1
|
Pourakbar N, Motamedi A, Pashapour M, Sharifi ME, Sharabiani SS, Fazlollahi A, Abdollahi H, Rahmim A, Rezaei S. Effectiveness of Artificial Intelligence Models in Predicting Lung Cancer Recurrence: A Gene Biomarker-Driven Review. Cancers (Basel) 2025; 17:1892. [PMID: 40507370 PMCID: PMC12153899 DOI: 10.3390/cancers17111892] [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: 04/08/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Lung cancer recurrence, particularly in NSCLC, remains a major challenge, with 30-70% of patients relapsing post-treatment. Traditional predictors like TNM staging and histopathology fail to account for tumor heterogeneity and immune dynamics. This review evaluates AI models integrating gene biomarkers (TP53, KRAS, FOXP3, PD-L1, and CD8) to enhance the recurrence prediction and improve the personalized risk stratification. METHODS Following the PRISMA guidelines, we systematically reviewed AI-driven recurrence prediction models for lung cancer, focusing on genomic biomarkers. Studies were selected based on predefined criteria, emphasizing AI/ML approaches integrating gene expression, radiomics, and clinical data. Data extraction covered the study design, AI algorithms (e.g., neural networks, SVM, and gradient boosting), performance metrics (AUC and sensitivity), and clinical applicability. Two reviewers independently screened and assessed studies to ensure accuracy and minimize bias. RESULTS A literature analysis of 18 studies (2019-2024) from 14 countries, covering 4861 NSCLC and small cell lung cancer patients, showed that AI models outperformed conventional methods. AI achieved AUCs of 0.73-0.92 compared to 0.61 for TNM staging. Multi-modal approaches integrating gene expression (PDIA3 and MYH11), radiomics, and clinical data improved accuracy, with SVM-based models reaching a 92% AUC. Key predictors included immune-related signatures (e.g., tumor-infiltrating NK cells and PD-L1 expression) and pathway alterations (NF-κB and JAK-STAT). However, small cohorts (41-1348 patients), data heterogeneity, and limited external validation remained challenges. CONCLUSIONS AI-driven models hold potential for recurrence prediction and guiding adjuvant therapies in high-risk NSCLC patients. Expanding multi-institutional datasets, standardizing validation, and improving clinical integration are crucial for real-world adoption. Optimizing biomarker panels and using AI trustworthily and ethically could enhance precision oncology, enabling early, tailored interventions to reduce mortality.
Collapse
Affiliation(s)
- Niloufar Pourakbar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran; (N.P.); (A.M.); (M.P.); (S.S.S.); (A.F.)
| | - Alireza Motamedi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran; (N.P.); (A.M.); (M.P.); (S.S.S.); (A.F.)
| | - Mahta Pashapour
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran; (N.P.); (A.M.); (M.P.); (S.S.S.); (A.F.)
| | - Mohammad Emad Sharifi
- Shariati Hospital Research Center, Tehran University of Medical Sciences, Tehran 1416634793, Iran;
| | - Seyedemad Seyedgholami Sharabiani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran; (N.P.); (A.M.); (M.P.); (S.S.S.); (A.F.)
| | - Asra Fazlollahi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran; (N.P.); (A.M.); (M.P.); (S.S.S.); (A.F.)
| | - Hamid Abdollahi
- Research Department of Integrative Oncology, BC Cancer Institute, Vancouver, BC V5Z 1L3, Canada; (H.A.); (A.R.)
| | - Arman Rahmim
- Research Department of Integrative Oncology, BC Cancer Institute, Vancouver, BC V5Z 1L3, Canada; (H.A.); (A.R.)
