1
|
Liu H, Zhang J, Cai J, Wu Q, Li G, Li W, Liu T, Yang P, Wang Z, Yi X. Extracellular vesicles derived from EZH2-high ovarian cancer cells facilitate omental metastasis by inducing Periostin + fibroblasts. Cell Signal 2025; 132:111773. [PMID: 40180166 DOI: 10.1016/j.cellsig.2025.111773] [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: 12/15/2024] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
The frequent omental metastasis of ovarian cancer (OvCa) at initial diagnosis is due to the omental premetastatic microenvironment, which is rich in activated fibroblasts. However, the molecular events driving the phenotypic transformation of omental fibroblasts that favor metastasis remain largely unexplored. Previously, we found that tumoral enhancer of zest homolog 2 (EZH2), a key epigenetic regulator catalyzing trimethylation at H3K27, played a crucial role in OvCa metastasis. In this study, we revealed that extracellular vesicles (EVs) derived from EZH2-high OvCa cells induce the expression of Periostin (POSTN), but not α-SMA, in omental fibroblasts, facilitating tumor metastasis. Nude mice with intraperitoneal injection of EVs before tumor cell inoculation showed that EVs derived from EZH2-high ovarian cancer cells promote omental metastasis. Human primary omental fibroblasts cocultured with EVs, especially those derived from EZH2-high OvCa cells, exhibited boosted migration, invasion capacities and conditioned medium from EV-activated fibroblasts promotes cancer cell migration, invasion and proliferation. These results may provide novel insight into EZH2-targeted therapy for ovarian carcinoma with intraperitoneal dissemination.
Collapse
Affiliation(s)
- Hongmei Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Gynecology, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China
| | - Jingni Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiulei Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guoqing Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenhan Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tong Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ping Yang
- Department of Gynecology, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China.
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaoqing Yi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
2
|
Lee JJ, Ng KY, Bakhtiar A. Extracellular matrix: unlocking new avenues in cancer treatment. Biomark Res 2025; 13:78. [PMID: 40426238 PMCID: PMC12117852 DOI: 10.1186/s40364-025-00757-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/05/2025] [Indexed: 05/29/2025] Open
Abstract
The extracellular matrix (ECM) plays a critical role in cancer progression by influencing tumor growth, invasion, and metastasis. This review explores the emerging therapeutic strategies that target the ECM as a novel approach in cancer treatment. By disrupting the structural and biochemical interactions within the tumor microenvironment, ECM-targeted therapies aim to inhibit cancer progression and overcome therapeutic resistance. We examine the current state of ECM research, focusing on key components such as collagen, laminin, fibronectin, periostin, and hyaluronic acid, and their roles in tumor biology. Additionally, we discuss the challenges associated with ECM-targeted therapies, including drug delivery, specificity, and potential side effects, while highlighting recent advancements and future directions. This review underscores the potential of ECM-focused strategies to enhance the efficacy of existing treatments and contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Jia Jing Lee
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Athirah Bakhtiar
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia.
| |
Collapse
|
3
|
Zhang J, Jin X, Hou Y, Gu B, Li H, Yi L, Wu W, Hu S. Comprehensive analysis of the critical role of the epithelial mesenchymal transition subtype - TAGLN-positive fibroblasts in colorectal cancer progression and immunosuppression. Cell Biosci 2025; 15:66. [PMID: 40413514 PMCID: PMC12102804 DOI: 10.1186/s13578-025-01405-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays a pivotal role in tumor metastasis and immune suppression in colorectal cancer (CRC). However, the specific mechanisms of EMT and its relationship with the clinical prognosis and immunotherapy response in CRC patients remain unclear. In this study, we identified TAGLN-positive fibroblasts (TAGLN⁺Fib) as a cancer-associated fibroblast (CAF) subtype within the tumor microenvironment (TME) that promotes tumor metastasis and immune evasion. High EMT scores, strongly associated with TAGLN expression, were correlated with advanced tumor stages, poor prognosis, and resistance to immunotherapy. Functional experiments demonstrated that TAGLN knockdown significantly reduced CRC cell proliferation, migration, and EMT phenotypes in vitro and suppressed tumor growth in vivo. Furthermore, TAGLN⁺Fib closely interacted with MMP7-positive tumor epithelial cells and SPP1-positive macrophages, forming a pro-metastatic and immunosuppressive network. An EMT-TME risk model constructed using TAGLN⁺Fib exhibited robust predictive power for CRC prognosis and immunotherapy response. This study reveals the association of EMT scores with CRC prognosis and immunotherapy response, highlights TAGLN⁺Fib's critical role in tumor progression, and develops an EMT-TME risk model, offering insights for personalized CRC treatment and precision medicine.
Collapse
Affiliation(s)
- Junli Zhang
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Anhui, China
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Xinxin Jin
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Yachao Hou
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Biao Gu
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China
| | - Hongwei Li
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China
| | - Li Yi
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China
| | - Wenjuan Wu
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, 233030, China.
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, 233030, China.
| | - Shangshang Hu
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China.
