1
|
Lin A, Song L, Wang Y, Yan K, Tang H. Future prospects of deep learning in esophageal cancer diagnosis and clinical decision support (Review). Oncol Lett 2025; 29:293. [PMID: 40271007 PMCID: PMC12016012 DOI: 10.3892/ol.2025.15039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Esophageal cancer (EC) is one of the leading causes of cancer-related mortality worldwide, still faces significant challenges in early diagnosis and prognosis. Early EC lesions often present subtle symptoms and current diagnostic methods are limited in accuracy due to tumor heterogeneity, lesion morphology and variable image quality. These limitations are particularly prominent in the early detection of precancerous lesions such as Barrett's esophagus. Traditional diagnostic approaches, such as endoscopic examination, pathological analysis and computed tomography, require improvements in diagnostic precision and staging accuracy. Deep learning (DL), a key branch of artificial intelligence, shows great promise in improving the detection of early EC lesions, distinguishing benign from malignant lesions and aiding cancer staging and prognosis. However, challenges remain, including image quality variability, insufficient data annotation and limited generalization. The present review summarized recent advances in the application of DL to medical images obtained through various imaging techniques for the diagnosis of EC at different stages. It assesses the role of DL in tumor pathology, prognosis prediction and clinical decision support, highlighting its advantages in EC diagnosis and prognosis evaluation. Finally, it provided an objective analysis of the challenges currently facing the field and prospects for future applications.
Collapse
Affiliation(s)
- Aiting Lin
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R. China
- Department of Thoracic Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai 200003, P.R. China
| | - Lirong Song
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R. China
| | - Ying Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R. China
| | - Kai Yan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, P.R. China
| | - Hua Tang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
2
|
Hao X, Zhang K, Hou Z, Guo J, Yang L, Sun S. Advances in natural polysaccharide/protein-based bioadhesive formulations for the potential application in esophagus: A review. Int J Biol Macromol 2025; 308:142513. [PMID: 40147657 DOI: 10.1016/j.ijbiomac.2025.142513] [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: 12/30/2024] [Revised: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
The esophagus is susceptible to various injuries or disorders, which can significantly impact quality of life and pose potentially life-threatening risks. The unique anatomical and physiological characteristics of the esophagus present challenges in achieving optimal bioavailability and efficacy during diagnosis and treatment. To address these challenges, polysaccharide- and protein-based bioadhesive formulations have been developed to adhere to esophageal tissue, thereby prolonging residence time and enhancing diagnostic accuracy and therapeutic outcomes. Natural polysaccharides and proteins have garnered attention in the medical field owing to their exceptional properties, including biocompatibility, bioavailability, biodegradability, and low toxicity. A substantial body of research has demonstrated the significant potential of polysaccharides and proteins in clinical applications for the esophagus. The objective of this review is to discuss the structural characteristics and biological activities of various polysaccharides, including chitosan, hyaluronic acid, alginate, cellulose, guar gum, gellan gum, and xanthan gum, as well as proteins such as gelatin and fibrin, and their utilization in esophageal bioadhesive formulations. The practical challenges and prospects associated with implementing polysaccharide and protein-based bioadhesives on the esophagus are also discussed.
Collapse
Affiliation(s)
- Xuanyu Hao
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Zhipeng Hou
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Jintao Guo
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Liqun Yang
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| | - Siyu Sun
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
3
|
Jiang H, Jin X, Gu H, Li B, Li Z, Sun Y. SPC25 upregulates CCND1 to promote the progression of esophageal squamous cell carcinoma by inhibiting MDM2-mediated E2F1 ubiquitination. Transl Oncol 2025; 53:102300. [PMID: 39919356 PMCID: PMC11849203 DOI: 10.1016/j.tranon.2025.102300] [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: 10/15/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/09/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is highly malignant worldwide. Despite significant advances in the treatment of ESCC, the prognosis remains unfavourable, necessitating research into its mechanisms and treatments. Spindle component 25 (SPC25) can ensure the fidelity of mitotic progression and the accurate segregation of chromosomes, thus plays an important role in the development of malignant tumors, but its role in ESCC is yet to be determined. In this study, the expression of SPC25 was assessed by IHC in 88 primary ESCC samples, with its expression being correlated with advanced clinical features. The function of SPC25 in the proliferation, migration and tumorigenicity of ESCC cells was verified in vitro and in vivo. Mechanistically, SPC25 facilitated tumorigenesis through promoting CCND1 expression. As the transcription factor for CCND1, E2F1 is stabilized by SPC25 through binding the ubiquitin ligase MDM2, resulting in enhanced E2F1 expression, which in turn promotes the expression of CCND1. In addition, overexpression of CCND1 counteracted the effects of SPC25 silencing. Collectively, we demonstrated that the aberrant expression of SPC25 inhibited E2F1 ubiquitination and promoted CCND1 expression, thus accelerating the progression of ESCC. These findings propose novel insights into the role of SPC25 in ESCC and provide potential therapeutic strategies for targeting SPC25 in ESCC patients.
Collapse
Affiliation(s)
- Haoyao Jiang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Xiangfeng Jin
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Haiyong Gu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Bin Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China.
| | - Yifeng Sun
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China.
| |
Collapse
|
4
|
Meng YQ, Feng HM, Li B, Xie Y, Li Z, Li ZQ, Li X. PYCR1 Promotes Esophageal Squamous Cell Carcinoma by Interacting With EGFR to Affecting the PI3K/Akt/mTOR Signaling Pathway. J Gene Med 2025; 27:e70017. [PMID: 40102683 DOI: 10.1002/jgm.70017] [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: 01/10/2025] [Revised: 02/18/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND The expression and functional role of pyrroline-5-carboxylate reductase 1 (PYCR1) in esophageal squamous cell carcinoma (ESCC) remain poorly understood. This study aimed to elucidate the role and underlying mechanisms of PYCR1 in ESCC. METHODS We utilized an ESCC tissue microarray coupled with immunohistochemical staining to assess variability in PYCR1 protein expression among ESCC patients and evaluate its clinical relevance. PYCR1 was silenced in ESCC cell lines with short hairpin RNA (shRNA), followed by functional assays (colony formation, caspase 3/7 activity, methylthiazol tetrazolium, wound healing, and migration/invasion assays) to evaluate its role in ESCC progression. In vivo, mouse tumor xenograft models were used to examine PYCR1's impact on tumor growth. To identify downstream targets and pathways, we conducted coimmunoprecipitation, mass spectrometry, immunofluorescence, and proteomic analyses, validated by western blotting and rescue experiments. RESULTS Our findings demonstrated a consistent upregulation of PYCR1 in ESCC tissues. Both in vitro and in vivo studies revealed that PYCR1 suppression significantly inhibited ESCC progression, impacting key processes such as proliferation, apoptosis, migration, and invasion. Mechanistically, PYCR1 was shown to interact with EGFR, promoting ESCC progression and metastasis by activating the PI3K/AKT/mTOR signaling pathways, which are integral to the aggressive behavior of the disease. Rescue experiments further confirmed that EGFR overexpression effectively reversed the inhibitory effects of PYCR1 knockdown in ESCC cells. CONCLUSION This study highlights the critical role of PYCR1 in driving ESCC progression and metastasis, underscoring its potential as a promising therapeutic target for managing this malignancy.
Collapse
Affiliation(s)
- Yu-Qi Meng
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
| | - Hai-Ming Feng
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
| | - Bin Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
| | - Yuan Xie
- Anesthesia Nursing Unit, Gansu Provincial Hospital of TCM, Lanzhou, China
| | - Zheng Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
| | - Zhen-Qing Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
| | - Xuan Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
| |
Collapse
|
5
|
Udupi A, Shetty S, Aranjani JM, Kumar R, Bharati S. Anticancer therapeutic potential of multimodal targeting agent- "phosphorylated galactosylated chitosan coated magnetic nanoparticles" against N-nitrosodiethylamine-induced hepatocellular carcinoma. Drug Deliv Transl Res 2025; 15:1023-1042. [PMID: 38990437 PMCID: PMC11782354 DOI: 10.1007/s13346-024-01655-1] [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] [Accepted: 06/19/2024] [Indexed: 07/12/2024]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) are extensively used as carriers in targeted drug delivery and has several advantages in the field of magnetic hyperthermia, chemodynamic therapy and magnet assisted radionuclide therapy. The characteristics of SPIONs can be tailored to deliver drugs into tumor via "passive targeting" and they can also be coated with tissue-specific agents to enhance tumor uptake via "active targeting". In our earlier studies, we developed HCC specific targeting agent- "phosphorylated galactosylated chitosan"(PGC) for targeting asialoglycoprotein receptors. Considering their encouraging results, in this study we developed a multifunctional targeting system- "phosphorylated galactosylated chitosan-coated magnetic nanoparticles"(PGCMNPs) for targeting HCC. PGCMNPs were synthesized by co-precipitation method and characterized by DLS, XRD, TEM, VSM, elemental analysis and FT-IR spectroscopy. PGCMNPs were evaluated for in vitro antioxidant properties, uptake in HepG2 cells, biodistribution, in vivo toxicity and were also evaluated for anticancer therapeutic potential against NDEA-induced HCC in mice model in terms of tumor status, electrical properties, antioxidant defense status and apoptosis. The characterization studies confirmed successful formation of PGCMNPs with superparamagnetic properties. The internalization studies demonstrated (99-100)% uptake of PGCMNPs in HepG2 cells. These results were also supported by biodistribution studies in which increased iron content (296%) was noted inside the hepatocytes. Further, PGCMNPs exhibited no in vivo toxicity. The anticancer therapeutic potential was evident from observation that PGCMNPs treatment decreased tumor bearing animals (41.6%) and significantly (p ≤ 0.05) lowered tumor multiplicity. Overall, this study indicated that PGCMNPs with improved properties are efficiently taken-up by hepatoma cells and has therapeutic potential against HCC. Further, this agent can be tagged with 32P and hence can offer multimodal cancer treatment options via radiation ablation as well as magnetic hyperthermia.
Collapse
Affiliation(s)
- Anushree Udupi
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sachin Shetty
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jesil Mathew Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rajesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Jodhpur, 342005, Rajasthan, India
| | - Sanjay Bharati
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
6
|
Xie L, Zhang Y, Niu X, Jiang X, Kang Y, Diao X, Fang J, Yu Y, Yao J. A nomogram for predicting cancer-specific survival in patients with locally advanced unresectable esophageal cancer: development and validation study. Front Immunol 2025; 16:1524439. [PMID: 40028339 PMCID: PMC11868048 DOI: 10.3389/fimmu.2025.1524439] [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: 11/07/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Background Immunotherapy research for esophageal cancer is progressing rapidly, particularly for locally advanced unresectable cases. Despite these advances, the prognosis remains poor, and traditional staging systems like AJCC inadequately predict outcomes. This study aims to develop and validate a nomogram to predict cancer-specific survival (CSS) in these patients. Methods Clinicopathological and survival data for patients diagnosed between 2010 and 2021 were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. Patients were divided into a training cohort (70%) and a validation cohort (30%). Prognostic factors were identified using the Least Absolute Shrinkage and Selection Operator (LASSO) regression. A nomogram was constructed based on the training cohort and evaluated using the concordance index (C-index), net reclassification improvement (NRI), integrated discrimination improvement (IDI), calibration plots, and area under the receiver operating characteristic curve (AUC). Kaplan-Meier survival curves were used to validate the prognostic factors. Results The study included 4,258 patients, and LASSO-Cox regression identified 10 prognostic factors: age, marital status, tumor location, tumor size, pathological grade, T stage, American Joint Committee on Cancer (AJCC) stage, SEER stage, chemotherapy, and radiotherapy. The nomogram achieved a C-index of 0.660 (training set) and 0.653 (validation set), and 1-, 3-, and 5-year AUC values exceeded 0.65. Calibration curves showed a good fit, and decision curve analysis (DCA), IDI, and NRI indicated that the nomogram outperformed traditional AJCC staging in predicting prognosis. Conclusions We developed and validated an effective nomogram model for predicting CSS in patients with locally advanced unresectable esophageal cancer. This model demonstrated significantly superior predictive performance compared to the traditional AJCC staging system. Future research should focus on integrating emerging biomarkers, such as PD-L1 expression and tumor mutational burden (TMB), into prognostic models to enhance their predictive accuracy and adapt to the evolving landscape of immunotherapy in esophageal cancer management.