- Departments of Radiology and Physics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Sahar Rezaei
- Department of Radiology, Medical School, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran
| |
Collapse
|
2
|
Diao B, Cai Y, Song D, Hu Y, Xie B, Kan Y, Hu X. A potential therapeutic molecule target: lncRNA AK023507 inhibits the metastasis of breast cancer by regulating the WNT/DOCK4/β-catenin axis. Breast Cancer Res Treat 2025; 211:727-741. [PMID: 40205246 DOI: 10.1007/s10549-025-07695-6] [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: 06/17/2024] [Accepted: 03/23/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE Breast cancer (BC) has become the most common malignant tumor in women worldwide. This study was carried out to find and validate a novel molecular therapeutic target for BC. METHODS Long non-coding RNA (lncRNA) AK023507 was selected as the study objects through microarray analysis. The function of lncRNA AK023507 was verified by various cell function experiments in vitro, subcutaneous tumorigenesis experiments, and lung metastasis model experiments in vivo. The RNA pull-down experiment and Western blot experiment were used to confirm the mechanism regulation pathway and the recovery experiment was used to verify it. TCGA datasets were used for clinical and immune function prediction analysis. RESULTS In vitro cell function tests and in vivo experiments suggested that overexpression of lncRNA AK023507 inhibited the proliferation and metastasis of BC cells. The RNA pull-down experiment and Western blot analysis validated that lncRNA AK023507 interacted with the dedicator of cytokinesis 4 (DOCK4) protein. Analysis of public databases predicted that DOCK4 is a potential prognostic risk factor associated with epithelial-mesenchymal transition (EMT) and central memory T cell (TCM) cellular immune infiltration. CONCLUSIONS LncRNA AK023507 inhibits the proliferation and metastasis of BC by regulating the DOCK4/β-catenin axis. This discovery will provide new potential therapeutic targets for BC.
Collapse
Affiliation(s)
- Biyu Diao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China
| | - Yangjun Cai
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China
- Department of Thyroid and Breast Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Dandan Song
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China
| | - Yingying Hu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China
| | - Bojian Xie
- Department of Thyroid and Breast Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Yang Kan
- Department of Thyroid and Breast Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Xiaoqu Hu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China.
| |
Collapse
|
3
|
Zhou Y, Chen T, Pan Y, Liu J. Exploring the mechanism of fibronectin extra domain B in the tumor microenvironment and implications for targeted immunotherapy and diagnostics (Review). Mol Med Rep 2025; 31:160. [PMID: 40211711 PMCID: PMC12015389 DOI: 10.3892/mmr.2025.13525] [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: 11/05/2024] [Accepted: 01/17/2025] [Indexed: 04/25/2025] Open
Abstract
Fibronectin extra domain B (FN‑EDB) is a unique domain of FN), whose expression is significantly upregulated in the tumor microenvironment (TME). FN‑EDB plays a key role in tumor cell adhesion, angiogenesis and invasion, and is closely related to tumor malignancy and poor prognosis. Moreover, the high expression of FN‑EDB in multiple cancer types makes it a potential therapeutic target. However, comprehensive studies of the mechanism of FN‑EDB in different cancer types and its potential as therapeutic targets are lacking. The present study aimed to explore the general role of FN‑EDB in multiple types of cancer and to integrate the knowledge of cell biology, molecular biology and immunology, so as to give a comprehensive understanding of the role of FN‑EDB in TME. Furthermore, by focusing on the use of FN‑EDB in clinical diagnosis and treatment, the potential of targeting FN‑EDB as a diagnostic and therapeutic target was evaluated and the progress in clinical trials of these drugs was discussed. By searching web sites such as PubMed and web of science, various high‑quality studies including RNA sequencing, drug experiments, cell experiments, animal models, clinical randomized controlled experiments and large‑scale cohort studies were collected, with sufficient evidence to support a comprehensive evaluation of the function and potential application of FN‑EDB. The present study revealed the general role of FN‑EDB in multiple types of cancer and evaluated its potential as a diagnostic and therapeutic target. It also provided a basis for future development of more effective and precise cancer therapies.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of General Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uyghur Autonomous Region 830000, P.R. China
| | - Tao Chen
- Department of Vascular Surgery, Jining Medical College, Jining, Shandong 272000, P.R. China
| | - Yawen Pan
- Department of Geriatric Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uyghur Autonomous Region 830000, P.R. China
| | - Jing Liu
- Department of General Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uyghur Autonomous Region 830000, P.R. China
| |
Collapse
|
4
|
Tang Q, Li L, Ge J, Wang D, Qu H, Wu J, Wang Q, Peng Z, Mo Y, Wang Y, Fan C, Yan Q, Chen P, Huang H, Guo W, Shi L, Zeng Z, Xiong W. m 6A modification-dependent upregulation of WNT2 facilitates M2-like macrophage polarization and perpetuates malignant progression of nasopharyngeal carcinoma. Oncogene 2025:10.1038/s41388-025-03452-7. [PMID: 40419792 DOI: 10.1038/s41388-025-03452-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/28/2025] [Accepted: 05/15/2025] [Indexed: 05/28/2025]
Abstract
The development and progression of nasopharyngeal carcinoma (NPC) involves intricate interactions between tumor cells and other surrounding cells in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) play pivotal roles in the progression of NPC, but their interactions remain largely unexplored. In this study, we revealed that NPC promoted M2-like polarization of TAMs through enhanced synthesis and secretion of WNT2. These M2-type macrophages, in turn, significantly boosted the proliferation and metastasis of NPC. This vicious cycle perpetuated the malignant progression of NPC. Mechanistically, elevated m6A modification of WNT2 in NPC stabilized its mRNA and facilitated its protein expression, which is coordinately regulated by the m6A "eraser" ALKBH5 and the "reader" YTHDF1. NPC promoted M2-like polarization of macrophages by activating the FZD2/β-catenin signaling axis through paracrine WNT2. Furthermore, elevated WNT2 can also trigger the WNT/β-catenin signaling pathway in NPC cells through autocrine signaling, synergically contributing to NPC development. The research reveals that WNT2 is upregulated in an m6A modification-dependent manner and promotes M2-like macrophages polarization of TAMs and malignant progression of NPC. This discovery provides novel potential molecular markers and therapeutic targets for the diagnosis and treatment of NPC.