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, 233030, China.
| |
Collapse
|
4
|
Li S, Zhou X, Feng H, Huang K, Chen M, Lin M, Lin H, Deng Z, Chen Y, Liao W, Zhang Z, Chen J, Guan B, Su T, Feng Z, Shu G, Yu A, Pan Y, Fu L. Deciphering the Immunomodulatory Function of GSN + Inflammatory Cancer-Associated Fibroblasts in Renal Cell Carcinoma Immunotherapy: Insights From Pan-Cancer Single-Cell Landscape and Spatial Transcriptomics Analysis. Cell Prolif 2025:e70062. [PMID: 40375605 DOI: 10.1111/cpr.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/13/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
The heterogeneity of cancer-associated fibroblasts (CAFs) could affect the response to immune checkpoint inhibitor (ICI) therapy. However, limited studies have investigated the role of inflammatory CAFs (iCAFs) in ICI therapy using pan-cancer single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics sequencing (ST-seq) analysis. We performed pan-cancer scRNA-seq and ST-seq analyses to identify the subtype of GSN+ iCAFs, exploring its spatial distribution characteristics in the context of ICI therapy. The pan-cancer scRNA-seq and bulk RNA-seq data are incorporated to develop the Caf.Sig model, which predicts ICI response based on CAF gene signatures and machine learning approaches. Comprehensive scRNA-seq analysis, along with in vivo and in vitro experiments, investigates the mechanisms by which GSN+ iCAFs influence ICI efficacy. The Caf.Sig model demonstrates well performances in predicting ICI therapy response in pan-cancer patients. A higher proportion of GSN+ iCAFs is observed in ICI non-responders compared to responders in the pan-cancer landscape and clear cell renal cell carcinoma (ccRCC). Using real-world immunotherapy data, the Caf.Sig model accurately predicts ICI response in pan-cancer, potentially linked to interactions between GSN+ iCAFs and CD8+ Tex cells. ST-seq analysis confirms that interactions and cellular distances between GSN+ iCAFs and CD8+ exhausted T (Tex) cells impact ICI efficacy. In a co-culture system of primary CAFs, primary tumour cells and CD8+ T cells, downregulation of GSN on CAFs drives CD8+ T cells towards a dysfunctional state in ccRCC. In a subcutaneously tumour-grafted mouse model, combining GSN overexpression with ICI treatment achieves optimal efficacy in ccRCC. Our study provides the Caf.Sig model as an outperforming approach for patient selection of ICI therapy, and advances our understanding of CAF biology and suggests potential therapeutic strategies for upregulating GSN in CAFs in cancer immunotherapy.
Collapse
Affiliation(s)
- Shan Li
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xinwei Zhou
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Haoqian Feng
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Kangbo Huang
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Minyu Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mingjie Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hansen Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zebing Deng
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
| | - Yuhang Chen
- Department of Geniturinary Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wuyuan Liao
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhengkun Zhang
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jinwei Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Bohong Guan
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tian Su
- Department of Pediatric Intensive Care Unit (PICU), Guangdong Provincial People's Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Zihao Feng
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Guannan Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou Institute of Pediatrics, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Anze Yu
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yihui Pan
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liangmin Fu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, Hunan, China
| |
Collapse
|
5
|
Xu W, Weng J, Zhao Y, Xie P, Xu M, Liu S, Yu Q, Yu M, Liang B, Chen J, Sun HC, Li H, Ye Q, Shen Y. FMO2 + cancer-associated fibroblasts sensitize anti-PD-1 therapy in patients with hepatocellular carcinoma. J Immunother Cancer 2025; 13:e011648. [PMID: 40316306 PMCID: PMC12049961 DOI: 10.1136/jitc-2025-011648] [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: 01/22/2025] [Accepted: 04/14/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND The efficacy of immune checkpoint inhibitors (ICIs) for hepatocellular carcinoma (HCC) is limited by heterogeneity in individual responses to therapy. The heterogeneous phenotypes and crucial roles of cancer-associated fibroblasts (CAFs) in immunotherapy resistance remain largely unclear. METHODS A specific CAF subset was identified by integrating comprehensive single-cell RNA sequencing, spatial transcriptomics and transcriptome profiling of patients with HCC with different responses to antiprogrammed cell death protein 1 (anti-PD-1) therapy. Mouse orthotopic HCC models and a coculture system were constructed, and cytometry by time-of-flight analysis was performed to investigate the functions and mechanisms of specific CAFs in the immune context of HCC. RESULTS We identified a distinct flavin-containing monooxygenase 2 (FMO2)+ CAF subset associated with a favorable response to anti-PD-1 therapy and better clinical outcomes. FMO2+ CAFs increase anti-PD-1 treatment efficacy by promoting tertiary lymphoid structure formation and increasing the infiltration of CD8+ T cells and M1-like macrophages through the C-C motif chemokine ligand 19 (CCL19)-C-C motif chemokine receptor 7 axis. Mechanistically, FMO2 promotes nuclear factor kappa B/p65-mediated CCL19 expression by competitively binding to glycogen synthase 1 (GYS1) with praja ring finger ubiquitin ligase 1 (PJA1), thereby suppressing the PJA1-mediated proteasomal degradation of GYS1. CCL19 treatment potentiated the therapeutic efficacy of anti-PD-1 therapy in mouse orthotopic HCC models. A favorable immunotherapy response was observed in patients with HCC with high serum levels of CCL19. CONCLUSIONS We identified a novel FMO2+ CAF subset that serves as a critical regulator of microenvironmental immune properties and a predictive biomarker of the immunotherapy response in patients with HCC. CCL19 in combination with anti-PD-1 therapy may constitute a novel therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Wenxin Xu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jialei Weng
- Department of Surgical Oncology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yufei Zhao
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Peiyi Xie
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Minghao Xu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shaoqing Liu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiang Yu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Mincheng Yu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Bugang Liang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Junbo Chen
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Hui Li
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Qinghai Ye
