1
|
Hao S, Shi L, Li J, Shi J, Kuang G, Liang G, Gao S. Biomacromolecular hydrogel scaffolds from microfluidics for cancer therapy: A review. Int J Biol Macromol 2024; 282:136738. [PMID: 39437954 DOI: 10.1016/j.ijbiomac.2024.136738] [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: 05/08/2024] [Revised: 09/29/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Traditional cancer treatment is confronted with the problem of limited therapeutic effect, tissue defects, and lack of drug screening. Hydrogel scaffolds from biological macromolecules based on microfluidic technology are a promising candidate, which can mimic tumor microenvironments to screen personalized drugs, promote the regeneration of healthy tissues, and deliver drugs for enhanced localized antitumor treatment. This review summarizes the latest research on the composition of biomacromolecular hydrogel scaffolds, the architecture of hydrogel scaffolds from microfluidic technology, and their application in cancer therapy, including anti-tumor drug screening, anti-tumor treatment, and anti-tumor treatment and tissue repair. In addition, the potential breakthroughs of this innovative platform in the clinical transformation of cancer therapy are further discussed. The insights revealed in this review are intended to guide the utilization of microfluidic technology-based biomacromolecular hydrogel scaffolds in cancer therapy.
Collapse
Affiliation(s)
- Siyu Hao
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, College of Basic Medicine and Forensic Medicine, Cancer Hospital, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and Technology, Luoyang 471003, China
| | - Linlin Shi
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, College of Basic Medicine and Forensic Medicine, Cancer Hospital, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and Technology, Luoyang 471003, China.
| | - Jiayi Li
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, College of Basic Medicine and Forensic Medicine, Cancer Hospital, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and Technology, Luoyang 471003, China
| | - Jiaming Shi
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, College of Basic Medicine and Forensic Medicine, Cancer Hospital, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and Technology, Luoyang 471003, China
| | - Gaizhen Kuang
- Department of Internal Medicine Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Gaofeng Liang
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, College of Basic Medicine and Forensic Medicine, Cancer Hospital, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and Technology, Luoyang 471003, China.
| | - Shegan Gao
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, College of Basic Medicine and Forensic Medicine, Cancer Hospital, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and Technology, Luoyang 471003, China.
| |
Collapse
|
2
|
Bhat N, Al-Mathkour M, Maacha S, Lu H, El-Rifai W, Ballout F. Esophageal adenocarcinoma models: a closer look. Front Mol Biosci 2024; 11:1440670. [PMID: 39600303 PMCID: PMC11589788 DOI: 10.3389/fmolb.2024.1440670] [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: 05/29/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Esophageal adenocarcinoma (EAC) is a subtype of esophageal cancer with significant morbidity and mortality rates worldwide. Despite advancements in tumor models, the underlying cellular and molecular mechanisms driving EAC pathogenesis are still poorly understood. Therefore, gaining insights into these mechanisms is crucial for improving patient outcomes. Researchers have developed various models to better understand EAC and evaluate clinical management strategies. However, no single model fully recapitulates the complexity of EAC. Emerging technologies, such as patient-derived organoids and immune-competent mouse models, hold promise for personalized EAC research and drug development. In this review, we shed light on the various models for studying EAC and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Nadeem Bhat
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Marwah Al-Mathkour
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Selma Maacha
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Heng Lu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Farah Ballout
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
3
|
Chen M, Qi Y, Zhang S, Du Y, Cheng H, Gao S. Molecular insights into programmed cell death in esophageal squamous cell carcinoma. PeerJ 2024; 12:e17690. [PMID: 39006030 PMCID: PMC11246021 DOI: 10.7717/peerj.17690] [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: 01/31/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is a deadly type of esophageal cancer. Programmed cell death (PCD) is an important pathway of cellular self-extermination and is closely involved in cancer progression. A detailed study of its mechanism may contribute to ESCC treatment. Methods We obtained expression profiling data of ESCC patients from public databases and genes related to 12 types of PCD from previous studies. Hub genes in ESCC were screened from PCD-related genes applying differential expression analysis, machine learning analysis, linear support vector machine (SVM), random forest and Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis. In addition, based on the HTFtarget and TargetScan databases, transcription factors (TFs) and miRNAs interacting with the hub genes were selected. The relationship between hub genes and immune cells were analyzed using the CIBERSORT algorithm. Finally, to verify the potential impact of the screened hub genes on ESCC occurrence and development, a series of in vitro cell experiments were conducted. Results We screened 149 PCD-related DEGs, of which five DEGs (INHBA, LRRK2, HSP90AA1, HSPB8, and EIF2AK2) were identified as the hub genes of ESCC. The area under the curve (AUC) of receiver operating characteristic (ROC) curve of the integrated model developed using the hub genes reached 0.997, showing a noticeably high diagnostic accuracy. The number of TFs and miRNAs regulating hub genes was 105 and 22, respectively. INHBA, HSP90AA1 and EIF2AK2 were overexpressed in cancer tissues and cells of ESCC. Notably, INHBA knockdown suppressed ECSS cell migration and invasion and altered the expression of important apoptotic and survival proteins. Conclusion This study identified significant molecules with promising accuracy for the diagnosis of ESCC, which may provide a new perspective and experimental basis for ESCC research.
Collapse
Affiliation(s)
- Min Chen
- School of Information Engineering, Henan University of Science and Technology, Luoyang, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College of Henan University of Science and Technology, Luoyang, China
| | - Yijun Qi
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College of Henan University of Science and Technology, Luoyang, China
| | - Shenghua Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College of Henan University of Science and Technology, Luoyang, China
| | - Yubo Du
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College of Henan University of Science and Technology, Luoyang, China
| | - Haodong Cheng
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College of Henan University of Science and Technology, Luoyang, China
| | - Shegan Gao
- School of Information Engineering, Henan University of Science and Technology, Luoyang, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
4
|
Gong K, Lin J, Chen X, Duan Y, Zhang J, Yu J, Wang J, Sun R, Li J, Duan Y. Thermosensitive gel-nano system against esophageal cancer via restoring p53 activity and boosting T-cell immunity. J Control Release 2024; 371:111-125. [PMID: 38782064 DOI: 10.1016/j.jconrel.2024.05.040] [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: 01/19/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
In esophageal cancer (EC), clinical specimen testing has uncovered a significant increase in BTB and CNC homolog 1 (BACH1) expression and a shift towards an immunosuppressive environment, alongside a notable decrease in p53 protein expression. Therefore, therapeutic strategies focusing on BACH1 inhibition and p53 upregulation appear promising. Traditional oral treatments for EC lack precision and efficacy. Here, we propose a novel approach employing tumor-targeted nanoparticles (NPs) for drug delivery. However, the formation of a drug reservoir at the esophageal site, crucial for the sustained release of therapeutics, presents significant challenges in nano-delivery systems for EC treatment. To address this, we developed a thermosensitive hydrogel composed of F127 and tannic acid, serving as a vehicle for NP loading. These NPs, synthesized through the emulsion/volatization methods of mPEG-PLGA-PLL-cRGD, facilitate in situ drug delivery. Upon contacting esophageal tissue, the hydrogel transitions to a gel, adhering to the lining and enabling sustained release of encapsulated therapeutics. The formulation encompasses NPs laden with small interfering RNA targeting BACH1 (siBACH1) and the p53 activator PRIMA-1, creating a cohesive gel-nano system. Preliminary biological assessments demonstrate that this injectable, thermosensitive gel-nano system adheres effectively to esophageal tissue and targets EC cells. For better modeling clinical outcomes, a patient-derived organoid xenograft (PDOX) model was innovated, involving transplantation of EC-derived organoids into humanized mice, reconstructed with peripheral blood mononuclear cells (PBMCs). Post-treatment analysis showed substantial EC growth inhibition (89.51% tumor inhibition rate), significant BACH1 level reduction, restored anti-tumor immune responses, and pronounced tumor apoptosis. In summary, our study introduces a thermosensitive gel-nano system for EC treatment via restoring p53 activity and boosting T-cell immunity, with potential for clinical application.
Collapse
Affiliation(s)
- Ke Gong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Jiangtao Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Xiaoyan Chen
- Department of Pathology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yi Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Jiali Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Jian Yu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Jing Wang
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, China
| | - Ruifang Sun
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, China.
| | - Jie Li
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, China.
| | - Yourong Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China.
| |
Collapse
|
5
|
Chen X, Cheng G, Zhu L, Liu T, Yang X, Liu R, Ou Z, Zhang S, Tan W, Lin D, Wu C. Alarmin S100A8 imparts chemoresistance of esophageal cancer by reprogramming cancer-associated fibroblasts. Cell Rep Med 2024; 5:101576. [PMID: 38776909 PMCID: PMC11228400 DOI: 10.1016/j.xcrm.2024.101576] [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: 12/20/2023] [Revised: 03/08/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Chemotherapy remains the first-line treatment for advanced esophageal cancer. However, durable benefits are achieved by only a limited subset of individuals due to the elusive chemoresistance. Here, we utilize patient-derived xenografts (PDXs) from esophageal squamous-cell carcinoma to investigate chemoresistance mechanisms in preclinical settings. We observe that activated cancer-associated fibroblasts (CAFs) are enriched in the tumor microenvironment of PDXs resistant to chemotherapy. Mechanistically, we reveal that cancer-cell-derived S100A8 triggers the intracellular RhoA-ROCK-MLC2-MRTF-A pathway by binding to the CD147 receptor of CAFs, inducing CAF polarization and leading to chemoresistance. Therapeutically, we demonstrate that blocking the S100A8-CD147 pathway can improve chemotherapy efficiency. Prognostically, we found the S100A8 levels in peripheral blood can serve as an indicator of chemotherapy responsiveness. Collectively, our study offers a comprehensive understanding of the molecular mechanisms underlying chemoresistance in esophageal cancer and highlights the potential value of S100A8 in the clinical management of esophageal cancer.
