1
|
Fu Y, Zhu D, Chen X, Qu L, Guo M, Zhang S, Xu G, Chen Z, Li M, Chen Y. Integrative Multi-Omics Approaches Reveal Selectivity Profiles and Molecular Mechanisms of FIIN-2, a Covalent FGFR Inhibitor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412578. [PMID: 39976135 PMCID: PMC11984845 DOI: 10.1002/advs.202412578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/02/2025] [Indexed: 02/21/2025]
Abstract
Fibroblast growth factor receptor (FGFR) inhibitors are emerged as an important class of targeted therapies in oncology, targeting key pathways associated with tumor growth, angiogenesis, and resistance to conventional treatments. FIIN-2, the first irreversible covalent pan-FGFR inhibitor, has shown promise in overcoming resistance due to gatekeeper mutations; however, its selectivity and molecular mechanisms in tumors remain poorly understood. In this study, an FIIN-2 chemical probe is designed and synthesized to identify both established and novel targets in hepatocellular carcinoma (HCC) via chemoproteomic profiling. An integrative multi-omics approach, including chemoproteomic, phosphoproteomic, transcriptomic, and proteomic analyses, is utilized to elucidate the full spectrum of target proteins, signaling pathways, and downstream effectors regulated by FIIN-2 in HCC. Notably, adenosine monophosphate-activated protein kinase α1 (AMPKα1) is identified as a novel target of FIIN-2, with Cys185 identified as its covalent binding site. These findings reveal that FIIN-2 can induce autophagy by directly binding to and activating AMPKα1, influencing its anti-tumor activity in HCC cells. Overall, this study greatly advances the understanding of FIIN-2's on- and off-target effects, offering a comprehensive view of its molecular mechanisms in cancer cells. The integrative multi-omics approach provides a valuable framework for the development and optimization of covalent kinase inhibitors.
Collapse
Affiliation(s)
- Ying Fu
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Dandan Zhu
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Xiaojuan Chen
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Lingzhi Qu
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Ming Guo
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Shuhong Zhang
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Guangyu Xu
- Key Laboratory of Chemical Biology and Traditional Chinese MedicineMinistry of Educational of ChinaKey Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan ProvinceCollege of Chemistry and Chemical EngineeringHunan Normal UniversityChangshaHunan410081China
| | - Zhuchu Chen
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Maoyu Li
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yongheng Chen
- Department of OncologyNHC Key Laboratory of Cancer Proteomics and State Local Joint Engineering Laboratory for Anticancer DrugsNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
2
|
Sun S, Zhu F, Xu Q, Hou X, Nie W, Su K, Wang L, Liu Z, Shan T, Shi C. Modified bFGF targeting connective tissue growth factor in the injured microenvironment improved cardiac repair after chronic myocardial ischemia. Regen Ther 2025; 28:438-450. [PMID: 39925966 PMCID: PMC11803136 DOI: 10.1016/j.reth.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 02/11/2025] Open
Abstract
Myocardial infarction (MI) was a cardiovascular emergency that led to heart failure, arrhythmia, and sudden death. Basic fibroblast growth factor (bFGF) was revealed to promote angiogenesis and protect cardiomyocytes against ischemic injury. But conventional delivery of bFGF in an uncontrolled manner was inefficient and diffusive, limiting its application in MI therapy. Currently, stimuli-responsive drug delivery is emphasized in tissue regeneration. The present study constructed a CFBP-bFGF recombinant protein, which could specifically target upregulated connective tissue growth factor (CTGF) and release bFGF in ischemic myocardium. In a rat model with MI, intravenous administration of CFBP-bFGF significantly accumulated in ischemic myocardium by targeting with CTGF. The responsive release of CFBP-bFGF effectively enhanced blood vessel regeneration, decreased cardiomyocyte apoptosis, and improved cardiac function recovery. In addition, the molecular mechanism was further explored by RNA sequencing and transcriptome analysis. Besides activating the pathways and genes related to angiogenesis and cardiac protection, CFBP-bFGF also decreased the expression of fibrosis-related pathways and genes, such as TGF-β. These results demonstrated that the CTGF-responsive CFBP-bFGF was effective for targeting release that promoted the functional recovery of MI.
Collapse
Affiliation(s)
- Shuwei Sun
- Department of School of Basic Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| | - Fengzheng Zhu
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong Province 266000, China
| | - Qingling Xu
- Department of School of Basic Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Weihong Nie
- Department of Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| | - Kaiyan Su
- Department of School of Basic Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| | - Li Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong Province 266000, China
| | - Zhuo Liu
- Department of School of Basic Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| | - Tao Shan
- Department of School of Basic Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| | - Chunying Shi
- Department of School of Basic Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| |
Collapse
|
3
|
Huang Y, Gao Y, Lin Z, Miao H. Involvement of the ubiquitin-proteasome system in the regulation of the tumor microenvironment and progression. Genes Dis 2025; 12:101240. [PMID: 39759114 PMCID: PMC11697063 DOI: 10.1016/j.gendis.2024.101240] [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: 06/29/2023] [Revised: 10/11/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2025] Open
Abstract
The tumor microenvironment is a complex environment comprising tumor cells, non-tumor cells, and other critical non-cellular components. Some studies about tumor microenvironment have recently achieved remarkable progress in tumor treatment. As a substantial part of post-translational protein modification, ubiquitination is a crucial player in maintaining protein stability in cell signaling, cell growth, and a series of cellular life activities, which are also essential for regulating tumor cells or other non-tumor cells in the tumor microenvironment. This review focuses on the role and function of ubiquitination and deubiquitination modification in the tumor microenvironment while discussing the prospect of developing inhibitors targeting ubiquity-related enzymes, thereby providing ideas for future research in cancer therapy.
Collapse
Affiliation(s)
- Yulan Huang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yuan Gao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
4
|
Chen J, Hu Q, Zhang C, Zhao A, Guan B, Wang Y, Zhang M, Li X, Chen B, Zeng L, Chen M, Wu B, Wang J, Yang Y, Ji J. Tendomodulin in pan-cancer analysis: exploring its impact on immune modulation and uncovering functional insights in colorectal cancer. BMC Cancer 2025; 25:239. [PMID: 39934677 DOI: 10.1186/s12885-025-13608-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: 10/21/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Tendomodulin (TNMD) is pivotal in various malignancies, including colorectal cancer (CRC). However, its comprehensive impact across cancers, particularly its immunomodulatory function in CRC, remains underexplored. This study explored the role of TNMD in CRC by focusing on its immunomodulatory functions through comprehensive molecular and clinical analyses. METHODS Multiple bioinformatics databases and analytical tools were utilized for the TNMD in pan-cancer analysis. To validate the role of TNMD in CRC, we performed experiments, including immunofluorescence (IF), immunohistochemistry (IHC), real-time quantitative reverse transcription PCR (qPCR), western blotting, and cell migration assays. RESULTS TNMD expression and gene mutation vary across cancers and offer high diagnostic value. Survival analysis found that TNMD is associated with prognosis in multiple cancers. Notably, in patients with high microsatellite instability (MSI-H) CRC, TNMD expression correlated positively with various immune cells, particularly natural killer (NK) cells, whereas it was inversely correlated with regulatory T cells (Tregs). Crucially, in patients with microsatellite stability (MSS) CRC, high TNMD expression was associated with better immunotherapy outcomes, indicating its potential as a biomarker for patient stratification and tailored treatment approaches. Furthermore, single-cell sequencing data revealed stronger interactions between TNMD-positive tumor cells and fibroblasts or macrophages in the tumor microenvironment. Finally, TNMD was overexpressed in CRC tumor tissues and cell lines, thereby promoting invasion and metastasis. CONCLUSIONS Our findings reveal a critical immunomodulatory role of TNMD in CRC, particularly in influencing tumor-immune interactions. Beyond its potential diagnostic and prognostic biomarker, TNMD promotes CRC metastasis and invasion, thus emerging as a promising therapeutic target. These findings highlight TNMD's significance in CRC and potentially other malignancies.
Collapse
Affiliation(s)
- Jingfeng Chen
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
- Anorectal surgery of The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Qin Hu
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Cong Zhang
- Department of Radiology, School of Medicine, Lishui Hospital of Zhejiang University, Lishui Central Hospital, Lishui, 323000, China
| | - Aiqi Zhao
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Bihua Guan
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Yifan Wang
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Min Zhang
- Department, Pathology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Xia Li
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Biao Chen
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Lulu Zeng
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Minjiang Chen
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
- Key Laboratory of Precision Medicine of Lishui, Lishui Central Hospital, Lishui, 323000, China
| | - Bing Wu
- Department, Pharmacy, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jianping Wang
- Anorectal surgery of The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Yang Yang
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China.
- Key Laboratory of Precision Medicine of Lishui, Lishui Central Hospital, Lishui, 323000, China.
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, 323000, Lishui, China.
| | - Jiansong Ji
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China.
- Department of Radiology, School of Medicine, Lishui Hospital of Zhejiang University, Lishui Central Hospital, Lishui, 323000, China.
- Key Laboratory of Precision Medicine of Lishui, Lishui Central Hospital, Lishui, 323000, China.
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, 323000, Lishui, China.
| |
Collapse
|
5
|
Yang J, Song X, Zhang H, Liu Q, Wei R, Guo L, Yuan C, Chen F, Xue K, Lai Y, Wang L, Shi J, Zhou C, Wang J, Yu Y, Mei Q, Hu L, Wang H, Zhang C, Zhang Q, Li H, Gu Y, Zhao W, Yu H, Wang J, Liu Z, Li H, Zheng S, Liu J, Yang L, Li W, Xu R, Chen J, Zhou Y, Cheng X, Yu Y, Wang D, Sun X, Yu H. Single-cell transcriptomic landscape deciphers olfactory neuroblastoma subtypes and intra-tumoral heterogeneity. NATURE CANCER 2024; 5:1919-1939. [PMID: 39543363 DOI: 10.1038/s43018-024-00855-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 10/07/2024] [Indexed: 11/17/2024]
Abstract
Olfactory neuroblastoma (ONB) is a rare malignancy known to originate from the olfactory epithelium. The complex tumor ecosystem of this pathology remains unclear. Here, we explored the cellular components within ten ONB tumors and one olfactory mucosa sample based on single-cell RNA profiles. We showed the intra-tumoral heterogeneity by identifying five unique expression programs among malignant epithelial cells. A distinct three-classification system (neural, basal, mesenchymal) for ONB was established according to the distinguished gene expression patterns. Biomarkers for categorizing bulk tumors into uncharacterized subtypes were elucidated. Different responses towards certain chemotherapy regimens could be cautiously inferred according to the molecular features representing the three tumor types, thus helping with precision chemotherapy. We also analyzed subclusters of the tumor microenvironment (TME) and the interactions among different cell types within the TME. The relative abundance of immunosuppressive tumor-associated macrophages suggests potential benefits of immunotherapies targeting macrophages.
Collapse
Affiliation(s)
- Jingyi Yang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- Olfactory Neuroblastoma Diagnosis and Treatment Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Xiaole Song
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- Mucosal Melanoma Diagnosis and Treatment Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Huankang Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Quan Liu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Ruoyan Wei
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Luo Guo
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, PR China
| | - Cuncun Yuan
- Department of Pathology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Fu Chen
- Department of Radiation Oncology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Kai Xue
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Yuting Lai
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Li Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Junfeng Shi
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Chengle Zhou
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Juan Wang
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Pharmaceutical Industry Research Institute, Shanghai, PR China
| | - Yingxuan Yu
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Pharmaceutical Industry Research Institute, Shanghai, PR China
| | - Qibing Mei
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Pharmaceutical Industry Research Institute, Shanghai, PR China
| | - Li Hu
- Department of Experimental Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Huan Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Chen Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Qianqian Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Houyong Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Ye Gu
- Department of Neurosurgery and Otolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Weidong Zhao
- Department of Neurosurgery and Otolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Huapeng Yu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Jingjing Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Zhuofu Liu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Han Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Shixing Zheng
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Juan Liu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Lu Yang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Wanpeng Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Rui Xu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Jiani Chen
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Yumin Zhou
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Xiankui Cheng
- Department of Pathology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| | - Yiqun Yu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- Olfactory Disorder Diagnosis and Treatment Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Dehui Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Xicai Sun
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- Olfactory Neuroblastoma Diagnosis and Treatment Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Hongmeng Yu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China.
- Research Unit of New Technologies of Endoscopic Surgery in Skull Base Tumor (2018RU003), Chinese Academy of Medical Sciences, Shanghai, PR China.
| |
Collapse
|
6
|
Athavale D, Balch C, Zhang Y, Yao X, Song S. The role of Hippo/YAP1 in cancer-associated fibroblasts: Literature review and future perspectives. Cancer Lett 2024; 604:217244. [PMID: 39260668 DOI: 10.1016/j.canlet.2024.217244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/27/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are activated fibroblasts that play a role in numerous malignant phenotypes, including hyperproliferation, invasion, and metastasis. These phenotypes correlate with activity of the Hippo pathway oncoprotein, Yes-associated protein-1 (YAP1), and its paralog, transcriptional coactivator with PDZ-binding motif (TAZ). YAP1/TAZ are normally involved in organ growth, under the regulation of various kinases and upon phosphorylation, are retained in the cytoplasm by chaperone proteins, leading to their proteasomal degradation. In CAFs and tumor cells, however, a lack of YAP1 phosphorylation results in its translocation to the nucleus, binding to TEAD transcription factors, and activation of mitogenic pathways. In this review we summarize the literature discussing the central role of YAP1 in CAF activation, the upstream cues that promote YAP1-mediated CAF activation and extracellular matrix remodeling, and how CAFs mediate tumor-stroma crosstalk to support progression, invasion and metastasis in various cancer models. We further highlight YAP1+CAFs functions in modulating an immunosuppressive tumor microenvironment and propose evaluation of several YAP1 targets regarding their role in regulating intra-tumoral immune landscapes. Finally, we propose that co-administration of YAP1- targeted therapies with immune checkpoint inhibitors can improve therapeutic outcomes in patients with advanced tumors.