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yinghao Shen
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
6
|
Lei K, Lei Y, Wang Z, Ye Z, Liu J, Chen W, Zhou C, Tan J, Chen S, Zhang Y, Tan J. Integrative multi-omics and Mendelian randomization analysis reveal SPP1 + tumor-associated macrophage-driven prognostic signature for hepatocellular carcinoma. Front Mol Biosci 2025; 12:1594610. [PMID: 40376263 PMCID: PMC12078150 DOI: 10.3389/fmolb.2025.1594610] [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: 03/16/2025] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
Background The SPP1+ tumor-associated macrophages (TAMs) have been implicated in tumor metastasis and immune evasion. However, the prognostic significance of SPP1+ TAMs in hepatocellular carcinoma (HCC) remains largely unexplored. This study aimed to identify SPP1+ TAMs-related genes and construct a model to predict overall survival (OS) in HCC patients. Methods Single-cell RNA sequencing (scRNA-seq) datasets from HCC patients were analyzed to identify SPP1+ TAMs. SPP1+ TAMs-related risk score (STRS) was developed using Mendelian randomization (MR) analysis and Least Absolute Shrinkage and Selection Operator (LASSO) regression. HCC patients from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) cohorts were stratified into high- and low-STRS groups based on STRS. Kaplan-Meier survival analysis, receiver operating characteristic (ROC) curve analysis, and functional enrichment analysis were performed to assess the prognostic value of STRS. Results SPP1+ TAMs exhibited strong associations with immunosuppressive functions. 16 SPP1+ TAMs-related genes were used to construct STRS. Patients in the high-STRS group had significantly worse OS than those in the low-STRS group (p < 0.001). ROC analysis demonstrated robust predictive power, with AUC values ranging from 0.685 to 0.748 for 1-year OS, 0.717 to 0.739 for 2-year OS, and 0.719 to 0.738 for 3-year OS. The STRS model also exhibited strong predictive capability for the distinction of drug resistance. Conclusion This study identified SPP1+ TAMs-related genes as key prognostic indicators in HCC. The STRS model provides an effective tool for predicting patient survival and may facilitate personalized treatment strategies for HCC. These findings enhance the understanding of TAMs-driven immune modulation in HCC and highlight potential therapeutic targets for improving patient outcomes.
Collapse
Affiliation(s)
- Kai Lei
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yichun Lei
- School of Nursing, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Zeyao Wang
- Department of General Surgery, Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen University, Huizhou, Guangdong, China
| | - Zhixin Ye
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiawei Liu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenhao Chen
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Caihong Zhou
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinmei Tan
- Department of Intensive Care Unit, Wuchuan People’s Hospital, Zhanjiang, Guangdong, China
| | - Shuxian Chen
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yifan Zhang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiehui Tan
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Miri H, Rahimzadeh P, Hashemi M, Nabavi N, Aref AR, Daneshi S, Razzaghi A, Abedi M, Tahmasebi S, Farahani N, Taheriazam A. Harnessing immunotherapy for hepatocellular carcinoma: Principles and emerging promises. Pathol Res Pract 2025; 269:155928. [PMID: 40184729 DOI: 10.1016/j.prp.2025.155928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
HCC is considered as one of the leadin causes of death worldwide, with the ability of resistance towards therapeutics. Immunotherapy, particularly ICIs, have provided siginficant insights towards harnessing the immune system. The present review introduces the concepts and possibilities of immunotherapy for HCC treatment, emphasizing its underlying mechanisms and capacity to enhance patient results, focusing on both pre-clinical and clinical insights. The functions of TME and immune evasion mechanisms typical of HCC would be evaluated along with how contemporary immunotherapeutic approaches are designed to address these challenges. Furthermore, the clinical application of immunotherapy in HCC is discussed, emphasizing recent trial findings demonstrating the effectiveness and safety of drugs. In addition, the problems caused by immune evasion and resistance would be discussed to increase potential of immunotherapy along with combination therapy.
Collapse
Affiliation(s)
- Hossein Miri
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University Of Medical Sciences, Jiroft, Iran
| | - Alireza Razzaghi
- Social Determinants of Health Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
8
|
Qiu Y, Wang Y, Liu J, Sun K, Liu B, Hou Q. Single-cell sequencing unveils the transcriptomic landscape of castration-resistant prostate cancer-associated fibroblasts and their association with prognosis and immunotherapy response. BMC Cancer 2025; 25:813. [PMID: 40307786 PMCID: PMC12044937 DOI: 10.1186/s12885-025-14212-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND The tumor microenvironment (TME) is increasingly acknowledged as a determinant in the malignant transformation and progression of castration-resistant prostate cancer (CRPC). Cancer-associated fibroblasts (CAFs), as a pivotal stromal cellular component in TME, are implicated in tumor progression and immune escape. However, the molecular characteristics and biological functions of CRPC-CAFs in prostate cancer necessitate further investigation. METHODS We ascertained the differential transcriptomic profiles between CRPC-CAFs and PCa-CAFs through single-cell RNA-sequencing (scRNA-seq). Bulk RNA-seq data were employed to assess the prognostic implications of CRPC-CAFs in PCa. In addition, we examined the impact of CRPC-CAFs on the efficacy of immunotherapy and the composition of the tumor immune milieu. Furthermore, a subcutaneous PCa model was applied to determine the potential of TGF-β signaling blockade to augment the response to immunotherapeutic interventions. RESULTS We observed a pronounced increase in the proportion of CAFs in CRPC compared to those in primary PCa. The functional pathways implicated in TGF-β signaling and ECM remodeling were remarkably upregulated in CRPC-CAFs. Moreover, gene regulatory network analysis uncovered substantial differences in the transcription factor activity profiles between CRPC-CAFs and PCa-CAFs. The elevated CRPC-CAFs abundance was associated with diminished recurrence-free survival and immunotherapy insensitivity. Substantially elevated infiltration of inhibitory immune cells and upregulated expression levels of immunosuppressive molecules were observed in patients with high CRPC-CAFs abundance. Importantly, administration of anti-TGF-β therapy remarkably potentiated the efficacy of anti-PD-1 immunotherapy through upregulating the anti-tumor immune response in the PCa model. CONCLUSION Our results highlighted the impact of CRPC-CAFs on clinical prognosis and immunosuppressive tumor milieu, indicating that CRPC-CAFs may function as a promising therapeutic target for CRPC.