Collapse
Affiliation(s)
- Liangyun Xie
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yafei Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiedong Niu
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiaomei Jiang
- Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Yuan Kang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xinyue Diao
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jinhai Fang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yilin Yu
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jun Yao
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
7
|
Yao L, Wu P, Yao F, Huang B, Zhong F, Wang X. ZCCHC4 regulates esophageal cancer progression and cisplatin resistance through ROS/c-myc axis. Sci Rep 2025; 15:5149. [PMID: 39934309 PMCID: PMC11814405 DOI: 10.1038/s41598-025-89628-3] [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: 09/21/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Zinc finger CCHC-type containing 4 (ZCCHC4) is a newly discovered N6-methyladenosine (m6A) RNA methyltransferase (MTase), which possesses an m6A MTase domain and an RNA-binding protein (RBP) Znf domain. Aberrantly expressed ZCCHC4 has been found to be correlated with poor prognosis and chemoresistance in various tumors, such as hepatocellular carcinoma, lung cancer and colorectal cancer. However, the expression and functional analysis of the role of ZCCHC4 in esophageal cancer (ESCA) is still elusive. The expression of ZCCHC4 in esophageal cancer tissues was evaluated by qPT-PCR and western blot. Serum esophageal tumor markers are detected by electrochemiluminescence immunoassay. Relationship between ZCCHC4 expression and pathway enrichment analysis were analyzed by R. The reactive oxygen species (ROS), cell proliferation, cell cycle and apoptosis of ZCCHC4 in esophageal squamous cell carcinoma (ESCC) cells tested by CCK8 assay and flow cytometry assay. Aberrant expression of ZCCHC4 is associated with cancer stages, lymph node metastasis (LNM), and tumor histology, and poorer Overall Survival (OS) in esophageal cancer. The mRNA level of ZCCHC4 in esophageal cancer patients correlates with serum carcinoembryonic antigen (CEA) levels, Squamous Cell Carcinoma (SCC) markers, and tissue polypeptide antigen (TPA) levels. Knockdown of ZCCHC4 induces DNA damage, leading to an elevation of reactive oxygen species (ROS), which in turn triggers S-phase arrest, enhances apoptosis, augments sensitivity to cisplatin treatment, and inhibits proliferation in esophageal cancer cells. Conversely, overexpression of ZCCHC4 promotes proliferation, inhibits apoptosis, and increases resistance to cisplatin in esophageal cancer cells. Furthermore, scavenging ROS reverses the effects of ZCCHC4 downregulation on both proliferation and apoptosis in esophageal cancer cells. Additionally, downregulation of ZCCHC4 inhibits the progression of esophageal cancer and reduces cisplatin resistance in vivo. In summary, downregulation of ZCCHC4 leads to increased sensitivity of ESCC cells to cisplatin, inhibits proliferation, and promotes apoptosis in esophageal cancer cells, potentially via the ROS/c-myc axis. The study suggests a potential adjunctive role for ZCCHC4 in the diagnosis and treatment of esophageal cancer and aids in further understanding the underlying mechanisms in ESCA progression.
Collapse
Affiliation(s)
- Lihua Yao
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China
| | - Piao Wu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Fangyi Yao
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China
| | - Fangmin Zhong
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China.
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
8
|
Ardalan Moghadam Al F, Forghanifard MM, Zarrinpour V. PYGO2 promotes resistance to chemotherapy via reducing apoptosis and G2/M cell cycle arrest in esophageal carcinoma cells. Med Oncol 2025; 42:45. [PMID: 39808374 DOI: 10.1007/s12032-024-02590-4] [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: 10/17/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
5-FU is a widely used chemotherapy drug for esophageal carcinomas, but therapy failure has been observed in 5-FU-resistant patients. Overcoming this resistance is a significant challenge in cancer treatment, requiring identifying and targeting important resistance mechanisms. PYGO2 expression is crucial in developing resistance to various chemotherapy drugs. In this study, we aimed to investigate the impact of PYGO2 overexpression on the sensitivity of YM-1 and KYSE-30 esophageal carcinoma cells against 5-FU. To do this, we compared cell viability, cell cycle arrest, apoptosis rate, and mRNA expressions of various apoptosis-related genes between pcDNA3-PYGO2 transfected and untransfected KYSE-30 and YM-1 esophageal carcinoma cells following treatment with 5-FU. We showed that PYGO2 expression reduces 5-FU sensitivity in YM-1 and KYSE-30 cells. PYGO2-overexpressing cells treated with 5-FU have exhibited a noteworthy reduction in both early and late apoptotic cells compared to controls. Furthermore, a significant decrease in the Bax/Bcl2 ratio and P53 gene expression was observed. 5-FU induces G2/M cell cycle arrest in YM-1 and KYSE-30 cells. However, PYGO2 overexpression impeded G2/M cell cycle arrest in 5-FU-treated cells, thereby suppressing the toxicity of 5-FU. PYGO2 may mediate its apoptotic effect by regulating cell cycle regulatory proteins, specifically cyclin D1 and p21. These results highlight PYGO2's capacity to alter how esophageal cancer cells respond to 5-FU therapy, emphasizing its importance as a potential focal point for treatment strategies.
Collapse
Affiliation(s)
| | | | - Vajiheh Zarrinpour
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
9
|
Sheng Y, Zhang L, Hu Z, Peng B. Prediction of Early Mortality in Esophageal Cancer Patients with Liver Metastasis Using Machine Learning Approaches. Life (Basel) 2024; 14:1437. [PMID: 39598235 PMCID: PMC11595315 DOI: 10.3390/life14111437] [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: 10/04/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Patients with esophageal cancer liver metastasis face a high risk of early mortality, making accurate prediction crucial for guiding clinical decisions. However, effective predictive tools are currently limited. In this study, we used clinicopathological data from 1897 patients diagnosed with esophageal cancer liver metastasis between 2010 and 2020, which were sourced from the SEER database. Prognostic factors were identified using univariate and multivariate logistic regression, and seven machine learning models, including extreme gradient boosting (XGBoost) and support vector machine (SVM), were developed to predict early mortality. The models were evaluated using Receiver Operating Characteristic (ROC) curves, calibration curves, decision curve analysis (DCA), and F1 scores. Results showed that 40% of patients experienced all-cause early mortality and 38% had cancer-specific early mortality. Key predictors of early mortality included age, location, chemotherapy, and lung metastasis. Among the models, XGBoost performed best in predicting all-cause early mortality, while SVM excelled in predicting cancer-specific early mortality. These findings demonstrate that machine learning models, particularly XGBoost and SVM, can serve as valuable tools for predicting early mortality in patients with esophageal cancer liver metastasis, aiding clinical decision making.
Collapse
Affiliation(s)
| | | | | | - Bin Peng
- School of Public Health, Chongqing Medical University, Chongqing 400016, China; (Y.S.); (L.Z.); (Z.H.)
| |
Collapse
|
10
|
Li B, Zhang J, Yu Y, Li Y, Chen Y, Zhao X, Li A, Zhao L, Li M, Wang Z, Lu X, Wu W, Zhang Y, Dong Z, Liu K, Jiang Y. Dronedarone inhibits the proliferation of esophageal squamous cell carcinoma through the CDK4/CDK6-RB1 axis in vitro and in vivo. Front Med 2024; 18:896-910. [PMID: 39266905 DOI: 10.1007/s11684-024-1062-x] [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: 07/04/2023] [Accepted: 01/17/2024] [Indexed: 09/14/2024]
Abstract
Treatment options for patients with esophageal squamous cell carcinoma (ESCC) often result in poor prognosis and declining health-related quality of life. Screening FDA-approved drugs for cancer chemoprevention is a promising and cost-efficient strategy. Here, we found that dronedarone, an antiarrhythmic drug, could inhibit the proliferation of ESCC cells. Moreover, we conducted phosphorylomics analysis to investigate the mechanism of dronedarone-treated ESCC cells. Through computational docking models and pull-down assays, we demonstrated that dronedarone could directly bind to CDK4 and CDK6 kinases. We also proved that dronedarone effectively inhibited ESCC proliferation by targeting CDK4/CDK6 and blocking the G0/G1 phase through RB1 phosphorylation inhibition by in vitro kinase assays and cell cycle assays. Subsequently, we found that knocking out CDK4 and CDK6 decreased the susceptibility of ESCC cells to dronedarone. Furthermore, dronedarone suppressed the growth of ESCC in patient-derived tumor xenograft models in vivo. Thus, our study demonstrated that dronedarone could be repurposed as a CDK4/6 inhibitor for ESCC chemoprevention.
Collapse
Affiliation(s)
- Bo Li
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450003, China
| | - Jing Zhang
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450003, China
| | - Yin Yu
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450003, China
| | - Yinhua Li
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yingying Chen
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaokun Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Ang Li
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Lili Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Mingzhu Li
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Zitong Wang
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Xuebo Lu
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenjie Wu
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yueteng Zhang
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Zigang Dong
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450003, China
- Basic Medicine Sciences Research Center, Zhengzhou University, Zhengzhou, 450052, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450001, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450001, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450001, China
| | - Kangdong Liu
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450003, China.
- Basic Medicine Sciences Research Center, Zhengzhou University, Zhengzhou, 450052, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450001, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450001, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450001, China.
| | - Yanan Jiang
- The Pathophysiology Department, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450003, China.
- Basic Medicine Sciences Research Center, Zhengzhou University, Zhengzhou, 450052, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450001, China.
| |
Collapse
|
11
|
Li Y, Ma L, Li P. Circ_FNDC3B Promotes Cell Proliferation and Metastasis in Esophageal Squamous Cell Carcinoma via Regulating MAPK1 by Binding to miR-136-5p. Biochem Genet 2024; 62:3803-3820. [PMID: 38228844 DOI: 10.1007/s10528-023-10585-5] [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: 02/08/2023] [Accepted: 11/01/2023] [Indexed: 01/18/2024]
Abstract
A handful of circular RNAs (circRNAs) associated with cancer progression have been indicated in esophageal squamous cell carcinoma (ESCC). The current study aimed to investigate the functional mechanism of circular RNA Fibronectin type III domain containing 3B (circ_FNDC3B) in ESCC. Circ_FNDC3B, FNDC3B, microRNA-136-5p (miR-136-5p) and mitogen-activated protein kinase 1 (MAPK1) were examined via the quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was evaluated by Cell Counting Kit-8 (CCK-8) and colony formation assays. Transwell assay was performed to measure cell migration and invasion. Protein analysis was implemented by western blot. Cell apoptosis was assessed via flow cytometry. Target interaction was affirmed using dual-luciferase reporter assay. The function analysis of circ_FNDC3B in vivo was explored by xenograft models. The upregulation of circ_FNDC3B was detected in ESCC tissues and cells. Functionally, ESCC cell proliferation and metastasis were repressed but apoptosis was promoted by circ_FNDC3B knockdown. Besides, circ_FNDC3B silence inhibited ESCC progression through MAPK1 downregulation. Further target analysis identified miR-136-5p as a target of circ_FNDC3B and an upstream control of MAPK1. Additionally, the regulation of si-circ_FNDC3B in ESCC was also dependent on targeting miR-136-5p. Moreover, circ_FNDC3B targeted miR-136-5p to affect MAPK1 level. Tumorigenesis in vivo was also suppressed by downregulating circ_FNDC3B to regulate miR-136-5p/MAPK1 axis. Circ_FNDC3B downregulation impeded the development of ESCC via the mediation of miR-136-5p/MAPK1 axis. This report afforded a novel insight into the functional mechanism of circ_FNDC3B in ESCC.
Collapse
Affiliation(s)
- Yuwei Li
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, People's Republic of China
| | - Lieting Ma
- Department of Laboratory, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Peng Li
- Department of Laboratory, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
12
|
Qi LW, Xie YF, Wang WN, Liu J, Yang KG, Chen K, Luo CH, Fei J, Hu JM. High microvessel and lymphatic vessel density predict poor prognosis in patients with esophageal squamous cell carcinoma. PeerJ 2024; 12:e18080. [PMID: 39351370 PMCID: PMC11441385 DOI: 10.7717/peerj.18080] [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: 05/03/2024] [Accepted: 08/20/2024] [Indexed: 10/04/2024] Open
Abstract
Background Microangiogenesis and lymphangiogenesis are essential for tumor growth in the tumor microenvironment, contributing to tumor invasion and metastasis. Limited literature exists on these processes in esophageal squamous cell carcinoma (ESCC). Therefore, the purpose of this study is to explore the impacts of microangiogenesis and lymphangiogenesis on the occurrence, progression, and prognosis assessment of ESCC. Methods Surgical specimens and paraffin-embedded human tissues were procured from ESCC patients, encompassing 100 ESCC tissues and 100 cancer-adjacent normal (CAN) tissues. CD34 and D2-40 were utilized as markers for microvessel endothelial cells and lymphatic vessel endothelial cells, respectively. Microvascular density (MVD) and lymphatic vessel density (LVD) were evaluated through immunohistochemical quantification. Results We found that tumor tissues in ESCC patients had significantly higher MVD and LVD than cancer-adjacent normal (CAN) tissues. High MVD and LVD were associated with lymph node metastasis and advanced tumor clinical stages. Additionally, both high MVD and high LVD were strongly linked to poorer prognosis among cancer patients. Furthermore, a positive correlation was found between high MVD and high LVD (p < 0.05). The presence of these markers individually indicated a worse prognosis, with their combined assessment showcasing enhanced prognostic value. Conclusions Overall, the increased MVD and LVD indicates higher invasion and metastasis of ESCC, closely correlating with unfavorablefor poor prognosis of ESCC patients.