Collapse
Affiliation(s)
- Xinjie Chen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Guoyu Cheng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Liang Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Xinyu Yang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Rucheng Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Zhengjie Ou
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Wen Tan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; CAMS Oxford Institute, Chinese Academy of Medical Sciences, Beijing 100006, China.
| |
Collapse
|
6
|
Bhat AA, Gupta G, Dahiya R, Thapa R, Gahtori A, Shahwan M, Jakhmola V, Tiwari A, Kumar M, Dureja H, Singh SK, Dua K, Kumarasamy V, Subramaniyan V. CircRNAs: Pivotal modulators of TGF-β signalling in cancer pathogenesis. Noncoding RNA Res 2024; 9:277-287. [PMID: 38505309 PMCID: PMC10945146 DOI: 10.1016/j.ncrna.2024.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 03/21/2024] Open
Abstract
The intricate molecular landscape of cancer pathogenesis continues to captivate researchers worldwide, with Circular RNAs (circRNAs) emerging as pivotal players in the dynamic regulation of biological functions. The study investigates the elusive link between circRNAs and the Transforming Growth Factor-β (TGF-β) signalling pathway, exploring their collective influence on cancer progression and metastasis. Our comprehensive investigation begins by profiling circRNA expression patterns in diverse cancer types, revealing a repertoire of circRNAs intricately linked to the TGF-β pathway. Through integrated bioinformatics analyses and functional experiments, we elucidate the specific circRNA-mRNA interactions that modulate TGF-β signalling, unveiling the regulatory controls governing this crucial pathway. Furthermore, we provide compelling evidence of the impact of circRNA-mediated TGF-β modulation on key cellular processes, including epithelial-mesenchymal transition (EMT), migration, and cell proliferation. In addition to their mechanistic roles, circRNAs have shown promise as diagnostic and prognostic biomarkers, as well as potential molecular targets for cancer therapy. Their ability to modulate critical pathways, such as the TGF-β signalling axis, underscores their significance in cancer biology and clinical applications. The intricate interplay between circRNAs and TGF-β is dissected, uncovering novel regulatory circuits that contribute to the complexity of cancer biology. This review unravels a previously unexplored dimension of carcinogenesis, emphasizing the crucial role of circRNAs in shaping the TGF-β signalling landscape.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
| | - Rajiv Dahiya
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad & Tobago
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Archana Gahtori
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, 248001, Uttarakhand, India
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates
| | - Vikas Jakhmola
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, India
| | - Abhishek Tiwari
- Pharmacy Academy, IFTM University, Lodhipur-Rajput, Moradabad, (U.P.), 244102, India
| | - Mahish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
7
|
Pu F, Guo H, Shi D, Chen F, Peng Y, Huang X, Liu J, Zhang Z, Shao Z. The generation and use of animal models of osteosarcoma in cancer research. Genes Dis 2024; 11:664-674. [PMID: 37692517 PMCID: PMC10491873 DOI: 10.1016/j.gendis.2022.12.021] [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: 02/27/2022] [Accepted: 12/16/2022] [Indexed: 09/12/2023] Open
Abstract
Osteosarcoma is the most common malignant bone tumor affecting children and adolescents. Currently, the most common treatment is surgery combined with neoadjuvant chemotherapy. Although the survival rate of patients with osteosarcoma has improved in recent years, it remains poor when the tumor(s) progress and distant metastases develop. Therefore, better animal models that more accurately replicate the natural progression of the disease are needed to develop improved prognostic and diagnostic markers, as well as targeted therapies for both primary and metastatic osteosarcoma. The present review described animal models currently being used in research investigating osteosarcoma, and their characteristics, advantages, and disadvantages. These models may help elucidate the pathogenic mechanism(s) of osteosarcoma and provide evidence to support and develop clinical treatment strategies.
Collapse
Affiliation(s)
- Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No.1 Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Haoyu Guo
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Deyao Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Fengxia Chen
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, China
- Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, China
| | - Yizhong Peng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xin Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jianxiang Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
8
|
Aslam M, Rajbdad F, Azmat S, Li Z, Boudreaux JP, Thiagarajan R, Yao S, Xu J. A novel method for detection of pancreatic Ductal Adenocarcinoma using explainable machine learning. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 245:108019. [PMID: 38237450 DOI: 10.1016/j.cmpb.2024.108019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND AND OBJECTIVE Pancreatic Ductal Adenocarcinoma (PDAC) is a form of pancreatic cancer that is one of the primary causes of cancer-related deaths globally, with less than 10 % of the five years survival rate. The prognosis of pancreatic cancer has remained poor in the last four decades, mainly due to the lack of early diagnostic mechanisms. This study proposes a novel method for detecting PDAC using explainable and supervised machine learning from Raman spectroscopic signals. METHODS An insightful feature set consisting of statistical, peak, and extended empirical mode decomposition features is selected using the support vector machine recursive feature elimination method integrated with a correlation bias reduction. Explicable features successfully identified mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS) and tumor suppressor protein53 (TP53) in the fingerprint region for the first time in the literature. PDAC and normal pancreas are classified using K-nearest neighbor, linear discriminant analysis, and support vector machine classifiers. RESULTS This study achieved a classification accuracy of 98.5% using a nonlinear support vector machine. Our proposed method reduced test time by 28.5 % and saved 85.6 % memory utilization, which reduces complexity significantly and is more accurate than the state-of-the-art method. The generalization of the proposed method is assessed by fifteen-fold cross-validation, and its performance is evaluated using accuracy, specificity, sensitivity, and receiver operating characteristic curves. CONCLUSIONS In this study, we proposed a method to detect and define the fingerprint region for PDAC using explainable machine learning. This simple, accurate, and efficient method for PDAC detection in mice could be generalized to examine human pancreatic cancer and provide a basis for precise chemotherapy for early cancer treatment.
Collapse
Affiliation(s)
- Murtaza Aslam
- Department of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Fozia Rajbdad
- Department of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Shoaib Azmat
- Department of Electrical and Computer Engineering, COMSATS University Islamabad, Pakistan
| | - Zheng Li
- Department of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - J Philip Boudreaux
- Department of Surgery, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ramcharan Thiagarajan
- Department of Surgery, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Shaomian Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Jian Xu
- Department of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
9
|
Yang Z, Guan F, Bronk L, Zhao L. Multi-omics approaches for biomarker discovery in predicting the response of esophageal cancer to neoadjuvant therapy: A multidimensional perspective. Pharmacol Ther 2024; 254:108591. [PMID: 38286161 DOI: 10.1016/j.pharmthera.2024.108591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/02/2023] [Accepted: 01/04/2024] [Indexed: 01/31/2024]
Abstract
Neoadjuvant chemoradiotherapy (NCRT) followed by surgery has been established as the standard treatment strategy for operable locally advanced esophageal cancer (EC). However, achieving pathologic complete response (pCR) or near pCR to NCRT is significantly associated with a considerable improvement in survival outcomes, while pCR patients may help organ preservation for patients by active surveillance to avoid planned surgery. Thus, there is an urgent need for improved biomarkers to predict EC chemoradiation response in research and clinical settings. Advances in multiple high-throughput technologies such as next-generation sequencing have facilitated the discovery of novel predictive biomarkers, specifically based on multi-omics data, including genomic/transcriptomic sequencings and proteomic/metabolomic mass spectra. The application of multi-omics data has shown the benefits in improving the understanding of underlying mechanisms of NCRT sensitivity/resistance in EC. Particularly, the prominent development of artificial intelligence (AI) has introduced a new direction in cancer research. The integration of multi-omics data has significantly advanced our knowledge of the disease and enabled the identification of valuable biomarkers for predicting treatment response from diverse dimension levels, especially with rapid advances in biotechnological and AI methodologies. Herein, we summarize the current status of research on the use of multi-omics technologies in predicting NCRT response for EC patients. Current limitations, challenges, and future perspectives of these multi-omics platforms will be addressed to assist in experimental designs and clinical use for further integrated analysis.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 15 West Changle Road, Xi'an, China
| | - Fada Guan
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Lawrence Bronk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 15 West Changle Road, Xi'an, China.
| |
Collapse
|
10
|
Abbas DB, Griffin M, Fahy EJ, Spielman AF, Guardino NJ, Pu A, Lintel H, Lorenz HP, Longaker MT, Wan DC. Establishing a Xenograft Model with CD-1 Nude Mice to Study Human Skin Wound Repair. Plast Reconstr Surg 2024; 153:121-128. [PMID: 36988644 DOI: 10.1097/prs.0000000000010465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
BACKGROUND A significant gap exists in the translatability of small-animal models to human subjects. One important factor is poor laboratory models involving human tissue. Thus, the authors have created a viable postnatal human skin xenograft model using athymic mice. METHODS Discarded human foreskins were collected following circumcision. All subcutaneous tissue was removed from these samples sterilely. Host CD-1 nude mice were then anesthetized, and dorsal skin was sterilized. A 1.2-cm-diameter, full-thickness section of dorsal skin was excised. The foreskin sample was then placed into the full-thickness defect in the host mice and sutured into place. Xenografts underwent dermal wounding using a 4-mm punch biopsy after engraftment. Xenografts were monitored for 14 days after wounding and then harvested. RESULTS At 14 days postoperatively, all mice survived the procedure. Grossly, the xenograft wounds showed formation of a human scar at postoperative day 14. Hematoxylin and eosin and Masson trichome staining confirmed scar formation in the wounded human skin. Using a novel artificial intelligence algorithm using picrosirius red staining, scar formation was confirmed in human wounded skin compared with the unwounded skin. Histologically, CD31 + immunostaining confirmed vascularization of the xenograft. The xenograft exclusively showed human collagen type I, CD26 + , and human nuclear antigen in the human scar without any staining of these human markers in the murine skin. CONCLUSION The proposed model demonstrates wound healing to be a local response from tissue resident human fibroblasts and allows for reproducible evaluation of human skin wound repair in a preclinical model. CLINICAL RELEVANCE STATEMENT Radiation-induced fibrosis is a widely prevalent clinical phenomenon without a well-defined treatment at this time. This study will help establish a small-animal model to better understand and develop novel therapeutics to treat irradiated human skin.
Collapse
Affiliation(s)
- Darren B Abbas
- From the Hagey Laboratory for Pediatric Regenerative Medicine
| | | | - Evan J Fahy
- From the Hagey Laboratory for Pediatric Regenerative Medicine
| | | | | | - Adrian Pu
- From the Hagey Laboratory for Pediatric Regenerative Medicine
| | - Hendrik Lintel
- From the Hagey Laboratory for Pediatric Regenerative Medicine
| | - H Peter Lorenz
- From the Hagey Laboratory for Pediatric Regenerative Medicine
| | - Michael T Longaker
- From the Hagey Laboratory for Pediatric Regenerative Medicine
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Derrick C Wan
- From the Hagey Laboratory for Pediatric Regenerative Medicine
| |
Collapse
|
11
|
Li L, Zhang P. Elevation of LEM Domain Containing 1 Predicts Poor Prognosis of NSCLC Patients and Triggers Malignant Stemness and Invasion of NSCLC Cells by Stimulating PI3K/AKT Pathway. Curr Mol Med 2024; 24:366-378. [PMID: 36967459 DOI: 10.2174/1566524023666230324135330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 12/16/2022] [Accepted: 01/09/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related death globally. LEM domain containing 1 (LEMD1) function has been identified in several cancers but not in NSCLC. OBJECTIVE This study aimed to investigate the LEMD1 function in NSCLC. METHODS NSCLC tissues were obtained from 66 patients, and LEMD1 expressions were measured using quantitative real-time PCR, immunohistochemical assay, and Western blot. Overall survival of NSCLC patients was estimated by the Kaplan-Meier method. Meanwhile, LEMD1 function and mechanism were assessed using Cell Counting Kit-8, 5-Ethynyl-2'-deoxyuridine analysis, Transwell, Sphere formation assay, and flow cytometry. Furthermore, LEMD1 function in vivo was evaluated by establishing a xenograft tumor model, hematoxylin-eosin staining, and immunohistochemical assay. RESULTS LEMD1 was highly expressed in NSCLC tissues and was interrelated to tumor differentiation, TNM stage, and lymph node metastasis of patients. Overall survival of NSCLC patients with high LEMD1 was found to be lower than that of patients with low LEMD1. Functionally, interference with LEMD1 restrained NSCLC cell proliferation, invasion, and stemness characteristics. Mechanistically, LEMD1 facilitated the malignant phenotype of NSCLC, and 740 Y-P reversed this impact, prompting that LEMD1 aggravated NSCLC by activating PI3K/AKT pathway. Furthermore, LEMD1 knockdown hindered NSCLC proliferation in vivo. Conclusion: LEMD1 accelerated NSCLC cell proliferation, invasion, and stemness characteristics via activating PI3K/AKT pathway.