Collapse
Affiliation(s)
- Yifeng Qiu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of Shenzhen University, Shenzhen, China
| | - Yuhan Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Jiahe Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Kai Sun
- Department of Radiology, the Third People's Hospital of Longgang District, Shenzhen, China
- Shenzhen Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518116, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
| | - Qi Hou
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of Shenzhen University, Shenzhen, China.
| |
Collapse
|
9
|
Jia H, Chen X, Zhang L, Chen M. Cancer associated fibroblasts in cancer development and therapy. J Hematol Oncol 2025; 18:36. [PMID: 40156055 PMCID: PMC11954198 DOI: 10.1186/s13045-025-01688-0] [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: 01/22/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are key players in cancer development and therapy, and they exhibit multifaceted roles in the tumor microenvironment (TME). From their diverse cellular origins, CAFs undergo phenotypic and functional transformation upon interacting with tumor cells and their presence can adversely influence treatment outcomes and the severity of the cancer. Emerging evidence from single-cell RNA sequencing (scRNA-seq) studies have highlighted the heterogeneity and plasticity of CAFs, with subtypes identifiable through distinct gene expression profiles and functional properties. CAFs influence cancer development through multiple mechanisms, including regulation of extracellular matrix (ECM) remodeling, direct promotion of tumor growth through provision of metabolic support, promoting epithelial-mesenchymal transition (EMT) to enhance cancer invasiveness and growth, as well as stimulating cancer stem cell properties within the tumor. Moreover, CAFs can induce an immunosuppressive TME and contribute to therapeutic resistance. In this review, we summarize the fundamental knowledge and recent advances regarding CAFs, focusing on their sophisticated roles in cancer development and potential as therapeutic targets. We discuss various strategies to target CAFs, including ECM modulation, direct elimination, interruption of CAF-TME crosstalk, and CAF normalization, as approaches to developing more effective treatments. An improved understanding of the complex interplay between CAFs and TME is crucial for developing new and effective targeted therapies for cancer.
Collapse
Affiliation(s)
- Hongyuan Jia
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Xingmin Chen
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Linling Zhang
- Department of Respiratory and Critical Care, Chengdu Third People's Hospital, Chengdu, China
| | - Meihua Chen
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
10
|
Xuan Z, Hu H, Xu J, Ling X, Zhang L, Li W, Li J, Zhu C, Song Y, Zhang X, Rao J, Wang Y, Cheng F. Adrenomedullin Inhibits the Efficacy of Combined Immunotherapy and Targeted Therapy in Biliary Tract Cancer by Disrupting Endothelial Cell Functions. J Cell Mol Med 2025; 29:e70460. [PMID: 40074697 PMCID: PMC11903196 DOI: 10.1111/jcmm.70460] [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: 08/27/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
The global incidence of biliary tract cancer (BTC) is on the rise, presenting a substantial healthcare challenge. The integration of immune checkpoint inhibitors (ICIs) with molecularly targeted therapies is emerging as a strategy to enhance immune responses. However, the efficacy and underlying mechanisms of these treatments in BTC are still largely unexplored. In this study, tissue samples from 19 BTC patients treated with camrelizumab and apatinib were analysed using the NanoString 289-panel to identify key molecular biomarkers. Comparative analyses and subsequent experimental validations, including cell-based assays and histopathological examinations, identified adrenomedullin (ADM) as a critical molecular marker associated with treatment efficacy and poor prognosis. ADM has been shown to promote BTC cell proliferation, migration and angiogenesis, primarily by interacting with vascular endothelial growth factor (VEGF) and increasing AKT phosphorylation. Furthermore, ADM disrupts endothelial cell barrier function via the calcitonin receptor-like receptor (CRLR) and vascular endothelial (VE)-cadherin signalling pathway. Preclinical inhibition of ADM or CRLR resulted in suppressed tumour growth. Additionally, elevated ADM expression was correlated with increased tumour-infiltrating immune cells and higher immune checkpoint expression. These findings suggest that ADM plays a pivotal role in resistance to immunotherapy and anti-angiogenic treatment in BTC, and thus, targeting ADM may offer a promising therapeutic approach to enhance treatment efficacy.
