1
|
Wei T, Fu G, Zhao J, Cao F, Guo D. Acyl-CoA dehydrogenase long chain acts as a tumor-suppressive factor in lung adenocarcinoma progression. Cell Adh Migr 2025; 19:2495676. [PMID: 40262559 DOI: 10.1080/19336918.2025.2495676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/12/2024] [Accepted: 04/05/2025] [Indexed: 04/24/2025] Open
Abstract
This study investigated the role of long-chain acyl-CoA dehydrogenase (ACADL) in lung adenocarcinoma (LUAD). ACADL was significantly downregulated in human LUAD tissues compared to normal lung tissues. In vitro, ectopic expression of ACADL in murine LLC cells decreased cell viability, migration, and invasion, while ACADL knockdown exhibited the opposite effect. In vivo, ACADL overexpression impeded tumor growth and metastasis. Mechanistically, ACADL hindered tumor progression by inducing cell cycle arrest, promoting apoptosis, and suppressing the epithelial-mesenchymal transition (EMT) process. These findings suggest ACADL acts as a tumor suppressor in LUAD progression.
Collapse
Affiliation(s)
- Tingju Wei
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Fu
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junjie Zhao
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fengan Cao
- Department of Respiratory Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Guo
- Henan Key Laboratory for Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Chen S, Yue S, Ba W, Fang J. A novel radial co-culture microfluidic device for parallel and control detection of tumor cell invasiveness. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 334:125942. [PMID: 40020497 DOI: 10.1016/j.saa.2025.125942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
Metastasis is responsible for the majority of cancer-related deaths, and tumor cell invasion is a critical step in the cancer metastatic cascade. Traditional invasion analysis using transwell assays and immunodeficient mouse models struggles to control tumor microenvironment factors, such as various biochemical signals and cell types. A microfluidic chip system has emerged as an important tool for invasion analysis, but the simultaneous parallel and controlled experiments within the same group of tumor cells remain challenging. Here, we developed a new three-dimensional co-culture microfluidic device to investigate tumor invasion. This device consists of three concentric circles and is divided into four identical regions. Each region includes a tumor cell region, an invasion channel and a co-culture channel. Additionally, the four identical regions allow for four specific groups of parallel or control analysis surrounding the same group of tumor cells. Thus, our device enables the comparison of invasion among the same group of cells under different conditions, avoiding the discrepancies in invasion that arise when the treatment factors differ across different groups of tumor cells. Using experimental examinations and numerical simulations, we verified the capability of the device for parallel and controlled analysis of tumor invasion in response to different stimuli, including chemokines, drugs and cellular factors secreted from co-cultured cells. Furthermore, we found that in a co-culture environment with cancer-associated fibroblasts, the invasiveness of LoVo cells and their resistance to 5-fluorouracil treatment were enhanced. This innovative approach allows for easy, parallel and controlled assays to study tumor cell invasion with the advantages of multiplexing and simplicity. The method provides straightforward, repeatable control over cell-biochemical signals and cell-cell interactions, making it a valuable tool for accurately evaluating tumor invasion in metastasis studies.
Collapse
Affiliation(s)
- Shuo Chen
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Shuai Yue
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Wei Ba
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Jin Fang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China.
| |
Collapse
|
3
|
Li X, Wu Y, Xie B, Xu M, Xie T, Yue W, Lin M, Lin Y, Chen Y. SPP1 Promotes NSCLC Brain Metastasis Via Sequestration of Ubiquitin Ligase RNF114 to Facilitate P85α Ubiquitination. Mol Carcinog 2025; 64:829-841. [PMID: 39918025 DOI: 10.1002/mc.23866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/04/2024] [Accepted: 12/02/2024] [Indexed: 04/12/2025]
Abstract
Brain metastasis (BM) is a significant factor contributing to the poor prognosis of patients with non-small cell lung cancer (NSCLC). Secreted phosphoprotein 1 (SPP1) is implicated in the progression and metastasis of several cancers. The role of SPP1 in NSCLC remains unclear, especially in NSCLC BM. This study aimed to identify genes associated with NSCLC BM and to investigate the involvement of SPP1 in NSCLC BM. Integrated genomic analysis was utilized to identify candidate genes in NSCLC. The expression levels of SPP1 were evaluated in NSCLC tissues and cell lines. In vitro and in vivo experiments were conducted to assess the effect of SPP1 on NSCLC cell behavior and BM. The potential mechanisms of SPP1 were demonstrated by CO-IP and liquid chromatography-mass spectrometry (LC-MS). The underlying mechanism involving the PI3K/AKT/mTOR pathway was explored. The results showed that SPP1 expression was upregulated in NSCLC tissues and cell lines. Depletion of SPP1 using shRNA inhibited cell proliferation, migration, and invasion in vitro and suppressed BM in vivo. Mechanistically, SPP1 facilitates the ubiquitination of P85α by interacting with the ubiquitin ligase RNF114, thus playing a role in regulating NSCLC BM through the PI3K/AKT/mTOR signaling pathway. Moreover, immunohistochemistry staining confirmed higher expression of SPP1 in NSCLC tissues with BM compared to those without BM. In summary, elevated SPP1 expression was associated with poor clinical outcomes in NSCLC patients. This study highlights the role of SPP1 as a regulator of cell metastasis and suggests its potential as a novel therapeutic target for BM in NSCLC.
Collapse
Affiliation(s)
- Xiaoqin Li
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| | - Yun Wu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Center for Geriatrics, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Baosong Xie
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| | - Mingxiao Xu
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai, China
| | - Tianjian Xie
- Xiapu County Hospital of Fujian Province, Ningde, China
| | - Wenxiang Yue
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Ming Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Ying Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, China
| | - Yusheng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| |
Collapse
|
4
|
Chan HY, Wang Q, Howie A, Bucci J, Graham P, Li Y. Extracellular vesicle biomarkers redefine prostate cancer radiotherapy. Cancer Lett 2025; 616:217568. [PMID: 39978570 DOI: 10.1016/j.canlet.2025.217568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Radiotherapy (RT) remains a cornerstone in the treatment of prostate cancer (PCa). Extracellular vesicles (EVs), nano-sized particles secreted by cells, play important roles in intercellular communication within the tumour microenvironment (TME) and contribute to tumour growth, metastasis, and therapy resistance. Recent advancements demonstrate the potential of EVs as biomarkers for cancer diagnosis, prognosis, and treatment monitoring. Accumulating evidence supports the role of EVs in modulating RT outcomes by shaping the TME, mediating radioresistance, and influencing cancer metastasis. Despite substantial progress, challenges remain, including the heterogeneity of EV biogenesis, variability in cargo composition, and the absence of standardised methods for EV isolation and characterisation. While the therapeutic and diagnostic prospects of EVs in PCa management are promising, further research is needed to clarify the mechanisms through which EVs impact RT and to translate these findings into clinical practice. Incorporating EV research into PCa treatment paradigms could enhance diagnostic accuracy, enable real-time monitoring of RT responses, and support the development of new targeted therapeutic strategies. This review discusses recent progress in understanding EVs in the context of RT for PCa, focuses on their roles in modulating tumour growth, contributing to radioresistance within the TME, and facilitating the monitoring of RT efficacy and recurrence. In addition, the potential of EVs as biomarkers for liquid biopsy and their applications in enhancing radiosensitivity or overcoming radioresistance is also explored.
Collapse
Affiliation(s)
- Hei Yeung Chan
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Qi Wang
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Andrew Howie
- Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Joseph Bucci
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Peter Graham
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Yong Li
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia.
| |
Collapse
|
5
|
Drago-Garcia D, Giri S, Chatterjee R, Simoni-Nieves A, Abedrabbo M, Genna A, Rios MLU, Lindzen M, Sekar A, Gupta N, Aharoni N, Bhandari T, Mayalagu A, Schwarzmüller L, Tarade N, Zhu R, Mohan-Raju HR, Karatekin F, Roncato F, Eyal-Lubling Y, Keidar T, Nof Y, Belugali Nataraj N, Bernshtein KS, Wagner B, Nair NU, Sanghvi N, Alon R, Seger R, Pikarsky E, Donzelli S, Blandino G, Wiemann S, Lev S, Prywes R, Barkan D, Rueda OM, Caldas C, Ruppin E, Shiloh Y, Dahlhoff M, Yarden Y. Re-epithelialization of cancer cells increases autophagy and DNA damage: Implications for breast cancer dormancy and relapse. Sci Signal 2025; 18:eado3473. [PMID: 40261955 DOI: 10.1126/scisignal.ado3473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/25/2024] [Accepted: 03/07/2025] [Indexed: 04/24/2025]
Abstract
Cellular plasticity mediates tissue development as well as cancer growth and progression. In breast cancer, a shift to a more epithelial phenotype (epithelialization) underlies a state of reversible cell growth arrest called tumor dormancy, which enables drug resistance, tumor recurrence, and metastasis. Here, we explored the mechanisms driving epithelialization and dormancy in aggressive mesenchymal-like breast cancer cells in three-dimensional cultures. Overexpressing either of the epithelial lineage-associated transcription factors OVOL1 or OVOL2 suppressed cell proliferation and migration and promoted transition to an epithelial morphology. The expression of OVOL1 (and of OVOL2 to a lesser extent) was regulated by steroid hormones and growth factors and was more abundant in tumors than in normal mammary cells. An uncharacterized and indirect target of OVOL1/2, C1ORF116, exhibited genetic and epigenetic aberrations in breast tumors, and its expression correlated with poor prognosis in patients. We further found that C1ORF116 was an autophagy receptor that directed the degradation of antioxidant proteins, including thioredoxin. Through C1ORF116 and unidentified mediators, OVOL1 expression dysregulated both redox homeostasis (in association with increased ROS, decreased glutathione, and redistribution of the transcription factor NRF2) and DNA damage and repair (in association with increased DNA oxidation and double-strand breaks and an altered interplay among the kinases p38-MAPK, ATM, and others). Because these effects, as they accumulate in cells, can promote metastasis and dormancy escape, the findings suggest that OVOLs not only promote dormancy entry and maintenance in breast cancer but also may ultimately drive dormancy exit and tumor recurrence.
Collapse
Affiliation(s)
- Diana Drago-Garcia
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Suvendu Giri
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rishita Chatterjee
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Arturo Simoni-Nieves
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maha Abedrabbo
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Alessandro Genna
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mary Luz Uribe Rios
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Moshit Lindzen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Arunachalam Sekar
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nitin Gupta
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Noa Aharoni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tithi Bhandari
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Agalyan Mayalagu
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Luisa Schwarzmüller
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg 69120, Germany
| | - Nooraldeen Tarade
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg 69120, Germany
| | - Rong Zhu
- MRC-Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
- School of Mathematics and Statistics, Beijing Institute of Technology, Beijing 100081, China
| | - Harsha-Raj Mohan-Raju
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Feride Karatekin
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Francesco Roncato
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yaniv Eyal-Lubling
- Cancer Research UK Cambridge Institute, Department of Oncology, University of Cambridge and the Cambridge Cancer Centre, Cambridge CB2 0RE, UK
| | - Tal Keidar
- Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Yam Nof
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nishanth Belugali Nataraj
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
- Bugworks Research India Pvt. Ltd., Center for Cellular and Molecular Platforms (C-CAMP), NCBS Campus, Bangalore 560 065, India
| | | | - Bettina Wagner
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna 1210, Austria
| | - Nishanth Ulhas Nair
- Cancer Data Science Lab, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Neel Sanghvi
- Cancer Data Science Lab, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Ronen Alon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rony Seger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eli Pikarsky
- Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Sara Donzelli
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg 69120, Germany
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Prywes
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Dalit Barkan
- Department of Human Biology, University of Haifa, Haifa 3103301, Israel
| | - Oscar M Rueda
- MRC-Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, Department of Oncology, University of Cambridge and the Cambridge Cancer Centre, Cambridge CB2 0RE, UK
| | - Eytan Ruppin
- Cancer Data Science Lab, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yosef Shiloh
- Department of Human Molecular Genetics and Biochemistry, Tel Aviv University School of Medicine, Tel Aviv 6997801, Israel
| | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna 1210, Austria
| | - Yosef Yarden
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
6
|
Gu Z, Heng Y, Fan R, Luo J, Ju L. Single-cell RNA sequencing reveals cellular and molecular heterogeneity in extensive-stage small cell lung cancer with different chemotherapy responses. Cancer Cell Int 2025; 25:157. [PMID: 40259334 DOI: 10.1186/s12935-025-03785-z] [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: 11/30/2024] [Accepted: 04/08/2025] [Indexed: 04/23/2025] Open
Abstract
Despite its rapid growth and early metastasis, small cell lung cancer (SCLC) is more chemosensitive than other lung cancers. However, some patients with extensive-stage SCLC (ES-SCLC) do not respond to first-line chemotherapy, resulting in poorer prognoses due to inter- and intratumoral heterogeneity. In this study, we conducted single-cell RNA sequencing of 9 treatment-naive ES-SCLC samples. Based on comprehensive imaging evidence collected before and after two cycles of first-line chemotherapy and sample types, the 9 samples were categorized into three groups: progressive disease with the pleural effusion sample (PD_PE group, n = 1), progressive disease with the primary tumor samples (PD_TU group, n = 2), and partial response with the primary tumor samples (PR_TU group, n = 6). Based on transcriptomic landscape and cell type composition, the PD samples represent a multicellular ecosystem distinct from PR samples. The immune response, along with the elevated expression of immune-related genes such as LTF, SLPI, SPARC and IGLV1-51, might correlate with a poor first-line chemotherapy response in ES-SCLC. We also observed that T cells, particularly effector T cells, were more abundant in PD_TU group, with TNFA signaling via NFκB being significantly enriched. The PD_TU group was strongly enriched with macrophages and tumor-associated macrophages (TAMs), and angiogenesis in TAMs was highly enriched. Immunomodulatory fibroblasts were highly abundant in PD_TU group, and the pathways of epithelial-mesenchymal transition and angiogenesis were upregulated. This study offers the first comprehensive insights into the cellular and molecular heterogeneity in treatment-naive patients with ES-SCLC with different chemotherapy responses.