Collapse
Affiliation(s)
- Li Li
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 402177, China
| | - Pei Zhang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 402177, China
| |
Collapse
|
12
|
Ding S, Liu J, Han X, Tang M. CRISPR/Cas9-Mediated Genome Editing in Cancer Therapy. Int J Mol Sci 2023; 24:16325. [PMID: 38003514 PMCID: PMC10671490 DOI: 10.3390/ijms242216325] [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: 10/08/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
The Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated protein 9 (CRISPR/Cas9) system, an RNA-based adaptive immune system found in bacteria and archaea, has catalyzed the development and application of a new generation of gene editing tools. Numerous studies have shown that this system can precisely target a wide range of human genes, including those associated with diseases such as cancer. In cancer research, the intricate genetic mutations in tumors have promoted extensive utilization of the CRISPR/Cas9 system due to its efficient and accurate gene editing capabilities. This includes improvements in Chimeric Antigen Receptor (CAR)-T-cell therapy, the establishment of tumor models, and gene and drug target screening. Such progress has propelled the investigation of cancer molecular mechanisms and the advancement of precision medicine. However, the therapeutic potential of genome editing remains underexplored, and lingering challenges could elevate the risk of additional genetic mutations. Here, we elucidate the fundamental principles of CRISPR/Cas9 gene editing and its practical applications in tumor research. We also briefly discuss the primary challenges faced by CRISPR technology and existing solutions, intending to enhance the efficacy of this gene editing therapy and shed light on the underlying mechanisms of tumors.
Collapse
Affiliation(s)
- Shuai Ding
- Department of Biochemistry and Molecular Biology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Jinfeng Liu
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Xin Han
- Department of Biochemistry and Molecular Biology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Mengfan Tang
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| |
Collapse
|
13
|
Yang X, Chen X, Zhang S, Fan W, Zhong C, Liu T, Cheng G, Zhu L, Liu Q, Xi Y, Tan W, Lin D, Wu C. Collagen 1-mediated CXCL1 secretion in tumor cells activates fibroblasts to promote radioresistance of esophageal cancer. Cell Rep 2023; 42:113270. [PMID: 37851572 DOI: 10.1016/j.celrep.2023.113270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/12/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023] Open
Abstract
Esophageal squamous-cell carcinoma (ESCC) is commonly treated with radiotherapy; however, radioresistance hinders its clinical effectiveness, and the underlying mechanism remains elusive. Here, we develop patient-derived xenografts (PDXs) from 19 patients with ESCC to investigate the mechanisms driving radioresistance. Using RNA sequencing, cytokine arrays, and single-cell RNA sequencing, we reveal an enrichment of cancer-associated fibroblast (CAF)-derived collagen type 1 (Col1) and tumor-cell-derived CXCL1 in non-responsive PDXs. Col1 not only promotes radioresistance by augmenting DNA repair capacity but also induces CXCL1 secretion in tumor cells. Additionally, CXCL1 further activates CAFs via the CXCR2-STAT3 pathway, establishing a positive feedback loop. Directly interfering with tumor-cell-derived CXCL1 or inhibiting the CXCL1-CXCR2 pathway effectively restores the radiosensitivity of radioresistant xenografts in vivo. Collectively, our study provides a comprehensive understanding of the molecular mechanisms underlying radioresistance and identifies potential targets to improve the efficacy of radiotherapy for ESCC.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Xinjie Chen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Wenyi Fan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100091, China; Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University (PKU), Beijing 100871, China
| | - Ce Zhong
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Guoyu Cheng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Liang Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Qingyi Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Yiyi Xi
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Wen Tan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; CAMS Oxford Institute, Chinese Academy of Medical Sciences, Beijing 100006, China.
| |
Collapse
|
14
|
Mahmoudian RA, Farshchian M, Golyan FF, Mahmoudian P, Alasti A, Moghimi V, Maftooh M, Khazaei M, Hassanian SM, Ferns GA, Mahaki H, Shahidsales S, Avan A. Preclinical tumor mouse models for studying esophageal cancer. Crit Rev Oncol Hematol 2023; 189:104068. [PMID: 37468084 DOI: 10.1016/j.critrevonc.2023.104068] [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: 05/10/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023] Open
Abstract
Preclinical models are extensively employed in cancer research because they can be manipulated in terms of their environment, genome, molecular biology, organ systems, and physical activity to mimic human behavior and conditions. The progress made in in vivo cancer research has resulted in significant advancements, enabling the creation of spontaneous, metastatic, and humanized mouse models. Most recently, the remarkable and extensive developments in genetic engineering, particularly the utilization of CRISPR/Cas9, transposable elements, epigenome modifications, and liquid biopsies, have further facilitated the design and development of numerous mouse models for studying cancer. In this review, we have elucidated the production and usage of current mouse models, such as xenografts, chemical-induced models, and genetically engineered mouse models (GEMMs), for studying esophageal cancer. Additionally, we have briefly discussed various gene-editing tools that could potentially be employed in the future to create mouse models specifically for esophageal cancer research.
Collapse
Affiliation(s)
- Reihaneh Alsadat Mahmoudian
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Farshchian
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Fatemeh Fardi Golyan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvaneh Mahmoudian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Alasti
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Moghimi
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq; Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
15
|
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
|
16
|
Liang M, Wang L, Xiao Y, Yang M, Mei C, Zhang Y, Shan H, Li D. Preclinical evaluation of a novel EGFR&c-Met bispecific near infrared probe for visualization of esophageal cancer and metastatic lymph nodes. Eur J Nucl Med Mol Imaging 2023; 50:2787-2801. [PMID: 37145165 DOI: 10.1007/s00259-023-06250-z] [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: 01/30/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
PURPOSE This study aimed to establish a near infrared fluorescent (NIRF) probe based on an EGFR&c-Met bispecific antibody for visualization of esophageal cancer (EC) and metastatic lymph nodes (mLNs). METHODS EGFR and c-Met expression were assessed by immunohistochemistry. EGFR&c-Met bispecific antibody EMB01 was labeled with IRDye800cw. The binding of EMB01-IR800 was assessed by enzyme linked immunosorbent assay, flow cytometry, and immunofluorescence. Subcutaneous tumors, orthotopic tumors, and patient-derived xenograft (PDX) were established for in vivo fluorescent imaging. PDX models using lymph nodes with or without metastasis were constructed to assess the performance of EMB01-IR800 in differential diagnosis of lymph nodes. RESULTS The prevalence of overexpressing EGFR or c-Met was significantly higher than single marker either in EC or corresponding mLNs. The bispecific probe EMB01-IR800 was successfully synthesized, with strong binding affinity. EMB01-IR800 showed strong cellular binding to both Kyse30 (EGFR overexpressing) and OE33 (c-Met overexpressing) cells. In vivo fluorescent imaging showed prominent EMB01-IR800 uptake in either Kyse30 or OE33 subcutaneous tumors. Likewise, EMB01-IR800 exhibited superior tumor enrichment in both thoracic orthotopic esophageal squamous cell carcinoma and abdominal orthotopic esophageal adenocarcinoma models. Moreover, EMB01-IR800 produced significantly higher fluorescence in patient-derived mLNs than in benign lymph nodes. CONCLUSION This study demonstrated the complementary overexpression of EGFR and c-Met in EC. Compared to single-target probes, the EGFR&c-Met bispecific NIRF probe can efficiently depict heterogeneous esophageal tumors and mLNs, which greatly increased the sensitivity of tumor and mLN identification.
Collapse
Affiliation(s)
- Mingzhu Liang
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Lizhu Wang
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Yitai Xiao
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Meilin Yang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Chaoming Mei
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Yaqin Zhang
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
- Center for Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
| | - Hong Shan
- Center for Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
| | - Dan Li
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
| |
Collapse
|
17
|
Hanson S, Dharan A, P. V. J, Pal S, Nair BG, Kar R, Mishra N. Paraptosis: a unique cell death mode for targeting cancer. Front Pharmacol 2023; 14:1159409. [PMID: 37397502 PMCID: PMC10308048 DOI: 10.3389/fphar.2023.1159409] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
Programmed cell death (PCD) is the universal process that maintains cellular homeostasis and regulates all living systems' development, health and disease. Out of all, apoptosis is one of the major PCDs that was found to play a crucial role in many disease conditions, including cancer. The cancer cells acquire the ability to escape apoptotic cell death, thereby increasing their resistance towards current therapies. This issue has led to the need to search for alternate forms of programmed cell death mechanisms. Paraptosis is an alternative cell death pathway characterized by vacuolation and damage to the endoplasmic reticulum and mitochondria. Many natural compounds and metallic complexes have been reported to induce paraptosis in cancer cell lines. Since the morphological and biochemical features of paraptosis are much different from apoptosis and other alternate PCDs, it is crucial to understand the different modulators governing it. In this review, we have highlighted the factors that trigger paraptosis and the role of specific modulators in mediating this alternative cell death pathway. Recent findings include the role of paraptosis in inducing anti-tumour T-cell immunity and other immunogenic responses against cancer. A significant role played by paraptosis in cancer has also scaled its importance in knowing its mechanism. The study of paraptosis in xenograft mice, zebrafish model, 3D cultures, and novel paraptosis-based prognostic model for low-grade glioma patients have led to the broad aspect and its potential involvement in the field of cancer therapy. The co-occurrence of different modes of cell death with photodynamic therapy and other combinatorial treatments in the tumour microenvironment are also summarized here. Finally, the growth, challenges, and future perspectives of paraptosis research in cancer are discussed in this review. Understanding this unique PCD pathway would help to develop potential therapy and combat chemo-resistance in various cancer.
Collapse
Affiliation(s)
- Sweata Hanson
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Aiswarya Dharan
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Jinsha P. V.
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Sanjay Pal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Bipin G. Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Rekha Kar
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, United States
| | - Nandita Mishra
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| |
Collapse
|
18
|
Kong M, Li Y, Wang K, Zhang S, Ji Y. Infantile hemangioma models: is the needle in a haystack? J Transl Med 2023; 21:308. [PMID: 37149592 PMCID: PMC10163722 DOI: 10.1186/s12967-023-04144-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/20/2023] [Indexed: 05/08/2023] Open
Abstract
Infantile hemangioma (IH) is the most prevalent benign vascular tumor in infants, with distinct disease stages and durations. Despite the fact that the majority of IHs can regress spontaneously, a small percentage can cause disfigurement or even be fatal. The mechanisms underlying the development of IH have not been fully elucidated. Establishing stable and reliable IH models provides a standardized experimental platform for elucidating its pathogenesis, thereby facilitating the development of new drugs and the identification of effective treatments. Common IH models include the cell suspension implantation model, the viral gene transfer model, the tissue block transplantation model, and the most recent three-dimensional (3D) microtumor model. This article summarizes the research progress and clinical utility of various IH models, as well as the benefits and drawbacks of each. Researchers should select distinct IH models based on their individual research objectives to achieve their anticipated experimental objectives, thereby increasing the clinical relevance of their findings.