Collapse
Affiliation(s)
- Dipti Athavale
- Coriell Institute for Medical Research, 403 Haddon Ave, Camden, NJ, 08103, USA
| | - Curt Balch
- Coriell Institute for Medical Research, 403 Haddon Ave, Camden, NJ, 08103, USA
| | - Yanting Zhang
- Coriell Institute for Medical Research, 403 Haddon Ave, Camden, NJ, 08103, USA
| | - Xiaodan Yao
- Coriell Institute for Medical Research, 403 Haddon Ave, Camden, NJ, 08103, USA
| | - Shumei Song
- Coriell Institute for Medical Research, 403 Haddon Ave, Camden, NJ, 08103, USA; MD Anderson Cancer Center at Cooper, Cooper University Hospital, 2 Cooper Plaza, Camden, NJ, 08103, USA; Departments of Surgery and Biomedical Sciences, Cooper Medical School of Rowan University, 401 Broadway, Camden, NJ, 08103, USA.
| |
Collapse
|
7
|
Yazdani F, Mottaghi-Dastjerdi N, Shahbazi B, Ahmadi K, Ghorbani A, Soltany-Rezaee-Rad M, Montazeri H, Khoshdel F, Guzzi PH. Identification of key genes and pathways involved in T-DM1-resistance in OE-19 esophageal cancer cells through bioinformatics analysis. Heliyon 2024; 10:e37451. [PMID: 39309859 PMCID: PMC11415672 DOI: 10.1016/j.heliyon.2024.e37451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Esophageal Cancer (EC) ranks among the most common malignancies worldwide. Most EC patients acquire drug resistance to chemotherapy either intrinsically or acquired after T-DM1 treatment, which shows that increasing or decreasing the expression of particular genes might influence chemotherapeutic sensitivity or resistance. Therefore, gaining a deeper understanding of the altered expression of genes involved in EC drug resistance and developing new therapeutic methods are essential targets for continued advancement in EC therapy. Methods The present study aimed to find critical regulatory genes/pathways in the progression of T-DM1 resistance in OE-19 EC cells. Expression datasets were extracted from GEO omnibus. Gene interactions were analyzed, and the protein-protein interaction network was drawn. Then, enrichment analysis of the hub genes and network cluster analysis of the hub genes was performed. Finally, the genes were screened in the DrugBank database as therapeutic targets and molecular docking analysis was done on the selected targets. Results In the current study, nine hub genes were identified in TDM-1-resistant EC cells (CTGF, CDH17, THBS1, CXCL8, NRP1, ITGB5, EDN1, FAT1, and PTGS2). The KEGG analysis highlighted the IL-17 signaling pathway and ECM-receptor interaction pathway as the most critical pathways; cluster analysis also showed the significance of these pathways. Therefore, the genes involved in these two pathways, including CXCL8, FSCN1, PTGS2, SERPINE2, LEF1, THBS1, CCN2, TAGLN, CDH11, and ITGA6, were searched in DrugBank as therapeutic targets. The DrugBank analysis suggests a potential role for Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) in reducing T-DM1 drug resistance in EC. The docking results revealed that NSAIDs, including Diclofenac, Mefenamic acid, Celecoxib, Naproxen, and Etoricoxib, significantly suppress resistant cancer cells. Conclusion This comprehensive bioinformatics analysis deeply explains the molecular mechanisms governing TDM-1 resistance in EC. The identified hub genes and their associated pathways offer potential targets for therapeutic interventions. Moreover, the possible role of NSAIDs in mitigating T-DM1 resistance presents an intriguing avenue for further investigation. This research contributes significantly to the field and establishes a basis for further research to enhance treatment efficacy for EC patients.
Collapse
Affiliation(s)
- Fateme Yazdani
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Negar Mottaghi-Dastjerdi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Behzad Shahbazi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
| | - Khadijeh Ahmadi
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Abozar Ghorbani
- Nuclear Agriculture Research School, Nuclear Science and Technology Research Institute (NSTRI), Karaj, Iran
| | | | - Hamed Montazeri
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Farzane Khoshdel
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Pietro Hiram Guzzi
- Department of Surgical and Medical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
8
|
Chang SL, Tsai YJ, Shieh JM, Wu WB. The novel thromboxane prostanoid receptor mediates CTGF production to drive human nasal fibroblast self-migration through NF-κB and PKCδ-CREB signaling pathways. J Cell Physiol 2024; 239:e31390. [PMID: 39104040 DOI: 10.1002/jcp.31390] [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: 03/11/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024]
Abstract
Chronic rhinosinusitis without nasal polyp (CRSsNP) is characterized by tissue repair/remodeling and the subepithelial stroma region in whose nasal mucosa has been reported by us to have thromboxane A2 (TXA2) prostanoid (TP) receptor and overexpress connective tissue growth factor (CTGF). Therefore, this study aimed to investigate the relationship between TP receptor activation and CTGF production/function in human CRSsNP nasal mucosa stromal fibroblasts. We found that TP agonists including U46619 and IBOP ([1S-[1α,2α(Z),3β(1E,3 S*),4α]]-7-[3-[3-hydroxy-4-(4-iodophenoxy)-1-butenyl]-7-oxabicyclo[2.2.1]hept-2-yl]-5-heptenoic acid) could promote CTGF protein/messenger RNA expression and secretion. The pharmacological intervention and TP activation assay with U46619 identified the possible participation of PKCμ, PKCδ, nuclear factor-κB (NF-κB), and cyclic AMP response element-binding protein (CREB) phosphorylation/activation in the CTGF induction. Moreover, a phorbol ester-phorbol-12-myristate 13-acetate (PMA) exhibited a similar cellular signaling and CTGF production profile to that elicited by TP activation. However, further small interfering RNA interference analysis revealed that only NF-κB and PKCδ-CREB pathways were necessarily required for TP-mediated CTGF production, which could not be completely supported by those findings from PMA. Finally, in a functional assay, although CTGF did not affect fibroblast proliferation, TP-mediated CTGF could drive novel self-migration in fibroblasts both in the scratch/wound healing and transwell apparatus assays. Meanwhile, the overall staining for stress fibers and formation of the lamellipodia and filopodia-like structures was concomitantly increased in the treated migrating cells. Collectively, we provided here that novel TP mediates CTGF production and self-migration in human nasal fibroblasts through NF-κB and PKCδ-CREB signaling pathways. More importantly, we also demonstrated that thromboxane, TP receptor, CTGF, and stromal fibroblasts may act in concert in the tissue remodeling/repair process during CRSsNP development and progression.
Collapse
Affiliation(s)
- Shih-Lun Chang
- Department of Otorhinolaryngology, Chi Mei Medical Center, Yongkang District, Tainan, Taiwan
- Department of Pet Care and Grooming, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Yih-Jeng Tsai
- Department of Otolaryngology Head and Neck Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Jiunn-Min Shieh
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Wen-Bin Wu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
9
|
Jing SY, Liu D, Feng N, Dong H, Wang HQ, Yan X, Chen XF, Qu MC, Lin P, Yi B, Feng F, Chen L, Wang HY, Li H, He YF. Spatial multiomics reveals a subpopulation of fibroblasts associated with cancer stemness in human hepatocellular carcinoma. Genome Med 2024; 16:98. [PMID: 39138551 PMCID: PMC11320883 DOI: 10.1186/s13073-024-01367-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are the prominent cell type in the tumor microenvironment (TME), and CAF subsets have been identified in various tumors. However, how CAFs spatially coordinate other cell populations within the liver TME to promote cancer progression remains unclear. METHODS We combined multi-region proteomics (6 patients, 24 samples), 10X Genomics Visium spatial transcriptomics (11 patients, 25 samples), and multiplexed imaging (92 patients, 264 samples) technologies to decipher the expression heterogeneity, functional diversity, spatial distribution, colocalization, and interaction of fibroblasts. The newly identified CAF subpopulation was validated by cells isolated from 5 liver cancer patients and in vitro functional assays. RESULTS We identified a liver CAF subpopulation, marked by the expression of COL1A2, COL4A1, COL4A2, CTGF, and FSTL1, and named F5-CAF. F5-CAF is preferentially located within and around tumor nests and colocalizes with cancer cells with higher stemness in hepatocellular carcinoma (HCC). Multiplexed staining of 92 patients and the bulk transcriptome of 371 patients demonstrated that the abundance of F5-CAFs in HCC was associated with a worse prognosis. Further in vitro experiments showed that F5-CAFs isolated from liver cancer patients can promote the proliferation and stemness of HCC cells. CONCLUSIONS We identified a CAF subpopulation F5-CAF in liver cancer, which is associated with cancer stemness and unfavorable prognosis. Our results provide potential mechanisms by which the CAF subset in the TME promotes the development of liver cancer by supporting the survival of cancer stem cells.
Collapse
Affiliation(s)
- Si-Yu Jing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Dan Liu
- Molecular Pathology Laboratory, National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Shanghai, 201800, People's Republic of China
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Na Feng
- Molecular Pathology Laboratory, National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Shanghai, 201800, People's Republic of China
| | - Hui Dong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, People's Republic of China
| | - He-Qi Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Xi Yan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Xu-Feng Chen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Min-Cheng Qu
- Molecular Pathology Laboratory, National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Shanghai, 201800, People's Republic of China
| | - Ping Lin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Bin Yi
- Department of Organ Transplantation, Eastern Hepatobiliary Surgery Hospital, Shanghai, 201800, People's Republic of China
| | - Feiling Feng
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, 201800, People's Republic of China
| | - Lei Chen
- National Center for Liver Cancer and International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai, 200438, People's Republic of China.
| | - Hong-Yang Wang
- National Center for Liver Cancer and International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai, 200438, People's Republic of China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education and Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai, 200438, People's Republic of China.
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China.
| | - Yu-Fei He
- Molecular Pathology Laboratory, National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Shanghai, 201800, People's Republic of China.
| |
Collapse
|
10
|
Fujita M, Sasada M, Iyoda T, Fukai F. Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases. Int J Mol Sci 2024; 25:6591. [PMID: 38928297 PMCID: PMC11204155 DOI: 10.3390/ijms25126591] [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: 05/29/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Senescence is a physiological and pathological cellular program triggered by various types of cellular stress. Senescent cells exhibit multiple characteristic changes. Among them, the characteristic flattened and enlarged morphology exhibited in senescent cells is observed regardless of the stimuli causing the senescence. Several studies have provided important insights into pro-adhesive properties of cellular senescence, suggesting that cell adhesion to the extracellular matrix (ECM), which is involved in characteristic morphological changes, may play pivotal roles in cellular senescence. Matricellular proteins, a group of structurally unrelated ECM molecules that are secreted into the extracellular environment, have the unique ability to control cell adhesion to the ECM by binding to cell adhesion receptors, including integrins. Recent reports have certified that matricellular proteins are closely involved in cellular senescence. Through this biological function, matricellular proteins are thought to play important roles in the pathogenesis of age-related diseases, including fibrosis, osteoarthritis, intervertebral disc degeneration, atherosclerosis, and cancer. This review outlines recent studies on the role of matricellular proteins in inducing cellular senescence. We highlight the role of integrin-mediated signaling in inducing cellular senescence and provide new therapeutic options for age-related diseases targeting matricellular proteins and integrins.
Collapse
Affiliation(s)
- Motomichi Fujita
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
| | - Manabu Sasada
- Clinical Research Center in Hiroshima, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8551, Japan
| | - Takuya Iyoda
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Doori, Sanyo-Onoda 756-0884, Yamaguchi, Japan
| | - Fumio Fukai
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
| |
Collapse
|
11
|
Marrero AD, Cárdenas C, Castilla L, Ortega-Vidal J, Quesada AR, Martínez-Poveda B, Medina MÁ. Antiangiogenic Potential of an Olive Oil Extract: Insights from a Proteomic Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:13023-13038. [PMID: 38809962 PMCID: PMC11181319 DOI: 10.1021/acs.jafc.3c08851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
Extra virgin olive oil (EVOO), a staple of the Mediterranean diet, is rich in phenolic compounds recognized for their potent bioactive effects, including anticancer and anti-inflammatory properties. However, its effects on vascular health remain relatively unexplored. In this study, we examined the impact of a "picual" EVOO extract from Jaén, Spain, on endothelial cells. Proteomic analysis revealed the modulation of angiogenesis-related processes. In subsequent in vitro experiments, the EVOO extract inhibited endothelial cell migration, adhesion, invasion, ECM degradation, and tube formation while inducing apoptosis. These results provide robust evidence of the extract's antiangiogenic potential. Our findings highlight the potential of EVOO extracts in mitigating angiogenesis-related pathologies, such as cancer, macular degeneration, and diabetic retinopathy.
Collapse
Affiliation(s)
- Ana Dácil Marrero
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER
de Enfermedades Raras (CIBERER), Instituto
de Salud Carlos III, E-28029 Madrid, Spain
| | - Casimiro Cárdenas
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Servicios
Centrales de Apoyo a la Investigación (SCAI), Universidad de Málaga, E-29071 Málaga, Spain
| | - Laura Castilla
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
| | - Juan Ortega-Vidal
- Departamento
de Química Inorgánica y Orgánica, Campus de Excelencia
Internacional Agroalimentaria ceiA3, Universidad
de Jaén, Jaén E- 23071, Spain
| | - Ana R. Quesada
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER
de Enfermedades Raras (CIBERER), Instituto
de Salud Carlos III, E-28029 Madrid, Spain
| | - Beatriz Martínez-Poveda
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER
de
Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Miguel Ángel Medina
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER
de Enfermedades Raras (CIBERER), Instituto
de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
12
|
D'Aguanno S, Brignone M, Scalera S, Chiacchiarini M, Di Martile M, Valentini E, De Nicola F, Ricci A, Pelle F, Botti C, Maugeri-Saccà M, Del Bufalo D. Bcl-2 dependent modulation of Hippo pathway in cancer cells. Cell Commun Signal 2024; 22:277. [PMID: 38755629 PMCID: PMC11097437 DOI: 10.1186/s12964-024-01647-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
INTRODUCTION Bcl-2 and Bcl-xL are the most studied anti-apoptotic members of Bcl-2 family proteins. We previously characterized both of them, not only for their role in regulating apoptosis and resistance to therapy in cancer cells, but also for their non-canonical functions, mainly including promotion of cancer progression, metastatization, angiogenesis, and involvement in the crosstalk among cancer cells and components of the tumor microenvironment. Our goal was to identify transcriptional signature and novel cellular pathways specifically modulated by Bcl-2. METHODS We performed RNAseq analysis of siRNA-mediated transient knockdown of Bcl-2 or Bcl-xL in human melanoma cells and gene ontology analysis to identify a specific Bcl-2 transcriptional signature. Expression of genes modulated by Bcl-2 and associated to Hippo pathway were validated in human melanoma, breast adenocarcinoma and non-small cell lung cancer cell lines by qRT-PCR. Western blotting analysis were performed to analyse protein expression of upstream regulators of YAP and in relation to different level of Bcl-2 protein. The effects of YAP silencing in Bcl-2 overexpressing cancer cells were evaluated in migration and cell viability assays in relation to different stiffness conditions. In vitro wound healing assays and co-cultures were used to evaluate cancer-specific Bcl-2 ability to activate fibroblasts. RESULTS We demonstrated the Bcl-2-dependent modulation of Hippo Pathway in cancer cell lines from different tumor types by acting on upstream YAP regulators. YAP inhibition abolished the ability of Bcl-2 to increase tumor cell migration and proliferation on high stiffness condition of culture, to stimulate in vitro fibroblasts migration and to induce fibroblasts activation. CONCLUSIONS We discovered that Bcl-2 regulates the Hippo pathway in different tumor types, promoting cell migration, adaptation to higher stiffness culture condition and fibroblast activation. Our data indicate that Bcl-2 inhibitors should be further investigated to counteract cancer-promoting mechanisms.