Collapse
Affiliation(s)
- Zhan Gu
- Department of Integrative Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongqing Heng
- Department of Integrative Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rui Fan
- Department of Integrative Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Luo
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lixia Ju
- Department of Integrative Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Wang Y, Duan Y, Yue K, Li L, Cao J, Shi W, Liu J, Wu Y, Wang X, Jing C. Blocking PSMD14-mediated E2F1/ERK/AKT signaling pathways suppresses the progression of anaplastic thyroid cancer. Cell Signal 2025:111826. [PMID: 40262717 DOI: 10.1016/j.cellsig.2025.111826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
Anaplastic thyroid cancer (ATC) is the most aggressive subtype of thyroid cancer with few effective therapeutic strategies. Recent studies have identified the deubiquitinating enzyme (DUB) 26S proteasome non-ATPase regulatory subunit 14 (PSMD14) as a promising therapeutic target for multiple cancers; however, the role of PSMD14 in ATC remains largely unknown. Here, we found that PSMD14 was upregulated in ATC tissues and that its aberrant expression was negatively associated with the overall survival of patients with ATC. Functionally, PSMD14 promotes the proliferation and invasiveness of ATC cells, whereas the depletion of PSMD14 or PSMD14 inhibitor thiolutin (THL) inhibits the growth, invasiveness, and epithelial-mesenchymal transition ((EMT) of ATC cells. In addition, the cell cycle was arrested and apoptosis was increased in PSMD14-depleted ATC or ATC cells treated with THL in vitro. An in vivo assay indicated that THL exerted a potent inhibitory effect on ATC xenografts. Mechanistically, PSMD14 increased E2F1 stabilization by binding to and deubiquitinating E2F1. PSMD14-regulated E2F1 improved the activation of the ERK and AKT signaling pathways, which are instrumental in ATC tumorigenesis and progression. Overall, our findings reveal the oncogenic role of PSMD14 in ATC and provide a promising therapeutic target for the treatment of ATC.
Collapse
Affiliation(s)
- Yuxuan Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Yuansheng Duan
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Kai Yue
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Linqi Li
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Jiayan Cao
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Weifeng Shi
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Jin Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Yansheng Wu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China
| | - Chao Jing
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Tianjin 300060, China.
| |
Collapse
|
8
|
Oliver S, Williams M, Jolly MK, Gonzalez D, Powathil G. Exploring the role of EMT in ovarian cancer progression using a multiscale mathematical model. NPJ Syst Biol Appl 2025; 11:36. [PMID: 40246908 PMCID: PMC12006308 DOI: 10.1038/s41540-025-00508-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) plays a key role in the progression of cancer tumours, significantly reducing the success of treatment. EMT occurs when a cell undergoes phenotypical changes, resulting in enhanced drug resistance, higher cell plasticity, and increased metastatic abilities. Here, we employ a 3D agent-based multiscale modelling framework using PhysiCell to explore the role of EMT over time in two cell lines, OVCAR-3 and SKOV-3. This approach allows us to investigate the spatiotemporal progression of ovarian cancer and the impacts of the conditions in the microenvironment. OVCAR-3 and SKOV-3 cell lines possess highly contrasting tumour layouts, allowing a wide range of different tumour dynamics and morphologies to be tested and studied. Along with performing sensitivity analysis on the model, simulation results capture the biological observations and trends seen in tumour growth and development, thus helping to obtain further insights into OVCAR-3 and SKOV-3 cell line dynamics.
Collapse
Affiliation(s)
- Samuel Oliver
- Department of Mathematics, Swansea University, Swansea, UK.
| | - Michael Williams
- Department of Biomedical Sciences, Swansea University, Swansea, UK
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore, India
| | | | - Gibin Powathil
- Department of Mathematics, Swansea University, Swansea, UK.
| |
Collapse
|
9
|
Duan Y, Ding L, Meng X, Lin J, Fu H, Zhu Y, Qiu Y, Cao J, Hu J, Dong Y, Duan Y, Chen J. A therapeutic strategy integrating ultrasound-guided microwave ablation with nanocomposite hydrogels to enhance autophagy and suppress tumor growth in hepatocellular carcinoma. Acta Biomater 2025:S1742-7061(25)00273-9. [PMID: 40246262 DOI: 10.1016/j.actbio.2025.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/25/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Microwave ablation (MWA) is widely recognized as an effective radical therapy for hepatocellular carcinoma (HCC). However, local ablation often results in a high risk of tumor recurrence. To address this challenge, we developed an effective anticancer drug delivery system comprising arsenic trioxide (As2O3)-loaded polyethylene glycol-dipalmitoylphosphatidylethanolamine (mPEG-DPPE) calcium phosphate nanoparticles (As2O3NPs) encapsulated within an injectable thermoresponsive hydrogel (ANPs-Gel). This study evaluated the therapeutic efficacy of MWA combined with ANPs-Gel in a rabbit hepatic VX2 tumor model. Ultrasound (US) and contrast-enhanced ultrasound (CEUS) were employed to assess tumor response and angiogenesis following treatment. The results demonstrated that MWA combined with ANPs-Gel significantly enhanced antitumor efficacy compared to other treatments, effectively inhibiting tumor growth and angiogenesis. Mechanistically, the therapeutic effects were associated with autophagy induced by MWA+ANPs-Gel, which played a critical role in promoting tumor cell death and suppressing epithelial-mesenchymal transition (EMT) both in vitro and in vivo. In vivo experiments further highlighted that the injectable thermoresponsive hydrogel system not only prolonged drug retention at the tumor site but also enhanced therapeutic efficacy by reducing EMT and preventing tumor recurrence. These findings suggest that MWA combined with ANPs-Gel provides a promising strategy for improving treatment outcomes in HCC through ultrasound-guided chemotherapy and targeted autophagy modulation. STATEMENT OF SIGNIFICANCE: This study introduces a potent therapeutic strategy that integrates ultrasound-guided microwave ablation (MWA) with a nanocomposite hydrogel to enhance autophagy and suppress tumor growth in hepatocellular carcinoma, as demonstrated in the rabbit VX2 hepatic tumor model. By combining advanced ultrasound guidance with a sophisticated nanomaterial platform, this approach significantly improves the efficacy of localized cancer therapy. Unlike conventional treatments, it not only ablates tumor cells but also regulates key cellular processes, such as autophagy, to amplify therapeutic outcomes. This work repurposes arsenic trioxide (Arsenic Trioxide) within a nanocomposite hydrogel delivery system and provides a detailed exploration of its therapeutic mechanisms when combined with MWA therapy. These findings pave the way for advanced clinical strategies in liver cancer management.
Collapse
Affiliation(s)
- Yi Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Li Ding
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Xianwei Meng
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiangtao Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Hao Fu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Yan Zhu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Yijie Qiu
- Department of Ultrasound, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China
| | - Jiaying Cao
- Department of Ultrasound, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China
| | - Jian Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Yi Dong
- Department of Ultrasound, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China.
| | - Yourong Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China.
| | - Jianhua Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
10
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [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: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
11
|
Liu S, Liao S, He J, Zhou Y, He Q. IGF2BP2: an m 6A reader that affects cellular function and disease progression. Cell Mol Biol Lett 2025; 30:43. [PMID: 40205577 PMCID: PMC11983839 DOI: 10.1186/s11658-025-00723-9] [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: 11/30/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
Insulin-like growth factor 2 messenger RNA (mRNA)-binding protein 2 (IGF2BP2) is a widely studied N6-methyladenosine (m6A) modification reader, primarily functioning to recognize and bind to m6A modification sites on the mRNA of downstream target genes, thereby enhancing their stability. Previous studies have suggested that the IGF2BP2-m6A modification plays an essential role in cellular functions and the progression of various diseases. In this review, we focus on summarizing the molecular mechanisms by which IGF2BP2 enhances the mRNA stability of downstream target genes through m6A modification, thereby regulating cell ferroptosis, epithelial-mesenchymal transition (EMT), stemness, angiogenesis, inflammatory responses, and lipid metabolism, ultimately affecting disease progression. Additionally, we update the related research progress on IGF2BP2. This article aims to elucidate the effects of IGF2BP2 on cell ferroptosis, EMT, stemness, angiogenesis, inflammatory responses, and lipid metabolism, providing a new perspective for a comprehensive understanding of the relationship between IGF2BP2 and cell functions such as ferroptosis and EMT, as well as the potential for targeted IGF2BP2 therapy for tumors and other diseases.
Collapse
Affiliation(s)
- Siyi Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, 410013, Hunan, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Shan Liao
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Junyu He
- Department of Clinical Laboratory, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, 410007, Hunan, People's Republic of China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China.
| | - Qian He
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, 410013, Hunan, China.
| |
Collapse
|
12
|
Jin Y, Hu H, Tian Y, Xu H, Yu Q, Cheng L, Guo X, Wang Z, Huang X, Wang X, Wang G. The role of LncRNA-MANCR induced by HIF-1α drive the malignant progression of pancreatic cancer by targeting miRNA-494/SIRT1 signaling axis under hypoxic conditions. Cancer Gene Ther 2025:10.1038/s41417-025-00900-0. [PMID: 40195439 DOI: 10.1038/s41417-025-00900-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/08/2025] [Accepted: 03/25/2025] [Indexed: 04/09/2025]
Abstract
This study revealed the prospective biological role and fundamental mechanisms of hypoxia-induced lncRNA-MANCR (MANCR), which is notably upregulated in pancreatic cancer (PC). This work uncovered the potential biological function and underlying mechanisms of hypoxia-induced MANCR, which is significantly elevated in PC. Microarray assays confirmed MANCR expression in the tissues of patients with PC and patients with chronic pancreatitis (CP), which positively correlated with sirtuin-1 (SIRT1) mRNA levels. Chromatin immunoprecipitation and luciferase assays were employed to gauge binding within the hypoxia-inducible factor-1α (HIF-1α)/MANCR/miRNA-494/SIRT1 pathway. Additionally, the association between MANCR expression and the clinical outcomes of patients with PC was confirmed. MANCR is significantly upregulated in PC cells under hypoxic conditions, which is closely linked to poor prognosis in patients with PC. Depletion of MANCR repressed in vitro proliferation, migration, and invasion of PC cells and in vivo growth of PC xenograft tumours. We further demonstrated that MANCR is localised in the cytoplasm and competitively binds miR-494, which directly targets SIRT1. Mechanically, the overexpression of SIRT1 improved the stability of the HIF-1α protein through deacetylation, leading to enhanced HIF-1α assembly. Moreover, MANCR underwent transcriptional regulation by HIF-1α in a hypoxic setting. This modulation was ascribed to HIF-1α binding to hypoxia response elements present in the MANCR promoter sequence. Data revealed the potential possibility of feedback between MANCR and HIF-1α, which may be conducive to hypoxia-induced oncogenicity and PC tumorigenesis, thereby providing a suitable therapeutic target.
Collapse
Affiliation(s)
- Yan Jin
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Hu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Yitong Tian
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Han Xu
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Qiao Yu
- Ultrasound medicine department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Long Cheng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyu Guo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zongwei Wang
- School of Medicine, Stanford University, San Francisco, CA, USA
| | - Xiaoxu Huang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
13
|
Montemurro M, Monier B, Suzanne M. The mechanical state of pre-tumoral epithelia controls subsequent Drosophila tumor aggressiveness. Dev Cell 2025; 60:1036-1052.e7. [PMID: 39765232 DOI: 10.1016/j.devcel.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/13/2024] [Accepted: 12/04/2024] [Indexed: 04/10/2025]
Abstract
Tumors evolve through the acquisition of increasingly aggressive traits associated with dysplasia. This progression is accompanied by alterations in tumor mechanical properties, especially through extracellular matrix remodeling. However, the contribution of pre-tumoral tissue mechanics to tumor aggressiveness remains poorly known in vivo. Here, we show that adherens junction tension in pre-tumoral tissues dictates subsequent tumor evolution in Drosophila. Increased cell contractility, observed in aggressive tumors before any sign of tissue overgrowth, proved sufficient to trigger dysplasia in normally hyperplastic tumors. In addition, high contractility precedes any changes in cell polarity and contributes to tumor evolution through cell death induction, which favors cell-cell junction weakening. Overall, our results highlight the need to re-evaluate the roles of tumoral cell death and identify pre-tumoral cell mechanics as an unsuspected early marker and key trigger of tumor aggressiveness.