Collapse
Affiliation(s)
- Meng Kong
- Division of Oncology, Department of Pediatric Surgery, West China Hospital, Sichuan University, #37# Guo-Xue-Xiang, Chengdu, 610041, China
| | - Yanan Li
- Division of Oncology, Department of Pediatric Surgery, West China Hospital, Sichuan University, #37# Guo-Xue-Xiang, Chengdu, 610041, China
| | - Kai Wang
- Division of Oncology, Department of Pediatric Surgery, West China Hospital, Sichuan University, #37# Guo-Xue-Xiang, Chengdu, 610041, China
| | - Shisong Zhang
- Department of Pediatric Surgery, Children's Hospital Affiliated to Shandong University, #23976# Jingshi Road, Jinan, 250022, China.
| | - Yi Ji
- Division of Oncology, Department of Pediatric Surgery, West China Hospital, Sichuan University, #37# Guo-Xue-Xiang, Chengdu, 610041, China.
| |
Collapse
|
19
|
Wang DP, Wu LH, Li R, He N, Zhang QY, Zhao CY, Jiang T. A Novel Aldisine Derivative Exhibits Potential Antitumor Effects by Targeting JAK/STAT3 Signaling. Mar Drugs 2023; 21:md21040218. [PMID: 37103357 PMCID: PMC10141377 DOI: 10.3390/md21040218] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
The JAK/STAT3 signaling pathway is aberrantly hyperactivated in many cancers, promoting cell proliferation, survival, invasiveness, and metastasis. Thus, inhibitors targeting JAK/STAT3 have enormous potential for cancer treatment. Herein, we modified aldisine derivatives by introducing the isothiouronium group, which can improve the antitumor activity of the compounds. We performed a high-throughput screen of 3157 compounds and identified compounds 11a, 11b, and 11c, which contain a pyrrole [2,3-c] azepine structure linked to an isothiouronium group through different lengths of carbon alkyl chains and significantly inhibited JAK/STAT3 activities. Further results showed that compound 11c exhibited the optimal antiproliferative activity and was a pan-JAKs inhibitor capable of inhibiting constitutive and IL-6-induced STAT3 activation. In addition, compound 11c influenced STAT3 downstream gene expression (Bcl-xl, C-Myc, and Cyclin D1) and induced the apoptosis of A549 and DU145 cells in a dose-dependent manner. The antitumor effects of 11c were further demonstrated in an in vivo subcutaneous tumor xenograft experiment with DU145 cells. Taken together, we designed and synthesized a novel small molecule JAKs inhibitor targeting the JAK/STAT3 signaling pathway, which has predicted therapeutic potential for JAK/STAT3 overactivated cancer treatment.
Collapse
Affiliation(s)
- Dong-Ping Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Li-Hong Wu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Rui Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Na He
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Qian-Yue Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Chen-Yang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Department of Cancer Biology, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
- Correspondence: (C.-Y.Z.); (T.J.)
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Correspondence: (C.-Y.Z.); (T.J.)
| |
Collapse
|
20
|
Chen S, Lin X, He R, Zhang W, Kang M, Xu R. PHLDA3 activated by BARX2 transcription, suppresses the malignant development of esophageal squamous cell carcinoma by downregulating PI3K/AKT levels. Exp Cell Res 2023; 426:113567. [PMID: 36965748 DOI: 10.1016/j.yexcr.2023.113567] [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: 05/23/2022] [Revised: 03/01/2023] [Accepted: 03/20/2023] [Indexed: 03/27/2023]
Abstract
BACKGROUND Low pleckstrin homology-like domain family A, member 3 (PHLDA3) expression has been reported to be associated with cancer specificity and disease-free survival in esophageal squamous cell carcinoma (ESCC), and was an independent predictor of postoperative recurrence. However, the specific mechanisms involved are still unclear. This paper aimed to explore the role and its mechanisms of PHLDA3 in ESCC. MATERIALS AND METHODS PHLDA3 and BarH-like homeobox 2 (BARX2) expressions in ESCC were predicted by Gene Expression Profiling Interactive Analysis (GEPIA) analysis and determined by quantitative real-time polymerase chain reaction (qRT-PCR) and western Blot. Western blot detected the expression of proteins associated with migration, angiogenesis and phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB/AKT) signaling pathway. The University of California Santa Cruz Genomics Institute (UCSC) database predicted that the relationship of BARX2 and PHLDA3 promoter and JASPAR identified the possible binding sites. Dual luciferase gene reporter verified PHLDA3 promoter activity, and the relationship of both was determined by chromatin immunoprecipitation (CHIP). Cell counting kit (CCK)-8, 5-ethynyl-2'-deoxyuridine (EDU) and colony formation were used to assess cell proliferation. Wound healing and transwell were used to detect cell migration and invasion ability. Tube formation assay was applied to assess angiogenesis. Mice were injected with transfected KYSE30 cells under the right axilla. Body weight and tumor volume and mass were recorded for each group of mice. Immunohistochemistry was performed to detect KI67 level in tumor tissues. RESULTS Both PHLDA3 and BARX2 were downregulated in ESCC. The upregulated PHLDA3 suppressed PI3K/AKT expression. In addition, BARX2 bound to the PHLDA3 promoter and transcriptionally activated PHLDA3. PHLDA3 overexpression inhibited ESCC cell proliferation, migration, invasion and angiogenesis, but this effect was reversed by BARX2 knockdown. In addition, BARX2 overexpression inhibited ESCC cell proliferation, migration, invasion and angiogenesis, but this effect was reversed by PHLDA3 knockdown. CONCLUSION PHLDA3 was transcriptionally activated by BARX2 and inhibited malignant progression of ESCC by downregulating PI3K/AKT levels.
Collapse
Affiliation(s)
- Shaogeng Chen
- Department of Thoracic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Xianzuan Lin
- Department of Thoracic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Rongqi He
- Department of Thoracic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Wanfei Zhang
- Department of Thoracic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Quanzhou, China
| | - Rongyu Xu
- Department of Thoracic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China.
| |
Collapse
|
21
|
Wu Q, Chen L, Huang X, Lin J, Gao J, Yang G, Wu Y, Wang C, Kang X, Yao Y, Wang Y, Xue M, Luan X, Chen X, Zhang Z, Sun S. A biomimetic nanoplatform for customized photothermal therapy of HNSCC evaluated on patient-derived xenograft models. Int J Oral Sci 2023; 15:9. [PMID: 36765028 PMCID: PMC9918549 DOI: 10.1038/s41368-022-00211-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/09/2022] [Accepted: 11/21/2022] [Indexed: 02/12/2023] Open
Abstract
Cancer cell membrane (CCM) derived nanotechnology functionalizes nanoparticles (NPs) to recognize homologous cells, exhibiting translational potential in accurate tumor therapy. However, these nanoplatforms are majorly generated from fixed cell lines and are typically evaluated in cell line-derived subcutaneous-xenografts (CDX), ignoring the tumor heterogeneity and differentiation from inter- and intra- individuals and microenvironments between heterotopic- and orthotopic-tumors, limiting the therapeutic efficiency of such nanoplatforms. Herein, various biomimetic nanoplatforms (CCM-modified gold@Carbon, i.e., Au@C-CCM) were fabricated by coating CCMs of head and neck squamous cell carcinoma (HNSCC) cell lines and patient-derived cells on the surface of Au@C NP. The generated Au@C-CCMs were evaluated on corresponding CDX, tongue orthotopic xenograft (TOX), immune-competent primary and distant tumor models, and patient-derived xenograft (PDX) models. The Au@C-CCM generates a photothermal conversion efficiency up to 44.2% for primary HNSCC therapy and induced immunotherapy to inhibit metastasis via photothermal therapy-induced immunogenic cell death. The homologous CCM endowed the nanoplatforms with optimal targeting properties for the highest therapeutic efficiency, far above those with mismatched CCMs, resulting in distinct tumor ablation and tumor growth inhibition in all four models. This work reinforces the feasibility of biomimetic NPs combining modular designed CMs and functional cores for customized treatment of HNSCC, can be further extended to other malignant tumors therapy.
Collapse
Affiliation(s)
- Qi Wu
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Lan Chen
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xiaojuan Huang
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China.
| | - Jiayi Lin
- grid.412540.60000 0001 2372 7462Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiamin Gao
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Guizhu Yang
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yaping Wu
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Chong Wang
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xindan Kang
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yanli Yao
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yujue Wang
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Mengzhu Xue
- grid.412523.30000 0004 0386 9086Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xin Luan
- grid.412540.60000 0001 2372 7462Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Chen
- grid.43169.390000 0001 0599 1243School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi’an Jiao Tong University, Xi’an, Shaanxi China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China.
| | - Shuyang Sun
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
22
|
Liu Z, Su R, Ahsan A, Liu C, Liao X, Tian D, Su M. Esophageal Squamous Cancer from 4NQO-Induced Mice Model: CNV Alterations. Int J Mol Sci 2022; 23:ijms232214304. [PMID: 36430789 PMCID: PMC9698903 DOI: 10.3390/ijms232214304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Squamous esophageal carcinoma is a common pathological type of esophageal carcinoma around the world. The prognosis of esophageal carcinoma is usually poor and diagnosed at late stages. Recently, research suggested that genomic instability occurred in esophageal cells during the development of esophageal squamous cell carcinoma (ESCC). Identifying prognostic and specific genomic characteristics, especially at the early hyperplasia stage, is critical. Mice were given 4-nitroquinoline 1-oxide (4NQO) with drinking water to induce esophageal cancer. The immortalized human esophageal epithelial cell line (NE2) was also treated with 4NQO. We performed histologic analyses, immunofluorescence, and immunohistochemical staining to detect DNA damage at different time points. Whole-exome sequencing was accomplished on the esophagus tissues at different pathological stages to detect single-nucleotide variants and copy number variation (CNV) in the genome. Our findings indicate that all mice were tumor-forming, and a series of changes from simple hyperplasia (ESSH) to intraepithelial neoplasia (IEN) to esophageal squamous cell carcinoma (ESCC) was seen at different times. The expression of γ-H2AX increased from ESSH to ESCC. In addition, mutations of the Muc4 gene were detected throughout the pathological stages. Furthermore, CNV burden appeared in the esophageal tissues from the beginning of ESSH and accumulated more in cancer with the deepening of the lesions. This study demonstrates that mutations caused by the early appearance of DNA damage may appear in the early stage of malignant tissue before the emergence of atypia. The detection of CNV and mutations of the Muc4 gene may be used as an ultra-early screening indicator for esophageal cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Su
- Correspondence: ; Fax: +86-0754-88900429
| |
Collapse
|
23
|
Advances in the development of chordoma models for drug discovery and precision medicine. Biochim Biophys Acta Rev Cancer 2022; 1877:188812. [DOI: 10.1016/j.bbcan.2022.188812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/31/2022] [Accepted: 09/28/2022] [Indexed: 12/24/2022]
|
24
|
Chemopreventive Properties of Black Raspberries and Strawberries in Esophageal Cancer Review. Antioxidants (Basel) 2022; 11:antiox11091815. [PMID: 36139889 PMCID: PMC9495642 DOI: 10.3390/antiox11091815] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/02/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
Esophageal cancer is one of the most fetal malignancies in the world. Esophageal squamous cell carcinoma (SCC) and esophageal adenocarcinoma (AC) are two main types of esophageal cancer and each with distinct epidemiological, etiological and histopathological characteristics. The continued global prevalence of tobacco use and alcohol consumption, coupled with limited intake of fresh fruits and vegetables, ensures that esophageal cancer will remain one of the major health threats. In addition to promoting quitting smoking and alcohol abuse, one of the strategies of cancer prevention is to identify foods, food components, or dietary patterns that can prevent or delay the onset of esophageal cancer. A food-based approach has the advantage of a complex of mixtures of bioactive components simultaneously targeting multiple processes in carcinogenesis. We have employed a preclinical rodent model of esophageal SCC to assess the effects of black raspberries (BRB) and strawberries. Our investigations demonstrate that BRB and strawberries are potent inhibitors of esophageal cancer. To prepare for this review, a literature search was performed to screen BRB and strawberries against esophageal cancer using electronic databases from PubMed, Science Direct and Google Scholar. Search was conducted covering the period from January 2000 to June 2022. Our present review has provided a systematic review about chemopreventive effects of BRB and strawberries in esophageal cancer by collecting and compiling diverse research findings from the above sources. In this review, we discussed the anti-tumor potentials of BRB and strawberries in esophageal SCC and esophageal AC separately. For each cancer type, we discuss animal models and research findings from both animal bioassays and human clinical studies. We also discuss the potential mechanisms of action of berries and their key bioactive components.