Collapse
Affiliation(s)
- Zhengfeng Xuan
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| | - Haoran Hu
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| | - Jian Xu
- Department of General SurgeryChangzhou Jintan District People's HospitalChangzhouJiangsuChina
| | - Xiaowei Ling
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| | - Long Zhang
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| | - Wenzhu Li
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| | - Junda Li
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| | - Chan Zhu
- Jiangsu Simcere Diagnostics Co. Ltd., Nanjing Simcere Medical Laboratory Science Co. Ltd.The State Key Laboratory of Neurology and Oncology Drug DevelopmentNanjingChina
| | - Yunjie Song
- Jiangsu Simcere Diagnostics Co. Ltd., Nanjing Simcere Medical Laboratory Science Co. Ltd.The State Key Laboratory of Neurology and Oncology Drug DevelopmentNanjingChina
| | - Xing Zhang
- Jiangsu Simcere Diagnostics Co. Ltd., Nanjing Simcere Medical Laboratory Science Co. Ltd.The State Key Laboratory of Neurology and Oncology Drug DevelopmentNanjingChina
| | - Jianhua Rao
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| | - Yong Wang
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| | - Feng Cheng
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjingJiangsuChina
| |
Collapse
|
11
|
Zhang YZ, Ma Y, Ma E, Chen X, Zhang Y, Yin B, Zhao J. Sophisticated roles of tumor microenvironment in resistance to immune checkpoint blockade therapy in hepatocellular carcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:10. [PMID: 40051497 PMCID: PMC11883234 DOI: 10.20517/cdr.2024.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/13/2025] [Accepted: 02/21/2025] [Indexed: 03/09/2025]
Abstract
Hepatocellular carcinoma (HCC) remains a serious threat to global health, with rising incidence and mortality rates. Therapeutic options for advanced HCC are quite limited, and the overall prognosis remains poor. Recent advancements in immunotherapy, particularly immune-checkpoint blockade (ICB) targeting anti-PD1/PD-L1 and anti-CTLA4, have facilitated a paradigm shift in cancer treatment, demonstrating substantial survival benefits across various cancer types, including HCC. However, only a subset of HCC patients exhibit a favorable response to ICB therapy, and its efficacy is often hindered by the development of resistance. There are many studies to explore the underlying mechanisms of ICB response. In this review, we compiled the latest progression in immunotherapies for HCC and systematically summarized the sophisticated mechanisms by which components of the tumor microenvironment (TME) regulate resistance to ICB therapy. Additionally, we also outlined some scientific rationale strategies to boost antitumor immunity and enhance the efficacy of ICB in HCC. These insights may serve as a roadmap for future research and help improve outcomes for HCC patients.
Collapse
Affiliation(s)
- Yi-Zhe Zhang
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Authors contributed equally
| | - Yunshu Ma
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Authors contributed equally
| | - Ensi Ma
- Liver Transplantation Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Organ Transplantation, Fudan University, Shanghai 200040, China
| | - Xizhi Chen
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yue Zhang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Baobing Yin
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Hepatobiliary surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian, China
| | - Jing Zhao
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Hepatobiliary surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian, China
- Cancer Metastasis Institute, Fudan University, Shanghai 201206, China
| |
Collapse
|
12
|
Liu Y, Dong G, Yu J, Liang P. Integration of single-cell and spatial transcriptomics reveals fibroblast subtypes in hepatocellular carcinoma: spatial distribution, differentiation trajectories, and therapeutic potential. J Transl Med 2025; 23:198. [PMID: 39966876 PMCID: PMC11837652 DOI: 10.1186/s12967-025-06192-0] [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: 11/22/2024] [Accepted: 02/01/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are key components of the hepatocellular carcinoma (HCC) tumor microenvironment (TME). regulating tumor proliferation, metastasis, therapy resistance, immune evasion via diverse mechanisms. A deeper understanding of the l diversity of CAFs is essential for predicting patient prognosis and guiding treatment strategies. METHODS We examined the diversity of CAFs in HCC by integrating single-cell, bulk, and spatial transcriptome analyses. RESULTS Using a training cohort of 88 HCC single-cell RNA sequencing (scRNA-seq) samples and a validation cohort of 94 samples, encompassing over 1.2 million cells, we classified three fibroblast subpopulations in HCC: HLA-DRB1 + CAF, MMP11 + CAF, and VEGFA + CAF based on highly expressed genes of which, which are primarily located in normal tissue, tumor boundaries, and tumor interiors, respectively. Cell trajectory analysis revealed that VEGFA + CAFs are at the terminal stage of differentiation, which, notably, is tumor-specific. VEGFA + CAFs were significantly associated with patient survival, and the hypoxic microenvironment was found to be a major factor inducing VEGFA + CAFs. Through cellular communication with capillary endothelial cells (CapECs), VEGFA + CAFs promoted intra-tumoral angiogenesis, facilitating tumor progression and metastasis. Additionally, a machine learning model developed using high-expression genes from VEGFA + CAFs demonstrated high accuracy in predicting prognosis and sorafenib response in HCC patients. CONCLUSIONS We characterized three fibroblast subpopulations in HCC and revealed their distinct spatial distributions within the tumor. VEGFA + CAFs, which was induced by hypoxic TME, were associated with poorer prognosis, as they promote tumor angiogenesis through cellular communication with CapECs. Our findings provide novel insights and pave the way for individualized therapy in HCC patients.
Collapse
Affiliation(s)
- Yue Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Ultrasound, Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
- Department of Interventional Ultrasound, First Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
| | - Guoping Dong
- Department of Ultrasound, Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
- Department of Interventional Ultrasound, First Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
| | - Jie Yu
- Department of Ultrasound, Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
- Department of Interventional Ultrasound, First Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
| | - Ping Liang
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of Ultrasound, Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China.