Collapse
Affiliation(s)
- Marianne Montemurro
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France
| | - Bruno Monier
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France.
| | - Magali Suzanne
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France.
| |
Collapse
|
14
|
Ciaramicoli LM, Kwon HY, Im CY, Kim N, Oh Y, Chang YT, Kang NY. Label-Free Enrichment of Highly Metastatic Tumor-Initiating Cells up to a Monoclonal State. Biomater Res 2025; 29:0168. [PMID: 40177028 PMCID: PMC11964298 DOI: 10.34133/bmr.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/30/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Affiliation(s)
- Larissa M. Ciaramicoli
- Department of Chemistry,
Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Haw-Young Kwon
- Department of Chemistry,
Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- SenPro Inc.,
Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Chun Y. Im
- New Drug Development Center,
Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), Daegu 41061, Republic of Korea
| | - Namhui Kim
- New Drug Development Center,
Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), Daegu 41061, Republic of Korea
| | - Yoojin Oh
- New Drug Development Center,
Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), Daegu 41061, Republic of Korea
| | - Young-Tae Chang
- Department of Chemistry,
Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- SenPro Inc.,
Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Nam-Young Kang
- SenPro Inc.,
Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
- Department of Convergence I.T. Engineering,
Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| |
Collapse
|
15
|
Chen H, Lin L, Qiao Z, Pei Y, Gao Y, Lou K, Yang L, Li C, He Y, Bai J, Zhang G. YTHDF3 drives tumor growth and metastasis by recruiting eIF4B to promote Notch2 translation in breast cancer. Cancer Lett 2025; 614:217534. [PMID: 39924078 DOI: 10.1016/j.canlet.2025.217534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
YTH domain family protein 3 (YTHDF3), an m6A RNA reader, is implicated in various cancers, but its role in breast cancer progression and metastasis remains unclear. In this study, we explore the oncogenic potential of YTHDF3 in breast cancer, focusing on its impact on epithelial-mesenchymal transition (EMT) and metastasis. We found that YTHDF3 is significantly upregulated in breast cancer tissues and associated with poor relapse-free survival (RFS). Functional studies demonstrated that YTHDF3 promotes EMT in breast cancer cell lines by enhancing cell migration, invasion, and metastasis in vivo. Mechanistically, we show that YTHDF3 regulates Notch2, a key driver of EMT, through an m6A-dependent mechanism. YTHDF3 binds to m6A-modified Notch2 mRNA and recruits eIF4B to facilitate its translation, leading to increased Notch2 translation and subsequent inducing EMT. Our findings highlight the importance of the YTHDF3-Notch2 axis in driving EMT and metastasis in breast cancer. Furthermore, targeting YTHDF3 with lipid nanoparticles (LNPs) encapsulating siRNA and indocyanine green (ICG) significantly suppressed tumor growth and lung metastasis while enabling real-time therapeutic monitoring via ICG fluorescence imaging. These findings establish YTHDF3 as a critical driver of EMT and metastasis through m6A-dependent Notch2 translation, highlighting its potential as a therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Hongyu Chen
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Linling Lin
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Zishan Qiao
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Yifei Pei
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Yiyang Gao
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Kangliang Lou
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Lulin Yang
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Chengxi Li
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Yueyang He
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Jingwen Bai
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; The Breast Center, Yunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, 650118, Kunming, China.
| | - Guojun Zhang
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China; The Breast Center, Yunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, 650118, Kunming, China.
| |
Collapse
|
16
|
Xie T, Guo J, Wang W. The Long Noncoding RNA Gall Bladder Cancer-Associated Suppressor of Pyruvate Carboxylase Inhibits the Proliferation, Migration, and Invasion of Colorectal Cancer Cells and Induces Their Apoptosis. Biochem Genet 2025; 63:1719-1733. [PMID: 38609669 DOI: 10.1007/s10528-024-10786-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 03/15/2024] [Indexed: 04/14/2024]
Abstract
This study aimed to determine the role of the long noncoding RNA (lncRNA) gall bladder cancer-associated suppressor of pyruvate carboxylase (SOD2-1) in the progression of colorectal cancer (CRC). A total of 23 pairs of specimens, including CRC tissues and adjacent normal tissues, were collected, and the expression of lncRNA SOD2-1 (lnc-SOD2-1) was measured. lnc-SOD2-1 function was examined using HCT15 and HCT116 cells. A lnc-SOD2-1 overexpression vector was designed and transfected into both cell lines. MTS and colony formation assays were used to determine cell viability. Flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling assays were performed to measure apoptosis. Cell migration and invasion were evaluated using the Transwell assay. Migration and invasion markers were validated using quantitative reverse transcription-polymerase chain reaction and western blot analysis. The results indicated that the expression of lnc-SOD2-1 was downregulated in CRC tissues. lnc-SOD2-1 overexpression evidently decreased cell viability and led to the formation of fewer cell colonies. lnc-SOD2-1 overexpression induced ~ twofold higher apoptosis than the control group. lnc-SOD2-1 overexpression reduced the proportion of migratory and invasive cells to 50% and 75% of the control group, respectively. lnc-SOD2-1 overexpression significantly decreased the expression of matrix metalloproteinase-2 and -9. In conclusion, lnc-SOD2-1 may act as a tumor suppressor that inhibits the proliferation, migration, and invasion of CRC cells and induces their apoptosis.
Collapse
Affiliation(s)
- Tingting Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianian Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No.250 Changgang East Road, Haizhu District, Guangzhou, 510260, China.
| |
Collapse
|
17
|
Li X, Lu J, Chen F, Yuan J, Zha Y, Li Y, Yan J, Li Q, Yuan J, Tong Q. Comprehensive proteomic analysis and multidimensional model construction of peritoneal metastasis in gastric cancer. Cancer Lett 2025; 614:217509. [PMID: 39914770 DOI: 10.1016/j.canlet.2025.217509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
Peritoneal metastasis following gastric cancer surgery is often associated with a poor prognosis. This study aimed to investigate the mechanisms underlying peritoneal metastasis and to develop a predictive model for the risk of postoperative peritoneal metastases in gastric cancer. We performed a comprehensive analysis of the protein mass spectra and tumor microenvironment in paraffin-embedded primary tumor sections from gastric cancer patients, both with and without postoperative peritoneal metastases. Using proteomic profiling, we identified 9595 proteins and stratified patients into three distinct proteomic subgroups (Pro1, Pro2, Pro3) based on differential protein expression. Simultaneously, immune cell profiling allowed us to classify patients into four immune subgroups (IG-I, IG-II, IG-III, IG-IV). The relationships between these proteomic, immune, and metastasis classifications were further explored to uncover potential associations and mechanisms driving metastasis. Building on these insights, we developed an integrative model combining proteomics, immunological, and radiomics data for predicting postoperative peritoneal metastases. This model demonstrated high predictive efficacy, offering a robust tool for identifying high-risk patients. Our findings provide a deeper understanding of the biological processes underlying peritoneal metastasis in gastric cancer, highlighting the interplay between proteomic and immune factors. By establishing novel patient subgroups and an effective prediction model, this study lays the groundwork for early diagnosis and tailored therapeutic strategies to improve outcomes for gastric cancer patients.
Collapse
Affiliation(s)
- Xiangpan Li
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jiatong Lu
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of WuhanUniversity, Wuhan, 430060, China
| | - Fangfang Chen
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jingwen Yuan
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of WuhanUniversity, Wuhan, 430060, China; Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yunfei Zha
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ying Li
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Junfeng Yan
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of WuhanUniversity, Wuhan, 430060, China
| | - Qiang Li
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of WuhanUniversity, Wuhan, 430060, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qiang Tong
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of WuhanUniversity, Wuhan, 430060, China.
| |
Collapse
|
18
|
Cheng XX, Lin GW, Zhou YQ, Li YQ, He S, Liu Y, Zeng YN, Guo YM, Liu SQ, Peng W, Wei PP, Luo CL, Bei JX. A rare KLHDC4 variant Glu510Lys is associated with genetic susceptibility and promotes tumor metastasis in nasopharyngeal carcinoma. J Genet Genomics 2025; 52:559-569. [PMID: 39706520 DOI: 10.1016/j.jgg.2024.12.008] [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: 09/05/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Various genetic association studies have identified numerous single nucleotide polymorphisms (SNPs) associated with nasopharyngeal carcinoma (NPC) risk. However, these studies have predominantly focused on common variants, leaving the contribution of rare variants to the "missing heritability" largely unexplored. Here, we integrate genotyping data from 3925 NPC cases and 15,048 healthy controls to identify a rare SNP, rs141121474, resulting in a Glu510Lys mutation in KLHDC4 gene linked to increased NPC risk. Subsequent analyses reveal that KLHDC4 is highly expressed in NPC and correlates with poorer prognosis. Functional characterizations demonstrate that KLHDC4 acts as an oncogene in NPC cells, enhancing their migratory and metastatic capabilities, with these effects being further augmented by the Glu510Lys mutation. Mechanistically, the Glu510Lys mutant exhibits increased interaction with Vimentin compared to the wild-type KLHDC4 (KLHDC4-WT), leading to elevated Vimentin protein stability and modulation of the epithelial-mesenchymal transition process, thereby promoting tumor metastasis. Moreover, Vimentin knockdown significantly mitigates the oncogenic effects induced by overexpression of both KLHDC4-WT and the Glu510Lys variant. Collectively, our findings highlight the critical role of the rare KLHDC4 variant rs141121474 in NPC progression and propose its potential as a diagnostic and therapeutic target for NPC patients.
Collapse
Affiliation(s)
- Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Guo-Wang Lin
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yi-Qi Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yan-Ni Zeng
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yun-Miao Guo
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, Guangdong 524045, China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Wan Peng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China.
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Sun Yat-sen University Institute of Advanced Studies Hong Kong, Science Park, Hong Kong SAR, China; Department of Medical Oncology, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
19
|
Zhang H, Jiao J, Long Y, Zhou L, Lv Y, Wei W, Sun Y, Han H, Chen C, Zhu Y, Zhang W. Targeting capture and eradicate circulating tumor cells by activated platelet derived vehicle for inhibiting triple-negative breast cancer metastasis. Mater Today Bio 2025; 31:101597. [PMID: 40092226 PMCID: PMC11910114 DOI: 10.1016/j.mtbio.2025.101597] [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: 12/22/2024] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Circulating tumor cells (CTCs) are cardinal intermediaries in the metastatic cascade, particularly in triple-negative breast cancer (TNBC), owing to their high-affinity interactions that bolster survival and dissemination. Addressing this pivotal mechanism, we have developed APEVs@DOX, a pioneering biomimetic delivery system. Utilizing activated platelet membranes as a scaffold, APEVs@DOX recapitulates the natural affinity between platelets and CTCs, enabling targeted delivery of doxorubicin. Our results, substantiated by meticulous in vitro and in vivo experimentation, revealed 78 % reduction in lung metastasis nodules in murine models relative to controls, affirming APEVs@DOX's proficiency in CTCs capture and eradication. This study not only illuminates the potential of CTCs-targeted therapies in the precision medicine armamentarium for TNBC but also contributes empirical data to guide the strategic design of anti-metastatic interventions. The therapeutic impact of APEVs@DOX in curtailing metastatic spread offers a beacon of hope for advancing TNBC treatment paradigms.
Collapse
Affiliation(s)
- Hongmei Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University , Nanjing, 210000, China
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210000, China
- Gansu Wuwei Institute of Medical Sciences, Gansu, 733000, China
| | - Jinlan Jiao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University , Nanjing, 210000, China
| | - Yongxuan Long
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University , Nanjing, 210000, China
| | - Lina Zhou
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Yinhua Lv
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Wenqian Wei
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Yuxiang Sun
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Hao Han
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of NanJing University Medical School, Nanjing, 210000, China
| | - Changrong Chen
- Department of Emergency Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Yun Zhu
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University , Nanjing, 210000, China
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210000, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, Jiangsu, 210000, China
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University , Nanjing, 210000, China
| |
Collapse
|
20
|
Kulkarni AM, Gayam PKR, Baby BT, Aranjani JM. Epithelial-Mesenchymal Transition in Cancer: A Focus on Itraconazole, a Hedgehog Inhibitor. Biochim Biophys Acta Rev Cancer 2025; 1880:189279. [PMID: 39938662 DOI: 10.1016/j.bbcan.2025.189279] [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: 07/22/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Cancer, and the resulting mortality from it, is an ever-increasing concern in global health. Cancer mortality stems from the metastatic progression of the disease, by dissemination of the tumor cells. Epithelial-Mesenchymal Transition, the major hypothesis purported to be the origin of metastasis, confers mesenchymal phenotype to epithelial cells in a variety of contexts, physiological and pathological. EMT in cancer leads to rise of cancer-stem-like cells, drug resistance, relapse, and progression of malignancy. Inhibition of EMT could potentially attenuate the mortality. While novel molecules for inhibiting EMT are underway, repurposing drugs is also being considered as a viable strategy. In this review, Itraconazole is focused upon, as a repurposed molecule to mitigate EMT. Itraconazole is known to inhibit Hedgehog signaling, and light is shed upon the existing evidence, as well as the questions remaining to be answered.
Collapse
Affiliation(s)
- Aniruddha Murahar Kulkarni
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576104, India.
| | - Prasanna Kumar Reddy Gayam
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576104, India.
| | - Beena Thazhackavayal Baby
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576104, India
| | - Jesil Mathew Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576104, India.
| |
Collapse
|
21
|
Wang Y, Huang J, Song Z, Zhang S, Guo H, Leng Q, Fang N, Ji S, Yang J. c-Jun promotes neuroblastoma cell differentiation by inhibiting APC formation via CDC16 and reduces neuroblastoma malignancy. Biol Direct 2025; 20:37. [PMID: 40149013 PMCID: PMC11948754 DOI: 10.1186/s13062-025-00630-1] [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: 12/24/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Neuroblastoma is a pediatric embryonal malignancy characterized by impaired neuronal differentiation. Differentiation status in neuroblastoma strongly affects the clinical outcome, thus, enforcement of differentiation becomes a treatment strategy for this disease. However, the molecular mechanisms that control neuroblastoma differentiation are poorly understood. As an extensively studied protein of the activator protein-1 (AP-1) complex, c-Jun is involved in numerous cell regulations such as proliferation, survival and differentiation. In the current study, we demonstrated that c-Jun expression was upregulated by retinoic acid (RA) and flow cytometry assay indicated c-Jun overexpression arrested cell cycle to G1 phase, which, in turn, promoted the initiation of neuroblastoma cell differentiation. Co-immunoprecipitation (co-IP) assay showed that c-Jun competitively interacted with CDC16, a key subunit in anaphase-promoting complex (APC), resulting in reduced APC formation and inhibition of cell cycle progression. Furthermore, EdU proliferation assay and transwell experiment showed that c-Jun overexpression inhibited neuroblastoma cell proliferation and migration via interacting and sequestering CDC16. These findings identify c-Jun as a key regulator of neuroblastoma cell cycle and differentiation and may represent a promising therapeutic target to induce neuroblastoma differentiation via the interaction between c-Jun and CDC16.