Collapse
Affiliation(s)
- Li Wen Qi
- Pathology, Shihezi University School of Medicine/The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
- Department of Oncology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Yu Fang Xie
- Pathology, Shihezi University School of Medicine/The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Wei Nan Wang
- Pathology, Shihezi University School of Medicine/The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Jia Liu
- Pathology, Shihezi University School of Medicine/The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Kai Ge Yang
- Pathology, Shihezi University School of Medicine/The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Kai Chen
- Pathology, Shihezi University School of Medicine/The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Cheng Hua Luo
- Pathology, Shihezi University School of Medicine/The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Jing Fei
- Department of Oncology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Jian Ming Hu
- Pathology, Shihezi University School of Medicine/The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
13
|
Hassan MS, Johnson C, Ponna S, Scofield D, Awasthi N, von Holzen U. Inhibition of Insulin-like Growth Factor 1 Receptor/Insulin Receptor Signaling by Small-Molecule Inhibitor BMS-754807 Leads to Improved Survival in Experimental Esophageal Adenocarcinoma. Cancers (Basel) 2024; 16:3175. [PMID: 39335147 PMCID: PMC11430189 DOI: 10.3390/cancers16183175] [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: 08/12/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The insulin-like growth factor-1 (IGF-1) and insulin axes are upregulated in obesity and obesity-associated esophageal adenocarcinoma (EAC). Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is a contemporary nanotechnology-based paclitaxel (PT) bound to human albumin, ensuring its solubility in water rather than a toxic solvent. Here, we examined the benefits of inhibiting insulin-like growth factor-1 receptor/insulin receptor (IGF-1/IR) signaling and the enhancement of nab-paclitaxel effects by inclusion of the small-molecule inhibitor BMS-754807 using both in vitro and in vivo models of EAC. Using multiple EAC cell lines, BMS-754807 and nab-paclitaxel were evaluated as mono and combination therapies for in vitro effects on cell proliferation, cell death, and cell movement. We then analyzed the in vivo anticancer potency with survival improvement with BMS-754807 and nab-paclitaxel mono and combination therapies. BMS-754807 monotherapy suppressed in vitro cell proliferation and wound healing while increasing apoptosis. BMS-754807, when combined with nab-paclitaxel, enhanced those effects on the inhibition of cell proliferation, increment in cell apoptosis, and inhibition of wound healing. BMS-754807 with nab-paclitaxel produced substantially greater antitumor effects by increasing in vivo apoptosis, leading to increased mice survival compared to those of BMS-754807 or nab-paclitaxel monotherapy. Our outcomes support the use of BMS-754807, alone and in combination with nab-paclitaxel, as an efficient and innovative treatment choice for EAC.
Collapse
Affiliation(s)
- Md Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Chloe Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Saisantosh Ponna
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Dimitri Scofield
- Department of Biology, Indiana University, South Bend, IN 47405, USA
| | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
- Goshen Center for Cancer Care, Goshen, IN 46526, USA
- School of Medicine, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
14
|
Su Y, Huang C, Yang C, Lin Q, Chen Z. Prediction of Survival in Patients With Esophageal Cancer After Immunotherapy Based on Small-Size Follow-Up Data. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2024; 5:769-782. [PMID: 39464488 PMCID: PMC11505867 DOI: 10.1109/ojemb.2024.3452983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/05/2024] [Accepted: 08/26/2024] [Indexed: 10/29/2024] Open
Abstract
Esophageal cancer (EC) poses a significant health concern, particularly among the elderly, warranting effective treatment strategies. While immunotherapy holds promise in activating the immune response against tumors, its specific impact and associated reactions in EC patients remain uncertain. Precise prognosis prediction becomes crucial for guiding appropriate interventions. This study, based on data from the First Affiliated Hospital of Xiamen University (January 2017 to May 2021), focuses on 113 EC patients undergoing immunotherapy. The primary objectives are to elucidate the effectiveness of immunotherapy in EC treatment and to introduce a stacking ensemble learning method for predicting the survival of EC patients who have undergone immunotherapy, in the context of small sample sizes, addressing the imperative of supporting clinical decision-making for healthcare professionals. Our method incorporates five sub-learners and one meta-learner. Leveraging optimal features from the training dataset, this approach achieved compelling accuracy (89.13%) and AUC (88.83%) in predicting three-year survival status, surpassing conventional techniques. The model proves efficient in guiding clinical decisions, especially in scenarios with small-size follow-up data.
Collapse
Affiliation(s)
- Yuhan Su
- School of Electronic Science and EngineeringXiamen UniversityXiamen361005China
- Shenzhen Research Institute of Xiamen UniversityShenzhen518057China
| | - Chaofeng Huang
- Institute of Artificial IntelligenceXiamen UniversityXiamen361005China
| | - Chen Yang
- First Affiliated Hospital of Xiamen UniversityXiamen361000China
| | - Qin Lin
- First Affiliated Hospital of Xiamen UniversityXiamen361000China
| | - Zhong Chen
- School of Electronic Science and EngineeringXiamen UniversityXiamen361005China
- Institute of Artificial IntelligenceXiamen UniversityXiamen361005China
| |
Collapse
|
15
|
Shao L, Li B. Synaptotagmin 13 Could Drive the Progression of Esophageal Squamous Cell Carcinoma Through Upregulating ACRV1. DNA Cell Biol 2024; 43:452-462. [PMID: 39046915 DOI: 10.1089/dna.2024.0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
SYT13 is one of the atypical members of the synaptotagmin (SYT) family whose function has attracted considerable attention in recent years. Although SYT13 has been studied in several types of human cancers, such as lung cancer, its role in esophageal squamous cell carcinoma (ESCC) is still unclear. It was demonstrated that SYT13 is significantly upregulated in ESCC tissues compared with normal ones and correlated with higher degree of malignancy. Knockdown of SYT13 could inhibit ESCC cell proliferation and migration, while promoting cell apoptosis. Meanwhile, ESCC cells with relatively lower SYT13 expression grew slower in vivo and finally formed smaller xenografts. Furthermore, acrosomal vesicular protein 1 was identified as a potential downstream target of SYT13, which regulates cell phenotypes of ESCC cells in cooperation with SYT13. All the in vitro and in vivo results in this study identified that SYT13 silencing could be an effective strategy to inhibit the development of ESCC, which could be considered as a promising therapeutic target in the treatment of ESCC.
Collapse
Affiliation(s)
- Longlong Shao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bin Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Chauhan A, Pathak VM, Yadav M, Chauhan R, Babu N, Chowdhary M, Ranjan A, Mathkor DM, Haque S, Tuli HS, Ramniwas S, Yadav V. Role of ursolic acid in preventing gastrointestinal cancer: recent trends and future perspectives. Front Pharmacol 2024; 15:1405497. [PMID: 39114347 PMCID: PMC11303223 DOI: 10.3389/fphar.2024.1405497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024] Open
Abstract
Gastrointestinal malignancies are one of the major worldwide health concerns. In the present review, we have assessed the plausible therapeutic implication of Ursolic Acid (UA) against gastrointestinal cancer. By modulating several signaling pathways critical in cancer development, UA could offer anti-inflammatory, anti-proliferative, and anti-metastatic properties. However, being of low oral bioavailability and poor permeability, its clinical value is restricted. To deliver and protect the drug, liposomes and polymer micelles are two UA nanoformulations that can effectively increase medicine stability. The use of UA for treating cancers is safe and appropriate with low toxicity characteristics and a predictable pharmacokinetic profile. Although the bioavailability of UA is limited, its nanoformulations could emerge as an alternative to enhance its efficacy in treating GI cancers. Further optimization and validation in the clinical trials are necessary. The combination of molecular profiling with nanoparticle-based drug delivery technologies holds the potential for bringing UA to maximum efficacy, looking for good prospects with GI cancer treatment.
Collapse
Affiliation(s)
- Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, Uttar Pradesh, India
| | | | - Monika Yadav
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Neelesh Babu
- Department of Microbiology, Baba Farid Institute of Technology, Dehradun, Uttarakhand, India
| | - Manish Chowdhary
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Ambala, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Mohali, India
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
17
|
Khan IR, Sadida HQ, Hashem S, Singh M, Macha MA, Al-Shabeeb Akil AS, Khurshid I, Bhat AA. Therapeutic implications of signaling pathways and tumor microenvironment interactions in esophageal cancer. Biomed Pharmacother 2024; 176:116873. [PMID: 38843587 DOI: 10.1016/j.biopha.2024.116873] [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: 03/24/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Esophageal cancer (EC) is significantly influenced by the tumor microenvironment (TME) and altered signaling pathways. Downregulating these pathways in EC is essential for suppressing tumor development, preventing metastasis, and enhancing therapeutic outcomes. This approach can increase tumor sensitivity to treatments, enhance patient outcomes, and inhibit cancer cell proliferation and spread. The TME, comprising cellular and non-cellular elements surrounding the tumor, significantly influences EC's development, course, and treatment responsiveness. Understanding the complex relationships within the TME is crucial for developing successful EC treatments. Immunotherapy is a vital TME treatment for EC. However, the heterogeneity within the TME limits the application of anticancer drugs outside clinical settings. Therefore, identifying reliable microenvironmental biomarkers that can detect therapeutic responses before initiating therapy is crucial. Combining approaches focusing on EC signaling pathways with TME can enhance treatment outcomes. This integrated strategy aims to interfere with essential signaling pathways promoting cancer spread while disrupting factors encouraging tumor development. Unraveling aberrant signaling pathways and TME components can lead to more focused and efficient treatment approaches, identifying specific cellular targets for treatments. Targeting the TME and signaling pathways may reduce metastasis risk by interfering with mechanisms facilitating cancer cell invasion and dissemination. In conclusion, this integrative strategy has significant potential for improving patient outcomes and advancing EC research and therapy. This review discusses the altered signaling pathways and TME in EC, focusing on potential future therapeutics.
Collapse
Affiliation(s)
- Inamu Rashid Khan
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine Doha 26999, Qatar
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu and Kashmir 192122, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Ibraq Khurshid
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar.
| |
Collapse
|
18
|
Sabatelle RC, Colson YL, Sachdeva U, Grinstaff MW. Drug Delivery Opportunities in Esophageal Cancer: Current Treatments and Future Prospects. Mol Pharm 2024; 21:3103-3120. [PMID: 38888089 PMCID: PMC11331583 DOI: 10.1021/acs.molpharmaceut.4c00246] [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] [Indexed: 06/20/2024]
Abstract
With one of the highest mortality rates of all malignancies, the 5-year survival rate for esophageal cancer is under 20%. Depending on the stage and extent of the disease, the current standard of care treatment paradigm includes chemotherapy or chemoradiotherapy followed by surgical esophagogastrectomy, with consideration for adjuvant immunotherapy for residual disease. This regimen has high morbidity, due to anatomic changes inherent in surgery, the acuity of surgical complications, and off-target effects of systemic chemotherapy and immunotherapy. We begin with a review of current treatments, then discuss new and emerging targets for therapies and advanced drug delivery systems. Recent and ongoing preclinical and early clinical studies are evaluating traditional tumor targets (e.g., human epidermal growth factor receptor 2), as well as promising new targets such as Yes-associated protein 1 or mammalian target of rapamycin to develop new treatments for this disease. Due the function and location of the esophagus, opportunities also exist to pair these treatments with a drug delivery strategy to increase tumor targeting, bioavailability, and intratumor concentrations, with the two most common delivery platforms being stents and nanoparticles. Finally, early results with antibody drug conjugates and chimeric antigenic receptor T cells show promise as upcoming therapies. This review discusses these innovations in therapeutics and drug delivery in the context of their successes and failures, with the goal of identifying those solutions that demonstrate the most promise to shift the paradigm in treating this deadly disease.
Collapse
Affiliation(s)
- Robert C. Sabatelle
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Uma Sachdeva
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
19
|
Hu J, Li P, Dan Y, Chen Z, Lu Y, Chen X, Yan S. COL8A1 Regulates Esophageal Squamous Carcinoma Proliferation and Invasion Through PI3K/AKT Pathway. Ann Surg Oncol 2024; 31:3502-3512. [PMID: 38429534 DOI: 10.1245/s10434-023-14370-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/15/2023] [Indexed: 03/03/2024]
Abstract
PURPOSE Esophageal squamous carcinoma (ESCC) is a gastrointestinal malignancy with a high mortality, but the tumorigenesis is still unclear, restricting the target therapy development of ESCC. We explored the role of COL8A1 in ESCC development. METHODS Tissue microarrays were used to investigate the expression level of COL8A1 in ESCC tissues. The association between COL8A1 and the overall survival of ESCC patients was assessed. The effect of differential COL8A1 expression on tumor growth was investigated by the xenograft model. The regulation of COL8A1 on tumor growth, migration, and invasion was studied by using ESCC cell lines. The signal transduction pathways involved in COL8A1 were bioinformatically profiled and validated. RESULTS The COL8A1 was significantly expressed in cancerous tissues and was associated with poor prognosis in patients with ESCC. In vivo, the tumor growth obviously declined after inhibition of the COL8A1 expression. The abilities of cell proliferation and invasion were both decreased when the expression of COL8A1 was knockdown in ESCC cell line. Furthermore, we found the inactivation of the PI3K/AKT pathway that was mediated by knockdown of COL8A1 in ESCC cells, which was reversed with COL8A1 overexpression, whereas the cell proliferation and invasion ability were restored. CONCLUSIONS This is the first report that COL8A1 promote ESCC progression, which hopefully will provide a theoretical basis for clinical targeting of ESCC.