Collapse
Affiliation(s)
- Simona D'Aguanno
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy.
| | - Matteo Brignone
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Stefano Scalera
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Martina Chiacchiarini
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Elisabetta Valentini
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | | | - Alessia Ricci
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, 66100, Italy
| | - Fabio Pelle
- Department of Surgery, Division of Breast Surgery, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Claudio Botti
- Department of Surgery, Division of Breast Surgery, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Marcello Maugeri-Saccà
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| |
Collapse
|
13
|
Qiu Y, Que Y, Ding Z, Zhang S, Wei R, Xia J, Lin Y. Drugs targeting CTGF in the treatment of pulmonary fibrosis. J Cell Mol Med 2024; 28:e18448. [PMID: 38774993 PMCID: PMC11109635 DOI: 10.1111/jcmm.18448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/24/2024] Open
Abstract
Pulmonary fibrosis represents the final alteration seen in a wide variety of lung disorders characterized by increased fibroblast activity and the accumulation of substantial amounts of extracellular matrix, along with inflammatory damage and the breakdown of tissue architecture. This condition is marked by a significant mortality rate and a lack of effective treatments. The depositing of an excessive quantity of extracellular matrix protein follows the damage to lung capillaries and alveolar epithelial cells, leading to pulmonary fibrosis and irreversible damage to lung function. It has been proposed that the connective tissue growth factor (CTGF) plays a critical role in the advancement of pulmonary fibrosis by enhancing the accumulation of the extracellular matrix and exacerbating fibrosis. In this context, the significance of CTGF in pulmonary fibrosis is examined, and a summary of the development of drugs targeting CTGF for the treatment of pulmonary fibrosis is provided.
Collapse
Affiliation(s)
- Yudan Qiu
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang ProvinceHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yueyue Que
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang ProvinceHangzhou Normal UniversityHangzhouZhejiangChina
| | - Zheyu Ding
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang ProvinceHangzhou Normal UniversityHangzhouZhejiangChina
| | - Shanshan Zhang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang ProvinceHangzhou Normal UniversityHangzhouZhejiangChina
| | - Rong Wei
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang ProvinceHangzhou Normal UniversityHangzhouZhejiangChina
| | - Jianing Xia
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang ProvinceHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yingying Lin
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang ProvinceHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
14
|
Ren M, Yao S, Chen T, Luo H, Tao X, Jiang H, Yang X, Zhang H, Yu S, Wang Y, Lu A, Zhang G. Connective Tissue Growth Factor: Regulation, Diseases, and Drug Discovery. Int J Mol Sci 2024; 25:4692. [PMID: 38731911 PMCID: PMC11083620 DOI: 10.3390/ijms25094692] [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: 02/27/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
In drug discovery, selecting targeted molecules is crucial as the target could directly affect drug efficacy and the treatment outcomes. As a member of the CCN family, CTGF (also known as CCN2) is an essential regulator in the progression of various diseases, including fibrosis, cancer, neurological disorders, and eye diseases. Understanding the regulatory mechanisms of CTGF in different diseases may contribute to the discovery of novel drug candidates. Summarizing the CTGF-targeting and -inhibitory drugs is also beneficial for the analysis of the efficacy, applications, and limitations of these drugs in different disease models. Therefore, we reviewed the CTGF structure, the regulatory mechanisms in various diseases, and drug development in order to provide more references for future drug discovery.
Collapse
Affiliation(s)
- Meishen Ren
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shanshan Yao
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Tienan Chen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hang Luo
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xiaohui Tao
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xin Yang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Huarui Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Sifan Yu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yin Wang
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
15
|
Chen K, Hong C, Kong W, Li G, Liu Z, Zhu K, Lu C, Si P, Gao P, Ning G, Zhang R. ACADL-YAP axis activity in non-small cell lung cancer carcinogenicity. Cancer Cell Int 2024; 24:86. [PMID: 38402174 PMCID: PMC10894480 DOI: 10.1186/s12935-024-03276-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/17/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND The role of Acyl-CoA dehydrogenase long chain (ACADL) in different tumor types had different inhibiting or promoting effect. However, its role in non-small cell lung cancer (NSCLC) carcinogenicity is not clear. METHOD In this study, we utilized The Cancer Genome Atlas (TCGA) database to analyze ACADL expression in NSCLC and its correlation with overall survival. Furthermore, we investigated the function of ACADL on cellular proliferation, invasion, colony, apoptosis, cell cycle in vitro with NSCLC cells. Mechanistically, we evaluated the regulatory effect of ACADL expression on its downstream factor yes-associated protein (YAP) by assessing YAP phosphorylation levels and its cellular localization. Finally, we verified the tumorigenic effect of ACADL on NSCLC cells through xenograft experiments in vivo. RESULTS Compared to adjacent non-cancerous samples, ACADL significantly down-regulated in NSCLC. Overexpression of ACADL, effectively reduced the proliferative, colony, and invasive capabilities of NSCLC cells, while promoting apoptosis and inducing cell cycle arrest. Moreover, ACADL overexpression significantly enhanced YAP phosphorylation and hindered its nuclear translocation. However, the inhibitory effect of the overexpression of ACADL in NSCLC cells mentioned above can be partially counteracted by YAP activator XMU-MP-1 application both in vitro and in vivo. CONCLUSION The findings suggest that ACADL overexpression could suppress NSCLC development by modulating YAP phosphorylation and limiting its nuclear shift. This role of ACADL-YAP axis provided novel insights into NSCLC carcinogenicity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Kegong Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, 230001, China
| | - Chunqiao Hong
- Department of Critical Care Medicine, Xiamen Chang Gung Hospital Hua Qiao University, Xiamen, 361013, China
| | - Weibo Kong
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
- Departments of Thoracic Surgery, Anhui Provincial Chest Hospital, Hefei, 230001, China
| | - Guanghua Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Zhuang Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230001, China
| | - Kechao Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
| | - Chen Lu
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
| | - Panpan Si
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
| | - Pan Gao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230001, China
| | - Guangyao Ning
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China.
| | - Renquan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China.
| |
Collapse
|
16
|
Kanai R, Norton E, Stern P, Hynes RO, Lamar JM. Identification of a Gene Signature That Predicts Dependence upon YAP/TAZ-TEAD. Cancers (Basel) 2024; 16:852. [PMID: 38473214 PMCID: PMC10930532 DOI: 10.3390/cancers16050852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Targeted therapies are effective cancer treatments when accompanied by accurate diagnostic tests that can help identify patients that will respond to those therapies. The YAP/TAZ-TEAD axis is activated and plays a causal role in several cancer types, and TEAD inhibitors are currently in early-phase clinical trials in cancer patients. However, a lack of a reliable way to identify tumors with YAP/TAZ-TEAD activation for most cancer types makes it difficult to determine which tumors will be susceptible to TEAD inhibitors. Here, we used a combination of RNA-seq and bioinformatic analysis of metastatic melanoma cells to develop a YAP/TAZ gene signature. We found that the genes in this signature are TEAD-dependent in several melanoma cell lines, and that their expression strongly correlates with YAP/TAZ activation in human melanomas. Using DepMap dependency data, we found that this YAP/TAZ signature was predictive of melanoma cell dependence upon YAP/TAZ or TEADs. Importantly, this was not limited to melanoma because this signature was also predictive when tested on a panel of over 1000 cancer cell lines representing numerous distinct cancer types. Our results suggest that YAP/TAZ gene signatures like ours may be effective tools to predict tumor cell dependence upon YAP/TAZ-TEAD, and thus potentially provide a means to identify patients likely to benefit from TEAD inhibitors.
Collapse
Affiliation(s)
- Ryan Kanai
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| | - Emily Norton
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| | - Patrick Stern
- Koch Institute for Integrative Cancer Research, at Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Richard O. Hynes
- Department of Biology, Koch Institute for Integrative Cancer Research, and Howard Hughes Medical Institute, at Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - John M. Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| |
Collapse
|
17
|
Li A, Zhang T, Zhang X, Xu Z, Liu H, Yuan M, Wei X, Zhu Y, Tu W, Jiang X, He Y. Flexocatalytic Reduction of Tumor Interstitial Fluid/Solid Pressure for Efficient Nanodrug Penetration. ACS NANO 2024. [PMID: 38330150 DOI: 10.1021/acsnano.3c09316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
The practical efficacy of nanomedicines for treating solid tumors is frequently low, predominantly due to the elevated interstitial pressure within such tumors that obstructs the penetration of nanomedicines. This increased interstitial pressure originates from both liquid and solid stresses related to an undeveloped vascular network and excessive fibroblast proliferation. To specifically resolve the penetration issues of nanomedicines for tumor treatment, this study introduces a holistic "dual-faceted" approach. A treatment platform predicated on the WS2/Pt Schottky heterojunction was adopted, and flexocatalysis technology was used to disintegrate tumor interstitial fluids, thus producing oxygen and reactive oxygen species and effectively mitigating the interstitial fluid pressure. The chemotherapeutic agent curcumin was incorporated to further suppress the activity of cancer-associated fibroblasts, minimize collagen deposition in the extracellular matrix, and alleviate solid stress. Nanomedicines achieve homologous targeting by enveloping the tumor cell membrane. It was found that this multidimensional strategy not only alleviated the high-pressure milieu of the tumor interstitium─which enhanced the efficiency of nanomedicine delivery─but also triggered tumor cell apoptosis via the generated reactive oxygen species and modulated the tumor microenvironment. This, in turn, amplified immune responses, substantially optimizing the therapeutic impacts of nanomedicines.
Collapse
Affiliation(s)
- Anshuo Li
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Heavy Metal Deep-Remediation in Water and Resource Reuse Key Lab of Hebei, Yanshan University, Qinhuangdao 066004, China
- Department of Prosthodontics, 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 Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China
| | - Tiantian Zhang
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Heavy Metal Deep-Remediation in Water and Resource Reuse Key Lab of Hebei, Yanshan University, Qinhuangdao 066004, China
| | - Xuwu Zhang
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Heavy Metal Deep-Remediation in Water and Resource Reuse Key Lab of Hebei, Yanshan University, Qinhuangdao 066004, China
| | - Zichuang Xu
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Heavy Metal Deep-Remediation in Water and Resource Reuse Key Lab of Hebei, Yanshan University, Qinhuangdao 066004, China
| | - Hengrui Liu
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Heavy Metal Deep-Remediation in Water and Resource Reuse Key Lab of Hebei, Yanshan University, Qinhuangdao 066004, China
| | - Meng Yuan
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Heavy Metal Deep-Remediation in Water and Resource Reuse Key Lab of Hebei, Yanshan University, Qinhuangdao 066004, China
| | - Xindi Wei
- Department of Oral and Maxillo-facial Implantology, 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 200011, China
| | - Yuhui Zhu
- Department of Prosthodontics, 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 Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China
| | - Wenkang Tu
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Heavy Metal Deep-Remediation in Water and Resource Reuse Key Lab of Hebei, Yanshan University, Qinhuangdao 066004, China
| | - Xinquan Jiang
- Department of Prosthodontics, 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 Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China
| | - Yuchu He
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Heavy Metal Deep-Remediation in Water and Resource Reuse Key Lab of Hebei, Yanshan University, Qinhuangdao 066004, China
| |
Collapse
|
18
|
Liu Q, Zhang C, Chen X, Han Z. Modern cancer therapy: cryoablation meets immune checkpoint blockade. Front Oncol 2024; 14:1323070. [PMID: 38384806 PMCID: PMC10881233 DOI: 10.3389/fonc.2024.1323070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
Cryoablation, as a minimally invasive technology for the treatment of tumors, destroys target tumors with lethal low temperatures. It simultaneously releases a large number of tumor-specific antigens, pro-inflammatory cytokines, and nucleoproteins, known as "danger signals", activating the body's innate and adaptive immune responses. However, tumor cells can promote the inactivation of immune effector cells by reprogramming immune checkpoints, leading to the insufficiency of these antigens to induce an immune response capable of eradicating the tumor. Immune checkpoint blockers rejuvenate exhausted T cells by blocking immune checkpoints that induce programmed death of T cells, and are therefore considered a promising therapeutic strategy to enhance the immune effects of cryoablation. In this review, we provide a detailed explanation of the immunological mechanisms of cryoablation and articulate the theoretical basis and research progress of the treatment of cancer with cryoablation combined with immune checkpoint blockers. Preliminary data indicates that this combined treatment strategy exhibits good synergy and has been proven to be safe and effective.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Chunyang Zhang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
19
|
Luo ZY, Tian Q, Cheng NM, Liu WH, Yang Y, Chen W, Zhang XZ, Zheng XY, Chen MS, Zhuang QY, Zhao BX, Liu CS, Liu XL, Li Q, Wang YC. Pien Tze Huang Inhibits Migration and Invasion of Hepatocellular Carcinoma Cells by Repressing PDGFRB/YAP/CCN2 Axis Activity. Chin J Integr Med 2024; 30:115-124. [PMID: 35947230 DOI: 10.1007/s11655-022-3533-8] [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] [Accepted: 01/28/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate the effects of Pien Tze Huang (PZH) on the migration and invasion of HCC cells and underlying molecular mechanism. METHODS Cell counting kit-8 (CCK-8) was applied to evaluate the cell viabilities of SMMC-7721, SK-Hep-1, C3A and HL-7702 (6 × 103 cells/well) co-incubated with different concentrations of PZH (0, 0.2, 0.4, 0.6, 0.8 mg/mL) for 24 h. Transwell, wound healing assay, CCK-8 and Annexin V-FITC/PI staining were conducted to investigate the effects of PZH on the migration, invasion, proliferation and apoptosis of SK-Hep-1 and SMMC-7721 cells (650 µ g/mL for SK-Hep-1 cells and 330 µ g/mL for SMMC-7721 cells), respectively. In vivo, lung metastasis mouse model constructed by tail vein injection of HCC cells was used for evaluating the anti-metastasis function of PZH. SK-Hep-1 cells (106 cells/200 µ L per mice) were injected into B-NDG mice via tail vein. Totally 8 mice were randomly divided into PZH and control groups, 4 mice in each group. After 2-d inoculation, mice in the PZH group were administered with PZH (250 mg/kg, daily) and mice in the control group received only vehicle (PBS) from the 2nd day after xenograft to day 17. Transcriptome analysis based on RNA-seq was subsequently used for deciphering anti-tumor mechanism of PZH. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were applied to verify RNA-seq results. Luciferase reporter assay was performed to examine the transcriptional activity of yes-associated protein (YAP). RESULTS PZH treatment significantly inhibited the migration, invasion, proliferation and promoted the apoptosis of HCC cells in vitro and in vivo (P<0.01). Transcriptome analysis indicated that Hippo signaling pathway was associated with anti-metastasis function of PZH. Mechanical study showed PZH significantly inhibited the expressions of platelet derived growth factor receptor beta (PDGFRB), YAP, connective tissue growth factor (CCN2), N-cadherin, vimentin and matrix metallopeptidase 2 (MMP2, P<0.01). Meanwhile, the phosphorylation of YAP was also enhanced by PZH treatment in vitro and in vivo. Furthermore, PZH played roles in inhibiting the transcriptional activity of YAP. CONCLUSION PZH restrained migration, invasion and epithelial-mesenchymal transition of HCC cells through repressing PDGFRB/YAP/CCN2 axis.