- Department of Interventional Ultrasound, First Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
13
|
Lou J, Zhang B, Cai J, Zhang L, Zhao Y, Zhao Z. Diabetes exacerbates periodontitis by disrupting IL-33-mediated interaction between periodontal ligament fibroblasts and macrophages. Int Immunopharmacol 2025; 147:113896. [PMID: 39740505 DOI: 10.1016/j.intimp.2024.113896] [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: 11/10/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
Tissue-resident fibroblasts with immunomodulatory properties have recently been identified as key players in inflammation. However, their roles within the periodontal niche in diabetes-associated periodontitis remain unclear. Interleukin (IL)-33, known as an "alarmin" in inflammatory responses, has recently emerged as a potential contributor to periodontitis. Herein, we show that IL-33 levels are reduced in periodontal ligament fibroblasts (PDLFs) in the in vivo models of diabetes-associated periodontitis and in vitro models of diabetic inflammation. In the in vitro co-culture model, overexpression of IL-33 in PDLFs promotes M2 macrophage polarization, while knockdown of IL-33 in PDLFs instigates M1 macrophage polarization. Notably, supplementation with IL-33 in vivosignificantly alleviates periodontal tissue destruction and enhances M2 macrophage infiltration, whereas targeting the IL-33/ST2 axis exacerbates tissue damage and promotes M1 macrophage polarization in diabetes-associated periodontitis. Additionally, theCUT&RUN assay confirms the direct regulation of IL-33 by Yes-associated protein (YAP). These findings demonstrate that IL-33 deficiency in PDLFs favors M1 macrophage polarization, thereby exacerbating the pathogenesis of diabetes-associated periodontitis. Our study underscores the essential immunomodulatory role of PDLFs in creating an inflammatory niche and unveils a novel interaction axis between PDLFs and macrophages in diabetes-associated periodontitis.
Collapse
Affiliation(s)
- Jingyang Lou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jingyi Cai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Linli Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yifan Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Deng Y, Chen Q, Guo C, Chen J, Li X, Li Z, Zhang Y, Zhao J, Zhou J, Cai J, Yan T, Wang X, Bi X, Huang Z, Zhao H. Comprehensive single-cell atlas of colorectal neuroendocrine tumors with liver metastases: unraveling tumor microenvironment heterogeneity between primary lesions and metastases. Mol Cancer 2025; 24:28. [PMID: 39838423 PMCID: PMC11748842 DOI: 10.1186/s12943-025-02231-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: 09/10/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Colorectal neuroendocrine tumors with liver metastases (CRNELM) are associated with a poorer prognosis compared to their nonmetastatic counterparts. A comprehensive understanding of the tumor microenvironment (TME) heterogeneity between primary lesions (PL) and liver metastases (LM) could provide crucial insights for enhancing clinical management strategies for these patients. METHODS We utilized single-cell RNA sequencing to analyze fresh tissue samples from CRNELM patients, aiming to elucidate the variations in TME between PL and LM. Complementary multidimensional validation was achieved through spatial transcriptomics, bulk RNA sequencing, and multiplex immunohistochemistry/immunofluorescence. RESULTS Our single-cell RNA sequencing analysis revealed that LM harboured a higher proportion of CD8 + T cells, CD4 + T cells, NK cells, NKT cells, and B cells exhibiting a stress-like phenotype compared to PL. RGS5 + pericytes may play a role in the stress-like phenotype observed in immune cells within LM. MCs in PL (PL_MCs) and LM (LM_MCs) exhibit distinct activation of tumor-associated signaling pathways. Notably, COLEC11 + matrix cancer-associated fibroblasts (COLEC11_mCAFs) were found to be significantly associated with LM_MCs. Cell communication analysis unveiled potential targetable receptor-ligand interactions between COLEC11_mCAFs and LM_MCs. Multidimensional validation confirmed the prominence of the characteristic stress-like phenotypes, including HSPA6_CD8_Tstr, HSPA6_NK, and COLEC11_mCAFs in LM. Moreover, a higher abundance of COLEC11_mCAFs correlated with poorer survival rates in the neuroendocrine tumor patient cohort. CONCLUSION Overall, our study provides the first single-cell analysis of the cellular and molecular differences between PL and LM in CRNELM patients. We identified distinct cell subsets and receptor-ligand interactions that may drive TME discrepancies and support metastatic tumor growth. These insights highlight potential therapeutic targets and inform strategies for better managing CRNELM patients.