Collapse
Affiliation(s)
- Yunyun Wang
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Science, Henan University, Kaifeng, Henan Province, 475004, China.
| | - Jingjing Huang
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Science, Henan University, Kaifeng, Henan Province, 475004, China
| | - Zhenhua Song
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Science, Henan University, Kaifeng, Henan Province, 475004, China
| | - Shuo Zhang
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Science, Henan University, Kaifeng, Henan Province, 475004, China
| | - Haojie Guo
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Science, Henan University, Kaifeng, Henan Province, 475004, China
| | - Qi Leng
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Science, Henan University, Kaifeng, Henan Province, 475004, China
| | - Na Fang
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Science, Henan University, Kaifeng, Henan Province, 475004, China
| | - Shaoping Ji
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Science, Henan University, Kaifeng, Henan Province, 475004, China.
- Zhengzhou Shuqing Medical College, Zhengzhou, Henan Province, 450064, China.
| | - Jian Yang
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
22
|
Chen Q, Wang H, Liu Q, Luo C. CTHRC1: a key player in colorectal cancer progression and immune evasion. Front Immunol 2025; 16:1579661. [PMID: 40201173 PMCID: PMC11975584 DOI: 10.3389/fimmu.2025.1579661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
The multifunctional secreted protein, collagen triple helix repeat containing 1 (CTHRC1), has recently emerged as a significant focus within oncology research. CTHRC1 expression in tumors is governed by a complex interplay of regulatory signals, including methylation, glycosylation, and notably, non-coding RNAs, which constitute its predominant regulatory mechanism. Colorectal cancer (CRC), a highly prevalent epithelial malignancy, sees CTHRC1 influencing tumor progression and metastasis through its modulation of several downstream signaling cascades, such as Wnt/PCP, TGF-β/Smad, and MEK/ERK pathways. Furthermore, CTHRC1 contributes to immune evasion in CRC via diverse mechanisms. It is intricately associated with macrophage phenotypic switching within the tumor microenvironment (TME), favoring M2 macrophage polarization and facilitating the infiltration of T cells and neutrophils. CTHRC1 is also instrumental in immune escape by driving the remodeling of the extracellular matrix through interactions with cancer-associated fibroblasts. Additionally, CTHRC1's roles extend to the regulation of hypoxia-related pathways, metabolism of glycolysis and fatty acids, and involvement in tumor angiogenesis, all of which support tumor immune evasion. Considering its multifaceted activities, CTHRC1 emerges as a promising therapeutic target in CRC, with the potential to enhance the outcomes of existing radiotherapeutic and immunotherapeutic regimens. This review endeavors to delineate the mechanistic and therapeutic landscapes of CTHRC1 in CRC. Through a comprehensive discussion of CTHRC1's diverse functions, we aim to provide insights that could pave the way for innovative approaches in cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Changjiang Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
23
|
Fan C, Wang Q, Krijger PHL, Cats D, Selle M, Khorosjutina O, Dhanjal S, Schmierer B, Mei H, de Laat W, Ten Dijke P. Identification of a SNAI1 enhancer RNA that drives cancer cell plasticity. Nat Commun 2025; 16:2890. [PMID: 40133308 PMCID: PMC11937597 DOI: 10.1038/s41467-025-58032-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Enhancer RNAs (eRNAs) are a pivotal class of enhancer-derived non-coding RNAs that drive gene expression. Here we identify the SNAI1 enhancer RNA (SNAI1e; SCREEM2) as a key activator of SNAI1 expression and a potent enforcer of transforming growth factor-β (TGF-β)/SMAD signaling in cancer cells. SNAI1e depletion impairs TGF-β-induced epithelial-mesenchymal transition (EMT), migration, in vivo extravasation, stemness, and chemotherapy resistance in breast cancer cells. SNAI1e functions as an eRNA to cis-regulate SNAI1 enhancer activity by binding to and strengthening the enrichment of the transcriptional co-activator bromodomain containing protein 4 (BRD4) at the local enhancer. SNAI1e selectively promotes the expression of SNAI1, which encodes the EMT transcription factor SNAI1. Furthermore, we reveal that SNAI1 interacts with and anchors the inhibitory SMAD7 in the nucleus, and thereby prevents TGF-β type I receptor (TβRI) polyubiquitination and proteasomal degradation. Our findings establish SNAI1e as a critical driver of SNAI1 expression and TGF-β-induced cell plasticity.
Collapse
Affiliation(s)
- Chuannan Fan
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Qian Wang
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Peter H L Krijger
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Davy Cats
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Miriam Selle
- Department of Medical Biochemistry and Biophysics, SciLifeLab and Karolinska Institute, Solna, Sweden
| | - Olga Khorosjutina
- Department of Medical Biochemistry and Biophysics, SciLifeLab and Karolinska Institute, Solna, Sweden
| | - Soniya Dhanjal
- Department of Medical Biochemistry and Biophysics, SciLifeLab and Karolinska Institute, Solna, Sweden
| | - Bernhard Schmierer
- Department of Medical Biochemistry and Biophysics, SciLifeLab and Karolinska Institute, Solna, Sweden
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter de Laat
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
24
|
Li J, Wang ZY, Jin Y, Xu J, Ya YJ, Wan TQ, Li X, Wang X. Transmembrane channel-like 5 drives hepatocellular carcinoma progression by regulating epithelial-mesenchymal transition. World J Clin Oncol 2025; 16:94091. [PMID: 40130046 PMCID: PMC11866081 DOI: 10.5306/wjco.v16.i3.94091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a difficult cancer to manage due to its highly invasive and metastatic nature. AIM To investigate the molecular function of transmembrane channel-like 5 (TMC5) in vitro and in vivo, with the objective of identifying novel diagnosis and treatment targets for HCC. METHODS The expression of TMC in cancer and normal tissues, along with its correlation with HCC prognosis, was analyzed using the GENT2, GEPIA database, and Human Protein Atlas. COX analysis was conducted to assess the relationship between TMC5 expression and overall survival in TCGA-LIHC patients. Further experiments were conducted to investigate the effect of TMC5 in cancer progression through loss- and gain-of-function assays in vitro and in vivo. RESULTS Bioinformatics revealed that TMC5 expression was generally higher in tumors than in normal tissues, and its expression was associated with poorer patient survival outcomes. TMC5 expression in HCC tissues and cells was consistent with the results of the bioinformatics analysis. Suppression of TMC5 expression reduced migration, invasion, and proliferation, while also decreasing the expression of epithelial-mesenchymal transition (EMT)-associated molecules in MHCC97-LM3 cells. Conversely, higher TMC5 expression significantly increased cell migration, invasion, proliferation, and EMT in MHCC97 L cells. TMC5 knockdown significantly decreased both the formation and spread of nodules in liver tissue, whereas TMC5 overexpression promoted them. CONCLUSION Our study provides compelling evidence that TMC5 is highly expressed in HCC and drives cancer progression through the activation of EMT-mediated invasion. TMC5 could represent a valuable molecular target for the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Jiao Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Zi-Yu Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Yan Jin
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Jing Xu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Yun-Jin Ya
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Ting-Qiu Wan
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Xi Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Xi Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| |
Collapse
|
25
|
Ye X, Song Q, Zhang L, Jing M, Fu Y, Yan W. Cysteine-rich intestinal protein family: structural overview, functional diversity, and roles in human disease. Cell Death Discov 2025; 11:114. [PMID: 40118853 PMCID: PMC11928533 DOI: 10.1038/s41420-025-02395-y] [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: 10/14/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
The cysteine-rich intestinal protein (CRIP) family, including CRIP1, CRIP2, and CRIP3, is a subfamily of the highly conserved Lin-1, Isl1, Mec3/double zinc finger protein family that exhibits diverse biological functions. The CRIP family is known to play an important role in cellular epithelial-mesenchymal transition, cell death, and tumor progression and participate in multiple signaling pathways. This article summarizes the roles and potential molecular mechanisms of the CRIP family in diseases, which will help to explore new research directions for this family and provide useful information for clinical applications such as disease diagnosis and treatment.
Collapse
Affiliation(s)
- Xilin Ye
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianben Song
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lumiao Zhang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengjia Jing
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yu Fu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
26
|
Liang W, Peng Z, Mingchu Z, Deshui Y. METTL3 mediated WISP1 m 6A modification promotes epithelial-mesenchymal transition and tumorigenesis in laryngeal squamous cell carcinoma via m 6A reader IGF2BP1. Gene 2025; 941:149222. [PMID: 39761803 DOI: 10.1016/j.gene.2025.149222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
OBJECT N6-methyladenosine (m6A), is well known as the most abundant epigenetic modification in messenger RNA, but its influence on laryngeal squamous cell carcinoma (LSCC) remains largely unexplored and poorly understood. This study was designed to explore the effects of m6A on WISP1-mediated epithelial-mesenchymal transition (EMT) and tumorigenesis in LSCC. METHODS m6A methylated and expression levels of WISP1 in LSCC tumor tissues and cells were measured by MeRIP-qPCR, qRT-PCR, and western blotting. The regulatory mechanism of m6A modification of WISP1 in LSCC was determined using MeRIP-qPCR, RIP, dual luciferase reporter assay, and RNA stability assay. Cell viability was assessed utilizing MTT method. The invasion and migration ability of LSCC cells were determined by transwell and wound healing method, respectively. Tumor xenograft models were used for the in vivo experiments. RESULTS The m6A methylation level of WISP1 was significantly enhanced in LSCC patients and LSCC cell lines. Overexpression of the m6A methyltransferase METTL3 significantly upregulated WISP1 expression by promoting its m6A methylation level, whereas METTL3 inhibition exhibited the opposite effect in LSCC cells. Functionally, we found that METTL3 accelerated the viability, invasion, migration, and EMT of LSCC cells by upregulating WISP1. Additionally, overexpression of METTL3 increased WISP1 expression and tumorigenesis were verified in in vivo experiments. Mechanistically, m6A-modified WISP1 was recognized by IGF2BP1, which enhanced the stability of WISP1 mRNA. CONCLUSION Our findings indicate that the m6A modification of WISP1 promotes EMT in LSCC by enhancing WISP1 mRNA stability via an IGF2BP1-dependent manner, which may highlight an m6A methylation-based approach for LSCC diagnosis and therapy.
Collapse
Affiliation(s)
- Wang Liang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zhang Peng
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhang Mingchu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Deshui
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
27
|
Ren R, Li Z, Fang Q. A disintegrin-like and metalloproteinase 15 facilitates glioblastoma proliferation and metastasis through activation of the protease-activated receptor 1. Cytojournal 2025; 22:34. [PMID: 40260066 PMCID: PMC12010881 DOI: 10.25259/cytojournal_92_2024] [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: 06/05/2024] [Accepted: 02/25/2025] [Indexed: 04/23/2025] Open
Abstract
Objective Glioblastoma hinders therapeutic interventions and prognostic outlooks. At the same time, a disintegrin-like and metalloproteinase 15 (ADAM15) influences cellular processes, such as adhesion and migration. Furthermore, protease-activated receptor 1 (PAR1), a vital receptor, impacts tumorigenesis and disease progression. This study aimed to investigate ADAM15 and PAR1 interaction in epithelial-mesenchymal transition (EMT) modulation in glioblastoma behavior and provide insights into therapeutic targets. Material and Methods The impacts of ADAM15 overexpression and PAR-1/2 inhibition on the proliferation, invasion, and migration of glioblastoma cells U251 and U87 were evaluated using transwell assays, EdU incorporation, clonogenic assay, Ki67 immunohistochemistry, and immunofluorescence staining. Real-time quantitative polymerase chain reaction and Western blot analysis were employed to investigate the impact of ADAM15 on PAR1 expression. Results After analyzing the impacts of ADAM15 overexpression on the migration, invasion, and proliferation of human glioblastoma cell lines U251 and U87, the results showed that ADAM15 overexpression significantly enhanced migration (P < 0.001) and invasion rates (P < 0.001), as confirmed by scratch and transwell assays, thus indicating its tumor-promoting effects. This study revealed a significant increase in colony formation (P < 0.001), EdU incorporation (P < 0.001), and Ki67-positive cells (P < 0.001) in the ADAM15 overexpressed group. PAR1 and EMT markers were significantly increased in the ADAM15 overexpressed group (P < 0.001). Treatment with the PAR-1 antagonist SCH79797 inhibited EMT (P < 0.01) and suppressed cell proliferation (P < 0.001), migration (P < 0.001), and invasion (P < 0.001) in U251 and U87 cells overexpressing ADAM15, indicating the involvement of PAR-1 signaling in the effects of ADAM15 on cell behaviors. In comparison, the PAR-2 antagonist FSLLRY-NH2 did not show significant effects on EMT or these cell behaviors. Conclusion ADAM15 drives glioblastoma cell lines U251 and U87 progression through PAR1.
Collapse
Affiliation(s)
- Rong Ren
- Department of Traditional Chinese Medicine, Qingdao Chengyang People’s Hospital, Qingdao, Shandong, China
| | - Zuowei Li
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Qingdao Medical College, Qing Dao University, Qingdao, Shandong, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Qiong Fang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
28
|
Liu M, Hernandez MO, Castven D, Lee HP, Wu W, Wang L, Forgues M, Hernandez JM, Marquardt JU, Ma L. Tumor cell villages define the co-dependency of tumor and microenvironment in liver cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.07.642107. [PMID: 40161587 PMCID: PMC11952337 DOI: 10.1101/2025.03.07.642107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Spatial cellular context is crucial in shaping intratumor heterogeneity. However, understanding how each tumor establishes its unique spatial landscape and what factors drive the landscape for tumor fitness remains significantly challenging. Here, we analyzed over 2 million cells from 50 tumor biospecimens using spatial single-cell imaging and single-cell RNA sequencing. We developed a deep learning-based strategy to spatially map tumor cell states and the architecture surrounding them, which we referred to as Spatial Dynamics Network (SDN). We found that different tumor cell states may be organized into distinct clusters, or 'villages', each supported by unique SDNs. Notably, tumor cell villages exhibited village-specific molecular co-dependencies between tumor cells and their microenvironment and were associated with patient outcomes. Perturbation of molecular co-dependencies via random spatial shuffling of the microenvironment resulted in destabilization of the corresponding villages. This study provides new insights into understanding tumor spatial landscape and its impact on tumor aggressiveness.