Collapse
|
25
|
Choi JH, Kang CM, Park JY. EGFR-targeted fluorescent imaging using the da Vinci® Firefly™ camera for gallbladder cancer. World J Surg Oncol 2022; 20:201. [PMID: 35701793 PMCID: PMC9199159 DOI: 10.1186/s12957-022-02675-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/05/2022] [Indexed: 11/29/2022] Open
Abstract
Background Fluorescent imaging may aid with the precise diagnosis and treatment of patients with gallbladder cancer. In this study, we sought to demonstrate whether the da Vinci® surgical system and Firefly™ camera could detect EGFR-targeted fluorescent images in orthotopic mouse models of gallbladder cancer. Methods An orthotopic mouse model of gallbladder cancer was created by injecting NOZ gallbladder cancer cells mixed with Matrigel into the gallbladder. In vivo imaging of subcutaneous and orthotopic gallbladder tumors was performed after the injection of DyLight 650- or 800-conjugated EGFR antibody. Results Western blotting, flow cytometry, and confocal microscopy showed the presence of EGFR in NOZ cells, but not in HEK293 cells. Subcutaneous NOZ cell tumors fluoresced after injection with fluorescent EGFR antibody, but subcutaneous HEK293 tumors did not. Fluorescent EGFR antibody made orthotopic NOZ tumors fluoresce, with an intensity stronger than that in the surrounding normal tissues. Histochemical examination confirmed the location of the tumors inside the gallbladder and adjacent liver parenchyma. Fluorescent signal was also detected in orthotopic gallbladder tumors with Firefly™ camera. Conclusion Our study showed that fluorescent EGFR antibodies and the Firefly camera in the da Vinci system can detect fluorescing gallbladder tumors, which demonstrates their potential use for molecular imaging-based prevision surgery in the near future.
Collapse
Affiliation(s)
- Jung Ha Choi
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Chang Moo Kang
- Division of HBP Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Pancreatobiliary Cancer Center, Yonsei Cancer Center, and Yonsei Institute of Gastroenterology, Severance Hospital, Seoul, South Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea. .,Pancreatobiliary Cancer Center, Yonsei Cancer Center, and Yonsei Institute of Gastroenterology, Severance Hospital, Seoul, South Korea.
| |
Collapse
|
26
|
|
27
|
PTK7, a Catalytically Inactive Receptor Tyrosine Kinase, Increases Oncogenic Phenotypes in Xenograft Tumors of Esophageal Squamous Cell Carcinoma KYSE-30 Cells. Int J Mol Sci 2022; 23:ijms23042391. [PMID: 35216506 PMCID: PMC8876147 DOI: 10.3390/ijms23042391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 12/24/2022] Open
Abstract
Protein tyrosine kinase 7 (PTK7), a catalytically defective receptor protein tyrosine kinase, is upregulated in tumor tissues and cell lines of esophageal squamous cell carcinoma (ESCC). We showed that PTK7 plays an oncogenic role in various ESCC cell lines. However, its role as an oncogene has not been demonstrated in vivo. Here, we examined the influence of PTK7 on the tumorigenic potential of ESCC KYSE-30 cells, which are known to establish xenograft tumors. Overexpression of PTK7 enhanced the proliferation, adhesion, wound healing, and migration of KYSE-30 cells, and these effects were reversed by the knockdown of PTK7. PTK7 overexpression and knockdown, respectively, increased and decreased the tyrosine phosphorylation of cellular proteins and the phosphorylation of ERK, AKT, and FAK, which are important for cell proliferation, survival, adhesion, and migration. Additionally, PTK7 overexpression and silencing, respectively, increased and decreased the weight, volume, and number of Ki-67-positive proliferating cells in xenograft tumors of KYSE-30 cells. Therefore, we propose that PTK7 plays an important role in the tumorigenesis of ESCC cells in vivo and is a potential therapeutic target for ESCC.
Collapse
|
28
|
Leung PKK, Lee LCC, Ip TKY, Liu HW, Yiu SM, Lee NP, Lo KKW. Luminescent rhenium(I) perfluorobiphenyl complexes as site-specific labels for peptides to afford photofunctional bioconjugates. Chem Commun (Camb) 2021; 57:11256-11259. [PMID: 34633395 DOI: 10.1039/d1cc04740j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We report herein new luminescent rhenium(I) perfluorobiphenyl complexes that reacted specifically with the cysteine residue of the π-clamp sequence (FCPF) to afford novel peptide-based imaging reagents, photosensitisers for singlet oxygen and enzyme sensors.
Collapse
Affiliation(s)
- Peter Kam-Keung Leung
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Tiffany Ka-Yan Ip
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Hua-Wei Liu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Shek-Man Yiu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Nikki P Lee
- Department of Surgery, The University of Hong Kong, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China. .,State Key Laboratory of Terahertz and Millimetre Waves, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.,Centre of Functional Photonics, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China
| |
Collapse
|
29
|
Wang F, Gu T, Chen Y, Chen Y, Xiong D, Zhu Y. Long non-coding RNA SOX21-AS1 modulates lung cancer progress upon microRNA miR-24-3p/PIM2 axis. Bioengineered 2021; 12:6724-6737. [PMID: 34511042 PMCID: PMC8806559 DOI: 10.1080/21655979.2021.1955578] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is a lethal cancer that threatens human health. Several studies have demonstrated the role of long non-coding RNAs (lncRNAs) in lung cancer. SOX21-AS1 is a newly discovered oncogenic lncRNA, but its molecular mechanism in lung cancer is not known. Here, the levels of SOX21-AS1, miR-24-3p, and PIM2 were examined in lung cancer and normal tissues. The relationships between miR-24-3p and SOX21-AS1 or PIM2 were predicted using bioinformatics tools and confirmed using a luciferase reporter assays. Colony formation, MTT, flow cytometry, and transwell assays were conducted to analyze cell proliferation, apoptosis, migration, and invasion abilities, respectively. Western blotting was used to measure PIM2 expression levels in cancer tissues and cells. SOX21-AS1 expression levels were high in lung cancer tissues and cells. In contrast, the amount of miR-24-3p bound to SOX21-AS1 was relatively low in cancerous tissues and cells. The knockdown of SOX21-AS1 decreased cell proliferation, activated apoptosis, and promoted cell migration and invasion. These effects were abolished by miR-24-3p inhibition. The oncogenic function of SOX21-AS1 mediated through targeting miR-24-3p was also demonstrated in animal models. PIM2 was targeted by miR-24-3p and showed increased levels in tumor tissues and cells. Furthermore, miR-24-3p overexpression inhibited the proliferation and promoted the apoptosis of lung cancer cells. In lung cancer cells, SOX21-AS1 negatively modulated the miR-24-3p/PIM2 axis to facilitate their proliferation, migration, and invasion. These findings offer a novel idea for future research on treating lung cancer at the molecular level.
Collapse
Affiliation(s)
- Fengfeng Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Respiration, People's Hospital of Jingjiang, Jingjiang, Jiangsu, China
| | - Tengfei Gu
- Department of Anesthesiology, People's Hospital of Lianshui County, Lianshui, Jiangsu, China
| | - Yao Chen
- Department of Respiration, People's Hospital of Jingjiang, Jingjiang, Jiangsu, China
| | - Yu Chen
- Department of Respiration, People's Hospital of Jingjiang, Jingjiang, Jiangsu, China
| | - Dan Xiong
- Department of Respiration, People's Hospital of Jingjiang, Jingjiang, Jiangsu, China
| | - Yehan Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
30
|
Choi JH, Park JY. Insulin-Like Growth Factor-1 Receptor Targeted Fluorescent Imaging for Gallbladder Cancer in Orthotopic Mouse Models. Gut Liver 2021; 16:606-612. [PMID: 34462395 PMCID: PMC9289833 DOI: 10.5009/gnl210164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/22/2021] [Accepted: 07/13/2021] [Indexed: 11/06/2022] Open
Abstract
Background/Aims Gallbladder cancer is fatal, but fluorescent imaging technology can facilitate timely diagnosis and improve patient outcomes. Fluorophore-conjugated insulin-like growth factor-1 receptor (IGF-1R) targeted antibodies were used to visualize gallbladder cancer in orthotopic tumor mouse models. Methods Western blotting, flow cytometric analysis, and confocal microscopy detected the expression of IGF-1R in SNU-308, SNU-478, and SNU-1196 bile duct cancer cells. In vivo imaging of SNU-478 and SNU-1196 subcutaneous tumors and orthotopic gallbladder tumor models of SNU-478 were performed after injection with DyLight 650-conjugated IGF-1R antibody. Results Western blotting and flow cytometric analysis showed that IGF-1R was expressed in bile duct cancer cells, and confocal microscopy demonstrated that IGF-1R antibody was able to bind to IGF-1R on the cell membrane. Fluorescent IGF-1R antibody injected into the mouse tail vein made subcutaneous tumors and orthotopic tumors become fluorescent. The intensity of fluorescence from the tumor was stronger than that from surrounding normal tissues. Histochemical examination confirmed that the tumor was located inside the gallbladder and adjacent liver parenchyma of mice. Conclusions Our study showed that a fluorescent IGF-1R-targeted antibody could help detect gallbladder tumors. Tumor-specific imaging technology can be applied to endoscopy, laparoscopy, and robotic surgery for better management of gallbladder cancer.
Collapse
Affiliation(s)
- Jung Ha Choi
- Division of Gastroenterology, Department of Internal Medicine and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
31
|
LaRocca CJ, Salzwedel AO, Sato-Dahlman M, Romanenko MV, Andrade R, Davydova J, Yamamoto M. Interferon Alpha-Expressing Oncolytic Adenovirus for Treatment of Esophageal Adenocarcinoma. Ann Surg Oncol 2021; 28:8556-8564. [PMID: 34324109 DOI: 10.1245/s10434-021-10382-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 06/11/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Esophageal adenocarcinoma (EAC) has increased in incidence in Western countries, and its poor prognosis necessitates the development of novel therapeutics. We previously reported the potential of conditionally replicative adenoviruses (CRAd) as a novel therapeutic treatment for this disease. To further augment the therapeutic effectiveness of our cyclooxygenase-2 (Cox2) controlled CRAd in EAC, we inserted an interferon alpha (IFN) transgene into the viral genome that is expressed upon viral replication. In this manuscript, we analyze the cytotoxic and oncolytic effects of an IFN-expressing oncolytic adenovirus in EAC and the role of the Cox2 promoter in providing for selective replication in human tissues. METHODS An infectivity-enhanced IFN-expressing CRAd (5/3 Cox2 CRAd ΔE3 ADP IFN) and other control viruses were first tested in vitro with cell lines. For the in vivo study, EAC xenografts in nude mice were treated with a single intratumoral dose of virus. An ex vivo analysis with live tissue slices was conducted using surgically resected EAC patient specimens. RESULTS Expression of IFN significantly enhanced the cytotoxic and oncolytic effect of a Cox2-promoter controlled CRAd. This virus showed significant tumor growth suppression in a xenograft model. Furthermore, in human EAC samples, the promoter-controlled virus demonstrated selective replication in cancerous tissues, leaving normal esophageal tissue unaffected. CONCLUSION An IFN-expressing CRAd driven by the Cox2 promoter has strong oncolytic effects as well as cancer-specific replication. Our novel vector possesses critical characteristics that make it a potential candidate for clinical translation to treat EAC.