Collapse
Affiliation(s)
- Zhi-Yi Luo
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province, 363099, China
| | - Qi Tian
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| | - Niang-Mei Cheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| | - Wen-Han Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Ye Yang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| | - Wei Chen
- Department of Internal Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Xiang-Zhi Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| | - Xiao-Yuan Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| | - Ming-Sheng Chen
- Department of Internal Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Qiu-Yu Zhuang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| | - Bi-Xing Zhao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| | - Cong-Sheng Liu
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province, 363099, China
| | - Xiao-Long Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| | - Qin Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China.
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China.
- Department of Internal Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China.
| | - Ying-Chao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- College of Biological Science and Engineering and Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, China
| |
Collapse
|
20
|
Fernandes S, Cassani M, Cavalieri F, Forte G, Caruso F. Emerging Strategies for Immunotherapy of Solid Tumors Using Lipid-Based Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305769. [PMID: 38054651 PMCID: PMC10885677 DOI: 10.1002/advs.202305769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/09/2023] [Indexed: 12/07/2023]
Abstract
The application of lipid-based nanoparticles for COVID-19 vaccines and transthyretin-mediated amyloidosis treatment have highlighted their potential for translation to cancer therapy. However, their use in delivering drugs to solid tumors is limited by ineffective targeting, heterogeneous organ distribution, systemic inflammatory responses, and insufficient drug accumulation at the tumor. Instead, the use of lipid-based nanoparticles to remotely activate immune system responses is an emerging effective strategy. Despite this approach showing potential for treating hematological cancers, its application to treat solid tumors is hampered by the selection of eligible targets, tumor heterogeneity, and ineffective penetration of activated T cells within the tumor. Notwithstanding, the use of lipid-based nanoparticles for immunotherapy is projected to revolutionize cancer therapy, with the ultimate goal of rendering cancer a chronic disease. However, the translational success is likely to depend on the use of predictive tumor models in preclinical studies, simulating the complexity of the tumor microenvironment (e.g., the fibrotic extracellular matrix that impairs therapeutic outcomes) and stimulating tumor progression. This review compiles recent advances in the field of antitumor lipid-based nanoparticles and highlights emerging therapeutic approaches (e.g., mechanotherapy) to modulate tumor stiffness and improve T cell infiltration, and the use of organoids to better guide therapeutic outcomes.
Collapse
Affiliation(s)
- Soraia Fernandes
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Marco Cassani
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Giancarlo Forte
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonSE5 9NUUK
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| |
Collapse
|
21
|
Choi KM, Kim B, Lee SM, Han J, Bae HS, Han SB, Lee D, Ham IH, Hur H, Kim E, Kim JY. Characterization of gastric cancer-stimulated signaling pathways and function of CTGF in cancer-associated fibroblasts. Cell Commun Signal 2024; 22:8. [PMID: 38167009 PMCID: PMC10763493 DOI: 10.1186/s12964-023-01396-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/12/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are key components of the tumor microenvironment (TME) that play an important role in cancer progression. Although the mechanism by which CAFs promote tumorigenesis has been well investigated, the underlying mechanism of CAFs activation by neighboring cancer cells remains elusive. In this study, we aim to investigate the signaling pathways involved in CAFs activation by gastric cancer cells (GC) and to provide insights into the therapeutic targeting of CAFs for overcoming GC. METHODS Alteration of receptor tyrosine kinase (RTK) activity in CAFs was analyzed using phospho-RTK array. The expression of CAFs effector genes was determined by RT-qPCR or ELISA. The migration and invasion of GC cells co-cultured with CAFs were examined by transwell migration/invasion assay. RESULTS We found that conditioned media (CM) from GC cells could activate multiple receptor tyrosine kinase signaling pathways, including ERK, AKT, and STAT3. Phospho-RTK array analysis showed that CM from GC cells activated PDGFR tyrosine phosphorylation, but only AKT activation was PDGFR-dependent. Furthermore, we found that connective tissue growth factor (CTGF), a member of the CCN family, was the most pronouncedly induced CAFs effector gene by GC cells. Knockdown of CTGF impaired the ability of CAFs to promote GC cell migration and invasion. Although the PDGFR-AKT pathway was pronouncedly activated in CAFs stimulated by GC cells, its pharmacological inhibition affected neither CTGF induction nor CAFs-induced GC cell migration. Unexpectedly, the knockdown of SRC and SRC-family kinase inhibitors, dasatinib and saracatinib, significantly impaired CTGF induction in activated CAFs and the migration of GC cells co-cultured with CAFs. SRC inhibitors restored the reduced expression of epithelial markers, E-cadherin and Zonula Occludens-1 (ZO-1), in GC cells co-cultured with CAFs, as well as CAFs-induced aggregate formation in a 3D tumor spheroid model. CONCLUSIONS This study provides a characterization of the signaling pathways and effector genes involved in CAFs activation, and strategies that could effectively inhibit it in the context of GC. Video Abstract.
Collapse
Affiliation(s)
- Kyoung-Min Choi
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, South Korea
| | - Boram Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, South Korea
| | - Su-Min Lee
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, South Korea
| | - Jisoo Han
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, South Korea
| | - Ha-Song Bae
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, South Korea
| | - Su-Bhin Han
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, South Korea
| | - Dagyeong Lee
- Department of Surgery, Ajou University School of Medicine, Suwon, South Korea
- Inflamm-Aging Translational Research Center, Ajou University School of Medicine, Suwon, South Korea
- AI-Super Convergence KIURI Translational Research Center, Suwon, South Korea
| | - In-Hye Ham
- Department of Surgery, Ajou University School of Medicine, Suwon, South Korea
- Inflamm-Aging Translational Research Center, Ajou University School of Medicine, Suwon, South Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, South Korea
- Inflamm-Aging Translational Research Center, Ajou University School of Medicine, Suwon, South Korea
| | - Eunjung Kim
- Natural Product Informatics Center, Korea Institute of Science and Technology (KIST), Gangneung, South Korea
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, South Korea.
| |
Collapse
|
22
|
Lu Q, Wang Y, Jiang X, Huang S. miR-584-5p Inhibits Osteosarcoma Progression by Targeting Connective Tissue Growth Factor. Cancer Biother Radiopharm 2023; 38:632-640. [PMID: 35041486 DOI: 10.1089/cbr.2021.0349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: miR-584-5p is a critical regulator in the progression of multiple cancers. However, its specific role and downstream targets in osteosarcoma are unclear. This research investigated the roles and underlying mechanisms of miR-769-5p and the Hippo pathway in osteosarcoma cells. Materials and Methods: RT-qPCR, CCK-8 and EdU and colony formation, wound-healing and transwell chamber, flow cytometry, and Western blot assay detected the expression of miR-584-5p and CTGF, cell proliferation, migration, invasion apoptosis and protein expression. Result: Their study illuminated that miR-584-5p overexpression repressed osteosarcoma cell migration/invasion and proliferation and facilitated apoptosis. Mechanistically, miR-584-5p targets negatively regulated connective tissue growth factor (CTGF). miR-584-5p inhibited osteosarcoma cell metastasis by regulating CTGF. In addition, miR-584-5p inactivated the Hippo pathway through CTGF in osteosarcoma. Conclusion: miR-584-5p inhibits osteosarcoma cell proliferation, migration, and invasion and promotes apoptosis by targeting CTGF, indicating that miR-584-5p acts as a promising diagnostic and predictive biomarker for osteosarcoma.
Collapse
Affiliation(s)
- Qian Lu
- Department of Orthopaedic Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, China
| | - Yongli Wang
- Department of Orthopaedic Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, China
| | - Xuesheng Jiang
- Department of Orthopaedic Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, China
| | - Sheng Huang
- Department of Orthopaedic Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
23
|
Di Carlo SE, Raffenne J, Varet H, Ode A, Granados DC, Stein M, Legendre R, Tuckermann J, Bousquet C, Peduto L. Depletion of slow-cycling PDGFRα +ADAM12 + mesenchymal cells promotes antitumor immunity by restricting macrophage efferocytosis. Nat Immunol 2023; 24:1867-1878. [PMID: 37798557 PMCID: PMC10602852 DOI: 10.1038/s41590-023-01642-7] [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: 06/16/2022] [Accepted: 09/07/2023] [Indexed: 10/07/2023]
Abstract
The capacity to survive and thrive in conditions of limited resources and high inflammation is a major driver of tumor malignancy. Here we identified slow-cycling ADAM12+PDGFRα+ mesenchymal stromal cells (MSCs) induced at the tumor margins in mouse models of melanoma, pancreatic cancer and prostate cancer. Using inducible lineage tracing and transcriptomics, we demonstrated that metabolically altered ADAM12+ MSCs induced pathological angiogenesis and immunosuppression by promoting macrophage efferocytosis and polarization through overexpression of genes such as Gas6, Lgals3 and Csf1. Genetic depletion of ADAM12+ cells restored a functional tumor vasculature, reduced hypoxia and acidosis and normalized CAFs, inducing infiltration of effector T cells and growth inhibition of melanomas and pancreatic neuroendocrine cancer, in a process dependent on TGF-β. In human cancer, ADAM12 stratifies patients with high levels of hypoxia and innate resistance mechanisms, as well as factors associated with a poor prognosis and drug resistance such as AXL. Altogether, our data show that depletion of tumor-induced slow-cycling PDGFRα+ MSCs through ADAM12 restores antitumor immunity.
Collapse
Affiliation(s)
- Selene E Di Carlo
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Jerome Raffenne
- INSERM U1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Hugo Varet
- Transcriptome and Epigenome Platform-Biomics Pole, Institut Pasteur, Université Paris Cité, Paris, France
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Anais Ode
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - David Cabrerizo Granados
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
- Laboratory for Disease Mechanisms in Cancer, KU Leuven, Leuven, Belgium
| | - Merle Stein
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Rachel Legendre
- Transcriptome and Epigenome Platform-Biomics Pole, Institut Pasteur, Université Paris Cité, Paris, France
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Corinne Bousquet
- INSERM U1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Lucie Peduto
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France.
| |
Collapse
|
24
|
Cirillo F, Talia M, Santolla MF, Pellegrino M, Scordamaglia D, Spinelli A, De Rosis S, Giordano F, Muglia L, Zicarelli A, Di Dio M, Rigiracciolo DC, Miglietta AM, Filippelli G, De Francesco EM, Belfiore A, Lappano R, Maggiolini M. GPER deletion triggers inhibitory effects in triple negative breast cancer (TNBC) cells through the JNK/c-Jun/p53/Noxa transduction pathway. Cell Death Discov 2023; 9:353. [PMID: 37749101 PMCID: PMC10520078 DOI: 10.1038/s41420-023-01654-0] [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: 05/10/2023] [Revised: 06/28/2023] [Accepted: 09/18/2023] [Indexed: 09/27/2023] Open
Abstract
The G protein-coupled estrogen receptor (GPER) mediates estrogen action in different pathophysiological conditions, including cancer. GPER expression and signaling have been found to join in the progression of triple-negative breast cancer (TNBC), even though controversial data have been reported. In present study, we aimed at providing new mechanistic and biological discoveries knocking out (KO) GPER expression by CRISPR/Cas9 technology in MDA-MB-231 TNBC cells. GPER KO whole transcriptome respect to wild type (WT) MDA-MB-231 cells was determined through total RNA sequencing (RNA-Seq) and gene ontology (GO) enrichment analysis. We ascertained that anti-proliferative and pro-apoptotic gene signatures characterize GPER KO MDA-MB-231 cells. Thereafter, we determined that these cells exhibit a reduced proliferative, clonogenic and self-renewal potential along with an increased mitochondria-dependent apoptosis phenotype. In addition, we recognized that decreased cAMP levels trigger the JNK/c-Jun/p53/Noxa axis, which in turn orchestrates the pro-apoptotic effects observed in GPER KO cells. In accordance with these data, survival analyses in TNBC patients of the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset indicated that high Noxa expression correlates with improved outcomes in TNBC patients. Furthermore, we demonstrated that GPER KO in TNBC cells impairs the expression and secretion of the well-acknowledged GPER target gene named CTGF, thus resulting in the inhibition of migratory effects in cancer-associated fibroblasts (CAFs). Overall, the present study provides novel mechanistic and biological insights on GPER KO in TNBC cells suggesting that GPER may be considered as a valuable target in comprehensive therapeutic approaches halting TNBC progression.
Collapse
Affiliation(s)
- Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Maria Francesca Santolla
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Michele Pellegrino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Domenica Scordamaglia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Asia Spinelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Salvatore De Rosis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Lucia Muglia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Azzurra Zicarelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Marika Di Dio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Damiano Cosimo Rigiracciolo
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milano, Italy
| | - Anna Maria Miglietta
- Breast and General Surgery Unit, Regional Hospital Cosenza, 87100, Cosenza, Italy
| | | | - Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122, Catania, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122, Catania, Italy
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| |
Collapse
|
25
|
Foda BM, Neubig RR. Role of Rho/MRTF in Aggressive Vemurafenib-Resistant Murine Melanomas and Immune Checkpoint Upregulation. Int J Mol Sci 2023; 24:13785. [PMID: 37762086 PMCID: PMC10531039 DOI: 10.3390/ijms241813785] [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: 08/04/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Cutaneous melanoma is the deadliest skin cancer. Most have Ras-MAPK pathway (BRAFV600E or NRAS) mutations and highly effective targeted therapies exist; however, they and immune therapies are limited by resistance, in part driven by small GTPase (Rho and Rac) activation. To facilitate preclinical studies of combination therapies to provide durable responses, we describe the first mouse melanoma lines resistant to BRAF inhibitors. Treatment of mouse lines, YUMM1.7 and YUMMER, with vemurafenib (Vem), the BRAFV600E-selective inhibitor, resulted in high-level resistance (IC50 shifts 20-30-fold). Resistant cells showed enhanced activation of Rho and the downstream transcriptional coactivator, myocardin-related transcription factor (MRTF). Resistant cells exhibited increased stress fibers, nuclear translocation of MRTF-A, and an increased MRTF-A gene signature. Pharmacological inhibition of the Rho/MRTF pathway using CCG-257081 reduced viability of resistant lines and enhanced sensitivity to Vem. Remarkably, co-treatment of parental lines with Vem and CCG-257081 eliminated resistant colony development. Resistant cells grew more slowly in vitro, but they developed highly aggressive tumors with a shortened survival of tumor-bearing mice. Increased expression of immune checkpoint inhibitor proteins (ICIs) in resistant lines may contribute to aggressive in vivo behavior. Here, we introduce the first drug-resistant mouse melanoma models for assessing combinations of targeted and immune therapies.