Collapse
Affiliation(s)
- Meng Liu
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Maria O. Hernandez
- Spatial Imaging Technology Resource, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Darko Castven
- Department of Medicine I, University Medical Center, Lübeck, Germany
| | - Hsin-Pei Lee
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Wenqi Wu
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Limin Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Marshonna Forgues
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jonathan M. Hernandez
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Jens U. Marquardt
- Department of Medicine I, University Medical Center, Lübeck, Germany
| | - Lichun Ma
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| |
Collapse
|
29
|
Cai J, Wang J, Wang Z, Wang J, Jia Y, Ma X. Perspectives on the α5 nicotinic acetylcholine receptor in lung cancer progression. Front Cell Dev Biol 2025; 13:1489958. [PMID: 40143965 PMCID: PMC11937065 DOI: 10.3389/fcell.2025.1489958] [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: 09/02/2024] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) are widely expressed in a variety of cell types and are involved in multiple physiological regulatory mechanisms in cells, tissues and systems. Increasing evidence suggests that the α5 nicotinic acetylcholine receptor (α5-nAChR), encoded by the CHRNA5 gene, is one of a key mediator involved in lung cancer development and immune responses. Several studies have shown that it is a regulator that stimulates processes via various signaling pathways, including STAT3 in lung cancer. In addition, α5-nAChR has a profound effect on lung immune response through multiple immune-related factor pathways. In this review, we focus on the perspectives on α5-nAChR in lung cancer progression, which indicates that targeting α5-nAChR could provide novel anticancer and immune therapy strategies for lung cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
30
|
Li J, Nagasaka Y, Shen H, Zhou X, Ma J, Trevisan-Silva D, Sherman NE, Ambati J, Gelfand BD, Guo LW. TMEM97 governs partial epithelial-mesenchymal transition of retinal pigment epithelial cells via the CTNND2-ADAM10 axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102460. [PMID: 39995975 PMCID: PMC11848774 DOI: 10.1016/j.omtn.2025.102460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is associated with retinal pigment epithelium (RPE) dysfunction in degenerative retinal diseases. However, the role of partial EMT (pEMT), a hybrid state exhibiting both epithelial and mesenchymal markers, remains poorly understood in this context. Our previous research demonstrated that TMEM97 ablation in mice worsens photoreceptor loss in an oxidant-induced RPE damage model. Here, we link TMEM97 to pEMT in RPE cells and explore the underlying molecular mechanisms. We found that re-expressing TMEM97 in the RPE of TMEM97-knockout mice, via subretinal lentiviral delivery, mitigated oxidant (NaIO3)-induced photoreceptor loss. Interestingly, TMEM97 knockout in ARPE19 cells in vitro led to upregulation of cadherin/adhesion-binding pathways, even without oxidant exposure. Integrated proteomic, transcriptomic, segmentation, and immunoblot analyses revealed that TMEM97 ablation induces pEMT, marked by the concurrent expression of epithelial E-cadherin and mesenchymal N-cadherin, a process reversed upon TMEM97 re-expression. Furthermore, TMEM97 negatively regulated CTNND2 protein (catenin δ-2), but not the known EMT driver β-catenin, and CTNND2 was found to promote ADAM10, which sustains both E- and N-cadherin protein levels. These findings identify TMEM97 as a novel regulator of RPE-cell pEMT through the CTNND2-ADAM10 axis, highlighting potential new targets for therapeutic intervention in RPE-related pathophysiology.
Collapse
Affiliation(s)
- Jing Li
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Yosuke Nagasaka
- Department of Ophthalmology, University of Virginia, Charlottesville, VA 22903, USA
| | - Hongtao Shen
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Xinyu Zhou
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Jianjie Ma
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Dilza Trevisan-Silva
- School of Medicine Core Facilities, University of Virginia, Charlottesville, VA 22903, USA
| | - Nicholas E. Sherman
- School of Medicine Core Facilities, University of Virginia, Charlottesville, VA 22903, USA
| | - Jayakrishna Ambati
- Department of Ophthalmology, University of Virginia, Charlottesville, VA 22903, USA
| | - Bradley D. Gelfand
- Department of Ophthalmology, University of Virginia, Charlottesville, VA 22903, USA
| | - Lian-Wang Guo
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
- Department of Ophthalmology, University of Virginia, Charlottesville, VA 22903, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
31
|
Song Q, Wu H, Jin Y, Hou J, Liu J, Zhang X, Hu W, Sun G, Zhang Z. Fruquintinib inhibits the migration and invasion of colorectal cancer cells by modulating epithelial-mesenchymal transition via TGF-β/Smad signaling pathway. Front Oncol 2025; 15:1503133. [PMID: 40134588 PMCID: PMC11932892 DOI: 10.3389/fonc.2025.1503133] [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: 09/28/2024] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Background Fruquintinib, a selective vascular endothelial growth factor receptor (VEGFR) inhibitor, has shown considerable efficacy in colorectal cancer (CRC) treatment. Despite its promising therapeutic effects, the precise molecular mechanisms underlying its therapeutic effects remain incompletely understood. In this study, we explored the functional roles and molecular mechanisms of fruquintinib in CRC therapy. Material and methods Human CRC cells (HCT-116 and LOVO) were cultured and treated with fruquintinib. Cell counting kit-8 assay kit (CCK-8) and colony formation assays were performed to investigate the effects of fruquintinib on cell proliferation. Wound healing and transwell assays were conducted to explore the role of fruquintinib on migration and invasion. RNA sequencing and bioinformatics analysis was used to investigate the potential mechanism of fruquintinib in the development of CRC. Western blot was used to measure the protein level. Results Fruquintinib significantly inhibited the proliferation, migration, and invasion of colorectal cancer cells. Bioinformatics analysis indicated that fruquintinib modulated the epithelial-mesenchymal transition (EMT) pathway, and experimental validation confirmed its regulatory effects on core EMT-associated protein biomarkers. Notably, fruquintinib treatment resulted in the upregulation of E-cadherin and the downregulation of N-cadherin, vimentin, and MMP9. Western blot analysis revealed that fruquintinib dose-dependently suppressed SMAD2/3 expression. Notably, treatment with the TGF-β receptor agonist KRFK TFA attenuated fruquintinib's effect, reversing the upregulation of E-cadherin as well as the downregulatin of N-cadherin and SMAD2/3. Additionally, KRFK TFA partially restored CRC cell migration and invasion in transwell assays, counteracting fruquintinib's inhibitory impact. Conclusion These findings indicate that Fruquintinib effectively hampers the migration and invasion of CRC cells by disrupting the EMT process via the TGF-β/Smad signaling pathway. This study sheds light on the mechanisms by which fruquintinib inhibits CRC progression and underscores its potential for further clinical investigation.
Collapse
Affiliation(s)
- Qinqin Song
- Department of Oncology, Hebei Medical University, Shijiazhuang, China
- Affliated Tangshan Gongren Hospital, Hebei Medical University, Tangshan, China
| | - Hongjiao Wu
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Ye Jin
- College of Clinical Medicine, North China University of Science and Technology, Tangshan, China
| | - Junzhi Hou
- Department of Oncology, Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Jiawei Liu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xuemei Zhang
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Wanning Hu
- Department of Oncology, Hebei Medical University, Shijiazhuang, China
- Affliated Tangshan Gongren Hospital, Hebei Medical University, Tangshan, China
| | - Guogui Sun
- Department of Hebei Key Laboratory of Medical-Industrial Intergration Precision Medicine, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Zhi Zhang
- Department of Oncology, Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
32
|
Wang X, Zhang Y, Wang Y, Shi L, Yuan C, Yin W, Teng Y, Li J, Mao Y. The correlation between epithelial-mesenchymal transition classification and MMP2 expression of circulating tumor cells and prognosis of advanced or metastatic nasopharyngeal carcinoma. Open Med (Wars) 2025; 20:20241074. [PMID: 40093514 PMCID: PMC11909577 DOI: 10.1515/med-2024-1074] [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: 03/08/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 03/19/2025] Open
Abstract
Background Epithelial-mesenchymal transition (EMT) and circulating tumor cells (CTCs) are key prognostic factors in nasopharyngeal carcinoma (NPC). However, the role of EMT status in CTCs for predicting outcomes in advanced NPC treated with radiotherapy after induction chemotherapy remains unclear. Methods A total of 143 CTC tests from 95 advanced/metastatic NPC patients were analyzed before, during, and after radiotherapy, with a 60-month follow-up. CTC count, matrix metalloproteinase 2 (MMP2)) protein expression, and EMT subtypes were examined. Results During radiotherapy, CTC counts increase but decrease afterward. Patients with higher pre-radiotherapy tumor-node-metastasis (TNM) stages have lower total and M-subtype CTC counts. Higher T and TNM stages during radiotherapy correlate with increased EMT-state CTCs, especially hybrid CTCs. EA/IgG-positive patients have a higher number of hybrid CTCs and E-type (epithelial + hybrid) CTCs, while EBV-EA-negative patients have more mesenchymal CTCs. A higher post-radiotherapy CTC count predicts relapse, and the positive rate of MMP2 expression on hybrid and epithelial CTCs is higher than that on mesenchymal CTCs. Conclusion EMT status, particularly in hybrid CTCs, is a potential prognostic marker for relapse in advanced NPC after radiotherapy.
Collapse
Affiliation(s)
- Xiaoju Wang
- Radiotherapy Department, Hangzhou Cancer Hospital, Hangzhou, 310005, Zhejiang, China
| | - Yuxin Zhang
- Radiotherapy Department, Hangzhou Cancer Hospital, Hangzhou, 310005, Zhejiang, China
| | - Yiqing Wang
- Radiotherapy Department, Hangzhou Cancer Hospital, Hangzhou, 310005, Zhejiang, China
| | - Lei Shi
- Radiotherapy Department, Hangzhou Cancer Hospital, Hangzhou, 310005, Zhejiang, China
| | - Caiqin Yuan
- Radiotherapy Department, Hangzhou Cancer Hospital, Hangzhou, 310005, Zhejiang, China
| | - Wei Yin
- Radiotherapy Department, Hangzhou Cancer Hospital, Hangzhou, 310005, Zhejiang, China
| | - Yaoshu Teng
- ENT Department, Hangzhou First People’s Hospital, Hangzhou, 310005, Zhejiang, China
| | - Jing Li
- ENT Department, Hangzhou First People’s Hospital, Hangzhou, 310005, Zhejiang, China
| | - Yanjiao Mao
- Radiotherapy Department, Hangzhou Cancer Hospital, Hangzhou, 310005, Zhejiang, China
| |
Collapse
|
33
|
Yu HC, Bai L, Jin L, Zhang YJ, Xi ZH, Wang DS. SLC25A35 enhances fatty acid oxidation and mitochondrial biogenesis to promote the carcinogenesis and progression of hepatocellular carcinoma by upregulating PGC-1α. Cell Commun Signal 2025; 23:130. [PMID: 40065301 PMCID: PMC11895242 DOI: 10.1186/s12964-025-02109-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 02/15/2025] [Indexed: 03/14/2025] Open
Abstract
Mitochondria dysfunction has been closely linked to a wide spectrum of human cancers, whereas the molecular basis has yet to be fully understood. SLC25A35 belongs to the SLC25 family of mitochondrial carrier proteins. However, the role of SLC25A35 in mitochondrial metabolism reprogramming, development and progression in human cancers remains unclear. Here, we found that SLC25A35 markedly reprogramed mitochondrial metabolism, characterized by increased oxygen consumption rate and ATP production and decreased ROS level, via enhancing fatty acid oxidation (FAO). Meanwhile, SLC25A35 also enhanced mitochondrial biogenesis characterized by increased mitochondrial mass and DNA content. Mechanistic studies revealed that SLC25A35 facilitated FAO and mitochondrial biogenesis through upregulating peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) via increasing acetyl-CoA-mediated acetylation of PGC-1α. Clinically, SLC25A35 was highly expressed in HCC and correlated with adverse patients' survival. Functionally, SLC25A35 promoted the proliferation and metastasis of HCC cells both in vitro and in vivo, as well as the carcinogenesis in a DEN-induced HCC mice model. Moreover, we found that SLC25A35 upregulation is caused, at least in part, by decreased miR-663a in HCC cells. Together, our results suggest a crucial oncogenic role of SLC25A35 in HCC by reprogramming mitochondrial metabolism and suggest SLC25A35 as a potential therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Heng-Chao Yu
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, 15 Changle Western Road, Xi'an, Shaanxi, 710032, China.
| | - Lu Bai
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xian, China
| | - Liang Jin
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, 15 Changle Western Road, Xi'an, Shaanxi, 710032, China
| | - Yu-Jia Zhang
- Department of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zi-Han Xi
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, 15 Changle Western Road, Xi'an, Shaanxi, 710032, China
| | - De-Sheng Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, 15 Changle Western Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
34
|
Sun C, Deng H, Wang Q, Long Y, Li T, Song C, Han C. The roles of HOXB9 and MMP12 in proliferation, migration, and invasion of human laryngeal cancer cells LCC and TU212. Biochem Biophys Res Commun 2025; 752:151391. [PMID: 39938450 DOI: 10.1016/j.bbrc.2025.151391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/22/2025] [Indexed: 02/14/2025]
Abstract
OBJECTIVE HOXB9 and MMP12 are involved in the initiation and progression of various tumors. This study aimed to investigate the roles of HOXB9 and MMP12 in the proliferation, migration, and invasion of human laryngeal cancer cells (LCC and TU212). METHOD The experiment was divided into five groups: control, sh-HOXB9, sh-MMP12, sh-HOXB9 + MMP12-OE, and sh-MMP12 + HOXB9-OE. Cell proliferation was assessed using the CCK-8 assay, migration was evaluated using the scratch assay, and invasion was measured using the Transwell assay. The mRNA and protein expression levels of HOXB9, MMP12, MMP-9, fibronectin, β-catenin, N-cadherin, vimentin, and Snail were detected by qPCR and western blotting. RESULTS Compared with the control group, sh-HOXB9 and sh-MMP12 groups exhibited significantly reduced proliferation, migration, and invasion capacities, accompanied by decreased expression of HOXB9, MMP12, MMP-9, fibronectin, β-catenin, N-cadherin, and vimentin in both LCC and TU212 cells (p < 0.05). In the sh-HOXB9 + MMP12-OE group, no significant change in proliferation, migration, or invasion was observed in LCC compared with that in the sh-HOXB9 group (p > 0.05), but a notable increase was observed in TU212 cells (p < 0.05). Additionally, MMP12, MMP-9, fibronectin, β-catenin, N-cadherin, and vimentin expression levels significantly increased in both LCC and TU212 cells (p < 0.05). In the sh-MMP12 + HOXB9-OE group, there was a significant increase in the proliferation, migration, and invasion of both LCC and TU212 cells compared with the sh-MMP12 group (p < 0.05), along with elevated expression of HOXB9, MMP12, MMP-9, fibronectin, β-catenin, N-cadherin, and vimentin (p < 0.05). CONCLUSION HOXB9 and MMP12 may modulate the Wnt/β-catenin signaling pathway and regulate the proliferation, migration, invasion, and EMT of LCC and TU212 cells.