Collapse
Affiliation(s)
- Jing Hu
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Pengbo Li
- The Affiliated Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Yanggang Dan
- Department of Cardiothoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Zhe Chen
- Department of Cardiothoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Yeting Lu
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Xue Chen
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Senxiang Yan
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Zhang X, Xu C, Wang C, Pei Y, He M, Wan Z, Hou J, Wang L. CD276 promotes epithelial-mesenchymal transition in esophageal squamous cell carcinoma through the TGF-β/SMAD signaling. Clin Exp Metastasis 2024; 41:81-90. [PMID: 38396262 DOI: 10.1007/s10585-024-10280-8] [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: 11/26/2023] [Accepted: 02/06/2024] [Indexed: 02/25/2024]
Abstract
OBJECTIVE Aberrant expression of CD276 has been reported in malignant tumors. However, the exact role and mechanisms of CD276 influence the progression of esophageal squamous cell carcinoma (ESCC) still need to be understood. METHODS Bioinformatics analysis of data from The Cancer Genome Atlas and Gene Expression Omnibus databases, along with immunohistochemistry staining, was used to explore the expression patterns of CD276 in ESCC. Cell counting kit-8 and Transwell assays were employed to evaluate the effects of CD276 expression on tumor cell proliferation and motility. Western blotting and Transwell assays were used to explore the potential pathways through which CD276 mediates the progression of ESCC. Moreover, the in vivo role of CD276 in tumor progression was investigated by establishing a lung metastasis mouse model. RESULTS A significant upregulation of CD276 was observed in ESCC tissues compared to adjacent tissues. The inhibition of CD276 had no evident impact on ESCC cell proliferation but notably hindered their migratory and invasive properties and the expression of epithelial-mesenchymal transition (EMT) markers. Inversely, overexpressing CD276 led to an upregulation of EMT markers, underscoring the capacity of CD276 to amplify the motility of ESCC cells. Furthermore, CD276 was found to enhance the migratory and invasive abilities of ESCC cells by activating the TGF-β/SMAD signaling but not the PI3K/AKT pathway. In vivo studies demonstrated that CD276 facilitates pulmonary metastasis. CONCLUSION CD276 is significant upregulation in ESCC tissues and facilitates the EMT process in ESCC cells via the TGF-β/SMAD signaling, thus promoting the progression of ESCC.
Collapse
Affiliation(s)
- Xiaoman Zhang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Cuicui Xu
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Cuicui Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Yuhui Pei
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Min He
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Zhicheng Wan
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jun Hou
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| | - Lianghai Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| |
Collapse
|
21
|
Yuan Q, Shi Y, Zhang Y, Shi Y, Hussain Z, Zhao J, Jiang Y, Qiao Y, Guo Y, Lu J, Dong Z, Dong Z, Wang J, Liu K. Domperidone inhibits cell proliferation via targeting MEK and CDK4 in esophageal squamous cell carcinoma. Cancer Cell Int 2024; 24:114. [PMID: 38528618 DOI: 10.1186/s12935-024-03291-8] [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: 11/24/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the leading causes of digestive system tumor related death in the world. Unfortunately, effective chemopreventive agent is lack for patients with ESCC in clinical practice, which leads to the extremely high mortality rate. METHODS A library of prescribed drugs was screened for finding critical anti-tumor properties in ESCC cells. The phosphoproteomics, kinase array, pulldown assay and drug affinity responsive target stabilization assay (DARTS) were applied to explore mechanisms and searched for synergistic targets. Established models of PDX in mice were used to determine the therapeutic effect of domperidone. RESULTS After screening a library of prescribed drugs, we discovered that domperidone has anti-tumor properties. Domperidone, acting as a gastroprokinetic agent, has been widely used in clinic for gastrointestinal motility disorders. Despite limited research, there are indications that domperidone may have anti-tumor properties. In this study, we determined that domperidone significantly inhibited ESCC proliferation in vitro and in vivo. We employed phosphoproteomics to reveal p-ERK, and p-SMAD3 down-regulation upon domperidone treatment. Then, the results of kinase assay and pulldown assay further validated that domperidone directly combined with MEK1/2 and CDK4, leading to the inhibition of their kinase activity. Furthermore, our results revealed that MEK/ERK and CDK4/SMAD3 signal pathway were major pathways in domperidone against ESCC. CONCLUSION Collectively, these findings suggest that domperidone serves as an effective "multi-target" inhibitor of MEK1/2 and CDK4, offering potential benefits for the chemoprevention of ESCC.
Collapse
Affiliation(s)
- Qiang Yuan
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yunshu Shi
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yuhan Zhang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yaqian Shi
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zubair Hussain
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jimin Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yan Qiao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yaping Guo
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jing Lu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Ziming Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zigang Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China.
| | - Junyong Wang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
22
|
Yu XW, She PW, Chen FC, Chen YY, Zhou S, Wang XM, Lin XR, Liu QL, Huang ZJ, Qiu Y. Metabolic subtypes and immune landscapes in esophageal squamous cell carcinoma: prognostic implications and potential for personalized therapies. BMC Cancer 2024; 24:230. [PMID: 38373930 PMCID: PMC10875771 DOI: 10.1186/s12885-024-11890-x] [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: 07/26/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND This study aimed to identify metabolic subtypes in ESCA, explore their relationship with immune landscapes, and establish a metabolic index for accurate prognosis assessment. METHODS Clinical, SNP, and RNA-seq data were collected from 80 ESCA patients from the TCGA database and RNA-seq data from the GSE19417 dataset. Metabolic genes associated with overall survival (OS) and progression-free survival (PFS) were selected, and k-means clustering was performed. Immune-related pathways, immune infiltration, and response to immunotherapy were predicted using bioinformatic algorithms. Weighted gene co-expression network analysis (WGCNA) was conducted to identify metabolic genes associated with co-expression modules. Lastly, cell culture and functional analysis were performed using patient tissue samples and ESCA cell lines to verify the identified genes and their roles. RESULTS Molecular subtypes were identified based on the expression profiles of metabolic genes, and univariate survival analysis revealed 163 metabolic genes associated with ESCA prognosis. Consensus clustering analysis classified ESCA samples into three distinct subtypes, with MC1 showing the poorest prognosis and MC3 having the best prognosis. The subtypes also exhibited significant differences in immune cell infiltration, with MC3 showing the highest scores. Additionally, the MC3 subtype demonstrated the poorest response to immunotherapy, while the MC1 subtype was the most sensitive. WGCNA analysis identified gene modules associated with the metabolic index, with SLC5A1, NT5DC4, and MTHFD2 emerging as prognostic markers. Gene and protein expression analysis validated the upregulation of MTHFD2 in ESCA. MTHFD2 promotes the progression of ESCA and may be a potential therapeutic target for ESCA. CONCLUSION The established metabolic index and identified metabolic genes offer potential for prognostic assessment and personalized therapeutic interventions for ESCA, underscoring the importance of targeting metabolism-immune interactions in ESCA. MTHFD2 promotes the progression of ESCA and may be a potential therapeutic target for ESCA.
Collapse
Affiliation(s)
- Xiao-Wan Yu
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China.
| | - Pei-Wei She
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, P. R. China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, 350001, Fuzhou, Fujian, P. R. China
| | - Fang-Chuan Chen
- Stomatology Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Ya-Yu Chen
- Stomatology Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Shuang Zhou
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Xi-Min Wang
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Xiao-Rong Lin
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Qiao-Ling Liu
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Zhi-Jun Huang
- Esophageal Surgery Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Yu Qiu
- Reproductive Center, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China.
| |
Collapse
|
23
|
Nguyen Vu TH, Kikuchi O, Ohashi S, Saito T, Ida T, Nakai Y, Cao Y, Yamamoto Y, Kondo Y, Mitani Y, Kataoka S, Kondo T, Katada C, Yamada A, Matsubara J, Muto M. Combination therapy with WEE1 inhibition and trifluridine/tipiracil against esophageal squamous cell carcinoma. Cancer Sci 2023; 114:4664-4676. [PMID: 37724648 PMCID: PMC10728021 DOI: 10.1111/cas.15966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/22/2023] [Accepted: 09/02/2023] [Indexed: 09/21/2023] Open
Abstract
Despite advanced therapeutics, esophageal squamous cell carcinoma (ESCC) remains one of the deadliest cancers. Here, we propose a novel therapeutic strategy based on synthetic lethality combining trifluridine/tipiracil and MK1775 (WEE1 inhibitor) as a treatment for ESCC. This study demonstrates that trifluridine induces single-strand DNA damage in ESCC cells, as evidenced by phosphorylated replication protein 32. The DNA damage response includes cyclin-dependent kinase 1 (CDK1) (Tyr15) phosphorylation as CDK1 inhibition and a decrease of the proportion of phospho-histone H3 (p-hH3)-positive cells, indicating cell cycle arrest at the G2 phase before mitosis entry. The WEE1 inhibitor remarkedly suppressed CDK1 phosphorylation (Try15) and reactivated CDK1, and also increased the proportion of p-hH3-positive cells, which indicates an increase of the number of cells into mitosis. Trifluridine combined with a WEE1 inhibitor increased trifluridine-mediated DNA damage, namely DNA double-strand breaks, as shown by increased γ-H2AX expression. Moreover, the combination treatment with trifluridine/tipiracil and a WEE1 inhibitor significantly suppressed tumor growth of ESCC-derived xenograft models. Hence, our novel combination treatment with trifluridine/tipiracil and a WEE1 inhibitor is considered a candidate treatment strategy for ESCC.
Collapse
Affiliation(s)
- Trang H. Nguyen Vu
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
- Endoscopy DepartmentCho Ray HospitalHo Chi Minh CityVietnam
| | - Osamu Kikuchi
- Department of Clinical Bio‐Resource CenterKyoto University HospitalKyotoJapan
- Division of Clinical Pharmacology and Cancer ImmunotherapyKyoto University Center for Cancer Immunotherapy and ImmunobiologyKyotoJapan
| | - Shinya Ohashi
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
- Preemptive Medicine and Lifestyle Disease Research CenterKyoto University HospitalKyotoJapan
| | - Tomoki Saito
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Tomomi Ida
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Yukie Nakai
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Yang Cao
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Yoshihiro Yamamoto
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Yuki Kondo
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Yosuke Mitani
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Shigeki Kataoka
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Tomohiro Kondo
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Chikatoshi Katada
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Atsushi Yamada
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Junichi Matsubara
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of MedicineKyoto UniversityKyotoJapan
- Department of Clinical Bio‐Resource CenterKyoto University HospitalKyotoJapan
| |
Collapse
|
24
|
Yao W, Cui X, Peng H, Zhang Y, Jia X, Wu S, Zhao J. IDO1 facilitates esophageal carcinoma progression by driving the direct binding of NF-κB and CXCL10. Cell Death Discov 2023; 9:403. [PMID: 37903782 PMCID: PMC10616276 DOI: 10.1038/s41420-023-01689-3] [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: 03/15/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 11/01/2023] Open
Abstract
Esophageal carcinoma (EC), one of the most lethal human malignancies, lacks effective targeted therapies. Indoleamine 2,3-dioxygenase 1 (IDO1) plays a key role in a variety of cancers, but its role and mechanism in EC are still unclear. Immunohistochemistry and qRT-PCR were used to analyze the expression of IDO1 in EC, and the prognostic value of IDO1 in EC was evaluated by Kaplan-Meier test. The in vitro and in vivo function loss/acquisition tests were performed to evaluate the biological effects of IDO1 in EC. The mechanism of action of IDO1-regulation EC was explored through Firefly luciferase & Renilla luciferase activity reporter, chromatin immunoprecipitation (ChIP) and immunofluorescence (IF) assays. Clinically, IDO1 expression was abnormally elevated in EC and positively correlated with overall survival. Functionally, IDO1 was contributed to the proliferation and migration of EC cells. Mechanically, IDO1 regulated the expression of chemokine C-X-C ligand 10 (CXCL10) by promoting the entry of NF-κB into the nucleus to combine with the promoter of CXCL10. Consistently, IDO1 facilitated EC progression may dependent on the presence of CXCL10. Moreover, NF-κB alleviated the inhibitory effect of IDO1 knockdown on EC. IDO1 drove the progression of EC by directly binding NF-κB and CXCL10, the finding that may provide an effective theoretical basis for precise therapies for EC.
Collapse
Affiliation(s)
- Wenjian Yao
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Zhengzhou, 450003, Henan province, China
| | - Xiaohai Cui
- Department of Thoracic Surgery, The first affiliated hospital of xi'an jiaotong university, No.277 Yanta West Road, Xi'an, 710061, Shanxi province, China
| | - Haodong Peng
- Department of Thoracic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, No.7 Weiwu Road, Zhengzhou, 45003, Henan province, China
| | - Yongkang Zhang
- Department of Thoracic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, No.7 Weiwu Road, Zhengzhou, 45003, Henan province, China
| | - Xiangbo Jia
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Zhengzhou, 450003, Henan province, China
| | - Sen Wu
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Zhengzhou, 450003, Henan province, China.
| | - Jian Zhao
- Department of Thoracic Surgery, Liaoning Cancer Hospital, No.44-3 Xiaohe Yan Road, Dadong District, Shenyang, 110000, Liaoning Province, China.
| |
Collapse
|
25
|
Acharya R, Mahapatra A, Verma HK, Bhaskar LVKS. Unveiling Therapeutic Targets for Esophageal Cancer: A Comprehensive Review. Curr Oncol 2023; 30:9542-9568. [PMID: 37999111 PMCID: PMC10670555 DOI: 10.3390/curroncol30110691] [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: 09/12/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 11/25/2023] Open
Abstract
Esophageal cancer is a highly aggressive and deadly disease, ranking as the sixth leading cause of cancer-related deaths worldwide. Despite advances in treatment, the prognosis remains poor. A multidisciplinary approach is crucial for achieving complete remission, with treatment options varying based on disease stage. Surgical intervention and endoscopic treatment are used for localized cancer, while systemic treatments like chemoradiotherapy and targeted drug therapy play a crucial role. Molecular markers such as HER2 and EGFR can be targeted with drugs like trastuzumab and cetuximab, and immunotherapy drugs like pembrolizumab and nivolumab show promise by targeting immune checkpoint proteins. Epigenetic modifications offer new avenues for targeted therapy. Treatment selection depends on factors like stage, tumor location, and patient health, with post-operative and rehabilitation care being essential. Early diagnosis, appropriate treatment, and supportive care are key to improving outcomes. Continued research is needed to develop effective targeted drugs with minimal side effects. This review serves as a valuable resource for clinicians and researchers dedicated to enhancing esophageal cancer treatment outcomes.