Collapse
Affiliation(s)
- Bardees M. Foda
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48823, USA;
- Molecular Genetics and Enzymology Department, National Research Centre, Dokki 12622, Egypt
| | - Richard R. Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48823, USA;
- Nicholas V. Perricone, M.D. Division of Dermatology, Department of Medicine, Michigan State University, East Lansing, MI 48823, USA
| |
Collapse
|
26
|
Zhai L, Yang X, Dong J, Qian L, Gao Y, Lv Y, Chen L, Chen B, Zhou F. O‑GlcNAcylation mediates endometrial cancer progression by regulating the Hippo‑YAP pathway. Int J Oncol 2023; 63:90. [PMID: 37350405 PMCID: PMC10552701 DOI: 10.3892/ijo.2023.5538] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/24/2023] [Indexed: 06/24/2023] Open
Abstract
The incidence of endometrial cancer (EC) is rapidly increasing worldwide. The majority of endometrial cancers are diagnosed at an early stage and are associated with a good prognosis; however, patients with advanced‑stage EC have a poor prognosis and present with invasive metastasis. The mechanisms responsible for the invasion and metastasis of endometrial cancer remain unknown. Here, the present study aimed to examine the effects of O‑GlcNAcylation on the malignancy of EC and its association with Yes‑associated protein (YAP). It was found that the expression of O‑GlcNAc transferase (OGT) and O‑GlcNAcylation were increased in EC tissues; the decrease in O‑GlcNAcylation levels was found to lead to the decreased proliferation, migration and invasion of EC cells. Mass spectrometric analysis revealed that OGT knockdown reduced the O‑GlcNAcylation of YAP. Furthermore, it was found that the reduction in the O‑GlcNAcylation of YAP promoted its phosphorylation, which in turn inhibited the access of YAP to the nucleus and downstream target gene activation, demonstrating that the level of O‑GlcNAcylation affects the development of EC. On the whole, the findings of the present study indicate that YAP is a key molecule linking the O‑GlcNAcylation and Hippo pathways, which together regulate the progression of EC.
Collapse
Affiliation(s)
- Lianghao Zhai
- Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032
| | - Xiaoshan Yang
- Stomatology Hospital, Southern Medical University, Guangzhou, Guangdong 510280
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi 710032
| | - Jian Dong
- Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032
| | - Luomeng Qian
- Department of Medicine, Nankai University, Tianjin 300071, P.R. China
| | - Yunge Gao
- Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032
| | - Yanhong Lv
- Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032
| | - Ligang Chen
- Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032
| | - Biliang Chen
- Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032
| | - Fuxing Zhou
- Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032
| |
Collapse
|
27
|
Yeger H. CCN proteins: opportunities for clinical studies-a personal perspective. J Cell Commun Signal 2023:10.1007/s12079-023-00761-y. [PMID: 37195381 DOI: 10.1007/s12079-023-00761-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/01/2023] [Indexed: 05/18/2023] Open
Abstract
The diverse members of the CCN family now designated as CCN1(CYR61), CCN2 (CTGF), CCN3(NOV), CCN4(WISP1), CCN5(WISP2), CCN6(WISP3) are a conserved matricellular family of proteins exhibiting a spectrum of functional properties throughout all organs in the body. Interaction with cell membrane receptors such as integrins trigger intracellular signaling pathways. Proteolytically cleaved fragments (constituting the active domains) can be transported to the nucleus and perform transcriptional relevant functional activities. Notably, as also found in other protein families some members act opposite to others creating a system of functionally relevant checks and balances. It has become apparent that these proteins are secreted into the circulation, are quantifiable, and can serve as disease biomarkers. How they might also serve as homeostatic regulators is just becoming appreciated. In this review I have attempted to highlight the most recent evidence under the subcategories of cancer and non-cancer relevant that could lead to potential therapeutic approaches or ideas that can be factored into clinical advances. I have added my own personal perspective on feasibility.
Collapse
Affiliation(s)
- Herman Yeger
- Developmental and Stem Cell Biology, Research Institute, SickKids, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Dhungel N, Youngblood R, Chu M, Carroll J, Dragoi AM. Assessing the epithelial-to-mesenchymal plasticity in a small cell lung carcinoma (SCLC) and lung fibroblasts co-culture model. Front Mol Biosci 2023; 10:1096326. [PMID: 36936987 PMCID: PMC10022497 DOI: 10.3389/fmolb.2023.1096326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
The tumor microenvironment (TME) is the source of important cues that govern epithelial-to-mesenchymal transition (EMT) and facilitate the acquisition of aggressive traits by cancer cells. It is now recognized that EMT is not a binary program, and cancer cells rarely switch to a fully mesenchymal phenotype. Rather, cancer cells exist in multiple hybrid epithelial/mesenchymal (E/M) states responsible for cell population heterogeneity, which is advantageous for the ever-changing environment during tumor development and metastasis. How are these intermediate states generated and maintained is not fully understood. Here, we show that direct interaction between small cell lung carcinoma cells and lung fibroblasts induces a hybrid EMT phenotype in cancer cells in which several mesenchymal genes involved in receptor interaction with the extracellular matrix (ECM) and ECM remodeling are upregulated while epithelial genes such as E-cadherin remain unchanged or slightly increase. We also demonstrate that several core EMT-regulating transcription factors (EMT-TFs) are upregulated in cancer cells during direct contact with fibroblasts, as is Yes-associated protein (YAP1), a major regulator of the Hippo pathway. Further, we show that these changes are transient and reverse to the initial state once the interaction is disrupted. Altogether, our results provide evidence that tumor cells' direct contact with the fibroblasts in the TME initiates a signaling cascade responsible for hybrid E/M states of cancer cells. These hybrid states are maintained during the interaction and possibly contribute to therapy resistance and immune evasion, while interference with direct contact will result in slow recovery and switch to the initial states.
Collapse
Affiliation(s)
- Nilu Dhungel
- Department of Molecular and Cellular Physiology, LSUHSC-Shreveport, Shreveport, LA, United States
| | - Reneau Youngblood
- Department of Molecular and Cellular Physiology, LSUHSC-Shreveport, Shreveport, LA, United States
| | - Min Chu
- Feist-Weiller Cancer Center, INLET Core, LSUHSC-Shreveport, Shreveport, LA, United States
| | - Jennifer Carroll
- Center for Emerging Viral Threats (CEVT), LSUHSC-Shreveport, Shreveport, LA, United States
| | - Ana-Maria Dragoi
- Department of Molecular and Cellular Physiology, LSUHSC-Shreveport, Shreveport, LA, United States
- Feist-Weiller Cancer Center, INLET Core, LSUHSC-Shreveport, Shreveport, LA, United States
| |
Collapse
|
29
|
Wang W, Tang X, Duan C, Tian S, Han C, Qian W, Jiang X, Hou X, Lin R. Intestinal epithelium-specific Fut2 deficiency promotes colorectal cancer through down-regulating fucosylation of MCAM. J Transl Med 2023; 21:82. [PMID: 36739428 PMCID: PMC9899399 DOI: 10.1186/s12967-023-03906-0] [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: 11/18/2022] [Accepted: 01/19/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Our previous study showed that fucosyltransferase 2 (Fut2) deficiency is closely related to colitis. Colitis increases the risk for the development of colorectal cancer (CRC). This study aimed to investigate the effect and underlying mechanism of action of Fut2 in CRC. METHODS Intestinal epithelium-specific Fut2 knockout (Fut2△IEC) mice were used in this study. CRC was induced using azoxymethane (AOM) and dextran sulfate sodium (DSS). Immunofluorescence was used to examine the fucosylation levels. Proteomics and N-glycoproteomics analyses, Ulex Europaeus Agglutinin I (UEA-I) affinity chromatography, immunoprecipitation, and rescue assay were used to investigate the mechanism of Fut2 in CRC. RESULTS The expression of Fut2 and α-1,2-fucosylation was lower in colorectal tumor tissues than in the adjacent normal tissues of AOM/DSS-induced CRC mice. More colorectal tumors were detected in Fut2△IEC mice than in control mice, and significant downregulation of melanoma cell adhesion molecule (MCAM) fucosylation was detected in the colorectal tumor tissues of Fut2△IEC mice. Overexpression of Fut2 inhibited cell proliferation, invasion and tumor metastasis in vivo and in vitro in SW480 and HCT116 cells. Moreover, fucosylation of MCAM may be a mediator of Fut2 in CRC. Peracetylated 2-F-Fuc, a fucosyltransferase inhibitor, repressed fucosylation modification of MCAM and reversed the inhibitory effects of Fut2 overexpression on SW480 cell proliferation, migration, and invasion. Our results indicate that Fut2 deficiency in the intestinal epithelium promotes CRC by downregulating the fucosylation of MCAM. CONCLUSIONS The regulation of fucosylation may be an potential therapy for CRC, especially in patients with Fut2 gene defects.
Collapse
Affiliation(s)
- Weijun Wang
- grid.33199.310000 0004 0368 7223Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Xuelian Tang
- grid.33199.310000 0004 0368 7223Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Caihan Duan
- grid.33199.310000 0004 0368 7223Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Shuxin Tian
- grid.33199.310000 0004 0368 7223Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,grid.411680.a0000 0001 0514 4044Department of Gastroenterology, The First Affiliated Hospital of Medical College, Shihezi University, Shihezi, 832008 China
| | - Chaoqun Han
- grid.33199.310000 0004 0368 7223Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Wei Qian
- grid.33199.310000 0004 0368 7223Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Xin Jiang
- grid.33199.310000 0004 0368 7223Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Xiaohua Hou
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
30
|
Janczi T, Fehrl Y, Kinne RW, Böhm B, Burkhardt H. The role of YAP1 target gene CTGF in the anoikis resistance of rheumatoid arthritis synovial fibroblasts. Rheumatology (Oxford) 2023; 62:850-860. [PMID: 35713503 DOI: 10.1093/rheumatology/keac354] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To analyse pro-survival mechanisms elicited in RA synovial fibroblasts (RASFs) upon detachment from their extracellular matrix dependent on the disintegrin metalloproteinase ADAM15 and Yes-associated protein kinase 1 (YAP1). METHODS Detachment-induced apoptosis was determined by caspase 3/7 assays. Immunofluorescent stainings, cell surface biotinylation and immunoblotting were applied to analyse phosphorylated kinases and subcellular localization of YAP1 and connective tissue growth factor (CTGF). Caspase and transwell transmigration assays served to study CTGF function. RESULTS Silencing of ADAM15 or YAP1 in RASFs leads to significantly increased levels of detachment-induced caspase activity. In non-silenced RASFs detachment causes simultaneous ADAM15-enhanced phosphorylation of YAP1 at S127, known for promoting its cytoplasmic localization, and Src-dependent phosphorylation at tyrosine Y357. The majority of nuclear YAP1 leaves the nucleus shortly after cell detachment, but prolonged detachment causes a marked nuclear re-entry of YAP1, resulting in significantly increased synthesis of CTGF. The newly synthesized CTGF, however, is not detectable in the supernatant, but is bound to the outside of the plasma membrane. In vitro studies demonstrated autocrine binding of CTGF to the EGF receptor and β1 integrin, with concomitant triggering of survival kinases, AKT1, ERK1/2, Src and focal adhesion kinase. Functional studies revealed anti-apoptotic effects of CTGF on detached RASFs and an enhancement of their potential for endothelial transmigration using HUVEC-coated transwells. CONCLUSION The elucidation of a new molecular mechanism that protects RASFs in the highly pro-apoptotic environment of inflamed RA joints by promoting anoikis-resistance and transendothelial migration via ADAM15/YAP1-mediated CTGF upregulation uncovers potentially new targets for future therapeutic intervention.
Collapse
Affiliation(s)
- Tomasz Janczi
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, Frankfurt am Main
| | - Yuliya Fehrl
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, Frankfurt am Main
| | - Raimund W Kinne
- Experimental Rheumatology Unit, Jena University Hospital, Waldkliniken Eisenberg GmbH, Eisenberg
| | - Beate Böhm
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, Frankfurt am Main
| | - Harald Burkhardt
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, Frankfurt am Main.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP.,Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Frankfurt am Main, Germany
| |
Collapse
|
31
|
Dong L, Wang H, Gao Y, Wang S, Wang W. Long non-coding RNA PVT1 promotes the proliferation, migration and EMT process of ovarian cancer cells by regulating CTGF. Oncol Lett 2022; 25:71. [PMID: 36688109 PMCID: PMC9843302 DOI: 10.3892/ol.2022.13657] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 11/01/2022] [Indexed: 01/01/2023] Open
Abstract
Ovarian cancer remains one of the most common gynecological malignancies with a poor prognosis. The present study investigated the roles of long non-coding RNA plasmacytoma variant translocation 1 (lncRNA PVT1) in the regulation of the malignant phenotype of ovarian cancer cells, including cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT). SKOV3 and CAOV3 cells were transfected with small interfering RNA (siRNA) targeting lncRNA PVT1 (si-PVT1) or control siRNA and the si-PVT1 transfected cells were co-cultured with recombinant human connective tissue growth factor (rhCTGF). The proliferation, migration and invasion abilities of the cells were examined via Cell Counting Kit-8, colony formation, wound-healing and Transwell assays. The relative expression levels of lncRNA PVT1, CTGF, E-cadherin and vimentin were analyzed using reverse transfection-quantitative polymerase chain reaction, and western blotting was employed to detect the protein levels of CTGF, E-cadherin and vimentin. The expression of lncRNA PVT1 was significantly reduced in SKOV3 and CAOV3 cells following transfection with si-PVT1. In addition, the proliferation, migration and invasion abilities of SKOV3 and CAOV3 cells were repressed following lncRNA PVT1 knockdown. The knockdown of lncRNA PVT1 also reduced the expression of CTGF and vimentin, and increased the expression of E-cadherin. The changes in the proliferation, migration and invasion of the cells induced by transfection with si-PVT1 were partially attenuated in the presence of rhCTGF. Furthermore, co-culture with rhCTGF reversed the si-PVT1-induced changes in the expression of EMT-associated proteins. In conclusion, lncRNA PVT1 promotes the proliferation, migration, invasiveness and EMT process of ovarian cancer cells, and CTGF contributes to the effect of lncRNA PVT1.