Collapse
Affiliation(s)
- Qiling Tang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
| | - Lvyuan Li
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
| | - Junshang Ge
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Dan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Hongke Qu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Jie Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, China
| | - Qian Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Zhouying Peng
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yumin Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunmei Fan
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Qijia Yan
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pan Chen
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - He Huang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
| | - Wenjia Guo
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, China
| | - Lei Shi
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China.
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China.
- FuRong Laboratory, Changsha, Hunan, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China.
- FuRong Laboratory, Changsha, Hunan, China.
| |
Collapse
|
5
|
Jin M, Lou Y, Wang X, Lv J, Wu Y, Huang G. Hyperoside suppresses NSCLC progression by inducing ATG13-mediated autophagy and apoptosis. Cell Immunol 2025; 411-412:104947. [PMID: 40250077 DOI: 10.1016/j.cellimm.2025.104947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Lung cancer is a leading cause for cancer-related mortality across the globe. In the last decade, significant advancements have been made in the research of non-small cell lung cancer (NSCLC). However, new biotherapeutic drugs urgently need to be developed. This study investigated the regulating effect of hyperoside on NSCLC progression. METHODS The colony formation assay and Cell Counting Kit-8 were used to detect cell proliferation. The Transwell assay was used to monitor cell migration. NSCLC growth in vivo was examined using a subcutaneous xenograft model. Proteomics, immunohistochemistry, and immunofluorescence analyses were used to detect anticancer regulatory mechanisms. RESULTS The results showed that hyperoside treatment inhibited cell migration, proliferation, and tumor growth in NSCLC in vivo and in vitro. Also, hyperoside treatment promoted apoptosis and cell cycle S-phase arrest. Proteomics, immunohistochemistry, and immunofluorescence detection also showed that hyperoside treatment promoted autophagy-related protein 13 (ATG13)-mediated autophagy, which further increased NSCLC apoptosis. CONCLUSION In summary, the findings illustrated that hyperoside treatment suppressed NSCLC progression by promotingATG13 expression and enhancing autophagy activation, finally promoting autophagy and apoptosis.
Collapse
Affiliation(s)
- Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yuqing Lou
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Xiaoshuo Wang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jia Lv
- Department of Obstetrics and Gynecology, Shanghai Fourth People's hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - Yue Wu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China..
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China..
| |
Collapse
|
6
|
Chalepaki AM, Gkoris M, Chondrou I, Kourti M, Georgakopoulos-Soares I, Zaravinos A. A multi-omics analysis of effector and resting treg cells in pan-cancer. Comput Biol Med 2025; 189:110021. [PMID: 40088713 DOI: 10.1016/j.compbiomed.2025.110021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Regulatory T cells (Tregs) are critical for maintaining the stability of the immune system and facilitating tumor escape through various mechanisms. Resting T cells are involved in cell-mediated immunity and remain in a resting state until stimulated, while effector T cells promote immune responses. Here, we investigated the roles of two gene signatures, one for resting Tregs (FOXP3 and IL2RA) and another for effector Tregs (FOXP3, CTLA-4, CCR8 and TNFRSF9) in pan-cancer. Using data from The Cancer Genome Atlas (TCGA), The Cancer Proteome Atlas (TCPA) and Gene Expression Omnibus (GEO), we focused on the expression profile of the two signatures, the existence of single nucleotide variants (SNVs) and copy number variants (CNVs), methylation, infiltration of immune cells in the tumor and sensitivity to different drugs. Our analysis revealed that both signatures are differentially expressed across different cancer types, and correlate with patient survival. Furthermore, both types of Tregs influence important pathways in cancer development and progression, like apoptosis, epithelial-to-mesenchymal transition (EMT) and the DNA damage pathway. Moreover, a positive correlation was highlighted between the expression of gene markers in both resting and effector Tregs and immune cell infiltration in adrenocortical carcinoma, while mutations in both signatures correlated with enrichment of specific immune cells, mainly in skin melanoma and endometrial cancer. In addition, we reveal the existence of widespread CNVs and hypomethylation affecting both Treg signatures in most cancer types. Last, we identified a few correlations between the expression of CCR8 and TNFRSF9 and sensitivity to several drugs, including COL-3, Chlorambucil and GSK1070916, in pan-cancer. Overall, these findings highlight new evidence that both Treg signatures are crucial regulators of cancer progression, providing potential clinical outcomes for cancer therapy.