Collapse
Affiliation(s)
- Rakesh Acharya
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur 495009, India; (R.A.); (A.M.)
| | - Ananya Mahapatra
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur 495009, India; (R.A.); (A.M.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany;
| | - L. V. K. S. Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur 495009, India; (R.A.); (A.M.)
| |
Collapse
|
26
|
Zhang Y, Zhu Q, Cao X, Ni B. RGS16 regulates Hippo-YAP activity to promote esophageal cancer cell proliferation and migration. Biochem Biophys Res Commun 2023; 675:122-129. [PMID: 37473526 DOI: 10.1016/j.bbrc.2023.04.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 07/22/2023]
Abstract
Esophageal Squamous Cell Carcinoma (ESCC) is a common malignant tumor of digestive tract, accounting for 90% of all pathological types of esophageal cancer. Despite the rapid development of multi-disciplinary treatment such as surgery, chemotherapy, radiotherapy and chemoradiotherapy, the prognosis of patients with ESCC is still poor. Regulators of G-protein signaling (RGSs) are involved in the processes of various cancers. The expression levels of its family member RGS16 are abnormally elevated in a variety of tumors, but its role in ESCC is still unclear. We found that RGS16 expression is aberrantly increased in ESCC tissues and correlated with poor prognosis of ESCC patients from The Cancer Genome Atlas (TCGA) database and our collected ESCC tissues. Moreover, knockdown of RGS16 in two ESCC cells could indeed inhibit their proliferation and migration. We further explored the molecular mechanism of RGS16 in ESCC, and the correlation analysis from TCGA database showed that the mRNA levels of RGS16 was positively correlated with that of CTGF and CYR61, two target genes of Hippo-YAP signaling. Consistently, RGS16- knockdown significantly inhibited the expression of CTGF and CYR61 in ESCC cells. We found that the phosphorylation levels of LATS1 and YAP were significantly increased and YAP translocated into the cytoplasm after depletion of RGS16 in ESCC cells. Also, RGS16-knockdown promoted the interaction between LATS1 and upstream kinase MST1. In addition, reintroduction of a constitutive active YAP5A mutant significantly rescued CTGF expression and cell proliferation in RGS16-knockdown cells. Together, our work revealed that RGS16 promoted YAP activity through disrupting the interaction between LATS1 and MST1, thus promoting the proliferation and migration of ESCC cells.
Collapse
Affiliation(s)
- Yanzhou Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Qing Zhu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xiufeng Cao
- Department of Cardiothoracic Surgery, Nanjing Yimin Hospital, Nanjing, 211103, Jiangsu, China
| | - Bin Ni
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
27
|
Zhou J, Wu J, Wu G, Huang J, Zhang Y, Che J, Zhu K, Geng J, Fan Q. TBX18 knockdown sensitizes esophageal squamous cell carcinoma to radiotherapy by blocking the CHN1/RhoA axis. Radiother Oncol 2023; 186:109788. [PMID: 37399907 DOI: 10.1016/j.radonc.2023.109788] [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: 02/21/2023] [Revised: 06/08/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVE Radioresistance is a challenge in the effective treatment of esophageal squamous cell carcinoma (ESCC). Herein, this research ascertained whether TBX18 reduced the radiosensitivity of ESCC. METHODS Bioinformatics analysis was utilized to retrieve differentially expressed genes. Then, the expression of corresponding candidate genes was tested using qRT-PCR in ESCC clinical specimens, and TBX18 was selected for subsequent experiments. The binding between TBX18 and CHN1 was evaluated by dual-luciferase reporter and ChIP assays, and the relationship between CHN1 and RhoA was identified by GST pull-down. Ectopic expression or knockdown experiments and radiation treatment were performed in cells and the nude mouse xenograft model to clarify the impacts of TBX18, CHN1, and RhoA on radiosensitivity in ESCC. RESULTS Bioinformatics analysis and qRT-PCR retrieved upregulated TBX18 in ESCC for the follow-up study. Additionally, TBX18 was positively correlated with CHN1 in ESCC clinical specimens. Mechanistically, TBX18 bound to the CHN1 promoter region to transcriptionally activate CHN1, thus elevating RhoA activity. Moreover, TBX18 knockdown reduced ESCC cell proliferation and migration while augmenting their apoptosis after radiation, which was negated by further overexpressing CHN1 or RhoA. CHN1 or RhoA knockdown diminished ESCC cell proliferation and migration, as well as enhanced cell apoptosis, subsequent to radiation. Likewise, TBX18 overexpression increased ESCC cell autophagy after radiation, which was partially reversed by knockdown of RhoA. The results of in vivo xenograft experiments in nude mice were concurrent with the in vitro results. CONCLUSION TBX18 knockdown lowered CHN1 transcription and thus reduced RhoA activity, which sensitized ESCC cells to radiotherapy.
Collapse
Affiliation(s)
- Jialiang Zhou
- Depatement of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Jia Wu
- Depatement of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Gang Wu
- Depatement of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Jianfeng Huang
- Depatement of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Yunxia Zhang
- Depatement of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Jun Che
- Depatement of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Koujun Zhu
- Depatement of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Jiqun Geng
- Depatement of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Qiang Fan
- Depatement of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
28
|
Luo J, Tian Z, Zhou Y, Xiao Z, Park SY, Sun H, Zhuang T, Wang Y, Li P, Zhao X. CircABCA13 acts as a miR-4429 sponge to facilitate esophageal squamous cell carcinoma development by stabilizing SRXN1. Cancer Sci 2023; 114:2835-2847. [PMID: 37017121 PMCID: PMC10323080 DOI: 10.1111/cas.15807] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/06/2023] Open
Abstract
Circular RNAs (circRNAs) play a pivotal role in the tumorigenesis and progression of various cancers. However, the role and mechanisms of circABCA13 in esophageal squamous cell carcinoma (ESCC) are largely unknown. Here, we reported that circABCA13, a novel circular RNA generated by back-splicing of the intron of the ABCA13 gene, is highly expressed in ESCC tumor tissues and cell lines. Upregulation of circABCA13 correlated with TNM stage and a poor prognosis in ESCC patients. While knockdown of circABCA13 in ESCC cells significantly reduced cell proliferation, migration, invasion, and anchorage-independent growth, overexpression of circABCA13 facilitated tumor growth both in vitro and in vivo. In addition, circABCA13 directly binds to miR-4429 and sequesters miR-4429 from its endogenous target, SRXN1 mRNA, which subsequently upregulates SRXN1 and promotes ESCC progression. Consistently, overexpression of miR-4429 or knockdown of SRXN1 abolished malignant behavior promotion of ESCC results from circABCA13 overexpression in vitro and in vivo. Collectively, our study uncovered the oncogenic role of circABCA13 and its mechanism in ESCC, suggesting that circABCA13 could be a potential therapeutic target and a predictive biomarker for ESCC patients.
Collapse
Affiliation(s)
- Junwen Luo
- Department of Thoracic SurgeryThe Second Hospital of Shandong UniversityJinanChina
| | - Zhongxian Tian
- Department of Thoracic SurgeryThe Second Hospital of Shandong UniversityJinanChina
- Key Laboratory of Chest CancerShandong University, The Second Hospital of Shandong UniversityJinanChina
| | - Yongjia Zhou
- Department of Thoracic SurgeryThe Second Hospital of Shandong UniversityJinanChina
| | - Zhaohua Xiao
- Department of Thoracic SurgeryThe Second Hospital of Shandong UniversityJinanChina
| | - Sun Young Park
- Department of Environmental MedicineNew York University Grossman School of MedicineNew YorkUSA
| | - Hong Sun
- Department of Environmental MedicineNew York University Grossman School of MedicineNew YorkUSA
| | - Ting Zhuang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory MedicineXinxiang Medical UniversityXinxiangChina
| | - Yongjie Wang
- Department of Thoracic SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Peiwei Li
- Institute of Medical SciencesThe Second Hospital of Shandong UniversityJinanChina
| | - Xiaogang Zhao
- Department of Thoracic SurgeryThe Second Hospital of Shandong UniversityJinanChina
- Key Laboratory of Chest CancerShandong University, The Second Hospital of Shandong UniversityJinanChina
| |
Collapse
|
29
|
Shen Z, Zhang P, Zhang W, Luo F, Xu H, Chen S, Kang M. IL-1RA inhibits esophageal carcinogenesis and lymphangiogenesis via downregulating VEGF-C and MMP9. Funct Integr Genomics 2023; 23:164. [PMID: 37198330 PMCID: PMC10191916 DOI: 10.1007/s10142-023-01049-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 05/19/2023]
Abstract
Interleukin-1 receptor antagonist (IL-1RA) has been shown to play an important role in cancer progression. However, its pathogenic effects and molecular mechanism in the malignant progression of esophageal squamous cell carcinoma (ESCC) remain largely unknown. This study was designed to explore the function of IL-1RA in ESCC and determine the relationship between IL-1RA and lymph node metastasis in ESCC patients. The clinical relevance of IL-1RA in relation to the clinicopathological features and prognosis of 100 ESCC patients was analyzed. The function and underlying mechanisms of IL-1RA in the growth, invasion, and lymphatic metastasis in ESCC were explored both in vitro and in vivo. The therapeutic effect of anakinra, an IL-1 receptor antagonist, on ESCC was also evaluated in animal experiments. Downregulation of IL-1RA was observed in ESCC tissues and cells and was found to be strongly correlated with pathological stage (P = 0.034) and lymphatic metastasis (P = 0.038). Functional assays demonstrated that upregulation of IL-1RA reduced cell proliferation, migration, and lymphangiogenesis both in vitro and in vivo. Mechanistic studies revealed that overexpression of IL-1RA activated the epithelial-to-mesenchymal transition (EMT) in the ESCC cells through activation of MMP9 and regulation of the expression and secretion of VEGF-C through the PI3K/NF-κB pathway. Anakinra treatment resulted in significant inhibition of tumor growth, lymphangiogenesis, and metastasis. IL-1RA inhibits lymph node metastasis of ESCC by regulating the EMT through activation of matrix metalloproteinase 9(MMP9) and lymphangiogenesis, driven by VEGF-C and the NF-κB signaling pathway. Anakinra may be an effective drug for the inhibition of ESCC tumor formation and lymph node metastasis.
Collapse
Affiliation(s)
- Zhimin Shen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Weiguang Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Fei Luo
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Hui Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Shuchen Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, 350122, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350122, Fujian, China.
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China.
| |
Collapse
|
30
|
Zhou L, Yao N, Yang L, Liu K, Qiao Y, Huang C, Du R, Yeung YT, Liu W, Cheng D, Dong Z, Li X. DUSP4 promotes esophageal squamous cell carcinoma progression by dephosphorylating HSP90β. Cell Rep 2023; 42:112445. [PMID: 37141098 DOI: 10.1016/j.celrep.2023.112445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 03/03/2023] [Accepted: 04/13/2023] [Indexed: 05/05/2023] Open
Abstract
The molecular and pathogenic mechanisms of esophageal squamous cell carcinoma (ESCC) development are still unclear, which hinders the development of effective treatments. In this study, we report that DUSP4 is highly expressed in human ESCC and is negatively correlated with patient prognosis. Knockdown of DUSP4 suppresses cell proliferation and patient-derived xenograft (PDX)-derived organoid (PDXO) growth and inhibits cell-derived xenograft (CDX) development. Mechanistically, DUSP4 directly binds to heat shock protein isoform β (HSP90β) and promotes the ATPase activity of HSP90β by dephosphorylating HSP90β on T214 and Y216. These dephosphorylation sites are critical for the stability of JAK1/2-STAT3 signaling and p-STAT3 (Y705) nucleus translocation. In vivo, Dusp4 knockout in mice significantly inhibits 4-nitrochinoline-oxide-induced esophageal tumorigenesis. Moreover, DUSP4 lentivirus or treatment with HSP90β inhibitor (NVP-BEP800) significantly impedes PDX tumor growth and inactivates the JAK1/2-STAT3 signaling pathway. These data provide insight into the role of the DUSP4-HSP90β-JAK1/2-STAT3 axis in ESCC progression and describe a strategy for ESCC treatment.
Collapse
Affiliation(s)
- Liting Zhou
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Ning Yao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Lu Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450000, China
| | - Yan Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450000, China
| | - Chuntian Huang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Ruijuan Du
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang 473004, China
| | - Yiu To Yeung
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Wenting Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Dan Cheng
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450000, China.
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
31
|
Symptom Experiences before Medical Help-Seeking and Psychosocial Responses of Patients with Esophageal Cancer: A Qualitative Study. Eur J Cancer Care (Engl) 2023. [DOI: 10.1155/2023/6506917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Objective. The purpose of this study was to investigate patients with esophageal cancer symptom experiences before seeking medical help and psychosocial responses. Methods. Within one month of diagnosis, semistructured interviews were held with 14 adult patients with esophageal cancer. To fully comprehend the symptom experiences that patients with esophageal cancer had prior to seeking medical help and the psychosocial responses they had to these experiences, thematic analysis and interpretative phenomenological analysis were used. Results. Patients with esophageal cancer recounted their symptom experiences, and psychosocial responses were categorized into three main themes: physical, psychological, and social. Three subthemes were included in the physiological aspect: early symptoms (warning signs), specific symptoms (the catalyst for seeking medical help), and concurrent symptoms (masked side effects). Four subthemes were associated with psychological aspects: unfairness, regret, worry, and acceptance. Two subthemes were related to the social element, namely, dragging down one’s family and societal self-isolation. Conclusion. Due to the self-remitting nature of early symptoms, the normalization of their interpretation, and concealment by other illnesses, patients with esophageal cancer may delay medical help-seeking. The findings could serve as a reference for healthcare professionals to implement tailored psychosocial interventions and help patients identify symptoms early in order to seek medical help.