Collapse
Affiliation(s)
- Lingling Dong
- Department of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China,Department of Oncology, Weifang Hospital of Traditional Chinese Medicine, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Huan Wang
- College of Life Science and Technology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Yun Gao
- College of Public Health and Management, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Shuai Wang
- Department of Radiotherapy, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Weibo Wang
- Department of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China,Correspondence to: Dr Weibo Wang, Department of Oncology, Shandong Provincial Hospital, Shandong University, 324 Jingwu Road, Huaiyin, Jinan, Shandong 250021, P.R. China, E-mail:
| |
Collapse
|
32
|
Tang SJ, Fan KH, You GR, Huang SF, Kang CJ, Huang YF, Huang YC, Chang JTC, Cheng AJ. Tumor Suppressor miRNA-503 Inhibits Cell Invasion in Head and Neck Cancer through the Wnt Signaling Pathway via the WNT3A/MMP Molecular Axis. Int J Mol Sci 2022; 23:15900. [PMID: 36555553 PMCID: PMC9786678 DOI: 10.3390/ijms232415900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Head and neck cancer (HNC) is the fifth most common cancer worldwide, and its incidence and death rates have been consistently high throughout the past decades. MicroRNAs (miRNAs) have recently gained significant attention because of their role in the regulation of a variety of biological processes via post-transcriptional silencing mechanisms. Previously, we determined a specific profile of miRNAs associated with HNC using a miRNA microarray analysis. Of the 23 miRNAs with highly altered expression in HNC cells, miR-503 was the most significantly downregulated miRNA. In this study, we confirmed that miR-503 acts as a tumor suppressor, as our results showed decreased levels of miR-503 in cancer cells and patients with HNC. We further characterized the role of miR-503 in the malignant functions of HNC. Although there was a minimal effect on cell growth, miR-503 was found to inhibit cellular invasion significantly. Algorithm-based studies identified multiple potential target genes and pathways associated with oncogenic mechanisms. The candidate target gene, WNT3A, was confirmed to be downregulated by miR-503 at both the mRNA and protein levels and validated by a reporter assay. Furthermore, miR-503 modulated multiple invasion-associated genes, including matrix metalloproteinases (MMPs), through the Wnt downstream signaling pathway. Overall, this study demonstrates that miR-503 suppresses HNC malignancy by inhibiting cell invasion through the Wnt signaling pathway via the WNT3A/MMP molecular axis. The modulation of miR-503 may be a novel therapeutic approach to intervene in cancer invasion.
Collapse
Affiliation(s)
- Shang-Ju Tang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Kang-Hsing Fan
- Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital, New Taipei City 236017, Taiwan
| | - Guo-Rung You
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shiang-Fu Huang
- Department of Otorhinolaryngology—Head and Neck Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Jan Kang
- Department of Otorhinolaryngology—Head and Neck Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
| | - Yi-Fang Huang
- Department of General Dentistry, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yu-Chen Huang
- Department of General Dentistry, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Joseph Tung-Chieh Chang
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
| |
Collapse
|
33
|
Jiang X, Zhang X, Jiang N, Sun Y, Li T, Zhang J, Shen Y, Cao J. The single-cell landscape of cystic echinococcosis in different stages provided insights into endothelial and immune cell heterogeneity. Front Immunol 2022; 13:1067338. [PMID: 36569953 PMCID: PMC9772464 DOI: 10.3389/fimmu.2022.1067338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Hydatid cysts and angiogenesis are the key characteristics of cystic echinococcosis, with immune cells and endothelial cells mediating essential roles in disease progression. Recent single-cell analysis studies demonstrated immune cell infiltration after Echinococcus granulosus infection, highlighting the diagnostic and therapeutic potential of targeting certain cell types in the lesion microenvironment. However, more detailed immune mechanisms during different periods of E. granulosus infection were not elucidated. Methods Herein, we characterized immune and endothelial cells from the liver samples of mice in different stages by single-cell RNA sequencing. Results We profiled the transcriptomes of 45,199 cells from the liver samples of mice at 1, 3, and 6 months after infection (two replicates) and uninfected wild-type mice. The cells were categorized into 26 clusters with four distinct cell types: natural killer (NK)/T cells, B cells, myeloid cells, and endothelial cells. An SPP1+ macrophage subset with immunosuppressive and pro-angiogenic functions was identified in the late infection stage. Single-cell regulatory network inference and clustering (SCENIC) analysis suggested that Cebpe, Runx3, and Rora were the key regulators of the SPP1+ macrophages. Cell communication analysis revealed that the SPP1+ macrophages interacted with endothelial cells and had pro-angiogenic functions. There was an obvious communicative relationship between SPP1+ macrophages and endothelial cells via Vegfa-Vegfr1/Vegfr2, and SPP1+ macrophages interacted with other immune cells via specific ligand-receptor pairs, which might have contributed to their immunosuppressive function. Discussion Our comprehensive exploration of the cystic echinococcosis ecosystem and the first discovery of SPP1+ macrophages with infection period specificity provide deeper insights into angiogenesis and the immune evasion mechanisms associated with later stages of infection.
Collapse
Affiliation(s)
- Xiaofeng Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Xiaofan Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China,Department of Laboratory Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nan Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Yeting Sun
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Teng Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Jing Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Yujuan Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China,*Correspondence: Yujuan Shen, ; Jianping Cao,
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China,School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Yujuan Shen, ; Jianping Cao,
| |
Collapse
|
34
|
Makino Y, Hikita H, Kato S, Sugiyama M, Shigekawa M, Sakamoto T, Sasaki Y, Murai K, Sakane S, Kodama T, Sakamori R, Kobayashi S, Eguchi H, Takemura N, Kokudo N, Yokoi H, Mukoyama M, Tatsumi T, Takehara T. STAT3 is Activated by CTGF-mediated Tumor-stroma Cross Talk to Promote HCC Progression. Cell Mol Gastroenterol Hepatol 2022; 15:99-119. [PMID: 36210625 PMCID: PMC9672888 DOI: 10.1016/j.jcmgh.2022.09.006] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Signal transducer and activator of transcription 3 (STAT3) is known as a pro-oncogenic transcription factor. Regarding liver carcinogenesis, however, it remains controversial whether activated STAT3 is pro- or anti-tumorigenic. This study aimed to clarify the significance and mechanism of STAT3 activation in hepatocellular carcinoma (HCC). METHODS Hepatocyte-specific Kras-mutant mice (Alb-Cre KrasLSL-G12D/+; KrasG12D mice) were used as a liver cancer model. Cell lines of hepatoma and stromal cells including stellate cells, macrophages, T cells, and endothelial cells were used for culture. Surgically resected 12 HCCs were used for human analysis. RESULTS Tumors in KrasG12D mice showed up-regulation of phosphorylated STAT3 (p-STAT3), together with interleukin (IL)-6 family cytokines, STAT3 target genes, and connective tissue growth factor (CTGF). Hepatocyte-specific STAT3 knockout (Alb-Cre KrasLSL-G12D/+ STAT3fl/fl) downregulated p-STAT3 and CTGF and suppressed tumor progression. In coculture with stromal cells, proliferation, and expression of p-STAT3 and CTGF, were enhanced in hepatoma cells via gp130/STAT3 signaling. Meanwhile, hepatoma cells produced CTGF to stimulate integrin/nuclear factor kappa B signaling and up-regulate IL-6 family cytokines from stromal cells, which could in turn activate gp130/STAT3 signaling in hepatoma cells. In KrasG12D mice, hepatocyte-specific CTGF knockout (Alb-Cre KrasLSL-G12D/+ CTGFfl/fl) downregulated p-STAT3, CTGF, and IL-6 family cytokines, and suppressed tumor progression. In human HCC, single cell RNA sequence showed CTGF and IL-6 family cytokine expression in tumor cells and stromal cells, respectively. CTGF expression was positively correlated with that of IL-6 family cytokines and STAT3 target genes in The Cancer Genome Atlas. CONCLUSIONS STAT3 is activated by CTGF-mediated tumor-stroma crosstalk to promote HCC progression. STAT3-CTGF positive feedback loop could be a therapeutic target.
Collapse
Affiliation(s)
- Yuki Makino
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Seiya Kato
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaya Sugiyama
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Minoru Shigekawa
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tatsuya Sakamoto
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoichi Sasaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuhiro Murai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sadatsugu Sakane
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryotaro Sakamori
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nobuyuki Takemura
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kokudo
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideki Yokoi
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Tomohide Tatsumi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
35
|
Sang Q, Dai W, Yu J, Chen Y, Fan Z, Liu J, Li F, Li J, Wu X, Hou J, Yu B, Feng H, Zhu ZG, Su L, Li YY, Liu B. Identification of prognostic gene expression signatures based on the tumor microenvironment characterization of gastric cancer. Front Immunol 2022; 13:983632. [PMID: 36032070 PMCID: PMC9411533 DOI: 10.3389/fimmu.2022.983632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 12/02/2022] Open
Abstract
Increasing evidence has elucidated that the tumor microenvironment (TME) shows a strong association with tumor progression and therapeutic outcome. We comprehensively estimated the TME infiltration patterns of 111 gastric cancer (GC) and 21 normal stomach mucosa samples based on bulk transcriptomic profiles based on which GC could be clustered as three subtypes, TME-Stromal, TME-Mix, and TME-Immune. The expression data of TME-relevant genes were utilized to build a GC prognostic model—GC_Score. Among the three GC TME subtypes, TME-Stomal displayed the worst prognosis and the highest GC_Score, while TME-Immune had the best prognosis and the lowest GC_Score. Connective tissue growth factor (CTGF), the highest weighted gene in the GC_Score, was found to be overexpressed in GC. In addition, CTGF exhibited a significant correlation with the abundance of fibroblasts. CTGF has the potential to induce transdifferentiation of peritumoral fibroblasts (PTFs) to cancer-associated fibroblasts (CAFs). Beyond characterizing TME subtypes associated with clinical outcomes, we correlated TME infiltration to molecular features and explored their functional relevance, which helps to get a better understanding of carcinogenesis and therapeutic response and provide novel strategies for tumor treatments.
Collapse
Affiliation(s)
- Qingqing Sang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wentao Dai
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, China
| | - Junxian Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunqin Chen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Zhiyuan Fan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jixiang Liu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Fangyuan Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfang Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiongyan Wu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junyi Hou
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beiqin Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoran Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Gang Zhu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Su
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan-Yuan Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, China
- *Correspondence: Bingya Liu, ; Yuan-Yuan Li,
| | - Bingya Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Bingya Liu, ; Yuan-Yuan Li,
| |
Collapse
|
36
|
Zhao Z, Li W, Zhu L, Xu B, Jiang Y, Ma N, Liu L, Qiu J, Zhang M. Construction and Verification of a Fibroblast-Related Prognostic Signature Model for Colon Cancer. Front Genet 2022; 13:908957. [PMID: 35910200 PMCID: PMC9329609 DOI: 10.3389/fgene.2022.908957] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Traditionally, cancer-associated fibroblasts (CAFs), an essential component of tumor microenvironment, were exert a crucial part in colon cancer progression. In this study, single-cell RNA-sequencing (scRNA-seq) data from 23 and bulk RNA-seq data from 452 colon cancer patients were extracted from the GEO database and TCGA-COAD and GEO databases, respectively. From single-cell analysis, 825 differentially expressed genes (DEGs) in CAFs were identified between each pair of six newly defined CAFs, named enCAF, adCAF, vaCAF, meCAF, erCAF, and cyCAF. Cell communication analysis with the iTALK package showed communication relationship between CAFs, including cell autocrine, cytokine, and growth factor subtypes, such as receptor-ligand pairs of TNFSF14-LTBR, IL6-F3, and IL6-IL6ST. Herein, we demonstrated the presence and prognostic value of adCAF and erCAF in colon cancer based on CIBERSORTx, combining single-cell marker genes and transcriptomics data. The prognostic significance of the enCAF and erCAF has been indirectly proved by both the correlation analysis with macrophages and CAFs, and the quantitative reverse transcription-polymerase chain reaction (qRT-PCR) experiment based on 20 paired tumor samples. A prognostic model was constructed with 10 DEGs using the LASSO Cox regression method. The model was validated using two testing datasets, indicate a significant survival accuracy (p < 0.0025). Correlation analyses between clinical information, such as age, gender, tumor stage and tumor features (tumor purity and immune score), and risk scores revealed our CAF-related model’s robustness and excellent performance. Cell infiltration analysis by xCell revealed that the interaction between CAFs and multiple non-specific immune cells such as macrophages and the dendritic cell was a vital factor affecting immune score and prognosis. Finally, we analyzed how common anti-cancer drugs, including camptothecin, docetaxel and bortezomib, and immunotherapy, such as anti-PD-1 treatment, could be different in low-risk and high-risk patients inferred from our CAF-related model. In conclusion, the study utilized refined colon cancer fibroblast subsets and established the prognostic effects from the interaction with nonspecific immune cell.