Collapse
Affiliation(s)
- Chuanhui Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China.
| | - Hua Deng
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Qian Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Yan Long
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550025, China
| | - Tao Li
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550025, China
| | - Can Song
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550025, China
| | - Changsong Han
- Department of Pathology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China.
| |
Collapse
|
35
|
Perelli L, Zhang L, Mangiameli S, Giannese F, Mahadevan KK, Peng F, Citron F, Khan H, Le C, Gurreri E, Carbone F, Russell AJC, Soeung M, Lam TNA, Lundgren S, Marisetty S, Zhu C, Catania D, Mohamed AMT, Feng N, Augustine JJ, Sgambato A, Tortora G, Draetta GF, Tonon G, Futreal A, Giuliani V, Carugo A, Viale A, Kim MP, Heffernan TP, Wang L, Kalluri R, Cittaro D, Chen F, Genovese G. Evolutionary fingerprints of epithelial-to-mesenchymal transition. Nature 2025:10.1038/s41586-025-08671-2. [PMID: 40044861 DOI: 10.1038/s41586-025-08671-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/17/2025] [Indexed: 04/13/2025]
Abstract
Mesenchymal plasticity has been extensively described in advanced epithelial cancers; however, its functional role in malignant progression is controversial1-5. The function of epithelial-to-mesenchymal transition (EMT) and cell plasticity in tumour heterogeneity and clonal evolution is poorly understood. Here we clarify the contribution of EMT to malignant progression in pancreatic cancer. We used somatic mosaic genome engineering technologies to trace and ablate malignant mesenchymal lineages along the EMT continuum. The experimental evidence clarifies the essential contribution of mesenchymal lineages to pancreatic cancer evolution. Spatial genomic analysis, single-cell transcriptomic and epigenomic profiling of EMT clarifies its contribution to the emergence of genomic instability, including events of chromothripsis. Genetic ablation of mesenchymal lineages robustly abolished these mutational processes and evolutionary patterns, as confirmed by cross-species analysis of pancreatic and other human solid tumours. Mechanistically, we identified that malignant cells with mesenchymal features display increased chromatin accessibility, particularly in the pericentromeric and centromeric regions, in turn resulting in delayed mitosis and catastrophic cell division. Thus, EMT favours the emergence of genomic-unstable, highly fit tumour cells, which strongly supports the concept of cell-state-restricted patterns of evolution, whereby cancer cell speciation is propagated to progeny within restricted functional compartments. Restraining the evolutionary routes through ablation of clones capable of mesenchymal plasticity, and extinction of the derived lineages, halts the malignant potential of one of the most aggressive forms of human cancer.
Collapse
Affiliation(s)
- Luigi Perelli
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Li Zhang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah Mangiameli
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Krishnan K Mahadevan
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fuduan Peng
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francesca Citron
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hania Khan
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney Le
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enrico Gurreri
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Andrew J C Russell
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Melinda Soeung
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Truong Nguyen Anh Lam
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sebastian Lundgren
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sujay Marisetty
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cihui Zhu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Desiree Catania
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alaa M T Mohamed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ningping Feng
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jithesh Jose Augustine
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Multiplex Spatial Imaging Facility, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giampaolo Tortora
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giovanni Tonon
- Center for Omics Sciences, IRCCS San Raffaele Institute, Milan, Italy
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Virginia Giuliani
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy P Heffernan
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center, UT Health Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Davide Cittaro
- Center for Omics Sciences, IRCCS San Raffaele Institute, Milan, Italy.
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
36
|
Yates J, Mathey-Andrews C, Park J, Garza A, Gagné A, Hoffman S, Bi K, Titchen B, Hennessey C, Remland J, Shannon E, Camp S, Balamurali S, Cavale SK, Li Z, Raghawan AK, Kraft A, Boland G, Aguirre AJ, Sethi NS, Boeva V, Van Allen E. Cell states and neighborhoods in distinct clinical stages of primary and metastatic esophageal adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.17.608386. [PMID: 39229240 PMCID: PMC11370330 DOI: 10.1101/2024.08.17.608386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Esophageal adenocarcinoma (EAC) is a highly lethal cancer of the upper gastrointestinal tract with rising incidence in western populations. To decipher EAC disease progression and therapeutic response, we performed multiomic analyses of a cohort of primary and metastatic EAC tumors, incorporating single-nuclei transcriptomic and chromatin accessibility sequencing, along with spatial profiling. We identified tumor microenvironmental features previously described to associate with therapy response. We identified five malignant cell programs, including undifferentiated, intermediate, differentiated, epithelial-to-mesenchymal transition, and cycling programs, which were associated with differential epigenetic plasticity and clinical outcomes, and for which we inferred candidate transcription factor regulons. Furthermore, we revealed diverse spatial localizations of malignant cells expressing their associated transcriptional programs and predicted their significant interactions with microenvironmental cell types. We validated our findings in three external single-cell RNA-seq and three bulk RNA-seq studies. Altogether, our findings advance the understanding of EAC heterogeneity, disease progression, and therapeutic response.
Collapse
Affiliation(s)
- Josephine Yates
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Camille Mathey-Andrews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Amanda Garza
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Andréanne Gagné
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Samantha Hoffman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Breanna Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
| | | | - Joshua Remland
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Erin Shannon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Sabrina Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Siddhi Balamurali
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Shweta Kiran Cavale
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Zhixin Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Akhouri Kishore Raghawan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Agnieszka Kraft
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Genevieve Boland
- Department of Surgery, Division of Gastrointestinal and Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Nilay S Sethi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Valentina Boeva
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
- Cochin Institute, Inserm U1016, CNRS UMR 8104, Paris Descartes University UMR-S1016, Paris 75014, France
| | - Eliezer Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
- Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Huang X, Arjsri P, Srisawad K, Umsumarng S, Yodkeeree S, Dejkriengkraikul P. Targeting MAPK Signaling: Loureirins A and B from Dracaena Loureiri Inhibit Epithelial-Mesenchymal Transition and Invasion in Non-Small Cell Lung Cancer Cell Lines. Life (Basel) 2025; 15:396. [PMID: 40141741 PMCID: PMC11943645 DOI: 10.3390/life15030396] [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: 01/27/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Metastasis remains the leading cause of death among patients with non-small cell lung cancer (NSCLC), emphasizing the urgent need for safer and more effective therapeutic options. Mitogen-activated protein kinase (MAPK) pathways play a crucial role in regulating EMT, migration, and invasion in NSCLC. Targeting these molecular mechanisms has become a key strategy in inhibiting NSCLC metastasis. Loureirin A and Loureirin B, flavonoids derived from the Thai traditional herb Dracaena loureiri, have shown potential pharmacological effects; however, their roles in NSCLC metastasis remain unexplored. This study aimed to elucidate the mechanisms by which Loureirin A and Loureirin B suppress EMT, migration, and invasion in NSCLC cells via the MAPK signaling pathway. The sulforhodamine B (SRB) assay showed that Loureirin A and Loureirin B, at concentrations ranging from 0 to 140 μM, were non-toxic to both A549 and H1299 cells. Additionally, Loureirins A and B exhibited no cytotoxic effects on primary human dermal fibroblast cells and did not induce hemolysis in red blood cells (RBCs). The wound-healing and trans-well assays were used to evaluate the anti-migratory and anti-invasion properties of Loureirin A and Loureirin B in NSCLC cell lines. Gelatin zymography was employed to investigate the activity of MMP-2 (gelatinase A) and MMP-9 (gelatinase B), while Western blot analysis was used to examine the expression of EMT markers and invasive proteins, and the phosphorylation of MAPK signaling molecules. Our results demonstrate that both Loureirin A and Loureirin B significantly suppressed the migration and invasion of A549 and H1299 cells. These compounds suppressed the activity of matrix metalloproteinases MMP-2 and MMP-9 and downregulated the expression of key invasive proteins including uPA, uPAR, and MT1-MMP. Additionally, they effectively suppressed the expression of EMT markers such as N-cadherin, Vimentin, and Fibronectin. Mechanistically, Loureirin A and Loureirin B inhibited the MAPK signaling pathway by downregulating the phosphorylation of ERK, JNK, and p38 proteins. In conclusion, these findings demonstrate that Loureirin A and Loureirin B exhibit potent anti-invasive properties and no cytotoxic effect on NSCLC cell lines, suggesting their potential as promising candidates for anti-cancer drug development. Furthermore, they may pave the way for the exploration of combination therapies with other anti-cancer drugs for clinical translation.
Collapse
Affiliation(s)
- Xiaomin Huang
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (X.H.); (P.A.); (K.S.); (S.Y.)
| | - Punnida Arjsri
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (X.H.); (P.A.); (K.S.); (S.Y.)
| | - Kamonwan Srisawad
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (X.H.); (P.A.); (K.S.); (S.Y.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sonthaya Umsumarng
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Supachai Yodkeeree
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (X.H.); (P.A.); (K.S.); (S.Y.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pornngarm Dejkriengkraikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (X.H.); (P.A.); (K.S.); (S.Y.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
38
|
Bangarh R, Saini RV, Saini AK, Singh T, Joshi H, Ramniwas S, Shahwan M, Tuli HS. Dynamics of epithelial-mesenchymal plasticity driving cancer drug resistance. CANCER PATHOGENESIS AND THERAPY 2025; 3:120-128. [PMID: 40182126 PMCID: PMC11963173 DOI: 10.1016/j.cpt.2024.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 04/05/2025]
Abstract
Epithelial-mesenchymal transition (EMT) promotes several cancers by increasing tumor cell motility, disrupting epithelial cell phenotypes, apical-basal polarity, and intracellular connections, and enhancing tumor resistance to immunotherapy and chemotherapy. Mesenchymal-epithelial transition (MET), the opposite of EMT, causes tumor metastasis. EMT drives primary tumor cells, whereas MET inhibits them. Importantly, the complex network of EMT includes cell-cell interactions in the tumor microenvironment. Transcription factors, post-translational regulation, cytokine-mediated signaling, and microRNAs control EMT. In this review, we discussed how molecular mechanisms, signaling networks, and epithelial/mesenchymal states affect cancer treatment resistance and the tumor microenvironment. Research on immunotherapy and chemotherapy problems associated with EMT suggests that targeting EMT might be a potential cancer treatment resistance strategy.
Collapse
Affiliation(s)
- Rashmi Bangarh
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India
| | - Reena V. Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India
| | - Adesh K. Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi 110007, India
| | - Hemant Joshi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Mohali 140413, India
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India
| |
Collapse
|
39
|
Giron-Michel J, Padelli M, Oberlin E, Guenou H, Duclos-Vallée JC. State-of-the-Art Liver Cancer Organoids: Modeling Cancer Stem Cell Heterogeneity for Personalized Treatment. BioDrugs 2025; 39:237-260. [PMID: 39826071 PMCID: PMC11906529 DOI: 10.1007/s40259-024-00702-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2024] [Indexed: 01/20/2025]
Abstract
Liver cancer poses a global health challenge with limited therapeutic options. Notably, the limited success of current therapies in patients with primary liver cancers (PLCs) may be attributed to the high heterogeneity of both hepatocellular carcinoma (HCCs) and intrahepatic cholangiocarcinoma (iCCAs). This heterogeneity evolves over time as tumor-initiating stem cells, or cancer stem cells (CSCs), undergo (epi)genetic alterations or encounter microenvironmental changes within the tumor microenvironment. These modifications enable CSCs to exhibit plasticity, differentiating into various resistant tumor cell types. Addressing this challenge requires urgent efforts to develop personalized treatments guided by biomarkers, with a specific focus on targeting CSCs. The lack of effective precision treatments for PLCs is partly due to the scarcity of ex vivo preclinical models that accurately capture the complexity of CSC-related tumors and can predict therapeutic responses. Fortunately, recent advancements in the establishment of patient-derived liver cancer cell lines and organoids have opened new avenues for precision medicine research. Notably, patient-derived organoid (PDO) cultures have demonstrated self-assembly and self-renewal capabilities, retaining essential characteristics of their respective in vivo tissues, including both inter- and intratumoral heterogeneities. The emergence of PDOs derived from PLCs serves as patient avatars, enabling preclinical investigations for patient stratification, screening of anticancer drugs, efficacy testing, and thereby advancing the field of precision medicine. This review offers a comprehensive summary of the advancements in constructing PLC-derived PDO models. Emphasis is placed on the role of CSCs, which not only contribute significantly to the establishment of PDO cultures but also faithfully capture tumor heterogeneity and the ensuing development of therapy resistance. The exploration of PDOs' benefits in personalized medicine research is undertaken, including a discussion of their limitations, particularly in terms of culture conditions, reproducibility, and scalability.