Collapse
Affiliation(s)
- Christopher J LaRocca
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA. .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| | | | - Mizuho Sato-Dahlman
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | | | - Rafael Andrade
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Julia Davydova
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,Institute of Molecular Virology, University of Minnesota, Minneapolis, MN, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,Institute of Molecular Virology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
32
|
Mahmoudian RA, Farshchian M, Abbaszadegan MR. Genetically engineered mouse models of esophageal cancer. Exp Cell Res 2021; 406:112757. [PMID: 34331909 DOI: 10.1016/j.yexcr.2021.112757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/10/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Esophageal cancer is the most common cause of cancer-related death worldwide with a diverse geographical distribution, poor prognosis, and diagnosis in advanced stages of the disease. Identification of the mechanisms involved in esophageal cancer development is evaluative to improve outcomes for patients. Genetically engineered mouse models (GEMMs) of cancer provide the physiologic, molecular, and histologic features of the human tumors to determine the pathogenesis and treatments for cancer, hence exhibiting a source of tremendous potential for oncology research. The advancement of cancer modeling in mice has improved to the extent that researchers can observe and manipulate the disease process in a specific manner. Despite the significant differences between mice and humans, mice can be great models for human oncology researches due to similarities between them at the molecular and physiological levels. Due to most of the existing esophageal cancer GEMMs do not propose an ideal system for pathogenesis of the disease, genetic risks, and microenvironment exposure, so identification of challenges in GEM modeling and well-developed technologies are required to obtain the most value for patients. In this review, we describe the biology of human and mouse, followed by the exciting esophageal cancer mouse models with a discussion of applicability and challenges of these models for generating new GEMMs in future studies.
Collapse
Affiliation(s)
| | - Moein Farshchian
- Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi, Mashhad, Iran.
| | | |
Collapse
|
33
|
Yang CS, Chen XL. Research on esophageal cancer: With personal perspectives from studies in China and Kenya. Int J Cancer 2021; 149:264-276. [PMID: 33270917 PMCID: PMC8141013 DOI: 10.1002/ijc.33421] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
The most common form of esophageal cancer (EC), esophageal squamous cell carcinoma (ESCC), is prevalent in many unindustrialized societies, among people with lower socioeconomic status and those who frequently use tobacco and alcohol. In some areas, ESCC mortality ranked top among all cancer. In this review, we begin with discussions of the extensive research on EC in Linxian in northern China that started 60 years ago and the recent studies in Kenya from our personal perspectives. Based on the results obtained from these studies and information from the literature, we summarize our current understanding about the risk factors for ESCC including lifestyle factors (smoking, alcohol, consumption of food and beverages at high temperature and other unhealthy habits), poor diet and nutritional insufficiencies and genetic susceptibility. Elimination or minimization of these environmental risk factors, as well as early detection and treatment of precancerous lesions, would be effective means for the prevention of ESCC. Current knowledge of molecular alterations in ESCC (gene mutations, hypermethylation and amplification or overexpression), as well as treatment of ESCC and the potential of targeted therapy, are also discussed. Finally, we propose effective approaches for the prevention of ESCC by adapting a healthy lifestyle, including a healthy diet that would also prevent other diseases. Community outreach, public education and international collaboration are important for achieving this public health goal.
Collapse
Affiliation(s)
- Chung S. Yang
- Department of Chemical BiologyErnest Mario School of Pharmacy, Rutgers, The State University of New JerseyPiscatawayNew JerseyUSA
| | - Xiaoxin Luke Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
34
|
Huang H, Yi JK, Lim SG, Park S, Zhang H, Kim E, Jang S, Lee MH, Liu K, Kim KR, Kim EK, Lee Y, Kim SH, Ryoo ZY, Kim MO. Costunolide Induces Apoptosis via the Reactive Oxygen Species and Protein Kinase B Pathway in Oral Cancer Cells. Int J Mol Sci 2021; 22:7509. [PMID: 34299129 PMCID: PMC8305390 DOI: 10.3390/ijms22147509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/23/2021] [Accepted: 07/02/2021] [Indexed: 12/28/2022] Open
Abstract
Oral cancer (OC) has been attracted research attention in recent years as result of its high morbidity and mortality. Costunolide (CTD) possesses potential anticancer and bioactive abilities that have been confirmed in several types of cancers. However, its effects on oral cancer remain unclear. This study investigated the potential anticancer ability and underlying mechanisms of CTD in OC in vivo and in vitro. Cell viability and anchorage-independent colony formation assays were performed to examine the antigrowth effects of CTD on OC cells; assessments for migration and invasion of OC cells were conducted by transwell; Cell cycle and apoptosis were investigated by flow cytometry and verified by immunoblotting. The results revealed that CTD suppressed the proliferation, migration and invasion of oral cancer cells effectively and induced cell cycle arrest and apoptosis; regarding the mechanism, CTD bound to AKT directly by binding assay and repressed AKT activities through kinase assay, which thereby downregulating the downstream of AKT. Furthermore, CTD remarkably promotes the generation of reactive oxygen species by flow cytometry assay, leading to cell apoptosis. Notably, CTD strongly suppresses cell-derived xenograft OC tumor growth in an in vivo mouse model. In conclusion, our results suggested that costunolide might prevent progression of OC and promise to be a novel AKT inhibitor.
Collapse
Affiliation(s)
- Hai Huang
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju 37224, Korea; (H.H.); (H.Z.); (E.K.)
| | - Jun-Koo Yi
- Gyeongbuk Livestock Research Institute, Yeongju 36052, Korea;
| | - Su-Geun Lim
- School of Life Science, Kyungpook National University, Daegu 41566, Korea; (S.-G.L.); (S.P.); (S.J.)
| | - Sijun Park
- School of Life Science, Kyungpook National University, Daegu 41566, Korea; (S.-G.L.); (S.P.); (S.J.)
| | - Haibo Zhang
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju 37224, Korea; (H.H.); (H.Z.); (E.K.)
| | - Eungyung Kim
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju 37224, Korea; (H.H.); (H.Z.); (E.K.)
| | - Soyoung Jang
- School of Life Science, Kyungpook National University, Daegu 41566, Korea; (S.-G.L.); (S.P.); (S.J.)
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju 58245, Korea;
| | - Kangdong Liu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450008, China;
| | - Ki-Rim Kim
- Department of Dental Hygiene, Kyungpook National University, Sangju 37224, Korea; (K.-R.K.); (E.-K.K.)
| | - Eun-Kyong Kim
- Department of Dental Hygiene, Kyungpook National University, Sangju 37224, Korea; (K.-R.K.); (E.-K.K.)
| | - Youngkyun Lee
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu 41566, Korea;
| | - Sung-Hyun Kim
- Department of Bio-Medical Analysis, Korea Polytechnic College, Chungnam 34134, Korea;
| | - Zae-Young Ryoo
- Gyeongbuk Livestock Research Institute, Yeongju 36052, Korea;
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju 37224, Korea; (H.H.); (H.Z.); (E.K.)
| |
Collapse
|
35
|
Lan T, Xue X, Dunmall LC, Miao J, Wang Y. Patient-derived xenograft: a developing tool for screening biomarkers and potential therapeutic targets for human esophageal cancers. Aging (Albany NY) 2021; 13:12273-12293. [PMID: 33903283 PMCID: PMC8109069 DOI: 10.18632/aging.202934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/23/2021] [Indexed: 04/15/2023]
Abstract
Esophageal cancer (EC) represents a human malignancy, diagnosed often at the advanced stage of cancer and resulting in high morbidity and mortality. The development of precision medicine allows for the identification of more personalized therapeutic strategies to improve cancer treatment. By implanting primary cancer tissues into immunodeficient mice for expansion, patient-derived xenograft (PDX) models largely maintain similar histological and genetic representations naturally found in patients' tumor cells. PDX models of EC (EC-PDX) provide fine platforms to investigate the tumor microenvironment, tumor genomic heterogeneity, and tumor response to chemoradiotherapy, which are necessary for new drug discovery to combat EC in addition to optimization of current therapeutic strategies for EC. In this review, we summarize the methods used for establishing EC-PDX models and investigate the utilities of EC-PDX in screening predictive biomarkers and potential therapeutic targets. The challenge of this promising research tool is also discussed.
Collapse
Affiliation(s)
- Tianfeng Lan
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xia Xue
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
- The Academy of Medical Science, Precision Medicine Center of the Second Affiliated Hospital of Zhengzhou University, Zhengzhou University, Henan, P.R. China
| | - Louisa Chard Dunmall
- Centre for Cancer Biomarkers and Biotherapeuitcs, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jinxin Miao
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, Henan, P.R. China
| | - Yaohe Wang
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
- Centre for Cancer Biomarkers and Biotherapeuitcs, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
36
|
Zhao J, Wang R, Zhang J, Zhao Y, Qiao S, Crouzier T, Yan H, Tian W. A novel 4D cell culture mimicking stomach peristalsis altered gastric cancer spheroids growth and malignance. Biofabrication 2021; 13. [PMID: 33836517 DOI: 10.1088/1758-5090/abf6bf] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/09/2021] [Indexed: 11/12/2022]
Abstract
In vitrocancer models that can largely mimic thein vivomicroenvironment are crucial for conducting more accurate research. Models of three-dimensional (3D) culture that can mimic some aspects of cancer microenvironment or cancer biopsies that can adequately represent tumor heterogeneity are intensely used currently. Those models still lack the dynamic stress stimuli in gastric carcinoma exposed to stomach peristalsisin vivo. This study leveraged a lab-developed four-dimensional (4D) culture model by a magnetic responsive alginate-based hydrogel to rotating magnets that can mimic stress stimuli in gastric cancer (GC). We used the 4D model to culture human GC cell line AGS and SGC7901, cells at the primary and metastasis stage. We revealed the 4D model altered the cancer cell growth kinetics mechanistically by alteringPCNAandp53expression compared to the 3D culture that lacks stress stimuli. We found the 4D model altered the cancer spheroids stemness as evidenced by enhanced cancer stem cells (CD44) marker expression in AGS spheroids but the expression was dampened in SGC7901 cells. We examined the multi-drug resistance (MDR1) marker expression and found the 4D model dampened the MDR1 expression in SGC7901 cell spheroids, but not in spheroids of AGS cells. Such a model provides the stomach peristalsis mimic and is promising for conducting basic or translational GC-associated research, drug screening, and culturing patient gastric biopsies to tailor the therapeutic strategies in precision medicine.