Collapse
Affiliation(s)
- Yiqiao Deng
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qichen Chen
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Chengyao Guo
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jinghua Chen
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xin Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiyu Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yefan Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianjun Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianguo Zhou
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiaobing Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhen Huang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
15
|
Bai H, Zhu X, Gao L, Feng S, Li H, Gu X, Xu J, Zong C, Hou X, Yang X, Jiang J, Zhao Q, Wei L, Zhang L, Han Z, Liu W, Qian J. ERG mediates the differentiation of hepatic progenitor cells towards immunosuppressive PDGFRα + cancer-associated fibroblasts during hepatocarcinogenesis. Cell Death Dis 2025; 16:26. [PMID: 39827226 PMCID: PMC11743139 DOI: 10.1038/s41419-024-07270-9] [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: 09/05/2024] [Revised: 11/03/2024] [Accepted: 11/27/2024] [Indexed: 01/22/2025]
Abstract
Cancer-associated fibroblasts (CAFs) play important roles in the occurrence and development of hepatocellular carcinoma (HCC) and are a key component of the immunosuppressive microenvironment. However, the origin of CAFs has not been fully elucidated. We employed single-cell sequencing technology to identify the dynamic changes in different subsets of fibroblasts at different time points in rat primary HCC model. Inflammation-associated CAFs (Pdgfrα+ CAFs) were subsequently identified, which demonstrated a significant correlation with the survival duration of HCC patients and a dual role in the tumour microenvironment (TME). On the one hand, they secrete the chemokines CCL3 and CXCL12, which recruit macrophages to the tumour site. On the other hand, they produce TGFβ, inducing the polarization of these macrophages towards an immunosuppressive phenotype. According to the in vitro and in vivo results, hepatic progenitor cells (HPCs) can aberrantly differentiate into PDGFRα+ CAFs upon stimulation with inflammatory cytokine. This differentiation is mediated by the activation of the MAPK signaling pathway and the downstream transcription factor ERG via the TLR4 receptor. Downregulating the expression of ERG in HPCs significantly reduces the number of PDGFRα+ CAFs and the infiltration of tumour-associated macrophages in HCC, thereby suppressing hepatocarcinogenesis. Collectively, our findings elucidate the distinct biological functions of PDGFRα+ cancer-associated fibroblasts (PDGFRα+ CAFs) within the TME. These insights contribute to our understanding of the mechanisms underlying the establishment of an immunosuppressive microenvironment in HCC, paving the way for the exploration of novel immunotherapeutic strategies tailored for HCC treatment.
Collapse
Affiliation(s)
- Haoran Bai
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyu Zhu
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China
| | - Lu Gao
- National Center for Liver Cancer, Shanghai, China
| | - Shiyao Feng
- Department of Urology, Chaohu Hospital of Anhui Medical University, HeFei, Anhui, China
| | - Hegen Li
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoqiang Gu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiahua Xu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Zong
- National Center for Liver Cancer, Shanghai, China
| | - Xiaojuan Hou
- National Center for Liver Cancer, Shanghai, China
| | - Xue Yang
- National Center for Liver Cancer, Shanghai, China
| | | | - Qiudong Zhao
- National Center for Liver Cancer, Shanghai, China
| | - Lixin Wei
- National Center for Liver Cancer, Shanghai, China
| | - Li Zhang
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China.
| | - Zhipeng Han
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- National Center for Liver Cancer, Shanghai, China.
| | - Wenting Liu
- National Center for Liver Cancer, Shanghai, China.
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Jianxin Qian
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
16
|
Zhao Y, Zhou C, Zuo L, Yan H, Gu Y, Liu H, Yu G, Zhou X. Identification of cancer cell-intrinsic biomarkers associated with tumor progression and characterization of SFTA3 as a tumor suppressor in lung adenocarcinomas. BMC Cancer 2025; 25:36. [PMID: 39780110 PMCID: PMC11707868 DOI: 10.1186/s12885-024-13395-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Recent advancements in contemporary therapeutic approaches have increased the survival rates of lung cancer patients; however, the long-term benefits remain constrained, underscoring the pressing need for novel biomarkers. Surfactant-associated 3 (SFTA3), a long non-coding RNA predominantly expressed in normal lung epithelial cells, plays a crucial role in lung development. Nevertheless, its function in lung adenocarcinoma (LUAD) remains inadequately understood. METHODS Single-cell RNA sequencing data were utilized to identify novel cancer cell-intrinsic gene signatures associated with the progression of LUAD, and their roles in LUAD were comprehensively analyzed. Serum samples were collected to quantify the expression levels of SFTA3 in LUAD patients. Furthermore, a series of biological experiments, including cell viability assays, scratch wound healing assays, and colony formation assays, were conducted to demonstrate the tumor-suppressive effects of SFTA3. RNA sequencing was performed to elucidate the molecular mechanisms underlying the role of SFTA3 in lung cancer cells. RESULTS We constructed a prognostic model comprising eight genes: ALDOA, ATP5MD, SERPINH1, SFTA3, SLK, U2SURP, SCGB1A1, and SCGB1A3. The model effectively stratified patients into high- and low-risk categories, revealing that low-risk patients experienced superior clinical outcomes, exhibited an immunologically hot tumor microenvironment (TME), and had a greater probability of responding to immunotherapy. In contrast, the high-risk group exhibited a cold TME and may benefit more from chemotherapy. Furthermore, our study revealed that a progressive decrease in SFTA3 expression in cancer cells was correlated with tumor advancement. Notably, the serum levels of SFTA3 significantly decreased in patients with LUAD, suggesting its potential utility in liquid biopsy for LUAD diagnosis. Additionally, the knockdown of SFTA3 enhances the proliferation and migration of lung cancer cells, whereas its overexpression inhibits these phenotypes. The epithelial-mesenchymal transition pathway was significantly enriched following SFTA3 silencing, suggesting that SFTA3 may impact tumor progression by modulating this process. We also identified key transcription factors and epigenetic mechanisms implicated in the downregulation of SFTA3 in LUAD. CONCLUSION We developed a robust prognostic model and identified SFTA3 as a novel biomarker with potential applications in the diagnosis, prognosis, and personalized treatment of LUAD. Additionally, our findings offer new insights into the mechanisms underlying LUAD tumorigenesis and immune evasion.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226000, China
| | - Chengcheng Zhou
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226000, China
- Department of Hematology, Affiliated Hospital and Medical School of Nantong University, Nantong, China
| | - Ling Zuo
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226000, China
| | - Haoming Yan
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226000, China
| | - Yuhan Gu
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226000, China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital and Medical School of Nantong University, Nantong, China
| | - Guiping Yu
- Department of Cardiothoracic Surgery, Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, China.