Collapse
Affiliation(s)
- Zhe Zhao
- Zhengzhou KingMed Center for Clinical Laboratory Co. Ltd., Zhengzhou, China
| | - Wenqi Li
- Department of Newborn Infants, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - LiMeng Zhu
- Zhengzhou KingMed Center for Clinical Laboratory Co. Ltd., Zhengzhou, China
| | - Bei Xu
- Zhengzhou KingMed Center for Clinical Laboratory Co. Ltd., Zhengzhou, China
| | - Yudong Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Nan Ma
- Zhengzhou KingMed Center for Clinical Laboratory Co. Ltd., Zhengzhou, China
| | - LiQun Liu
- Zhengzhou KingMed Center for Clinical Laboratory Co. Ltd., Zhengzhou, China
| | - Jie Qiu
- Department of Newborn Infants, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Min Zhang
- Zhengzhou KingMed Center for Clinical Laboratory Co. Ltd., Zhengzhou, China
- *Correspondence: Min Zhang,
| |
Collapse
|
37
|
Human Mesenchymal Stromal Cells Do Not Cause Radioprotection of Head-and-Neck Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:ijms23147689. [PMID: 35887032 PMCID: PMC9323822 DOI: 10.3390/ijms23147689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/03/2022] [Accepted: 07/09/2022] [Indexed: 02/01/2023] Open
Abstract
Radiotherapy of head-and-neck squamous cell carcinoma (HNSCC) can cause considerable normal tissue injuries, and mesenchymal stromal cells (MSCs) have been shown to aid regeneration of irradiation-damaged normal tissues. However, utilization of MSC-based treatments for HNSCC patients undergoing radiotherapy is hampered by concerns regarding potential radioprotective effects. We therefore investigated the influence of MSCs on the radiosensitivity of HNSCCs. Several human papillomavirus (HPV)-negative and HPV-positive HNSCCs were co-cultured with human bone marrow-derived MSCs using two-dimensional and three-dimensional assays. Clonogenic survival, proliferation, and viability of HNSCCs after radiotherapy were assessed depending on MSC co-culture. Flow cytometry analyses were conducted to examine the influence of MSCs on irradiation-induced cell cycle distribution and apoptosis induction in HNSCCs. Immunofluorescence stainings of γH2AX were conducted to determine the levels of residual irradiation-induced DNA double-strand breaks. Levels of connective tissue growth factor (CTGF), a multifunctional pro-tumorigenic cytokine, were analyzed using enzyme-linked immunosorbent assays. Neither direct MSC co-culture nor MSC-conditioned medium exerted radioprotective effects on HNSCCs as determined by clonogenic survival, proliferation, and viability assays. Consistently, three-dimensional microwell arrays revealed no radioprotective effects of MSCs. Irradiation resulted in a G2/M arrest of HNSCCs at 96 h independently of MSC co-culture. HNSCCs’ apoptosis rates were increased by irradiation irrespective of MSCs. Numbers of residual γH2AX foci after irradiation with 2 or 8 Gy were comparable between mono- and co-cultures. MSC mono-cultures and HNSCC-MSC co-cultures exhibited comparable CTGF levels. We did not detect radioprotective effects of human MSCs on HNSCCs. Our results suggest that the usage of MSC-based therapies for radiotherapy-related toxicities in HNSCC patients may be safe in the context of absent radioprotection.
Collapse
|
38
|
Pouliquen DL, Boissard A, Henry C, Coqueret O, Guette C. Curcuminoids as Modulators of EMT in Invasive Cancers: A Review of Molecular Targets With the Contribution of Malignant Mesothelioma Studies. Front Pharmacol 2022; 13:934534. [PMID: 35873564 PMCID: PMC9304619 DOI: 10.3389/fphar.2022.934534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022] Open
Abstract
Curcuminoids, which include natural acyclic diarylheptanoids and the synthetic analogs of curcumin, have considerable potential for fighting against all the characteristics of invasive cancers. The epithelial-to-mesenchymal transition (EMT) is a fundamental process for embryonic morphogenesis, however, the last decade has confirmed it orchestrates many features of cancer invasiveness, such as tumor cell stemness, metabolic rewiring, and drug resistance. A wealth of studies has revealed EMT in cancer is in fact driven by an increasing number of parameters, and thus understanding its complexity has now become a cornerstone for defining future therapeutic strategies dealing with cancer progression and metastasis. A specificity of curcuminoids is their ability to target multiple molecular targets, modulate several signaling pathways, modify tumor microenvironments and enhance the host’s immune response. Although the effects of curcumin on these various parameters have been the subject of many reviews, the role of curcuminoids against EMT in the context of cancer have never been reviewed so far. This review first provides an updated overview of all EMT drivers, including signaling pathways, transcription factors, non-coding RNAs (ncRNAs) and tumor microenvironment components, with a special focus on the most recent findings. Secondly, for each of these drivers the effects of curcumin/curcuminoids on specific molecular targets are analyzed. Finally, we address some common findings observed between data reported in the literature and the results of investigations we conducted on experimental malignant mesothelioma, a model of invasive cancer representing a useful tool for studies on EMT and cancer.
Collapse
Affiliation(s)
- Daniel L. Pouliquen
- Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
- *Correspondence: Daniel L. Pouliquen,
| | - Alice Boissard
- ICO, Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
| | - Cécile Henry
- ICO, Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
| | - Olivier Coqueret
- Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
| | - Catherine Guette
- ICO, Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
| |
Collapse
|
39
|
Rovira E, Moreno B, Razquin N, Hjerpe R, Gonzalez-Lopez M, Barrio R, Ruiz de los Mozos I, Ule J, Pastor F, Blazquez L, Fortes P. U1A is a positive regulator of the expression of heterologous and cellular genes involved in cell proliferation and migration. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:831-846. [PMID: 35664701 PMCID: PMC9136276 DOI: 10.1016/j.omtn.2022.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/07/2022] [Indexed: 11/16/2022]
Abstract
Here, we show that direct recruitment of U1A to target transcripts can increase gene expression. This is a new regulatory role, in addition to previous knowledge showing that U1A decreases the levels of U1A mRNA and other specific targets. In fact, genome-wide, U1A more often increases rather than represses gene expression and many U1A-upregulated transcripts are directly bound by U1A according to individual nucleotide resolution crosslinking and immunoprecipitation (iCLIP) studies. Interestingly, U1A-mediated positive regulation can be transferred to a heterologous system for biotechnological purposes. Finally, U1A-bound genes are enriched for those involved in cell cycle and adhesion. In agreement with this, higher U1A mRNA expression associates with lower disease-free survival and overall survival in many cancer types, and U1A mRNA levels positively correlate with those of some oncogenes involved in cell proliferation. Accordingly, U1A depletion leads to decreased expression of these genes and the migration-related gene CCN2/CTGF, which shows the strongest regulation by U1A. A decrease in U1A causes a strong drop in CCN2 expression and CTGF secretion and defects in the expression of CTGF EMT targets, cell migration, and proliferation. These results support U1A as a putative therapeutic target for cancer treatment. In addition, U1A-binding sequences should be considered in biotechnological applications.
Collapse
Affiliation(s)
- Eric Rovira
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Beatriz Moreno
- Department of Molecular Therapy, Aptamer Unit, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Nerea Razquin
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Roland Hjerpe
- Department of Functional Genomics, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Monika Gonzalez-Lopez
- Department of Functional Genomics, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Rosa Barrio
- Department of Functional Genomics, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Igor Ruiz de los Mozos
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, WC1B5EH London, UK
- RNA Networks Lab, The Francis Crick Institute, NW11BF London, UK
| | - Jernej Ule
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, WC1B5EH London, UK
- RNA Networks Lab, The Francis Crick Institute, NW11BF London, UK
| | - Fernando Pastor
- Department of Molecular Therapy, Aptamer Unit, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Lorea Blazquez
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, WC1B5EH London, UK
- RNA Networks Lab, The Francis Crick Institute, NW11BF London, UK
- Neurosciences Area, Biodonostia Health Research Institute, 20014 San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- Corresponding author. Lorea Blazquez, Department of Neuromuscular Diseases, Institute of Neurology, UCL, WC1B5EH London, UK.
| | - Puri Fortes
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Spain
- Spanish Network for Advanced Therapies (TERAV ISCIII), Spain
- Corresponding author. Puri Fortes, Neurosciences Area, Biodonostia Health Research Institute, 20014 San Sebastian, Spain.
| |
Collapse
|
40
|
Zhou YT, Yu YQ, Yang H, Yang H, Huo YF, Huang Y, Tian XX, Fang WG. Extracellular ATP promotes angiogenesis and adhesion of TNBC cells to endothelial cells via up-regulation of CTGF. Cancer Sci 2022; 113:2457-2471. [PMID: 35441763 PMCID: PMC9277410 DOI: 10.1111/cas.15375] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 12/01/2022] Open
Abstract
Our previous works have indicated that extracellular ATP is an important prometastasis factor. However, the molecular mechanism involved needs to be further studied. We demonstrated that extracellular ATP treatment could upregulate the expression of connective tissue growth factor (CTGF) in both triple‐negative breast cancer (TNBC) cells and endothelial cells (ECs). Extracellular ATP stimulated the migration of TNBC cells and ECs, and angiogenesis of ECs via the P2Y2––YAP‐CTGF axis. Furthermore, we demonstrated that adenosine triphosphate (ATP) stimulated TNBC cell adhesion to ECs and transmigration through the EC layer via CTGF by upregulation of integrin β1 on TNBC cells and VCAM‐1 on ECs. Both apyrase (ATP‐diphosphohydrolase) and CTGF shRNA treatments could inhibit the metastasis of inoculated tumors to lung and liver in a mouse model, and these treated tumors had fewer blood vessels. Collectively, our data indicated that extracellular ATP promotes tumor angiogenesis and the interactions between TNBC cells and ECs through upregulation of CTGF, thereby stimulating TNBC metastasis. The pleiotropic effects of ATP in angiogenesis and cell adhesion suggest that extracellular ATP or CTGF could be an effective target for TNBC therapy.
Collapse
Affiliation(s)
- Yan-Ting Zhou
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Yu-Qing Yu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Hui Yang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Han Yang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Yan-Fei Huo
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Yang Huang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Xin-Xia Tian
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Wei-Gang Fang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| |
Collapse
|
41
|
Fu M, Peng D, Lan T, Wei Y, Wei X. Multifunctional regulatory protein connective tissue growth factor (CTGF): A potential therapeutic target for diverse diseases. Acta Pharm Sin B 2022; 12:1740-1760. [PMID: 35847511 PMCID: PMC9279711 DOI: 10.1016/j.apsb.2022.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Connective tissue growth factor (CTGF), a multifunctional protein of the CCN family, regulates cell proliferation, differentiation, adhesion, and a variety of other biological processes. It is involved in the disease-related pathways such as the Hippo pathway, p53 and nuclear factor kappa-B (NF-κB) pathways and thus contributes to the developments of inflammation, fibrosis, cancer and other diseases as a downstream effector. Therefore, CTGF might be a potential therapeutic target for treating various diseases. In recent years, the research on the potential of CTGF in the treatment of diseases has also been paid more attention. Several drugs targeting CTGF (monoclonal antibodies FG3149 and FG3019) are being assessed by clinical or preclinical trials and have shown promising outcomes. In this review, the cellular events regulated by CTGF, and the relationships between CTGF and pathogenesis of diseases are systematically summarized. In addition, we highlight the current researches, focusing on the preclinical and clinical trials concerned with CTGF as the therapeutic target.
Collapse
|
42
|
The value of plasma hypoxia markers for predicting imaging-based hypoxia in patients with head-and-neck cancers undergoing definitive chemoradiation. Clin Transl Radiat Oncol 2022; 33:120-127. [PMID: 35243023 PMCID: PMC8881198 DOI: 10.1016/j.ctro.2022.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
Higher osteopontin plasma levels correlate with more hypoxic tumors at baseline. Increased baseline osteopontin levels are associated with residual tumor hypoxia. Absent early hypoxia response is linked with higher VEGF and CTGF levels in week 5. Plasma hypoxic markers may serve as biomarkers favoring radiotherapy personalization.
Background Methods Results Conclusion
Collapse
|
43
|
Filipe A, Katopodis P, Chudasama D, Kerslake R, Jeyaneethi J, Anikin V, Silva E, Kyrou I, Randeva HS, Sisu C, Hall M, Karteris E. Differential Expression of RAD51AP1 in Ovarian Cancer: Effects of siRNA In Vitro. J Pers Med 2022; 12:jpm12020201. [PMID: 35207688 PMCID: PMC8876735 DOI: 10.3390/jpm12020201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/08/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Background: DNA double strand breaks can affect genome integrity potentially leading to cancer. RAD51-associated protein 1 (RAD51AP1), an accessory protein to RAD51, is critical for homologous recombination, a key DNA damage response pathway. Emerging studies indicate a novel role for RAD51AP1 in carcinogenesis. Here we provide additional insight into the role of RAD51AP1 in ovarian cancer (OvCa). Methods: Gene expression and patient phenotype data were obtained from TCGA and GTEX project consortia for bioinformatics analysis. Immunohistochemistry of OvCa tissue microarray was undertaken. Functional analyses were performed in a SKOV3 OvCa cell line with down-regulation of RAD51AP1 using siRNA. Results: RAD51AP1 is overexpressed at gene level in primary and recurrent OvCa compared to controls. At protein level, RAD51AP1 was up-regulated in low grade serous tumors compared to high grade OvCa. There was higher expression of RAD51AP1 in OvCa metastatic to lymph nodes compared to primary cancer samples. Gene enrichment analyses identified 12 differentially expressed genes (DEGs) related to OvCa, eight of which are also common in tissue from patients with type 2 diabetes mellitus (T2DM). Conclusions: RAD51AP1 is overexpressed in OvCa, Given the link between OvCa and T2DM, the eight-gene signature shows potential for predictive value.
Collapse
Affiliation(s)
- Alice Filipe
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
| | - Periklis Katopodis
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
| | - Dimple Chudasama
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
| | - Rachel Kerslake
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
| | - Jeyarooban Jeyaneethi
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
| | - Vladimir Anikin
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
- Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State, Medical University, 119146 Moscow, Russia
| | - Elisabete Silva
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (I.K.); (H.S.R.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Department of Food Science & Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (I.K.); (H.S.R.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Cristina Sisu
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
| | - Marcia Hall
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Mount Vernon Cancer Centre, Northwood, London HA6 2RN, UK
- Correspondence: (M.H.); (E.K.)
| | - Emmanouil Karteris
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
- Correspondence: (M.H.); (E.K.)
| |
Collapse
|
44
|
Zhang Z, Lin J, Hu J, Liu L. Liquiritigenin Blocks Breast Cancer Progression by Inhibiting Connective Tissue Growth Factor Expression via Up-Regulating miR-383-5p. Int J Toxicol 2022; 41:5-15. [PMID: 35045746 DOI: 10.1177/10915818211059470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The objective of this study was to investigate the effect of liquiritigenin (LQ) on breast cancer (BC) and its mechanism. After BC cell lines and normal mammary epithelial cells were cultured with LQ, CCK-8, and Scratch, Transwell assays and flow cytometry were applied to test the effect of LQ on cell proliferation, migration, invasion, and apoptosis. The effect of LQ on the expression of microRNA-383-5p (miR-383-5p) and connective tissue growth factor (CTGF) was measured by qRT-PCR and Western blotting. Bioinformatics prediction was used to evaluate the binding relationship between miR-383-5p and CTGF, which was verified by dual-luciferase reporter assay. After miR-383-5p and/or CTGF expression was upregulated through cell transfection, the relationship between miR-383-5p and CTGF, as well as their effects on BC, was further assessed. The results showed that LQ can significantly inhibit CTGF expression and the proliferative, migratory, and invasive abilities of BC cells, while facilitating apoptosis of BC cells and miR-383-5p expression. The inhibiting effect of LQ was dose-dependently enhanced in BC cells. Dual-luciferase reporter assay verified that miR-383-5p targeted CTGF. CTGF expression was inversely regulated by miR-383-5p. CTGF upregulation repressed the suppressive effect of miR-385-5p on BC cell development. In conclusion, LQ can inhibit CTGF expression by upregulating miR-383-5p, thereby inhibiting proliferative, migratory, and invasive abilities and promoting apoptosis of BC cells.