Collapse
Affiliation(s)
- Juzhi Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| | - Ruiqi Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| | - Jinyu Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| | - Yufang Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| | - Shupei Qiao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| | - Thomas Crouzier
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden.,AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Hongji Yan
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden.,AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Weiming Tian
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| |
Collapse
|
37
|
Chen X, Shen C, Wei Z, Zhang R, Wang Y, Jiang L, Chen K, Qiu S, Zhang Y, Zhang T, Chen B, Xu Y, Feng Q, Huang J, Zhong Z, Li H, Che G, Xiao K. Patient-derived non-small cell lung cancer xenograft mirrors complex tumor heterogeneity. Cancer Biol Med 2021; 18:184-198. [PMID: 33628593 PMCID: PMC7877179 DOI: 10.20892/j.issn.2095-3941.2020.0012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 06/28/2020] [Indexed: 02/05/2023] Open
Abstract
Objective Patient-derived xenograft (PDX) models have shown great promise in preclinical and translational applications, but their consistency with primary tumors in phenotypic, genetic, and pharmacodynamic heterogeneity has not been well-studied. This study aimed to establish a PDX repository for non-small cell lung cancer (NSCLC) and to further elucidate whether it could preserve the heterogeneity within and between tumors in patients. Methods A total of 75 surgically resected NSCLC specimens were implanted into immunodeficient NOD/SCID mice. Based on the successful establishment of the NSCLC PDX model, we compared the expressions of vimentin, Ki67, EGFR, and PD-L1 proteins between cancer tissues and PDX models using hematoxylin and eosin staining and immunohistochemical staining. In addition, we detected whole gene expression profiling between primary tumors and PDX generations. We also performed whole exome sequencing (WES) analysis in 17 first generation xenografts to further assess whether PDXs retained the patient heterogeneities. Finally, paclitaxel, cisplatin, doxorubicin, atezolizumab, afatininb, and AZD4547 were used to evaluate the responses of PDX models to the standard-of-care agents. Results A large collection of serially transplantable PDX models for NSCLC were successfully developed. The histology and pathological immunohistochemistry of PDX xenografts were consistent with the patients' tumor samples. WES and RNA-seq further confirmed that PDX accurately replicated the molecular heterogeneities of primary tumors. Similar to clinical patients, PDX models responded differentially to the standard-of-care treatment, including chemo-, targeted- and immuno-therapeutics. Conclusions Our established PDX models of NSCLC faithfully reproduced the molecular, histopathological, and therapeutic characteristics, as well as the corresponding tumor heterogeneities, which provides a clinically relevant platform for drug screening, biomarker discovery, and translational research.
Collapse
Affiliation(s)
- Xuanming Chen
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Cheng Shen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Zhe Wei
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Rui Zhang
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Yongsheng Wang
- GCP Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Ke Chen
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Shuang Qiu
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Yuanli Zhang
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Ting Zhang
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
- Laboratory of Nonhuman Primate Disease Modeling Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Bin Chen
- Center for Infectious Diseases, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yanjun Xu
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Qiyi Feng
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Jinxing Huang
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Zhihui Zhong
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
- Laboratory of Nonhuman Primate Disease Modeling Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Hongxia Li
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Guowei Che
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Kai Xiao
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| |
Collapse
|
38
|
Li H, Lv J, Guo J, Wang S, Liu S, Ma Y, Liang Z, Wang Y, Qi W, Qiu W. 5-Fluorouracil enhances the chemosensitivity of gastric cancer to TRAIL via inhibition of the MAPK pathway. Biochem Biophys Res Commun 2021; 540:108-115. [PMID: 33476960 DOI: 10.1016/j.bbrc.2021.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 01/04/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has the ability to selectively trigger cancer cell apoptosis and can be used as a target for tumor therapy. However, gastric cancer cells are usually insensitive to TRAIL so reducing this drug resistance may improve the treatment of gastric cancer. In this study, we used Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) experiments to determine the effects of 5-fluorouracil (5-FU) and TRAIL on the proliferation of gastric cancer cells. An Annexin V/propidium iodide (PI) staining experiment was used to detect apoptosis, and Western blotting was used to analyze the expression levels of apoptosis-related proteins and mitogen-activated protein kinase (MAPK) pathway proteins. The antitumor effects of 5-FU and TRAIL were verified in vivo using a nude mouse tumorigenesis experiment, and a TUNEL assay was performed to evaluate apoptosis in tumor tissue from the nude mice. We found the combination of 5-FU and TRAIL had a greater inhibitory effect on the proliferation of gastric cancer cells than 5-FU or TRAIL alone both in vivo and in vitro. 5-FU enhanced TRAIL-induced gastric cancer cell apoptosis by inactivating the MAPK pathway. Overall, our analysis firstly provided new insights into the role of 5-FU in increasing sensitivity to TRAIL. 5-FU can be used as a sensitizer for TRAIL, and its administration is a potential strategy for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Hui Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Jing Lv
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Jing Guo
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Shihai Liu
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Yingji Ma
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Zhiwei Liang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Yunyun Wang
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Weiwei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China.
| | - Wensheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China.
| |
Collapse
|
39
|
Bai Y, Zhang H, Wang Y, Zhu L, Shi T, Wei H, Xiao J, Zhang Y, Wang Z. Novel Oxovanadium Complex VO(hntdtsc)(NPIP): Anticancer Activity and Mechanism of Action on HeLa Cells. Front Pharmacol 2021; 11:608218. [PMID: 33628179 PMCID: PMC7897675 DOI: 10.3389/fphar.2020.608218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 12/21/2020] [Indexed: 01/17/2023] Open
Abstract
Oxovanadium complexes, particularly vanadyl (IV) derivatives with hybrid ligands of Schiff base and polypyridyl, have been demonstrated to possess great anticancerous therapeutic efficacy. However, most of the studies on the activity of these oxovanadium complexes have mainly focused on in vitro studies, and animal studies in vivo are extremely scarce. Based on the antitumor test results of four novel oxovanadium complexes in our previous work, this work further conducted a comprehensive antitumor activity study in vitro and in vivo on VO(hntdtsc)(NPIP), which owned the strongest inhibitory activity in vitro on multiple tumor cell proliferation. The cellular mechanism study suggested that VO(hntdtsc)(NPIP) inhibited the cell proliferation via arresting the cell cycle at G0/G1 phase through the p16-cyclin D1-CDK4-p-Rb pathway and inducing cell apoptosis through mitochondrial-dependent apoptosis pathway on HeLa cells. Inconsistent with the effects in vitro, VO(hntdtsc)(NPIP) significantly inhibited the growth of tumor and induced the apoptosis of cancer cells in mice xenograft models according to the results of nude mice in vivo image detection, H&E pathological examination, and immunohistochemical detection of p16/Ki-67 protein expression. Collectively, all the results, particularly studies in vivo, demonstrated that VO(hntdtsc)(NPIP) hold a potential to be the lead compound and further to be an anticervical cancer drug.
Collapse
Affiliation(s)
- Yinliang Bai
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yali Wang
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, China
| | - Longqing Zhu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Tao Shi
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hangzhi Wei
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Jiyuan Xiao
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, China
| | - Youcheng Zhang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhen Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
40
|
Liu Q, He J, Zhou X, Han M, Li J, Liu C, Yuan H. ACP-5862 suppresses esophageal squamous cell carcinoma growth through inducing apoptosis via activation of endoplasmic reticulum stress and ROS production. Biochem Biophys Res Commun 2021; 534:995-1002. [PMID: 33168189 DOI: 10.1016/j.bbrc.2020.10.052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/17/2020] [Indexed: 01/14/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common type of human oral malignancy with poor survival. Presently, it is necessary to find new and effective drugs for clinical therapy. This study aimed to identify the potential anti-tumor effects of ACP-5862, a major metabolite of acalabrutinib, on human ESCC progression, and to reveal the underlying mechanisms. Our findings suggested that ACP-5862 treatments markedly reduced the cell proliferation of ESCC cell lines in a time- and dose-dependent manner, while had no significant cytotoxicity to normal cells. Cell cycle arrest in G2/M phase was markedly induced by ACP-5862 in ESCC cells. Furthermore, apoptosis and endoplasmic reticulum (ER) stress were detected in ESCC cells treated with ACP-5862. Intriguingly, ACP-5862-induced apoptotic cell death was partly dependent on ER stress. Moreover, reactive oxygen species (ROS) was greatly triggered in ACP-5862-incubated ESCC cells, which was closely involved in apoptosis and ER stress mediated by ACP-5862. In addition, we showed that the expression of nuclear factor-erythroid 2-related factor-2 (Nrf-2) was considerably reduced in ACP-5862-treated cells. Importantly, ACP-5862 combined with Nrf-2 knockdown could further induce apoptosis and ER stress in ESCC cells compared with the ACP-5862 single group. Animal studies confirmed that repressing Nrf-2 promoted the anti-tumor effect of ACP-5862 on ESCC growth. Taken together, these findings demonstrated that ACP-5862 exerted anti-cancer effects on ESCC through inducing ER stress-mediated apoptosis via the ROS production. Meanwhile, ACP-5862 co-treated with Nrf-2 inhibitors may supply new and effective therapeutic strategies for ESCC treatment in future.
Collapse
Affiliation(s)
- Qiong Liu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Jingjing He
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Xuejun Zhou
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Mingkun Han
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Jianhui Li
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China; Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital Sanya 572000, China
| | - Chenqing Liu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Hu Yuan
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China.
| |
Collapse
|
41
|
Qin B, Dong M, Wang Z, Wan J, Xie Y, Jiao Y, Yan D. Long non‑coding RNA CASC15 facilitates esophageal squamous cell carcinoma tumorigenesis via decreasing SIM2 stability via FTO‑mediated demethylation. Oncol Rep 2020; 45:1059-1071. [PMID: 33650646 PMCID: PMC7860005 DOI: 10.3892/or.2020.7917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/27/2020] [Indexed: 01/18/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are involved in the regulation of esophageal squamous cell carcinoma (ESCC) progression. However, the function and mechanism of lncRNA cancer susceptibility candidate 15 (CASC15) are poorly defined. In the present study, tumor and normal adjacent tissues were collected from 45 patients with ESCC. Expression levels of CASC15, fat mass and obesity-associated (FTO) protein and single-minded 2 (SIM2) were examined via reverse transcription-quantitative PCR and western blot assays. Cell proliferation and apoptosis were evaluated via MTT, flow cytometry and caspase-3 activity assays, respectively. Additionally, an ESCC mouse xenograft model was used to assess the function of CASC15 in vivo. The interaction between FTO and CASC15/SIM2 was analyzed via RNA immunoprecipitation and RNA pull-down assays. The results revealed that CASC15 expression was elevated in ESCC tissues, and patients with ESCC exhibiting high CASC15 expression had a poor prognosis. CASC15-knockdown inhibited ESCC cell proliferation and facilitated apoptosis. Additionally, CASC15-knockdown decreased the growth of ESCC xenograft tumors. CASC15 decreased SIM2 stability via FTO-mediated demethylation. Additionally, FTO loss markedly weakened CASC15-mediated pro-proliferative and anti-apoptotic effects in ESCC cells. SIM2 downregulation weakened the effect of CASC15-knockdown on cell proliferation and inhibited the increase of the apoptotic rate and caspase-3 activity induced by CASC15 depletion in ESCC cells. In conclusion, CASC15 promoted ESCC tumorigenesis by decreasing SIM2 stability via FTO-mediated demethylation.