Collapse
Affiliation(s)
- Anna-Maria Chalepaki
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Marios Gkoris
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Irene Chondrou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| |
Collapse
|
7
|
Lai PH, Tyrer JP, Pharoah P, Gayther SA, Jones MR, Peng PC. Characterizing somatic mutations in ovarian cancer germline risk regions. Commun Biol 2025; 8:676. [PMID: 40301634 PMCID: PMC12041368 DOI: 10.1038/s42003-025-08072-1] [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: 08/29/2024] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
Epithelial ovarian cancer (EOC) genetics research has been focused on germline or somatic mutations independently. Emerging evidence suggests that the somatic mutational landscape can be shaped by the germline genetic background. In this study, we aim to unravel the role of somatic alterations within EOC germline susceptibility regions by incorporating functional annotations. We investigate somatic events, including mutational signatures, point mutations, copy number alterations, and transcription factor binding disruptions, within 33 EOC germline susceptibility regions. Our analysis identifies significant associations between candidate germline susceptibility genes and somatic mutational signatures known to be key risk factors for EOC, such as mismatch repair deficiency, age-related mutagenesis, and homologous recombination deficiency. In addition, we find somatic point mutations and copy number alterations are significantly enriched in histotype-specific active enhancers and promoters within EOC risk loci. Furthermore, we examine the impact of germline variants and somatic mutations on transcription factor binding sites, identifying cancer developmental transcription factor motifs frequently affected by both types of mutations. Overall, our study highlights the importance of integrating germline and somatic mutations with regulatory and epigenomic data to gain insights into the genetic basis of EOC.
Collapse
Affiliation(s)
- Ping-Hung Lai
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West Hollywood, CA, USA
| | - Jonathan P Tyrer
- CR-UK Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Paul Pharoah
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West Hollywood, CA, USA
| | - Simon A Gayther
- Center for Inherited Oncogenesis, Department of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Michelle R Jones
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Pei-Chen Peng
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West Hollywood, CA, USA.
| |
Collapse
|
8
|
Varlamova EG. Roles of selenium-containing glutathione peroxidases and thioredoxin reductases in the regulation of processes associated with glioblastoma progression. Arch Biochem Biophys 2025; 766:110344. [PMID: 39956249 DOI: 10.1016/j.abb.2025.110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Glioblastoma remains the most common and aggressive primary tumor of the central nervous system in adults. Current treatment options include standard surgical resection combined with radiation/chemotherapy, but such protocol most likely only delays the inevitable. Therefore, the problem of finding therapeutic targets to prevent the occurrence and development of this severe oncological disease is currently acute. It is known that the functions of selenoproteins in the regulation of carcinogenesis processes are not unambiguous. Either they exhibit cytotoxic activity on cancer cells, or cytoprotective. A special place in the progression of oncological diseases of various etiologies is occupied by proteins of the thioredoxin and glutathione systems. These are two cellular antioxidant systems that regulate redox homeostasis, counteracting the increased production of reactive oxygen species in cells. The review reflects the latest data on the role of key enzymes of these redox systems in the regulation of processes associated with the progression of glioblastoma. A thorough consideration of these issues will expand fundamental knowledge about the functions of selenium-containing thioredoxin reductases and glutathione peroxidases in the therapy of glioblastomas and provide an understanding of the prospects for the treatment of this aggressive oncological disease.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", St. Institutskaya 3, Pushchino, 142290, Russia.