Collapse
Affiliation(s)
- Zhanwei Zhang
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
- Department of Chinese Traditional Surgery, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Jie Lin
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Jinhui Hu
- Department of Breast Surgery, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Lifang Liu
- Department of Breast Surgery, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
45
|
Yang Z, Li W, Song C, Leng H. CTGF as a multifunctional molecule for cartilage and a potential drug for osteoarthritis. Front Endocrinol (Lausanne) 2022; 13:1040526. [PMID: 36325449 PMCID: PMC9618584 DOI: 10.3389/fendo.2022.1040526] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2022] Open
Abstract
CTGF is a multifunctional protein and plays different roles in different cells and under different conditions. Pamrevlumab, a monoclonal antibody against CTGF, is an FDA approved drug for idiopathic pulmonary fibrosis (IPF) and Duchenne muscular dystrophy (DMD). Recent studies have shown that CTGF antibodies may potentially serve as a new drug for osteoarthritis (OA). Expression of CTGF is significantly higher in OA joints than in healthy counterparts. Increasing attention has been attracted due to its interesting roles in joint homeostasis. Joint homeostasis relies on normal cellular functions and cell-cell interactions. CTGF is essential for physiological activities of chondrocytes. Abnormal CTGF expression may cause cartilage degeneration. In this review, the physiological functions of CTGF in chondrocytes and related mechanisms are summarized. Changes in the related signaling pathways due to abnormal CTGF are discussed, which are contributing factors to inflammation, cartilage degeneration and synovial fibrosis in OA. The possibility of CTGF as a potential therapeutic target for OA treatment are reviewed.
Collapse
Affiliation(s)
- Zihuan Yang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing Municipal Science & Technology Commission, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- *Correspondence: Huijie Leng,
| |
Collapse
|
46
|
Lobe C, Vallette M, Arbelaiz A, Gonzalez-Sanchez E, Izquierdo L, Pellat A, Guedj N, Louis C, Paradis V, Banales JM, Coulouarn C, Housset C, Vaquero J, Fouassier L. Zinc Finger E-Box Binding Homeobox 1 Promotes Cholangiocarcinoma Progression Through Tumor Dedifferentiation and Tumor-Stroma Paracrine Signaling. Hepatology 2021; 74:3194-3212. [PMID: 34297412 DOI: 10.1002/hep.32069] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 06/29/2021] [Accepted: 07/11/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Zinc finger E-box binding homeobox 1 (ZEB1) is a transcription factor that promotes metastatic and stem cell features, which has been associated with poor prognosis in cholangiocarcinoma (CCA), a desmoplastic cancer enriched in cancer-associated fibroblasts (CAFs). We aimed to define ZEB1 regulatory functions in malignant and stromal compartments of CCA. APPROACH AND RESULTS Bioinformatic and immunohistochemical analyses were performed to determine correlations between ZEB1 and markers of progressiveness in human intrahepatic CCA (iCCA). Gain-of-function and loss-of-function models were generated in CCA cells and liver myofibroblasts as a model of CAFs. Conditioned media (CM) was used to unravel tumor-stroma interplay. In vivo experiments were performed using a xenograft CCA model. ZEB1 expression in tumor cells of human iCCA was associated with undifferentiated tumor and vascular invasion. In vitro, ZEB1 promoted epithelial-mesenchymal transition and stemness in tumor cells, leading to cell migration and spheroid formation. In vivo, ZEB1-overexpressing CCA cells formed larger tumors with more abundant stroma. Expression of cellular communication network factor 2 (CCN2, encoding connective tissue growth factor [CTGF]) was increased in tumor cells from ZEB1-overexpressing xenografts and correlated with ZEB1 expression in human tumors. In vitro, CM from ZEB1-overexpressing tumor cells or recombinant CTGF induced myofibroblast proliferation. ZEB1 was also expressed by CAFs in human CCA, and its expression correlated with CCN2 in myofibroblasts and CCA stroma. In mice, cotransplantation of CCA cells with ZEB1-depleted myofibroblasts reduced CCA progressiveness compared to CCA cells/ZEB1-expressing myofibroblasts. Furthermore, ZEB1 controls the expression of paracrine signals (i.e., HGF and IL6) in tumor cells and myofibroblasts. CONCLUSIONS ZEB1 plays a key role in CCA progression by regulating tumor cell-CAF crosstalk, leading to tumor dedifferentiation and CAF activation.
Collapse
Affiliation(s)
- Cindy Lobe
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Marie Vallette
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Ander Arbelaiz
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Ester Gonzalez-Sanchez
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France.,TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, Barcelona, Spain.,National Biomedical Research Institute on Liver and Gastrointestinal Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Izquierdo
- National Biomedical Research Institute on Liver and Gastrointestinal Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute-Donostia University Hospital, University of the Basque Country, San Sebastián, Spain
| | - Anna Pellat
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Nathalie Guedj
- Service d'Anatomie Pathologique Hôpital Beaujon, Clichy, France
| | - Corentin Louis
- INSERM, UMR 1149, Centre de Recherche sur l'Inflammation, Paris, France
| | - Valérie Paradis
- Service d'Anatomie Pathologique Hôpital Beaujon, Clichy, France.,INSERM, UMR 1149, Centre de Recherche sur l'Inflammation, Paris, France
| | - Jesus M Banales
- National Biomedical Research Institute on Liver and Gastrointestinal Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute-Donostia University Hospital, University of the Basque Country, San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | | | - Chantal Housset
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France.,Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, ERN Rare-Liver, Paris, France
| | - Javier Vaquero
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France.,TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, Barcelona, Spain.,National Biomedical Research Institute on Liver and Gastrointestinal Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,LPP, Sorbonne Université, CNRS, Ecole Polytechnique, Université Paris-Sud, Observatoire de Paris, Université Paris-Saclay, PSL Research University, Paris, France
| | - Laura Fouassier
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| |
Collapse
|
47
|
Wang M, Li XZ, Zhang MX, Ye QY, Chen YX, Chang X. Atractylenolide-I Sensitizes Triple-Negative Breast Cancer Cells to Paclitaxel by Blocking CTGF Expression and Fibroblast Activation. Front Oncol 2021; 11:738534. [PMID: 34692516 PMCID: PMC8526898 DOI: 10.3389/fonc.2021.738534] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/20/2021] [Indexed: 12/18/2022] Open
Abstract
This investigation was conducted to elucidate whether atractylenolide-I (ATL-1), which is the main component of Atractylodes macrocephala Koidz, can sensitize triple-negative breast cancer (TNBC) cells to paclitaxel and investigate the possible mechanism involved. We discovered that ATL-1 could inhibit tumor cell migration and increase the sensitivity of tumor cells to paclitaxel. ATL-1 downregulated the expression and secretion of CTGF in TNBC cells. Apart from inhibiting TNBC cell migration via CTGF, ATL-1 downregulated the expression of CTGF in fibroblasts and decreased the ability of breast cancer cells to transform fibroblasts into cancer-associated fibroblasts (CAFs), which in turn increased the sensitivity of TNBC cells to paclitaxel. In a mouse model, we found that ATL-1 treatments could enhance the chemotherapeutic effect of paclitaxel on tumors and reduce tumor metastasis to the lungs and liver. Primary cultured fibroblasts derived from inoculated tumors in mice treated with ATL-1 combined with paclitaxel expressed relatively low levels of CAF markers. Collectively, our data indicate that ATL-1 can sensitize TNBC cells to paclitaxel by blocking CTGF expression and fibroblast activation and could be helpful in future research to determine the value of ATL-1 in the clinical setting.
Collapse
Affiliation(s)
- Meng Wang
- First Department of Surgery, Panyu Hospital of Chinese Medicine, Guangzhou, China
| | - Xue-Zhen Li
- Department of Breast Surgery, Guangdong Second Hospital of Traditional Chinese Medicine, Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ming-Xing Zhang
- Department of Mammary Disease, Panyu Hospital of Chinese Medicine, Guangzhou, China
| | - Qian-Yu Ye
- Department of Mammary Disease, Panyu Hospital of Chinese Medicine, Guangzhou, China
| | - Ying-Xia Chen
- Department of Mammary Disease, Panyu Hospital of Chinese Medicine, Guangzhou, China
| | - Xu Chang
- Department of Mammary Disease, Panyu Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
48
|
Bai Y, Yu T, Deng J, Yang Y, Tan J, Dai Q, Zhang Z, Dong S, Xu J. Connective Tissue Growth Factor From Periosteal Tartrate Acid Phosphatase-Positive Monocytes Direct Skeletal Stem Cell Renewal and Fate During Bone Healing. Front Cell Dev Biol 2021; 9:730095. [PMID: 34595178 PMCID: PMC8476870 DOI: 10.3389/fcell.2021.730095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/20/2021] [Indexed: 11/25/2022] Open
Abstract
The periosteum is critical for bone healing. Studies have shown that the periosteum contains periosteal stem cells (PSCs) with multidirectional differentiation potential and self-renewal ability. PSCs are activated in early fracture healing and are committed to the chondrocyte lineage, which is the basis of callus formation. However, the mechanism by which PSCs are activated and committed to chondrocytes in bone regeneration remains unclear. Here, we show that tartrate acid phosphatase (TRAP)-positive monocytes secrete CTGF to activate PSCs during bone regeneration. The loss function of TRAP-positive monocytes identifies their specific role during bone healing. Then, the secreted CTGF promotes endochondral ossification and activates PSCs in mouse bone fracture models. The secreted CTGF enhances PSC renewal by upregulating the expression of multiple pluripotent genes. CTGF upregulates c-Jun expression through αVβ5 integrin. Then, c-Jun transcription activates the transcription of the pluripotent genes Sox2, Oct4, and Nanog. Simultaneously, CTGF also activates the transcription and phosphorylation of Smad3 through αVβ5 integrin, which is the central gene in chondrogenesis. Our study indicates that TRAP-positive monocyte-derived CTGF promotes bone healing by activating PSCs and directing lineage commitment and that targeting PSCs may be an effective strategy for preventing bone non-union.
Collapse
Affiliation(s)
- Yun Bai
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Tao Yu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiezhong Deng
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yusheng Yang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiulin Tan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qijie Dai
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zehua Zhang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shiwu Dong
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
49
|
Fraunhoffer NA, Closa D, Folch-Puy E, Abuelafia AM, Calvo EL, Chuluyan E, Iovanna J. Targeting REG3β limits pancreatic ductal adenocarcinoma progression through CTGF downregulation. Cancer Lett 2021; 521:64-70. [PMID: 34450197 DOI: 10.1016/j.canlet.2021.08.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/03/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022]
Abstract
The crosstalk between the transformed tumoral cells and their microenvironment is a key aspect for pancreatic ductal adenocarcinoma (PDAC) progression. This molecular dialog is intensively studied because it may result in an efficient therapeutic target. Contrary to this near microenvironment, the stromal portion in direct contact with the transformed cells, a far microenvironment, placed at the periphery of the tumor mass, produces factors signaling tumors. Among these factors, REG3β, produced by this part of the pancreas, is an important factor in promoting tumor progression. This paper demonstrated that targeting REG3β protein with specific antibodies limits the PDAC tumor growth in an orthotopic, syngeneic mice model induced by injection of Panc02 cells. Then, we showed that CTGF is over-expressed in response to REG3β in PDAC-derived cells. Moreover, inactivation of REG3β by treating tumors with anti-REG3β antibodies results in a strong decrease of CTGF in PDAC tumors. Lastly, we demonstrated that forced expression of CTGF in xenografted Panc02 cells abolishes the therapeutic effect of the anti-REG3β antibody treatment. Altogether, these results indicate that the effect of REG3β in promoting PDAC progression is mediated by CTGF over-activation. Thus, REG3β is a promising therapeutic target to treat PDAC with an original rationale. In conclusion, we demonstrated that the far microenvironment is essential for PDAC progression by producing active secretory factors, and some of them could be used as therapeutic targets.
Collapse
Affiliation(s)
- Nicolás A Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288, Marseille, France; Laboratory of Immunomodulators, School of Medicine, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Daniel Closa
- Experimental Pathology Department, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Emma Folch-Puy
- Experimental Pathology Department, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Analía Meilerman Abuelafia
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288, Marseille, France
| | - Ezequiel Luis Calvo
- Centre Génomique du Centre de Recherche du CHUL Research Center, Ville de Québec, Quebec, Canada
| | - Eduardo Chuluyan
- Laboratory of Immunomodulators, School of Medicine, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288, Marseille, France.
| |
Collapse
|
50
|
Zheng D, Xia K, Yu L, Gong C, Shi Y, Li W, Qiu Y, Yang J, Guo W. A Novel Six Metastasis-Related Prognostic Gene Signature for Patients With Osteosarcoma. Front Cell Dev Biol 2021; 9:699212. [PMID: 34368151 PMCID: PMC8343004 DOI: 10.3389/fcell.2021.699212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/02/2021] [Indexed: 11/18/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor, and although there has been significant progress in its management, metastases often herald incurable disease. Here we defined genes differentially expressed between primary and metastatic osteosarcoma as metastasis-related genes (MRGs) and used them to construct a novel six-MRG prognostic signature for overall survival of patients with osteosarcoma. Validation in internal and external datasets confirmed satisfactory accuracy and generalizability of the prognostic model, and a nomogram based on the signature and clinical variables was constructed to aid clinical decision-making. Of the six MRGs, FHIT is a well-documented tumor suppressor gene that is poorly defined in osteosarcoma. Consistent with tumor suppressor function, FHIT was downregulated in osteosarcoma cells and human osteosarcoma samples. FHIT overexpression inhibited osteosarcoma proliferation, migration, and invasion both in vitro and in vivo. Mechanistically, FHIT overexpression upregulate the epithelial marker E-cadherin while repressing the mesenchymal markers N-cadherin and vimentin. Our six-MRG signature represents a novel and clinically useful prognostic biomarker for patients with osteosarcoma, and FHIT might represent a therapeutic target by reversing epithelial to mesenchymal transition.
Collapse
Affiliation(s)
- Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kezhou Xia
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling Yu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Changtian Gong
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yubo Shi
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonglong Qiu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Yang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|