pancreatic cancer. Front Pharmacol 2023; 14:1193791. [PMID: 37324492 PMCID: PMC10267405 DOI: 10.3389/fphar.2023.1193791] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Resistance to gemcitabine is common and critically limits its therapeutic efficacy in pancreatic ductal adenocarcinoma (PDAC). Methods: We constructed 17 patient-derived xenograft (PDX) models from PDAC patient samples and identified the most notable responder to gemcitabine by screening the PDX sets in vivo. To analyze tumor evolution and microenvironmental changes pre- and post-chemotherapy, single-cell RNA sequencing (scRNA-seq) was performed. Results: ScRNA-seq revealed that gemcitabine promoted the expansion of subclones associated with drug resistance and recruited macrophages related to tumor progression and metastasis. We further investigated the particular drug-resistant subclone and established a gemcitabine sensitivity gene panel (GSGP) (SLC46A1, PCSK1N, KRT7, CAV2, and LDHA), dividing PDAC patients into two groups to predict the overall survival (OS) in The Cancer Genome Atlas (TCGA) training dataset. The signature was successfully validated in three independent datasets. We also found that 5-GSGP predicted the sensitivity to gemcitabine in PDAC patients in the TCGA training dataset who were treated with gemcitabine. Discussion and conclusion: Our study provides new insight into the natural selection of tumor cell subclones and remodeling of tumor microenvironment (TME) cells induced by gemcitabine. We revealed a specific drug resistance subclone, and based on the characteristics of this subclone, we constructed a GSGP that can robustly predict gemcitabine sensitivity and prognosis in pancreatic cancer, which provides a theoretical basis for individualized clinical treatment.
Collapse
Affiliation(s)
- Zelin Hou
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiajing Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuan Ma
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Haizhong Fang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuwei Wu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhijiang Chen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xianchao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fengchun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shi Wen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | | | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
5
|
Zhao Y, Cai H, Ding X, Zhou X. An integrative analysis of the single-cell transcriptome identifies DUSP4 as an exhaustion-associated gene in tumor-infiltrating CD8+ T cells. Funct Integr Genomics 2023; 23:136. [PMID: 37086337 DOI: 10.1007/s10142-023-01056-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 04/23/2023]
Affiliation(s)
- Yu Zhao
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Huihui Cai
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Xiaoling Ding
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.
- Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, China.
| | - Xiaorong Zhou
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.
| |
Collapse
|
6
|
Xu W, Nie C, Chen X. DUSP4 inhibits autophagic cell death and apoptosis in colorectal cancer by regulating BCL2-Beclin1/Bax signaling. Mol Biol Rep 2023; 50:3229-3239. [PMID: 36705792 DOI: 10.1007/s11033-023-08270-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023]
Abstract
BACKGROUND The DUSP4 gene plays an important role in the carcinogenesis of colorectal cancer (CRC). However, the underlying mechanism of DUSP4-regulated colorectal carcinogenesis is unknown. DUSP4 is a negative regulator of the MAP kinase (MAPK) JNK, and JNK-mediated BCL2 phosphorylation is associated with apoptosis and autophagic cell death. Our study aimed to explore the significance of BCL2 phosphorylation-dependent autophagy and apoptosis in DUSP4-promoted colorectal carcinogenesis. METHODS We first investigated the roles of DUSP4 in the survival of HCT116 and SW480 CRC cell lines using gene-silencing and -overexpression techniques. Next, we explored the effects of DUSP4 on the BCL2 phosphorylation, autophagy and apoptosis of HCT116 and SW480 cells. Ultimately, with the help of pharmacological inhibitors of Beclin1 and BCL2 (spautin-1 and ABT-737), the relationship between BCL2-Beclin1/Bax signaling and DUSP4-regulated autophagy, apoptosis, survival and migration in HCT116 cells was clarified. RESULTS Our results first confirmed the contribution of DUSP4 to the survival of HCT116 and SW480 cells. In addition, DUSP4 silencing resulted in BCL2 phosphorylation and the enhancement in autophagy and apoptosis in HCT116 and SW480 cells, while DUSP4 overexpression showed the opposite effect. Moreover, DUSP4 silencing inhibited the protein interaction between BCL2 and Beclin1 or Bax in HCT116 cells. Moreover, the survival and migration of HCT116 cells inhibited by DUSP4 silencing were blocked by autophagy inhibition with spautin-1. Notably, the survival and migration of HCT116 cells promoted by DUSP4 overexpression were reversed by ABT-737. CONCLUSIONS It was indicated that DUSP4 can maintain the survival and function of CRC cells by inhibiting BCL2 phosphorylation-dependent autophagic cell death and apoptosis.
Collapse
Affiliation(s)
- Weifeng Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, No.127 Dong Ming Road, Zhengzhou, 450008, Henan, People's Republic of China
| | - Caiyun Nie
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, No.127 Dong Ming Road, Zhengzhou, 450008, Henan, People's Republic of China
| | - Xiaobing Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, No.127 Dong Ming Road, Zhengzhou, 450008, Henan, People's Republic of China.
| |
Collapse
|