| | - Xiaorong Zhou
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226000, China.
| |
Collapse
|
17
|
De Martin E, Fulgenzi CAM, Celsa C, Laurent-Bellue A, Torkpour A, Lombardi P, D'Alessio A, Pinato DJ. Immune checkpoint inhibitors and the liver: balancing therapeutic benefit and adverse events. Gut 2024:gutjnl-2024-332125. [PMID: 39658265 DOI: 10.1136/gutjnl-2024-332125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
Immune checkpoint inhibitors (ICI) have led to breakthrough improvements in the management of malignancy including hepatocellular (HCC) and biliary tract cancer, improving decades-old standards of care and increasing patient survival. In both liver tumour types, which commonly arise in the context of liver inflammation and underlying functional impairment, the lack of validated predictors of response underscores the need to balance predicted gains in survival with risk of treatment-related hepatoxicity and decompensation of underlying chronic liver disease.In addition, the liver is implicated in the toxicity associated with ICI therapy for non-liver cancers, which exhibits a high degree of variability in presentation and severity. An accurate assessment is mandatory for the diagnosis and management of ICI-induced liver injury.In this Recent Advances article, we provide an overview of the mechanisms of efficacy and toxicity of anticancer immunotherapy in liver tumours and liver toxicity in extrahepatic malignancies.We compare and contrast characteristics, management strategies and outcomes from immune-related liver injury in patients with chronic hepatitis/cirrhosis or with an underlying healthy liver and discuss the latest findings on how toxicity and decompensation may impact the outlook of patients with liver tumours and extrahepatic malignancies offering insights into the future directions of clinical research and practice in the field.
Collapse
Affiliation(s)
- Eleonora De Martin
- Centre Hepatobiliaire, Paul Brousse Hospital, Villejuif, France
- Paris-Saclay University, Faculty of Medicine, Le Kremlin-Bicetre, France
| | | | - Ciro Celsa
- Surgery & Cancer, Imperial College London, London, UK
- Department of Health Promotion, Mother & Child Care, Internal Medicine & Medical Specialties, Gastroenterology and Hepatology Unit, Palermo, Italy
| | - Astrid Laurent-Bellue
- Hôpital Kremlin Bicêtre, Anatomie & Cytologie Pathologiques, Le Kremlin Bicetre, France
| | - Aria Torkpour
- Surgery & Cancer, Imperial College London, London, UK
| | - Pasquale Lombardi
- Surgery & Cancer, Imperial College London, London, UK
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Antonio D'Alessio
- Surgery & Cancer, Imperial College London, London, UK
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - David J Pinato
- Surgery & Cancer, Imperial College London, London, UK
- Imperial College London, University of Eastern Piedmont Amedeo Avogadro, Department of Translational Medicine, Novara, Italy
| |
Collapse
|
18
|
Zhang S, Cao G, Shen S, Wu Y, Tan X, Jiang X. CAF-derived miR-642a-3p supports migration, invasion, and EMT of hepatocellular carcinoma cells by targeting SERPINE1. PeerJ 2024; 12:e18428. [PMID: 39544420 PMCID: PMC11562775 DOI: 10.7717/peerj.18428] [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: 06/07/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Background Cancer-associated fibroblasts (CAFs) and hepatocellular carcinoma (HCC) cells interact to promote HCC progression, but the underlying mechanisms remain unclear. Serpin family E member 1 (SERPINE1) has conflicting roles in HCC, and microRNAs (miRNAs) are known to regulate tumor progression through intercellular communication. Therefore, we investigated the potential involvement of miRNA/SERPINE1 axis in crosstalk between CAFs and HCC cells. Methods In this study, candidate miRNAs targeting SERPINE1 3' UTR were predicted using multiple miRNA databases. The miRNAs and SERPINE1 mRNA expression in Huh7 cells was assessed after co-culture with CAFs using RT-qPCR. Huh7 cell proliferation and invasion were detected after SERPINE1 siRNA. The functions of the CAF-derived miR-642a-3p/SERPINE1 axis in HCC cells were examined using CCK-8, wound healing, transwell assays, western blot, and dual-luciferase reporter assays. Moreover, a orthotopic xenograft model was used to investigate the contribution of miR-642a-3p knockdown in HCC. Results SERPINE1 mRNA expression decreased, while miR-642a-3p expression increased in Huh7 cells co-cultured with CAFs. SERPINE1 knockdown enhanced Huh7 cell proliferation and invasion as well as miR-642a-3p expression. miR-642a-3p overexpression promoted migration, invasion, and epithelial-mesenchymal transition (EMT) in Huh7 cells by targeting SERPINE1, while miR-642a-3p knockdown yielded the opposite effect. Rescue experiments confirmed that SERPINE1 knockdown attenuated the inhibitory effects of miR-642a-3p knockdown on migration, invasion, and EMT in Huh7 cells. Importantly, miR-642a-3p knockdown suppressed growth and EMT in orthotopic liver tumors. Conclusion CAF-derived miR-642a-3p/SERPINE1 axis facilitated migration, invasion, and EMT in the HCC cells, suggesting miR-642a-3p/SERPINE1 axis can be a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Pharmacy, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Gang Cao
- Office of the Dean, Nantong Maternal and Child Health Care Hospital, Nantong, China
| | - Shuijie Shen
- Department of Science and Education, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Yu Wu
- Department of Science and Education, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Xiying Tan
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyan Jiang
- Department of Pharmacy, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| |
Collapse
|