Collapse
|
32
|
He X, Zhou Y, Chen W, Zhao X, Duan L, Zhou H, Li M, Yu Y, Zhao J, Guo Y, Gu H, Jiang Y, Dong Z, Liu K. Repurposed pizotifen malate targeting NRF2 exhibits anti-tumor activity through inducing ferroptosis in esophageal squamous cell carcinoma. Oncogene 2023; 42:1209-1223. [PMID: 36841865 DOI: 10.1038/s41388-023-02636-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 02/27/2023]
Abstract
Targeted therapy attempts are needed to enhance esophageal squamous cell carcinoma (ESCC) patients' overall survival and satisfaction of life. Nuclear factor erythroid 2-related factor 2 (NRF2), as a high-confidence cancer driver gene, controls the antioxidant response, metabolic balance and redox homeostasis in cancer and is regarded as a potent molecular target for cancer treatment. Here, we attempted to find a new NRF2 inhibitor and study the underlying molecular mechanism in ESCC. We found that up-regulated NRF2 protein was negatively correlated with patient prognosis and promoted tumor proliferation in ESCC. Moreover, Pizotifen malate (PZM), a FDA-approved medication, bound to the Neh1 domain of NRF2 and prevented NRF2 protein binding to the ARE motif of target genes, suppressing transcription activity of NRF2. PZM treatment suppressed tumor development in ESCC PDX model by inducing ferroptosis via down-regulating the transcription of GPX4, GCLC, ME1 and G6PD. Our study illustrates that the over expression of NRF2 indicates poor prognosis and promotes tumor proliferation in ESCC. PZM, as a novel NRF2 inhibitor, inhibits the tumor growth by inducing ferroptosis and elucidates a potent NRF2-based therapy strategy for patients with ESCC.
Collapse
Affiliation(s)
- Xinyu He
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yubing Zhou
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Wenjing Chen
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Xiaokun Zhao
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Lina Duan
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Hao Zhou
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mingzhu Li
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yin Yu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Jimin Zhao
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China.,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China
| | - Yaping Guo
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Huihui Gu
- Basic Medicine Sciences Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Zigang Dong
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China. .,Basic Medicine Sciences Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
33
|
Cheng J, Wu K, Yang Q, Zhu Z, Zhao H. RNF6 activates TGF-β1/c-Myb pathway to promote EMT in esophageal squamous cell carcinoma. Front Oncol 2023; 13:1081333. [PMID: 36845743 PMCID: PMC9948393 DOI: 10.3389/fonc.2023.1081333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Objective This study aimed to investigate RING-Finger Protein 6 (RNF6) expression in esophageal squamous cell carcinoma (ESCC) cells and whether it affects cell proliferation, invasion, and migration by regulating the TGF-β1/c-Myb pathway. Methods TCGA database was used to analyze RNF6 expression in normal tissues and esophageal cancer tissues. Kaplan-Meier method was used to examine the correlation between RNF6 expression and patient prognosis. SiRNA interference vector and RNF6 overexpression plasmid were constructed, and RNF6 was transfected into Eca-109 and KYSE-150 esophageal cancer cell line. In vitro scratch assay and Transwell assay were conducted to investigate the effects of RNF6 on the migration and invasion of Eca-109 and KYSE-150 cells. RT-PCR detected the expression of Snail, E-cadherin, and N-cadherin, and TUNEL detected the apoptosis of cells. Results RNF6 up-regulation promoted the progression of esophageal cancer and predicted poor prognosis. RNF6 also enhanced the migration and invasion of ESCC cells in vitro. RNF6 silencing inhibited the migration and invasion of ESCC cells. TGF-β inhibitors reversed the oncogenic effects of RNF6. RNF6 regulated the migration and invasion of ESCC cells by activating the TGF-β pathway. RNF6/TGF-β1 promoted esophageal cancer progression through c-Myb. Conclusion RNF6 promotes the proliferation, invasion, and migration of ESCC cells possibly by activating the TGF-β1/c-Myb pathway and affects the progression of ESCC.
Collapse
Affiliation(s)
- Jingge Cheng
- The Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Kun Wu
- The Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qian Yang
- The Department of Thoracic Surgery, Han Dan Central Hospital, Handan, China
| | - Ziming Zhu
- The Department of Thoracic Surgery, The First Hospital of Xingtai, Xingtai, China
| | - Hongye Zhao
- The Department of Dermatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,*Correspondence: Hongye Zhao,
| |
Collapse
|
34
|
lncRNA SSTR5-AS1 Predicts Poor Prognosis and Contributes to the Progression of Esophageal Cancer. DISEASE MARKERS 2023; 2023:5025868. [PMID: 36726845 PMCID: PMC9886483 DOI: 10.1155/2023/5025868] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/19/2022] [Accepted: 11/24/2022] [Indexed: 01/24/2023]
Abstract
Esophageal cancer (ESCA), as a common cancer worldwide, is a main cause of cancer-related mortality. Long noncoding RNAs (lncRNAs) have been shown in an increasing number of studies to be capable of playing an important regulatory function in human malignancies. Our study is aimed at delving into the prognostic value and potential function of lncRNA SSTR5-AS1 (SSTR5-AS1) in ESCA. The gene expression data of 182 ESCA samples from TCGA and 653 nontumor specimens from GTEx. The expressions of SSTR5-AS1 were analyzed. We investigated whether there was a correlation between the expression of SSTR5-AS1 and the clinical aspects of ESCA. In order to compare survival curves, the Kaplan-Meier method together with the log-rank test was utilized. The univariate and multivariate Cox regression models were used to analyze the data in order to determine the SSTR5-AS1 expression's significance as a prognostic factor in ESCA patients. In order to investigate the level of SSTR5-AS1 expression in ESCA cells, RT-PCR was utilized. CCK-8 trials served as a model for the loss-of-function tests. In this study, we found that the expressions of SSTR5-AS1 were increased in ESCA specimens compared with nontumor specimens. According to the ROC assays, high SSTR5-AS1 expression had an AUC value of 0.7812 (95% CI: 0.7406 to 0.8217) for ESCA. Patients who had a high level of SSTR5-AS1 expression had a lower overall survival rate than those who had a low level of SSTR5-AS1 expression. In addition, multivariate analysis suggested that SSTR5-AS1 was an independent predictor of overall survival for ESCA patients. Moreover, RT-PCR experiments indicated that SSTR5-AS1 expression was distinctly increased in three ESCA cells compared with HET1A cells. CCK-8 experiments indicated that silence of SSTR5-AS1 distinctly inhibited the proliferation of ESCA cells. Overall, ESCA patients with elevated SSTR5-AS1 had a worse chance of survival, suggesting it could be used as a prognostic and diagnostic biomarker for ESCA.
Collapse
|
35
|
Shang QX, Kong WL, Huang WH, Xiao X, Hu WP, Yang YS, Zhang H, Yang L, Yuan Y, Chen LQ. Identification of m6a-related signature genes in esophageal squamous cell carcinoma by machine learning method. Front Genet 2023; 14:1079795. [PMID: 36733344 PMCID: PMC9886874 DOI: 10.3389/fgene.2023.1079795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Background: We aimed to construct and validate the esophageal squamous cell carcinoma (ESCC)-related m6A regulators by means of machine leaning. Methods: We used ESCC RNA-seq data of 66 pairs of ESCC from West China Hospital of Sichuan University and the transcriptome data extracted from The Cancer Genome Atlas (TCGA)-ESCA database to find out the ESCC-related m6A regulators, during which, two machine learning approaches: RF (Random Forest) and SVM (Support Vector Machine) were employed to construct the model of ESCC-related m6A regulators. Calibration curves, clinical decision curves, and clinical impact curves (CIC) were used to evaluate the predictive ability and best-effort ability of the model. Finally, western blot and immunohistochemistry staining were used to assess the expression of prognostic ESCC-related m6A regulators. Results: 2 m6A regulators (YTHDF1 and HNRNPC) were found to be significantly increased in ESCC tissues after screening out through RF machine learning methods from our RNA-seq data and TCGA-ESCA database, respectively, and overlapping the results of the two clusters. A prognostic signature, consisting of YTHDF1 and HNRNPC, was constructed based on our RNA-seq data and validated on TCGA-ESCA database, which can serve as an independent prognostic predictor. Experimental validation including the western and immunohistochemistry staining were further successfully confirmed the results of bioinformatics analysis. Conclusion: We constructed prognostic ESCC-related m6A regulators and validated the model in clinical ESCC cohort as well as in ESCC tissues, which provides reasonable evidence and valuable resources for prognostic stratification and the study of potential targets for ESCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Long-Qi Chen
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
36
|
Ma J, Luo Y, Liu Y, Chen C, Chen A, Liang L, Wang W, Song Y. Exosome-mediated lnc-ABCA12-3 promotes proliferation and glycolysis but inhibits apoptosis by regulating the toll-like receptor 4/nuclear factor kappa-B signaling pathway in esophageal squamous cell carcinoma. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:61-73. [PMID: 36575934 PMCID: PMC9806635 DOI: 10.4196/kjpp.2023.27.1.61] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/23/2022] [Accepted: 11/03/2022] [Indexed: 12/29/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a kind of malignant tumor with high incidence and mortality in the digestive system. The aim of this study is to explore the function of lnc-ABCA12-3 in the development of ESCC and its unique mechanisms. RT-PCR was applied to detect gene transcription levels in tissues or cell lines like TE-1, EC9706, and HEEC cells. Western blot was conducted to identify protein expression levels of mitochondrial apoptosis and toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) signaling pathway. CCK-8 and EdU assays were carried out to measure cell proliferation, and cell apoptosis was examined by flow cytometry. ELISA was used for checking the changes in glycolysis-related indicators. Lnc-ABCA12-3 was highly expressed in ESCC tissues and cells, which preferred it to be a candidate target. The TE-1 and EC9706 cells proliferation and glycolysis were obviously inhibited with the downregulation of lnc-ABCA12-3, while apoptosis was promoted. TLR4 activator could largely reverse the apoptosis acceleration and relieved the proliferation and glycolysis suppression caused by lnc-ABCA12-3 downregulation. Moreover, the effect of lnc-ABCA12-3 on ESCC cells was actualized by activating the TLR4/NF-κB signaling pathway under the mediation of exosome. Taken together, the lnc-ABCA12-3 could promote the proliferation and glycolysis of ESCC, while repressing its apoptosis probably by regulating the TLR4/NF-κB signaling pathway under the mediation of exosome.
Collapse
Affiliation(s)
- Junliang Ma
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China,Correspondence Junliang Ma, E-mail:
| | - Yijun Luo
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yingjie Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of Zuinyi Medical University, Zunyi, Guizhou 563003, China
| | - Cheng Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Anping Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Lubiao Liang
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Wenxiang Wang
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410031, China
| | - Yongxiang Song
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| |
Collapse
|
37
|
Zhang L, Li L, Chen X, Yuan S, Xu T, Zhao W, Li M, Wang F, Hoffman RM, Jia L. Evodiamine inhibits ESCC by inducing M-phase cell-cycle arrest via CUL4A/p53/p21 axis and activating noxa-dependent intrinsic and DR4-dependent extrinsic apoptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154493. [PMID: 36265256 DOI: 10.1016/j.phymed.2022.154493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/17/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a malignancy with high incidence in several regions of China, and the prognosis of patients with ESCC is unfavorable. Evodiamine (Evo), a small molecule derived from the traditional Chinese herb Evodia rutaecarpa, has shown anti-cancer efficacy in numerous human malignancies but not in ESCC. PURPOSE To determine whether Evo induces cell-cycle arrest and apoptosis in ESCC in vitro and in vivo and elucidate the underlying mechanisms. METHODS ATPlite and colony formation assay were used to validate the inhibiting effect of Evo on three ESCC cells in vitro; Two subcutaneous tumor models of ESCC cells were used to evaluate the anti-ESCC effect of Evo and assess the biosafety of Evo in vivo; RNAseq and Database of KEGG pathway analysis provided a direction for the mechanistic study of Evo; FACS was used to detect Evo-induced cell cycle arrest and cell apoptosis in ESCC cells; Western blot and QPCR were respectively used to detect the level of related genes and proteins in Evo-treated ESCC cells; SiRNA and other experimental techniques were used to identify the molecular mechanism of Evo-induced ESCC cell cycle arrest and cell apoptosis. RESULTS Evo significantly suppressed the growth of ESCC both in vitro and in vivo. Mechanistically, Evo induced M-phase cell-cycle arrest by inactivation of CUL4A E3 ligase, which mediates degradation blockage of p53 and transcriptional activation of p21. With the prolonged treatment time, Evo triggered both Noxa-dependent intrinsic and DR4-dependent extrinsic cell apoptosis in two ESCC cell lines. CONCLUSION Our findings revealed the anti-tumor efficacy and mechanisms of Evo, providing a solid scientific basis for Evo as an attractive choice for ESCC treatment.