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France.
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France.
| | - Maël Padelli
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- Department of Biochemistry and Oncogenetics, Paul Brousse Hospital, AP-HP, Villejuif, France
| | - Estelle Oberlin
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Hind Guenou
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Jean-Charles Duclos-Vallée
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- INSERM UMR-S 1193, Paul Brousse Hospital, Villejuif, France
- Hepato-Biliary Department, Paul Brousse Hospital, APHP, Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hepatinov, Villejuif, France
| |
Collapse
|
40
|
Kim KP, Lemmon CA. Fibrotic extracellular matrix preferentially induces a partial Epithelial-Mesenchymal Transition phenotype in a 3-D agent based model of fibrosis. Math Biosci 2025; 381:109375. [PMID: 39832653 PMCID: PMC11925401 DOI: 10.1016/j.mbs.2025.109375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
One of the main drivers of fibrotic diseases is epithelial-mesenchymal transition (EMT): a transdifferentiation process in which cells undergo a phenotypic change from an epithelial state to a pro-migratory state. The cytokine transforming growth factor-β1 (TGF-β1) has been previously shown to regulate EMT. TGF-β1 binds to fibronectin (FN) fibrils, which are the primary extracellular matrix (ECM) component in renal fibrosis. We have previously demonstrated experimentally that inhibition of FN fibrillogenesis and/or TGF-β1 tethering to FN inhibits EMT. However, these studies have only been conducted on 2-D cell monolayers, and the role of TGF-β1-FN tethering in 3-D cellular environments is not clear. As such, we sought to develop a 3-D computational model of epithelial spheroids that captured both EMT signaling dynamics and TGF-β1-FN tethering dynamics. We have incorporated the bi-stable EMT switch model developed by Tian et al. (2013) into a 3-D multicellular model to capture both temporal and spatial TGF-β1 signaling dynamics. We showed that the addition of increasing concentrations of exogeneous TGF-β1 led to faster EMT progression, indicated by increased expression of mesenchymal markers, decreased cell proliferation and increased migration. We then incorporated TGF-β1-FN fibril tethering by locally reducing the TGF-β1 diffusion coefficient as a function of EMT to simulate the reduced movement of TGF-β1 when tethered to FN fibrils during fibrosis. We showed that incorporation of TGF-β1 tethering to FN fibrils promoted a partial EMT state, independent of exogenous TGF-β1 concentration, indicating a mechanism by which fibrotic ECM can promote a partial EMT state.
Collapse
Affiliation(s)
- Kristin P Kim
- Department of Biomedical Engineering, Virginia Commonwealth University, 410 West Main St., Richmond, VA, 23284, USA.
| | - Christopher A Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, 410 West Main St., Richmond, VA, 23284, USA.
| |
Collapse
|
41
|
Xu X, Xu Z, Cai Y, Chen X, Huang C. CKIP-1 inhibits M2 macrophage polarization to suppress the progression of gastric cancer by inactivating JAK/STAT3 signaling. Cell Biochem Biophys 2025; 83:1289-1298. [PMID: 39470944 DOI: 10.1007/s12013-024-01562-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 11/01/2024]
Abstract
Gastric cancer (GC) is a frequently occurring malignancy with poor prognosis. Casein kinase 2 interacting protein-1 (CKIP-1) is a PH domain-containing protein implicated in regulating tumorigenesis and macrophage homeostasis. This study aimed to elucidate the role and potential mechanism of CKIP-1 in the progression of GC. CKIP-1 expression in GC tumor and para-carcinoma tissues was detected using RT-qPCR. Then, human monocyte cell line THP-1 was treated with PMA, interleukin (IL)-4 and IL-13 to induce M2-polarized macrophages. CD206, arginase-1 (Arg-1) and transforming growth factorβ1 (TGFβ1) expression in M2-polarized macrophages with or without CKIP-1 overexpression was evaluated. Moreover, GC cell lines (MKN45 and HGC27 cells) were co-cultured with CKIP-1-overexpressed M2-polarized macrophages, and the viability, migration and invasion of GC cells were measured. Additionally, immunoblotting assessed the expression of JAK/STAT3 signaling-related proteins and STAT3 agonist Colivelin was used to treat GC cells to perform the rescue experiments to analyze the changes of malignant phenotypes of GC cells. Results showed that CKIP-1 was downregulated in GC tissues and M2-polarized macrophages. CKIP-1 overexpression inhibited M2 macrophage polarization and decreased TGFβ1 secretion. Besides, elevated CKIP-1 expression in M2-polarized macrophages inhibited the viability, migration and invasion of GC cells. Furthermore, CKIP-1 overexpression inactivated JAK2/STAT3 signaling in GC cells by inhibiting TGFβ1 level. Specifically, Colivelin treatment abrogated the influences of CKIP-1 upregulation on the malignant phenotypes of GC cells. Collectively, CKIP-1 inhibits M2 macrophage polarization to suppress the progression of GC by inactivating JAK/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Xuefeng Xu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, 350122, China
- Department of Gastrointestinal Surgery, The First Hospital of Putian City, Putian, Fujian, 351100, China
- Cardia Cancer Institute, Putian University, Putian, Fujian, 351100, China
| | - Zihong Xu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Yaowu Cai
- Department of Gastrointestinal Surgery, The First Hospital of Putian City, Putian, Fujian, 351100, China
- Cardia Cancer Institute, Putian University, Putian, Fujian, 351100, China
| | - Xintong Chen
- Department of Gastrointestinal Surgery, The First Hospital of Putian City, Putian, Fujian, 351100, China
- Cardia Cancer Institute, Putian University, Putian, Fujian, 351100, China
| | - Chaoqing Huang
- Department of Gastrointestinal Surgery, The First Hospital of Putian City, Putian, Fujian, 351100, China.
- Cardia Cancer Institute, Putian University, Putian, Fujian, 351100, China.
| |
Collapse
|
42
|
Severino MB, Morelli AP, Pavan ICB, Mancini MCS, Góis MM, Borges RJ, Braga RR, da Silva LGS, Quintero-Ruiz N, Costa MM, Oliveira WDL, Bezerra RMN, Ropelle ER, Simabuco FM. A CRISPR-edited isoform of the AMPK kinase LKB1 improves the response to cisplatin in A549 lung cancer cells. J Biol Chem 2025; 301:108308. [PMID: 39955067 PMCID: PMC11952844 DOI: 10.1016/j.jbc.2025.108308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 01/23/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025] Open
Abstract
Lung cancer presents the highest mortality rate in the world when compared to other cancer types and often presents chemotherapy resistance to cisplatin. The A549 nonsmall cell lung cancer line is widely used as a model for lung adenocarcinoma studies since it presents a high proliferative rate and a nonsense mutation in the STK11 gene. The LKB1 protein, encoded by the STK11 gene, is one of the major regulators of cellular metabolism through AMPK activation under nutrient deprivation. Mutation in the STK11 gene in A549 cells potentiates cancer hallmarks, such as deregulation of cellular metabolism, aside from the Warburg effect, mTOR activation, autophagy inhibition, and NRF2 and redox activation. In this study, we investigated the integration of these pathways associated with the metabolism regulation by LKB1/AMPK to improve cisplatin response in the A549 cell line. We first used the CRISPR/Cas9 system to generate cell lines with a CRISPR-edited LKB1 isoform (called Super LKB1), achieved through the introduction of a +1 adenine insertion in the first exon of the STK11 gene after NHEJ-mediated repair. This insertion led to the expression of a higher molecular weight protein containing an alternative exon described in the Peutz-Jeghers Syndrome. Through metabolic regulation by Super LKB1 expression and AMPK activation, we found an increase in autophagy flux (LC3 GFP/RFP p < 0.05), as well as a reduction in the phosphorylation of mTORC1 downstream targets (S6K2 phospho-serine 423; p < 0.05; and S6 ribosomal protein phospho-serine 240/244; p < 0.03). The NRF2 protein exhibited increased levels and more nuclear localization in A549 WT cells compared to the edited cells (p < 0.01). We also observed lower levels of H2O2 in the WT A549 cells, as a possible result of NRF2 activation, and a higher requirement of cisplatin to achieve the IC50 (WT: 10 μM; c2SL+: 5.5 μM; c3SL+: 6 μM). The data presented here suggests that the regulation of molecular pathways by the novel Super LKB1 in A549 cells related to metabolism, mTORC1, and autophagy promotes a better response of lung cancer cells to cisplatin. This NHEJ-CRISPR-based approach may be potentially used for lung cancer gene therapy.
Collapse
Affiliation(s)
- Matheus Brandemarte Severino
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Ana Paula Morelli
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil; Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Camargo Silva Mancini
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Mariana Marcela Góis
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Rafael Junqueira Borges
- Department of Physics and Biophysics, Biosciences Institute, State University of São Paulo, Botucatu, Brazil; Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, Brazil; Center for Medicinal Chemistry (CQMED), University of Campinas, Campinas, Brazil
| | - Renata Rosseto Braga
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Nathalia Quintero-Ruiz
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Maíra Maftoum Costa
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Wesley de Lima Oliveira
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Rosângela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil; Applied Molecular Signaling Laboratory (LabSIMA), Department of Biochemistry, Federal University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
43
|
Morin C, Paraqindes H, Van Long FN, Isaac C, Thomas E, Pedri D, Pulido-Vicuna CA, Morel AP, Marchand V, Motorin Y, Carrere M, Auclair J, Attignon V, Pommier RM, Ruiz E, Bourdelais F, Catez F, Durand S, Ferrari A, Viari A, Marine JC, Puisieux A, Diaz JJ, Moyret-Lalle C, Marcel V. Specific modulation of 28S_Um2402 rRNA 2'- O-ribose methylation as a novel epitranscriptomic marker of ZEB1-induced epithelial-mesenchymal transition in different mammary cell contexts. NAR Cancer 2025; 7:zcaf001. [PMID: 39877292 PMCID: PMC11773364 DOI: 10.1093/narcan/zcaf001] [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: 05/07/2024] [Accepted: 01/27/2025] [Indexed: 01/31/2025] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a dynamic transdifferentiation of epithelial cells into mesenchymal cells. EMT programs exhibit great diversity, based primarily on the distinct impact of molecular activities of the EMT transcription factors. Using a panel of cancer cell lines and a series of 71 triple-negative primary breast tumors, we report that the EMT transcription factor ZEB1 modulates site-specific chemical modifications of ribosomal RNA (rRNA). Overexpression of ZEB1 and ZEB2, but not TWIST1, decreased the level of 2'-O-ribose methylation (2'Ome) of 28S rRNA at position Um2402. ZEB1 overexpression specifically reduced the expression of the corresponding C/D box small nucleolar RNAs (snoRNAs) SNORD143/144, which guide the rRNA 2'Ome complex at the 28S_Um2402 site. During ZEB1-induced EMT induction/reversion, the levels of both 2'Ome at 28S_Um2402 and SNORD143/144 were dynamically comodulated. Taken together, these data demonstrate that 2'Ome rRNA epitranscriptomics is a novel marker of ZEB1-induced EMT.
Collapse
Affiliation(s)
- Chloé Morin
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Hermes Paraqindes
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
- Bioinformatics Platform Gilles Thomas, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Synergie Lyon Cancer Fondation, 69008 Lyon, France
| | - Flora Nguyen Van Long
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Caroline Isaac
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Emilie Thomas
- Bioinformatics Platform Gilles Thomas, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Synergie Lyon Cancer Fondation, 69008 Lyon, France
| | - Dennis Pedri
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, 3001 Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU 3000 Leuven, Belgium
| | - Carlos Ariel Pulido-Vicuna
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, 3001 Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU 3000 Leuven, Belgium
| | - Anne-Pierre Morel
- EMT and Cancer Cell Plasticity Team, Centre Léon Bérard, 69008 Lyon, France
| | - Virginie Marchand
- UMS2008 IBSLor CNRS-INSERM-Lorraine University, Biopôle, 9 avenue de la forêt de haye, 54505 Vandoeuvre-les-Nancy, France
| | - Yuri Motorin
- UMS2008 IBSLor CNRS-INSERM-Lorraine University, Biopôle, 9 avenue de la forêt de haye, 54505 Vandoeuvre-les-Nancy, France
- IMoPA, UMR 7365 CNRS-UL, Biopole UL, 54500 Vandoeuvre-les-Nancy, France
| | - Marjorie Carrere
- Cancer Genomic Platform, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Jessie Auclair
- Cancer Genomic Platform, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Valéry Attignon
- Cancer Genomic Platform, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Roxane M Pommier
- Bioinformatics Platform Gilles Thomas, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Synergie Lyon Cancer Fondation, 69008 Lyon, France
| | - Emmanuelle Ruiz
- Department of Pathobiological Sciences, School of Veterinary and Medicine, Louisiana State University, 70802 Baton Rouge, LA, United States
| | - Fleur Bourdelais
- RibosOMICS Platform, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Frédéric Catez
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Sébastien Durand
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
- RibosOMICS Platform, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Anthony Ferrari
- Bioinformatics Platform Gilles Thomas, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Synergie Lyon Cancer Fondation, 69008 Lyon, France
| | - Alain Viari
- Bioinformatics Platform Gilles Thomas, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Synergie Lyon Cancer Fondation, 69008 Lyon, France
- INRIA Grenoble Rhône-Alpes, Montbonnot-Saint-Martin 38334, France
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, 3001 Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU 3000 Leuven, Belgium
| | - Alain Puisieux
- Institut Curie, PSL Research University, 75005 Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR3666, INSERM U1143, Paris, France
| | - Jean-Jacques Diaz
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Caroline Moyret-Lalle
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Virginie Marcel
- Ribosome, Translation and Cancer Team, LaEx DEVweCAN, Institut Convergence Plascan, LYriCAN+, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
- RibosOMICS Platform, Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| |
Collapse
|
44
|
Tu J, Jiang F, Liu J, Ong HH, Huang Z, Wang K, Luo Q, Shi L, Ye J, Wang D. Increased expression of epithelial-mesenchymal transition markers associated with recurrence of sinonasal inverted papilloma. Int Forum Allergy Rhinol 2025; 15:354-358. [PMID: 39652071 DOI: 10.1002/alr.23501] [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: 06/21/2024] [Revised: 10/17/2024] [Accepted: 11/26/2024] [Indexed: 03/04/2025]
Abstract
KEY POINTS Sinonasal inverted papilloma (SNIP) is a benign epithelial proliferative disease with a high recurrence rate. The role of epithelial-mesenchymal transition (EMT) in the pathogenesis of SNIP remains unclear. EMT marker expression is elevated in SNIP tissues and is associated with its recurrence.