Collapse
Affiliation(s)
- Bo Qin
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Meng Dong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhengyang Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jiajia Wan
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yingying Xie
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yi Jiao
- Yongcheng Coal & Electricity Holding Group Co., Ltd., Shangqiu, Henan 476000, P.R. China
| | - Dan Yan
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
42
|
TAp63α targeting of Lgr5 mediates colorectal cancer stem cell properties and sulforaphane inhibition. Oncogenesis 2020; 9:89. [PMID: 33040081 PMCID: PMC7548006 DOI: 10.1038/s41389-020-00273-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) have an established role in cancer progression and therapeutic resistance. The p63 proteins are important transcription factors which belong to the p53 family, but their function and mechanism in CSCs remain elusive. Here, we investigated the role of TAp63α in colorectal CSCs and the effects of sulforaphane on TAp63α. We found that TAp63α was upregulated in spheres with stem cell properties compared to the parental cells. Overexpression of TAp63α promoted self-renewal capacity and enhanced CSC markers expression in colorectal sphere-forming cells. Furthermore, we showed that TAp63α directly bound to the promoter region of Lgr5 to enhance its expression and activate its downstream β-catenin pathway. Functional experiments revealed that sulforaphane suppressed the stemness of colorectal CSCs both in vitro and in vivo. Upregulation of TAp63α attenuated the inhibitory effect of sulforaphane on colorectal CSCs, indicating the role of TAp63α in sulforaphane suppression of the stemness in colorectal cancer. The present study elucidated for the first time that TAp63α promoted CSCs through targeting Lgr5/β-catenin axis and participated in sulforaphane inhibition of the stem cell properties in colorectal cancer.
Collapse
|
43
|
Contartese D, Salamanna F, Veronesi F, Fini M. Relevance of humanized three-dimensional tumor tissue models: a descriptive systematic literature review. Cell Mol Life Sci 2020; 77:3913-3944. [PMID: 32285137 PMCID: PMC11104864 DOI: 10.1007/s00018-020-03513-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022]
Abstract
Despite numerous advances in tumor screening, diagnosis, and treatment, to date, tumors remain one of the leading causes of death, principally due to metastasis and the physiological damage produced by tumor growth. Among the main limits related to the study of tumor physiology there is the complex and heterogeneity nature of its environment and the absence of relevant, simple and inexpensive models able to mimic the biological processes occurring in patients allowing the correct clinical translation of results. To enhance the understanding of the mechanisms of tumors and to develop and evaluate new therapeutic approaches the set-up of advanced and alternative models is mandatory. One of the more translational approaches seems to be the use of humanized three-dimensional (3D) tissue culture. This model allows to accurately mimic tumor morphology and biology, maintaining the native microenvironment without any manipulation. However, little is still known on the real clinical relevance of these models for the study of tumor mechanisms and for the screening of new therapy. The aim of this descriptive systematic literature review was to evaluate and summarize the current knowledge on human 3D tumor tissue culture models. We reviewed the strategies employed by researchers to set-up these systems, also considering the different approaches and culture conditions used. All these aspects greatly contribute to the existing knowledge on tumors, providing a specific link to clinical scenarios and making the humanized 3D tumor tissue models a more attractive tool both for researchers and clinicians.
Collapse
Affiliation(s)
- D Contartese
- Laboratory Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136, Bologna, Italy
| | - Francesca Salamanna
- Laboratory Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136, Bologna, Italy.
| | - F Veronesi
- Laboratory Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136, Bologna, Italy
| | - M Fini
- Laboratory Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136, Bologna, Italy
| |
Collapse
|
44
|
Huang C, Ogawa R. Systemic factors that shape cutaneous pathological scarring. FASEB J 2020; 34:13171-13184. [DOI: 10.1096/fj.202001157r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Chenyu Huang
- Department of Dermatology Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University Beijing China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery Nippon Medical School Tokyo Japan
| |
Collapse
|
45
|
Basili T, Dopeso H, Kim SH, Ferrando L, Pareja F, Da Cruz Paula A, da Silva EM, Stylianou A, Maroldi A, Marchiò C, Rubin BP, Papotti M, Weigelt B, Moreira Ferreira CG, Lapa E Silva JR, Reis-Filho JS. Oncogenic properties and signaling basis of the PAX8-GLIS3 fusion gene. Int J Cancer 2020; 147:2253-2264. [PMID: 32383186 DOI: 10.1002/ijc.33040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/01/2020] [Accepted: 04/24/2020] [Indexed: 12/20/2022]
Abstract
Hyalinizing trabecular tumors of the thyroid are rare and mostly benign epithelial neoplasms of follicular cell origin, which have recently been shown to be underpinned by the PAX8-GLIS3 fusion gene. In our study, we sought to investigate the potential oncogenic mechanisms of the PAX8-GLIS3 fusion gene. Forced expression of PAX8-GLIS3 was found to increase proliferation, clonogenic potential and migration of human nonmalignant thyroid (Nthy-ori 3-1) and embryonic kidney (HEK-293) cells. Moreover, in xenografts, Nthy-ori 3-1 PAX8-GLIS3 expressing cells generated significantly larger and more proliferative tumors compared to controls. These oncogenic effects were found to be mediated through activation of the Sonic Hedgehog (SHH) pathway. Targeting of smoothened (SMO), a key protein in the SHH pathway, using the small molecule inhibitor Cyclopamine partially reversed the increased proliferation, colony formation and migration in PAX8-GLIS3 expressing cells. Our data demonstrate that the oncogenic effects of the PAX8-GLIS3 fusion gene are, at least in part, due to an increased activation of the SHH pathway.
Collapse
Affiliation(s)
- Thais Basili
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Higinio Dopeso
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sarah H Kim
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lorenzo Ferrando
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Arnaud Da Cruz Paula
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Edaise M da Silva
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Anthe Stylianou
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ana Maroldi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Caterina Marchiò
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - Brian P Rubin
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mauro Papotti
- Department of Oncology, University of Turin, at Città della Salute Hospital, Torino, Italy
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Carlos Gil Moreira Ferreira
- Oncoclinicas Institute for Research and Education, Sao Paulo, Brazil
- Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
46
|
Li HM, Yu YK, Liu Q, Wei XF, Zhang J, Zhang RX, Sun HB, Wang ZF, Xing WQ, Li Y. LncRNA SNHG1 Regulates the Progression of Esophageal Squamous Cell Cancer by the miR-204/HOXC8 Axis. Onco Targets Ther 2020; 13:757-767. [PMID: 32158227 PMCID: PMC6986417 DOI: 10.2147/ott.s224550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022] Open
Abstract
Objective Long noncoding RNA small nucleolar RNA host gene 1 (SNHG1) has been reported to be aberrantly expressed and plays an important role in human cancers, including esophageal squamous cell cancer. However, the regulatory mechanism underlying SNHG1 in the progression of esophageal squamous cell cancer is poorly defined. Materials and Methods Fifty-three esophageal squamous cell cancer patients were recruited and overall survival was analyzed. EC9706 and KYSE150 cells were cultured for study in vitro. The expression levels of SNHG1, microRNA (miR)-204 and homeobox c8 (HOXC8) were detected by quantitative real-time polymerase chain reaction and Western blot. Cell cycle distribution, apoptosis, migration and invasion were determined by flow cytometry and transwell assays, respectively. The target interaction among SNHG1, miR-204 and HOXC8 was validated by luciferase reporter assay and RNA immunoprecipitation. Xenograft model was established to investigate the role of SNHG1 in vivo. Results High expression of SNHG1 was exhibited in esophageal squamous cell cancer and indicated poor outcomes of patients. SNHG1 silence led to cell cycle arrest at G0-G1 phase, inhibition of migration and invasion and increase of apoptosis. miR-204 was validated to sponge by SNHG1 and target HOXC8 in esophageal squamous cell cancer cells. miR-204 knockdown or HOXC8 restoration reversed the inhibitive role of SNHG1 silence in the progression of esophageal squamous cell cancer cells. Furthermore, inhibiting SNHG1 decreased xenograft tumor growth by regulating miR-204 and HOXC8. Conclusion SNHG1 knockdown suppresses migration and invasion but induces apoptosis of esophageal squamous cell cancer cells by increasing miR-204 and decreasing HOXC8.
Collapse
Affiliation(s)
- Hao Miao Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yong Kui Yu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Qi Liu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xiu Feng Wei
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jun Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Rui Xiang Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hai Bo Sun
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zong Fei Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Wen Qun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yin Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Department of Thoracic Surgery, The Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
47
|
He JY, Wei XH, Li SJ, Liu Y, Hu HL, Li ZZ, Kuang XH, Wang L, Shi X, Yuan ST, Sun L. Adipocyte-derived IL-6 and leptin promote breast Cancer metastasis via upregulation of Lysyl Hydroxylase-2 expression. Cell Commun Signal 2018; 16:100. [PMID: 30563531 PMCID: PMC6299564 DOI: 10.1186/s12964-018-0309-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/25/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Adipocytes make up the major component of breast tissue, accounting for 90% of stromal tissue. Thus, the crosstalk between adipocytes and breast cancer cells may play a critical role in cancer progression. Adipocyte-breast cancer interactions have been considered important for the promotion of breast cancer metastasis. However, the specific mechanisms underlying these interactions are unclear. In this study, we investigated the mechanisms of adipocyte-mediated breast cancer metastasis. METHODS Breast cancer cells were cocultured with mature adipocytes for migration and 3D matrix invasion assays. Next, lentivirus-mediated loss-of-function experiments were used to explore the function of lysyl hydroxylase (PLOD2) in breast cancer migration and adipocyte-dependent migration of breast cancer cells. The role of PLOD2 in breast cancer metastasis was further confirmed using orthotopic mammary fat pad xenografts in vivo. Clinical samples were used to confirm that PLOD2 expression is increased in tumor tissue and is associated with poor prognosis of breast cancer patients. Cells were treated with cytokines and pharmacological inhibitors in order to verify which adipokines were responsible for activation of PLOD2 expression and which signaling pathways were activated in vitro. RESULTS Gene expression profiling and Western blotting analyses revealed that PLOD2 was upregulated in breast cancer cells following coculture with adipocytes; this process was accompanied by enhanced breast cancer cell migration and invasion. Loss-of-function studies indicated that PLOD2 knockdown suppressed cell migration and disrupted the formation of actin stress fibers in breast cancer cells and abrogated the migration induced by following coculture with adipocytes. Moreover, experiments performed in orthotopic mammary fat pad xenografts showed that PLOD2 knockdown could reduce metastasis to the lung and liver. Further, high PLOD2 expression correlated with poor prognosis of breast cancer patients. Mechanistically, adipocyte-derived interleukin-6 (IL-6) and leptin may facilitate PLOD2 upregulation in breast cancer cells and promote breast cancer metastasis in tail vein metastasis assays. Further investigation revealed that adipocyte-derived IL-6 and leptin promoted PLOD2 expression through activation of the JAK/STAT3 and PI3K/AKT signaling pathways. CONCLUSIONS Our study reveals that adipocyte-derived IL-6 and leptin promote PLOD2 expression by activating the JAK/STAT3 and PI3K/AKT signaling pathways, thus promoting breast cancer metastasis.
Collapse
Affiliation(s)
- Jin-Yong He
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiao-Hui Wei
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Si-Jing Li
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yang Liu
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Hao-Lin Hu
- Breast Disease Center, Zhong-Da Hospital, Southeast University, Nanjing, China
| | - Zheng-Zheng Li
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Xin-Hong Kuang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Lai Wang
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xin Shi
- Department of General Surgery, Zhong-Da Hospital, Southeast University, Nanjing, China
| | - Sheng-Tao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China.
| | - Li Sun
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|