| |
Collapse
|
9
|
Hu C, Zhang JQ, Zhuang XY, Zhao ZD, Qian XJ, Chen Y. The hidden potential of solasonine: Targeting non-small cell lung cancer (NSCLC) metastasis through GATM and Smad2 pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156455. [PMID: 39922150 DOI: 10.1016/j.phymed.2025.156455] [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: 10/26/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/10/2025]
Abstract
PURPOSE To study the pharmacological effects of solasonine (Sol) in inhibiting non-small cell lung cancer (NSCLC) metastasis. METHODS The pharmacological effects of solasonine were examined in vitro by continuous induction of NSCLC cells by no treatment, or TGF-β, or creatine. Cell migration and invasion were assessed via wound healing and transwell assays, and the levels of epithelial-mesenchymal transition (EMT) markers were determined using immunofluorescence and Western Blot. Solasonine's ability to bind to TGF-β and creatine transferases and isoenzymes was assessed by TCGA, KMPLOT database, cellular thermal shift assay (CETSA), and molecular docking assay. In vivo, two types of NSCLC cells were injected into mice using the tail vein. Solasonine's effect on tumor metastasis was determined using in vivo fluorescent probes, hematoxylin-eosin staining assay, and immunofluorescence. RESULTS Unlike traditional therapies targeting TGF-β receptors, Sol was shown to inhibit NSCLC metastasis through a novel mechanism independent of TGF-β signaling. Creatine was found to promote NSCLC metastasis in vitro by enhancing cell migration, invasion, and EMT-related protein expression, and in vivo by increasing metastasis to the liver. These effects were mediated by the activation of Smad2 phosphorylation and EMT independently of TGF-β. Sol effectively counteracted these effects by targeting guanidinoacetate methyltransferase (GATM) with high binding affinity, thereby suppressing the TTK-mediated Smad2 pathway and significantly reducing metastasis to the lungs and liver, as confirmed by HE staining and fluorescence imaging. CONCLUSIONS This study identifies solasonine as a novel inhibitor of NSCLC metastasis, acting through the GATM and Smad2 pathways to counter creatine-induced metastasis. These findings underscore the potential of Sol as a therapeutic candidate for metastatic NSCLC.
Collapse
Affiliation(s)
- Cheng Hu
- Science and Technology Experiment Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jia-Qi Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Xiao-Yu Zhuang
- Science and Technology Experiment Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhen-Duo Zhao
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Xiao-Jing Qian
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Ying Chen
- Nanxiang Branch of Ruijin Hospital, Shanghai 201802, China.
| |
Collapse
|
10
|
Zhang D, Heng Y, Jin QY, Tang D, Zhu XK, Lu LM, Wu CP, Tao L. Prognostic significance of cytotoxic-T-lymphocytes to immunosuppressive lymphocytes ratio (CIL) in laryngeal squamous cell carcinoma. Cancer Immunol Immunother 2025; 74:157. [PMID: 40126590 PMCID: PMC11933541 DOI: 10.1007/s00262-025-04008-0] [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: 11/06/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
Immunoscore (IS), based on CD3/CD8, has been proposed to characterize the immune landscape of the tumor immune microenvironment and has demonstrated an association with the prognosis of laryngeal squamous cell carcinoma (LSCC). However, traditional IS does not include immunosuppressive cells. The purpose of this study is to evaluate the prognostic performance of cytotoxic-T-lymphocytes to immunosuppressive cells ratio (CIL) in laryngeal squamous cell carcinoma (LSCC) patients. Two cohorts were included in this study: The training cohort (N = 75) consisted of tumor tissue microarrays from LSCC patients in our department, and the validation cohort (N = 116) utilized bulk RNA-seq data from the TCGA database. Patients with high IS or CIL showed significantly prolonged overall survival and disease-free survival in both cohorts. Upon analyzing the relative contribution of each parameter, it was found that CIL exhibited the highest significance among the factors examined. It emerged as the strongest predictor of overall survival, emphasizing its crucial influence in determining the outcomes. The prognostic ability of IS-TCGA was similar to the original IS. Additionally, high CILM2-TCGA was associated with prolonged survival of patients with LSCC in the TCGA dataset. CIL, which is easier to construct than IS, proves to be reliable in predicting survival outcomes for patients with LSCC.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Eye & ENT Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Yu Heng
- Department of Eye & ENT Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Qiu-Yan Jin
- Department of Eye & ENT Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Di Tang
- Department of Eye & ENT Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Xiao-Ke Zhu
- Department of Eye & ENT Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Li-Ming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, People's Republic of China.