Collapse
Affiliation(s)
- Li Zhang
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihui Li
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xihui Chen
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuying Yuan
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Xu
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weili Zhao
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Li
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fengying Wang
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Robert M Hoffman
- Department of Surgery, University of California, La Jolla, San Diego, CA, United States; AntiCancer Inc., San Diego, CA, United States
| | - Lijun Jia
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
38
|
Tian X, Hou Y, Guo J, Wu H, Nie L, Wang H, Zhang Y, Lv Y. Effect of intensity modulated radiotherapy on lymphocytes in patients with esophageal squamous cell carcinoma and its clinical significance. Front Oncol 2023; 13:1096386. [PMID: 36959779 PMCID: PMC10028288 DOI: 10.3389/fonc.2023.1096386] [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: 11/12/2022] [Accepted: 02/24/2023] [Indexed: 03/09/2023] Open
Abstract
Background Radiotherapy usually leads to a decrease in the total number of lymphocytes in patients with esophageal cancer. The factors that causing lymphopenia and the clinical significance of lymphopenia are studied in this article. Patients and methods 110 patients with esophageal squamous cell carcinoma who had undergo intensity-modulated radiation therapy were enrolled. Statistical methods were used to analyze the correlation between lymphopenia and total survival in patients with esophageal cancer during radiotherapy, and analyze the correlations between nutritional factors and lymphopenia. Results There were 11 patients with the lowest lymphocyte value with level 1-2 during radiotherapy, accounting for 10% of all the patients, and 110 patients with level 3-4, accounting for 90% of all the patient. In all the enrolled patients, the incidence of lymphocyte nadir G1, G2, G3 and G4 MinALC during radiotherapy accounted for 0.91%, 9.09%, 62.73% and 27.27%, respectively.KM survival analysis showed that the overall survival of patients in the group (MinALC ≤ 0.41×109/L) was significantly lower than that of the patients in the other group (MinALC>0.43×109/L). Nutritional indicators were positively correlated with the decline degree of lymphocytes. The minimal value of lymphocyte can predict the occurrence of grade 3-4 radiation pneumonitis. Conclusion Lymphopenia induced by radiotherapy can predict survival and radiation pneumonitis. Nutritional factors such as hemoglobin and albumin were positively correlated with total lymphocytes numbers induced by radiotherapy.
Collapse
Affiliation(s)
- Xiufang Tian
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong, China
| | - Yong Hou
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong, China
| | - Jianping Guo
- Department of Oncology, Maternal and Child Health Care Hospital of Zibo, Zibo, Shandong, China
| | - Haiyan Wu
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Limin Nie
- Department of Pathology, Caoxian People's Hospital, Shandong, Heze, China
| | - Hang Wang
- Department of Graduate, Shandong First Medical University, Jinan, Shandong, China
| | - Yan Zhang
- Department of Clinical Medicine, Shandong University, Jinan, Shandong, China
| | - Yajuan Lv
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong, China
- *Correspondence: Yajuan Lv,
| |
Collapse
|
39
|
Wang X, Liu Y, Lu Y, Chen S, Xing Y, Yang H, Wang X, Zhang Y, Pan T, Li J, Wang M, Zhang N, Liang M, Zhou F. Clinical impact of Fn-induced high expression of KIR2DL1 in CD8 T lymphocytes in oesophageal squamous cell carcinoma. Ann Med 2022; 54:51-62. [PMID: 34935568 PMCID: PMC8725851 DOI: 10.1080/07853890.2021.2016942] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND To analyze the correlation between the inducing effect of Fusobacterium nucleatum (Fn) on the surface expression of the inhibitory receptor KIR2DL1 on CD8+ T cells in oesophageal squamous cell carcinoma (ESCC) and the clinicopathological features and survival prognosis and to explore its clinical significance. METHODS The inducing effect of Fn on CD8+ T cell surface inhibitory receptor KIR2DL1 expression was analyzed in a coculture system of human CD8+ T cells and ESCC cells infected with Fn. Fn infection and the expression of KIR2DL1 on CD8+ T cells were detected by RNAscope and immunohistochemistry in ESCC tissues, and the correlations between the inducing effect of Fn on KIR2DL1 expression on CD8+ T cells and clinicopathological features were analyzed. COX regression was used to analyze the influence of each factor on the prognosis of ESCC. Survival curves were plotted by the Kaplan-Meier method, and the effect of KIR2DL1 induction on survival time was analyzed by the log-rank test. RESULTS In the coculture system, KIR2DL1 expression on the surface of CD8+ T cells increased with increasing Fn infection time. In ESCC tissues, Fn infection was significantly correlated with high KIR2DL1 expression on CD8+ T cells. The Fn + CD8+KIR2DL1 positive patients were predominantly males who were smokers and alcohol drinkers. Moreover, patients with Fn infection were characterized by poor tumour differentiation, advanced clinical stage, and a short survival time. Meanwhile, Fn + CD8+KIR2DL1 positive group was independent risk factor affecting the prognosis of ESCC patients. CONCLUSIONS Long-term drinking and smoking lead to an extremely unhealthy oral environment in which Fn infection and colonization are more likely to occur, thus inducing high expression of KIR2DL1 on the surface of CD8+ T cells, which can weaken the antitumour immune response and promote the malignant progression of ESCC.HIGHLIGHTSFn induced high expression of KIR2DL1 CD8+ T cells in a time-dependent manner.Fn can reduce the response of tumour cells to CDDP.The inducing effect of Fn on CD8+ T cell surface KIR2DL1 expression was significantly associated with the poor prognosis of ESCC patients.
Collapse
Affiliation(s)
- Xiaopeng Wang
- Anyang Tumor Hospital (The Fourth Affiliated Hospital of Henan University of Science and Technology), Anyang, Henan, China.,The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yiwen Liu
- College of Clinical Medicine, Laboratory of Molecular Biology of the First Affiliated Hospital, Cancer Institute of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China
| | - Yannan Lu
- Graduate School of Dalian Medical University, Dalian, Liaoning, China
| | - Simo Chen
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yaoping Xing
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Haijun Yang
- Anyang Tumor Hospital (The Fourth Affiliated Hospital of Henan University of Science and Technology), Anyang, Henan, China
| | - Xiaojun Wang
- Basic Medical School of Henan University of Science and Technology, Luoyang, Henan, China
| | - Yaowen Zhang
- Anyang Tumor Hospital (The Fourth Affiliated Hospital of Henan University of Science and Technology), Anyang, Henan, China
| | - Tao Pan
- Department of Lymphoma & Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Junkuo Li
- Anyang Tumor Hospital (The Fourth Affiliated Hospital of Henan University of Science and Technology), Anyang, Henan, China
| | - Min Wang
- Anyang Tumor Hospital (The Fourth Affiliated Hospital of Henan University of Science and Technology), Anyang, Henan, China.,The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Ning Zhang
- College of Clinical Medicine, Laboratory of Molecular Biology of the First Affiliated Hospital, Cancer Institute of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China
| | - Mengxia Liang
- College of Clinical Medicine, Laboratory of Molecular Biology of the First Affiliated Hospital, Cancer Institute of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China
| | - Fuyou Zhou
- Anyang Tumor Hospital (The Fourth Affiliated Hospital of Henan University of Science and Technology), Anyang, Henan, China.,The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.,College of Clinical Medicine, Laboratory of Molecular Biology of the First Affiliated Hospital, Cancer Institute of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China
| |
Collapse
|
40
|
Gui Z, Hu W, Kong Q, Liu C, Xu Y, Wang F. Esophageal stenosis as an independent factor of poor prognosis in patients with ESCC treated with definitive chemoradiotherapy. Future Oncol 2022; 18:4193-4207. [PMID: 36651337 DOI: 10.2217/fon-2022-0125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aim: To evaluate the clinical outcome and elucidate the prognostic factors in patients with esophageal squamous cell carcinoma (ESCC) treated with definitive chemoradiotherapy (CRT). Patients: Data for patients newly diagnosed with ESCC receiving definitive CRT at our institution between 2012 and 2018 were retrospectively reviewed. Results: A total of 201 patients were included. Severe stenosis after radiotherapy was an independent factor relevant to prognosis. Maximal esophageal wall thickness, short-term responses, severe stenosis at diagnosis and a high neutrophil-to-lymphocyte ratio were independent risk factors for the occurrence of severe stenosis after radiotherapy. Conclusion: Severe stenosis after radiotherapy is a useful predictive indicator in patients with ESCC receiving definitive CRT. Further studies are needed to verify these findings.
Collapse
Affiliation(s)
- Zhongxuan Gui
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Wenjun Hu
- Department of Radiation Oncology, Anhui Chest Hospital, Hefei, Anhui, 230022, People's Republic of China
| | - Qi Kong
- Department of Radiation Oncology, Anhui Second People's Hospital, Hefei, Anhui, 230012, People's Republic of China
| | - Can Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yuechen Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Fan Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| |
Collapse
|
41
|
Drug Repurposing Applications to Overcome Male Predominance via Targeting G2/M Checkpoint in Human Esophageal Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14235854. [PMID: 36497337 PMCID: PMC9741366 DOI: 10.3390/cancers14235854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/29/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is strongly characterized by a male predominance with higher mortality rates and worse responses to treatment in males versus females. Despite the role of sex hormones, other causes that may contribute to sex bias in ESCC remain largely unknown, especially as age increases and the hormone difference begins to diminish between sexes. In this study, we analyzed genomics, transcriptomics, and epigenomics from 663 ESCC patients and found that G2/M checkpoint pathway-related sex bias and age bias were significantly present in multi-omics data. In accordance with gene expression patterns across sexes, ten compounds were identified by applying drug repurposing from three drug sensitivity databases: The Connective Map (CMap), Genomics of Drug Sensitivity in Cancer (GDSC), and The Cancer Therapeutic Response Portal (CTRP). MK1775 and decitabine showed better efficacy in two male ESCC cell lines in vitro and in vivo. The drugs' relevance to the transition between G2 and M was especially evident in male cell lines. In our study, we first validated the sex bias of the G2/M checkpoint pathway in ESCC and then determined that G2/M targets may be included in combination therapy for male patients to improve the efficacy of ESCC treatment.
Collapse
|
42
|
Ye ZM, Xu Z, Zeng FY, Tang ZQ, Zhou Q. Cost-Effectiveness Analysis of Sintilimab Combined with Chemotherapy Versus Chemotherapy Alone as the First-Line Treatment for Advanced Esophageal Cancer. Front Pharmacol 2022; 13:934275. [PMID: 36518659 PMCID: PMC9742528 DOI: 10.3389/fphar.2022.934275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 09/15/2023] Open
Abstract
Background: Esophageal cancer has a poor prognosis and currently ranks sixth in global cancer mortality rates. The ORIENT-15 trial showed sintilimab plus chemotherapy significantly improved survival when compared to chemotherapy alone. This study aimed to evaluate the cost-effectiveness of sintilimab, a programmed death-ligand 1 (PD-L1) inhibitor, plus chemotherapy in treating patients with esophageal cancer compared with chemotherapy alone. Methods: A Markov model with a 10-year horizon was developed based on the perspective of the Chinese healthcare payers. We conducted a cost-effectiveness analysis for sintilimab combined with chemotherapy based on a questionnaire. Patients were grouped into the sintilimab group based on a positive score of 10 or more (combined positive score (CPS) ≥ 10 groups), and those with any other PD-L1 expression were randomized into patient groups. We estimated the cost and the effectiveness of sintilimab on the quality-adjusted life-years (QALYs), and the incremental cost-effectiveness ratio (ICER) was computed. One-way and probabilistic sensitivity analyses were conducted to explore the impact of uncertainties on the cost-effectiveness results. Results: In the base-case analysis, compared with chemotherapy alone, the ICER of sintilimab plus chemotherapy for all patients was $21024.05 per QALY, and in the CPS≥10 group, it was $20974.23 per QALY. This was lower than $37653 per QALY. One-way sensitivity analysis demonstrated that ICERs were most sensitive to the price of sintilimab. Conclusion: The study demonstrated that sintilimab plus chemotherapy for advanced esophageal cancer as its first-line treatment would be more cost-effective than chemotherapy alone in Chinese patients.
Collapse
Affiliation(s)
- Zhuo-Miao Ye
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhe Xu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Fan-Yuan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zi-Qing Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
43
|
Akinlalu AO, Njoku PC, Nzekwe CV, Oni RO, Fojude T, Faniyi AJ, Olagunju AS. Recent developments in the significant effect of mRNA modification (M6A) in glioblastoma and esophageal cancer. SCIENTIFIC AFRICAN 2022. [DOI: 10.1016/j.sciaf.2022.e01347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
44
|
Cheng J, Ma H, Yan M, Zhang Z, Xing W. Circ_0007624 suppresses the development of esophageal squamous cell carcinoma via targeting miR-224-5p/CPEB3 to inactivate the EGFR/PI3K/AKT signaling. Cell Signal 2022; 99:110448. [PMID: 35998761 DOI: 10.1016/j.cellsig.2022.110448] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/03/2022]
Abstract
Circular RNAs (circRNAs) have been confirmed to be involved in the regulation of esophageal squamous cell carcinoma (ESCC) progression. According to GEO datasets (GSE112496 and GSE150476), we identified that circ_0007624 was abnormally down-regulated in ESCC. However, there is still no reports regarding the function and mechanism of circ_0007624 in ESCC development. Here, we found that circ_0007624 was significantly underexpressed in ESCC tissues, and low expression of circ_0007624 was indicative of a poor prognosis. Overexpressing circ_0007624 or silencing miR-224-5p suppressed cell proliferation, metastasis, epithelial-mesenchymal transition (EMT), and promoted apoptosis in vitro. Also, circ_0007624 up-regulation slowed ESCC tumor growth in vivo. Mechanistically, circ_0007624 could serve as a competing endogenous RNA (ceRNA) by sponging miR-224-5p to antagonize its inhibitory effect on the target cytoplasmic polyadenylation element binding protein 3 (CPEB3). Rescue experiments showed that the anti-cancer properity role of circ_0007624 in ESCC is partly reversed by the restoration of miR-224-5p or down-regulation of CPEB3. Furthermore, EGFR/PI3K/AKT pathway was involved in the regulation of circ_0007624/miR-224-5p/CPEB3 axis in ESCC. Together, our findings demonstrate for the first time that circ_0007624/miR-224-5p/CPEB3 suppresses ESCC progression by inactivating EGFR/PI3K/AKT signaling, providing a basis for developing circ_0007624-targeted therapies for ESCC patients.