Collapse
Affiliation(s)
- Junhao Tu
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Fan Jiang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jing Liu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Hsiao Hui Ong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Zhiqun Huang
- Allergy Department, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Keshuang Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
- Department of Otorhinolaryngology, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, China
| | - Qing Luo
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Li Shi
- Department of Otolaryngology, The Second Hospital of Shandong University, Jinan, China
| | - Jing Ye
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Allergy Department, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Otorhinolaryngology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Deyun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| |
Collapse
|
45
|
Zhang R, Ren Y, Ju Y, Zhang Y, Zhang Y, Wang Y. FAM20C: A key protein kinase in multiple diseases. Genes Dis 2025; 12:101179. [PMID: 39790934 PMCID: PMC11714710 DOI: 10.1016/j.gendis.2023.101179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/23/2023] [Accepted: 10/31/2023] [Indexed: 01/12/2025] Open
Abstract
Family with sequence similarity 20 C (FAM20C) is a Golgi protein kinase that phosphorylates the serine residue in the S-x-E/pS motif of target proteins. FAM20C phosphorylates most secreted proteins, which play important roles in multiple biological processes, including cancer progression, biomineralization, and lipid homeostasis. Numerous studies have documented the potential contribution of FAM20C to the growth, invasion, and metastasis of glioma, breast cancer, and other cancers, as well as to the mineralization process of teeth and bone. In addition, FAM20C has been found to be associated with the occurrence and development of certain cardiovascular diseases and endocrine metabolism disorders. It raises hopes that understanding the disease-specific mechanisms of FAM20C may hold the key to developing new strategies for these diseases. This review comprehensively covers the existing literature to provide a summary of the structure and biological functions of FAM20C, with a particular focus on its roles in the disease context.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanming Ren
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Ju
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuekang Zhang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
46
|
Tang L, Peng S, Zhuang X, He Y, Song Y, Nie H, Zheng C, Pan Z, Lam AK, He M, Shi X, Li B, Xu WW. Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention. MEDCOMM – ONCOLOGY 2025; 4. [DOI: 10.1002/mog2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/04/2025]
Abstract
ABSTRACTMetastasis remains a leading cause of cancer‐related deaths, defined by a complex, multi‐step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post‐translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.
Collapse
Affiliation(s)
- Lin Tang
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Shao‐Cong Peng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Xiao‐Wan Zhuang
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Yan He
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Yu‐Xiang Song
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Hao Nie
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Can‐Can Zheng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Zhen‐Yu Pan
- Department of Radiation Oncology, The Affiliated Huizhou Hospital Guangzhou Medical University Huizhou China
| | - Alfred King‐Yin Lam
- Cancer Molecular Pathology and Griffith Medical School Griffith University Gold Coast Queensland Australia
| | - Ming‐Liang He
- Department of Biomedical Sciences City University of Hong Kong Hong Kong China
| | - Xing‐Yuan Shi
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Bin Li
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Wen Wen Xu
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| |
Collapse
|
47
|
Chen Z, Gong Y, Chen F, Lee HJ, Qian J, Zhao J, Zhang W, Li Y, Zhou Y, Xu Q, Xia Y, Zhou L, Cheng J. Orchestrated desaturation reprogramming from stearoyl-CoA desaturase to fatty acid desaturase 2 in cancer epithelial-mesenchymal transition and metastasis. Cancer Commun (Lond) 2025; 45:245-280. [PMID: 39722173 PMCID: PMC11947613 DOI: 10.1002/cac2.12644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Adaptative desaturation in fatty acid (FA) is an emerging hallmark of cancer metabolic plasticity. Desaturases such as stearoyl-CoA desaturase (SCD) and fatty acid desaturase 2 (FADS2) have been implicated in multiple cancers, and their dominant and compensatory effects have recently been highlighted. However, how tumors initiate and sustain their self-sufficient FA desaturation to maintain phenotypic transition remains elusive. This study aimed to explore the molecular orchestration of SCD and FADS2 and their specific reprogramming mechanisms in response to cancer progression. METHODS The potential interactions between SCD and FADS2 were explored by bioinformatics analyses across multiple cancer cohorts, which guided subsequent functional and mechanistic investigations. The expression levels of desaturases were investigated with online datasets and validated in both cancer tissues and cell lines. Specific desaturation activities were characterized through various isomer-resolved lipidomics methods and sensitivity assays using desaturase inhibitors. In-situ lipid profiling was conducted using multiplex stimulated Raman scattering imaging. Functional assays were performed both in vitro and in vivo, with RNA-sequencing employed for the mechanism verification. RESULTS After integration of the RNA-protein-metabolite levels, the data revealed that a reprogramming from SCD-dependent to FADS2-dependent desaturation was linked to cancer epithelial-mesenchymal transition (EMT) and progression in both patients and cell lines. FADS2 overexpression and SCD suppression concurrently maintained EMT plasticity. A FADS2/β-catenin self-reinforcing feedback loop facilitated the degree of lipid unsaturation, membrane fluidity, metastatic potential and EMT signaling. Moreover, SCD inhibition triggered a lethal apoptosis but boosted survival plasticity by inducing EMT and enhancing FA uptake via adenosine monophosphate-activated protein kinase activation. Notably, this desaturation reprogramming increased transforming growth factor-β2, effectively sustaining aggressive phenotypes and metabolic plasticity during EMT. CONCLUSIONS These findings revealed a metabolic reprogramming from SCD-dependent to FADS2-dependent desaturation during cancer EMT and progression, which concurrently supports EMT plasticity. Targeting desaturation reprogramming represents a potential vulnerability for cancer metabolic therapy.
Collapse
Affiliation(s)
- Zhicong Chen
- Department of Obstetrics and GynecologyCenter for Reproductive MedicineGuangdong Provincial Key Laboratory of Major Obstetric DiseasesGuangdong Provincial Clinical Research Center for Obstetrics and GynecologyGuangdong‐Hong Kong‐Macao Greater Bay Area Higher Education Joint Laboratory of Maternal‐Fetal MedicineThe Third Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongP. R. China
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Yanqing Gong
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Fukai Chen
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
| | - Hyeon Jeong Lee
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
- College of Biomedical Engineering & Instrument ScienceKey Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jinqin Qian
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Jing Zhao
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua UniversityBeijingP. R. China
| | - Wenpeng Zhang
- State Key Laboratory of Precision Measurement Technology and InstrumentsDepartment of Precision InstrumentTsinghua UniversityBeijingP. R. China
| | - Yamin Li
- Department of Biomedical EngineeringTufts UniversityMedfordMassachusettsUSA
| | - Yihui Zhou
- College of Biomedical Engineering & Instrument ScienceKey Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qiaobing Xu
- Department of Biomedical EngineeringTufts UniversityMedfordMassachusettsUSA
| | - Yu Xia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua UniversityBeijingP. R. China
| | - Liqun Zhou
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Ji‐Xin Cheng
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
48
|
Wang L, Li N, Chen Y, Qiao Y, Song Y, Zhang X, Zhao H, Ran W, Li G, Xing X. GPSM1 interacts and cooperates with MMP19 to promote proliferation and EMT in colorectal cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119903. [PMID: 39855604 DOI: 10.1016/j.bbamcr.2025.119903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Among patients with colorectal cancer (CRC), metastasis accounts for the majority of deaths, and epithelial-mesenchymal transition (EMT) is important in the metastatic process. However, the mechanism underlying the correlation between the two in CRC is unknown. Here, we verified that a receptor-independent protein, G-protein signaling modulator 1 (GPSM1), was increased in CRC and had a significant positive correlation with matrix metalloproteinase 19 (MMP19). GPSM1 and MMP19 knockdown or overexpression decreased and increased proliferation, migration and invasion of CRC cells, respectively. In addition, overexpression or knockdown of GPSM1 and MMP19 upregulated and inhibited EMT, respectively. Interfering with MMP19 reversed EMT activation via GPSM1 overexpression. Apoptosis was induced by GPSM1 and MMP19 knockdown and activated the caspase3/Bcl-2/Bax signaling pathway. In conclusion, these results support the role of GPSM1 and MMP19 in CRC progression.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Na Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Chen
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yehua Qiao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaolin Song
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangyan Zhang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Han Zhao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenwen Ran
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guangqi Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoming Xing
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
49
|
Bayasgalan T, Kanda M, Sato Y, Zhu H, Hamrah MH, Martinez FEG, Shinozuka T, Ito Y, Sasahara M, Shimizu D, Umeda S, Inokawa Y, Hattori N, Hayashi M, Tanaka C, Kodera Y. SPOCD1 Enhances Cancer Cell Activities and Serves as a Prognosticator in Esophageal Squamous Cell Carcinoma. Cancer Genomics Proteomics 2025; 22:306-325. [PMID: 39993802 PMCID: PMC11880929 DOI: 10.21873/cgp.20503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/02/2025] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND/AIM Comprehensive transcriptome analysis has revealed SPOC Domain Containing 1 (SPOCD1) as a potential biomarker for esophageal squamous cell carcinoma (ESCC). However, the expression and oncological roles of SPOCD1 in ESCC remains underexplored. We aimed to evaluate the role of SPOCD1 in oncogenesis and prognosis of ESCC in vitro and in vivoMaterials and Methods: The Cancer Cell Line Encyclopedia (CCLE) database was utilized to evaluate correlations between SPOCD1 expression and oncogenes in ESCC. mRNA and protein levels were measured by qRT-PCR and Simple Western assays, respectively. siRNA-mediated knockdown and overexpression experiments assessed the effects of SPOCD1 expression on proliferation, migration, and invasion of ESCC cell lines. In vivo, siRNA knockdown effects on tumor growth were tested in mouse xenograft models. SPOCD1 mRNA levels in 164 resected tissues were correlated with clinicopathological parameters and survival, while a cohort of 177 patients was analyzed for protein expression and survival. RESULTS SPOCD1 mRNA expression varied widely among ESCC cell lines and correlated with epithelial-mesenchymal transition-related genes. Knockdown significantly suppressed proliferation, migration, and invasion (p<0.001), while overexpression increased proliferation (p<0.001). In vivo, siRNA knockdown reduced tumor growth compared to both si-control (p=0.005) and untransfected groups (p<0.001). High SPOCD1 mRNA expression was linked to poor disease-specific survival (p=0.009, HR=1.965, 95% CI=1.187-3.252) and disease-free survival (p=0.047, HR=1.602, 95% CI=1.007-2.549). Similarly, elevated protein levels were associated with unfavorable disease-specific (p=0.013, HR=1.860, 95% CI=1.137-3.041) and disease-free survival (p=0.032, HR=1.618, 95% CI=1.042-2.513). CONCLUSION SPOCD1 expression correlates with the aggressiveness of ESCC cells, and its expression levels in tumor tissues may serve as a prognostic factor for ESCC patients.
Collapse
Affiliation(s)
- Tuvshin Bayasgalan
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan;
| | - Yusuke Sato
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Haote Zhu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mohammad Hussain Hamrah
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Takahiro Shinozuka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Ito
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Sasahara
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshikuni Inokawa
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
50
|
Zhang Y, Zhang Y, Gong R, Liu X, Zhang Y, Sun L, Ma Q, Wang J, Lei K, Ren L, Zhao C, Zheng X, Xu J, Ren H. Label-Free Prediction of Tumor Metastatic Potential via Ramanome. SMALL METHODS 2025; 9:e2400861. [PMID: 39558758 DOI: 10.1002/smtd.202400861] [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: 06/08/2024] [Revised: 09/02/2024] [Indexed: 11/20/2024]
Abstract
Assessing metastatic potential is crucial for cancer treatment strategies. However, current methods are time-consuming, labor-intensive, and have limited sample accessibility. Therefore, this study aims to investigate the urgent need for rapid and accurate approaches by proposing a Ramanome-based metastasis index (RMI) using machine learning of single-cell Raman spectra to rapidly and accurately assess tumor cell metastatic potential. Validation with various cultured tumor cells and a mouse orthotopic model of pancreatic ductal adenocarcinoma show a Kendall rank correlation coefficient of 1 compared to Transwell experiments and histopathological assessments. Significantly, lipid-related Raman peaks are most influential in determining RMI. The lipidomic analysis confirmed strong correlations between metastatic potential and phosphatidylcholine, phosphatidylethanolamine, cholesteryl ester, ceramide, and bis(monoacylglycero)phosphate, crucial in cell membrane composition or signal transduction. Therefore, RMI is a valuable tool for predicting tumor metastatic potential and providing insights into metastasis mechanisms.
Collapse
Affiliation(s)
- Yuxing Zhang
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yanmei Zhang
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Ruining Gong
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Xiaolan Liu
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yu Zhang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Luyang Sun
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Qingyue Ma
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Jia Wang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Ke Lei
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Linlin Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Chenyang Zhao
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Xiaoshan Zheng
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Jian Xu
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - He Ren
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| |
Collapse
|