Collapse
Affiliation(s)
- Jiwei Cheng
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China
| | - Haibo Ma
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China
| | - Ming Yan
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China
| | - Zhen Zhang
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China.; Department of Anesthesiology, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China..
| | - Wenqun Xing
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China..
| |
Collapse
|
45
|
Geng C, Wang Q, Xing PF, Wang M, Tong SD, Zhou JY. Effects and mechanisms of GSG2 in esophageal cancer progression. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04260-2. [PMID: 35939116 DOI: 10.1007/s00432-022-04260-2] [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: 05/16/2022] [Accepted: 08/02/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Esophageal cancer was recognized as one of the malignant tumors with poor prognosis. Germ cell associated 2 (GSG2) has been reported to be of great significance in cell growth and tumor formation. This study aimed to investigate the biological function and molecular mechanism of GSG2 in esophageal cancer. METHODS First, relationship between GSG2 expression and tumor characteristics in esophageal cancer patients was analyzed through immunohistochemical (IHC) staining. MTT assay, flow cytometry, cloning formation assay, wound-healing assay and Transwell assay were used to determine proliferation, apoptosis and migration of esophageal cancer cell with GSG2 knockdown in vitro. Expression of apoptosis related proteins and downstream pathway proteins after GSG2 knockdown were detected through Human Apoptosis Antibody Array and western blot analysis. The GSG2 knockdown function in vivo was explored through a xenograft tumor model. RESULTS GSG2 was highly expressed in tumor tissues, which has clinical significance in predicting the malignant degree of patients with esophageal cancer. In addition, GSG2 knockdown significantly inhibited a variety of malignant biological behaviors of esophageal cancer cells, such as inhibiting proliferation, reducing colony formation, promoting apoptosis, hindering migration. The decrease of GSG2 expression in esophageal cancer cells can inhibit the xenograft tumor growth. CONCLUSIONS In conclusion, GSG2 was involved in esophageal cancer progression and development, which may provide an effective molecular target for the treatment of esophageal cancer in the future.
Collapse
Affiliation(s)
- Chong Geng
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Jiangsu Province, No.188 Shizi street, Suzhou, 215006, China
- Department of Radiation Oncology, Xuzhou Cancer Hospital, Xuzhou, 221005, China
| | - Qiang Wang
- Department of Radiation Oncology, Xuzhou Cancer Hospital, Xuzhou, 221005, China
| | - Peng-Fei Xing
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Min Wang
- Department of Radiation Oncology, Xuzhou Cancer Hospital, Xuzhou, 221005, China
| | - Shao-Dong Tong
- Department of Radiation Oncology, Xuzhou Cancer Hospital, Xuzhou, 221005, China
| | - Ju-Ying Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Jiangsu Province, No.188 Shizi street, Suzhou, 215006, China.
| |
Collapse
|
46
|
Li ZC, Sun YT, Lai MY, Zhou YX, Qiu MZ. Efficacy and safety of PD-1 inhibitors combined with chemotherapy as first-line therapy for advanced esophageal cancer: A systematic review and network meta-analysis. Int Immunopharmacol 2022; 109:108790. [DOI: 10.1016/j.intimp.2022.108790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 01/22/2023]
|
47
|
Liao F, Yu S, Zhou Y, Feng B. A machine learning model predicting candidates for surgical treatment modality in patients with distant metastatic esophageal adenocarcinoma: A propensity score-matched analysis. Front Oncol 2022; 12:862536. [PMID: 35936753 PMCID: PMC9354694 DOI: 10.3389/fonc.2022.862536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo explore the role of surgical treatment modality on prognosis of metastatic esophageal adenocarcinoma (mEAC), as well as to construct a machine learning model to predict suitable candidates.MethodAll mEAC patients pathologically diagnosed between January 2010 and December 2018 were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. A 1:4 propensity score-matched analysis and a multivariate Cox analysis were performed to verify the prognostic value of surgical treatment modality. To identify suitable candidates, a machine learning model, classification and regression tree (CART), was constructed, and its predictive performance was evaluated by the area under receiver operating characteristic curve (AUC).ResultsOf 4520 mEAC patients, 2901 (64.2%) were aged over 60 years and 4012 (88.8%) were males. There were 411 (9.1%) patients receiving surgical treatment modality. In the propensity score-matched analysis, surgical treatment modality was significantly associated with a decreased risk of death (HR: 0.47, 95% CI: 0.40-0.55); surgical patients had almost twice as much median survival time (MST) as those without resection (MST with 95% CI: 23 [17-27] months vs. 11 [11-12] months, P <0.0001). The similar association was also observed in the multivariate Cox analysis (HR: 0.47, 95% CI: 0.41-0.53). Then, a CART was constructed to identify suitable candidates for surgical treatment modality, with a relatively good discrimination ability (AUC with 95% CI: 0.710 [0.648-0.771]).ConclusionSurgical treatment modality may be a promising strategy to prolong survival of mEAC patients. The CART in our study could serve as a useful tool to predict suitable candidates for surgical treatment modality. Further creditable studies are warranted to confirm our findings.
Collapse
Affiliation(s)
- Fang Liao
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, China
| | - Shuangbin Yu
- Department of Medical Administration, Sichuan Academy of Medical Sciences and Sichuan People’s Hospital, Chengdu, China
| | - Ying Zhou
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, China
| | - Benying Feng
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, China
- *Correspondence: Benying Feng,
| |
Collapse
|
48
|
Ainiwaer J, Zhang L, Niyazi M, Awut E, Zheng S, Sheyhidin I, Dai J. Alpha Protein Kinase 2 Promotes Esophageal Cancer via Integrin Alpha 11. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7676582. [PMID: 35813220 PMCID: PMC9259355 DOI: 10.1155/2022/7676582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/10/2022] [Accepted: 05/27/2022] [Indexed: 12/24/2022]
Abstract
Background As a common disease around the world, esophageal cancer (EC) primarily includes two subclasses: esophageal adenocarcinoma and esophageal squamous cell carcinoma. Mortality has been rising over the years; hence, exploring the mechanism of EC development has become critical. Among the alpha protein kinases, alpha protein kinase 2 (ALPK2) presumably has a connection with EC, but it has never been revealed before. Methods In this study, IHC analysis was used for ALPK2 expression quantification in ES tissues. TE-1 and Eca-109, which are both human EC cell lines, were used for in vitro analysis of cell proliferation, migration, apoptosis, and colony formation. Results ALPK2 was found to have an abundant expression within EC tissues (P < 0.001), as well as in the two selected human EC cell lines (P < 0.05). The data showed that ALPK2 depletion suppressed EC cell proliferation, migration, and colony formation, meanwhile stimulating apoptosis (P < 0.001). The in vivo experiments also displayed inhibitory effects caused by ALPK2 depletion on EC tumorigenesis (P < 0.001). It was further validated that ALPK2 depletion made the phosphorylation of Akt and mTOR, as well as CDK6 and PIK3CA levels downregulated (P < 0.001). Mechanistically, we identified integrin alpha 11 (ITGA11) as a downstream gene of ALPK2 regulating EC. More importantly, we found that ITGA11 elevation promoted cell proliferation and migration and rescued the suppression effects caused by ALPK2 depletion (P < 0.001). Conclusions ALPK2 promotes esophageal cancer via integrin its downstream gene alpha 11; ALPK2 can potentially act as a target for the treatment of EC.
Collapse
Affiliation(s)
- Julaiti Ainiwaer
- School of Public Health, Xinjiang Medical University, China
- Department of Thoracic Surgery, First Affiliated Hospital of Xinjiang Medical University, China
| | - Liwei Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Xinjiang Medical University, China
| | - Maidiniyeti Niyazi
- The Clinical Medicine Research Institute, First Affiliated Hospital of Xinjiang Medical University, China
| | - Edris Awut
- Department of Thoracic Surgery, First Affiliated Hospital of Xinjiang Medical University, China
| | - Shutao Zheng
- The Clinical Medicine Research Institute, First Affiliated Hospital of Xinjiang Medical University, China
| | - Ilyar Sheyhidin
- Department of Thoracic Surgery, First Affiliated Hospital of Xinjiang Medical University, China
| | - JiangHong Dai
- School of Public Health, Xinjiang Medical University, China
| |
Collapse
|
49
|
Chen L, Fang B, Qiao L, Zheng Y. Discovery of Anticancer Activity of Amentoflavone on Esophageal Squamous Cell Carcinoma: Bioinformatics, Structure-Based Virtual Screening, and Biological Evaluation. J Microbiol Biotechnol 2022; 32:718-729. [PMID: 35484963 PMCID: PMC9628896 DOI: 10.4014/jmb.2203.03050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/28/2022] [Indexed: 12/15/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common primary esophageal malignancy with poor prognosis. Here, due to the necessity for exploring potential therapies against ESCC, we obtained the gene expression data on ESCC from the TCGA and GEO databases. Venn diagram analysis was applied to identify common targets. The protein-protein interaction network was constructed by Cytoscape software, and the hub targets were extracted from the network via cytoHubba. The potential hub nodes as drug targets were found by pharmacophore-based virtual screening and molecular modeling, and the antitumor activity was evaluated through in vitro studies. A total of 364 differentially expressed genes (DEGs) in ESCC were identified. Pathway enrichment analyses suggested that most DEGs were mainly involved in the cell cycle. Three hub targets were retrieved, including CENPF, CCNA2 (cyclin A), and CCNB1 (cyclin B1), which were highly expressed in esophageal cancer and associated with prognosis. Moreover, amentoflavone, a promising drug candidate found by pharmacophore-based virtual screening, showed antiproliferative and proapoptotic effects and induced G1 in esophageal squamous carcinoma cells. Taken together, our findings suggested that amentoflavone could be a potential cell cycle inhibitor targeting cyclin B1, and is therefore expected to serve as a great therapeutic agent for treating esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Lei Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Bo Fang
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang 325015, P.R. China
| | - Liman Qiao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Yihui Zheng
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China,Corresponding author Phone/Fax : 86-0577-6288-2358 E-mail:
| |
Collapse
|
50
|
Gao Y, Shen L, Dong T, Yang X, Cui H, Guo Y, Ma Y, Kong P, Cheng X, Zhang L, Cui Y. An N-glycoproteomic site-mapping analysis reveals glycoprotein alterations in esophageal squamous cell carcinoma. J Transl Med 2022; 20:285. [PMID: 35752862 PMCID: PMC9233802 DOI: 10.1186/s12967-022-03489-2] [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: 01/14/2022] [Accepted: 06/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background Aberrant glycosylation has been recognized as a hallmark of cancer and N-glycosylation is one of the main types of glycosylation in eukaryotes. Although N-glycoproteomics has made contributions to the discovery of biomarkers in a variety of cancers, less is known about the abnormal glycosylation signatures in esophageal squamous cell carcinoma (ESCC). Methods In this study, we reported the proteomics and N-glycoproteomic site-mapping analysis of eight pairs of ESCC tissues and adjacent normal tissues. With zic-HILIC enrichment, TMT-based isobaric labeling, LC–MS/MS analysis, differentially expressed N-glycosylation was quantitatively characterized. Lectin affinity enrichment combined with western blot was used to validate the potential biomarkers in ESCC. Results A series of differentially expressed glycoproteins (e.g., LAMP2, PLOD2) and enriched signaling pathways (e.g., metabolism-related pathway, ECM-receptor interaction, focal adhesion) were identified. Besides that, seven significantly enriched motifs were found from the identified N-glycosylation sites. Three clusters were identified after conducting the dynamic profiling analysis of glycoprotein change during lymph node metastasis progression. Further validation found that the elevated fucosylation level of ITGB1, CD276 contributed to the occurrence and development of ESCC, which might be the potential biomarkers in ESCC. Conclusion In summary, we characterized the N-glycosylation and N-glycoprotein alterations associated with ESCC. The typical changes in glycoprotein expression and glycosylation occupancy identified in our study will not only be used as ESCC biomarkers but also improve the understanding of ESCC biology. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03489-2.
Collapse
Affiliation(s)
- Yingzhen Gao
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Liuyi Shen
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Tianyue Dong
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Xin Yang
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Heyang Cui
- Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, People's Republic of China
| | - Yanlin Guo
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yanchun Ma
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Pengzhou Kong
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Xiaolong Cheng
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Ling Zhang
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China. .,Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, People's Republic of China.
| | - Yongping Cui
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China. .,Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, People's Republic of China.
| |
Collapse
|