| | - Chun-Ping Wu
- Department of Eye & ENT Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China.
| | - Lei Tao
- Department of Eye & ENT Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China.
| |
Collapse
|
11
|
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
|
12
|
Ge S, Cen J, Liu X, Hong Y, Tang Y, Yu Y, Li H, Xie T, Wang C, Cai M, Qiu Y, Zeng X, Peng T, Li Q, Li Q, Wu X, Song XL, Zhao SC. TGFβ-activated Asporin interacts with STMN1 to promote prostate cancer docetaxel chemoresistance and metastasis by upregulating the Wnt/β-catenin signaling pathway. Drug Resist Updat 2025; 81:101227. [PMID: 40073743 DOI: 10.1016/j.drup.2025.101227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
AIMS Prostate cancer (PCa) remains a significant challenge in oncology due to high rates of drug resistance following standard treatment with docetaxel-based chemotherapy. Asporin (ASPN) has been regarded as an oncogene and its upregulation is closely associated with malignant behavior and poor prognosis in multiple cancers. Studies indicated that abnormal activation of the Wnt/β-catenin signaling pathway is prevalent in PCa. This study investigated the important role of ASPN in regulating Wnt/β-catenin signaling pathway in docetaxel resistance and metastasis of PCa. METHODS The impacts of ASPN on the docetaxel chemoresistance and metastasis of PCa cells were investigated in vitro and in vivo assays. Lastly, the underlying mechanism of ASPN was revealed by Western blot, protein immunocoprecipitation, Immunofluorescence, Immunohistochemical staining, liquid chromatography-mass spectrometry, and rescue experiments. RESULTS In present study, we reported that ASPN is highly expressed in PCa cells and tissues. Functional and molecular analyses showed that ASPN is activated by TGFβ and interacts with STMN1. ASPN increases the expression of β-catenin and promotes its nuclear accumulation by mediating the activation of the Wnt/β-catenin signaling pathway, thereby enhancing the stemness and epithelial-mesenchymal transition (EMT) of PCa cells, ultimately facilitating the docetaxel resistance and metastasis of PCa cells. CONCLUSIONS Our findings identify ASPN as a novel upstream regulatory factor of Wnt/β-catenin signaling pathway, suggesting that targeting the ASPN/STMN1/β-catenin axis could be a promising strategy for PCa intervention.
Collapse
Affiliation(s)
- Shengdong Ge
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China; Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Jinpeng Cen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Xiaofeng Liu
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Yaying Hong
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361101, PR China
| | - Yuting Tang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Yuzhong Yu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Haolin Li
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, PR China
| | - Tao Xie
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Chong Wang
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Maoping Cai
- Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, PR China
| | - Yang Qiu
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Xianzi Zeng
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Tianming Peng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Qu Li
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Qianyi Li
- First Clinical Medical College, Southern Medical University, Guangzhou, China Contact Information, PR China
| | - Xingcheng Wu
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100720, PR China.
| | - Xian-Lu Song
- Department of Radiotherapy, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510095, PR China.
| | - Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China; Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China.
| |
Collapse
|
13
|
Xu Y, Xie K, Li L, Li Z, Lu Q, Feng J. FOXN3 Downregulation in Colorectal Cancer Enhances Tumor Cell Stemness by Promoting EP300-Mediated Epigenetic Upregulation of SOX12. Mol Carcinog 2025; 64:410-424. [PMID: 39607349 DOI: 10.1002/mc.23852] [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: 06/12/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024]
Abstract
Cancer stemness plays a crucial role in promoting the progression of colorectal cancer (CRC). Forkhead box N3 (FOXN3) is a tumor suppressor protein. Herein, we investigated the role of FOXN3 in the regulation of CRC cell stemness. Cell viability, proliferation, migration, and invasion were assessed utilizing cell counting kit-8 assay, 5-ethynyl-20-deoxyuridine assay, and Transwell assay, respectively. Cell-sphere formation was assessed using a sphere-forming assay. The enrichment of H3K27ac modifications at the SRY-related HMG-box 12 (SOX12) promoter, interactions among FOXN3, SOX12, and E1A binding protein p300 (EP300) were analyzed using chromatin immunoprecipitation or dual luciferase reporter assays. We found that FOXN3 overexpression inhibited CRC cell proliferation, migration, invasion, stemness, and tumor formation in mice by inactivating the Wnt/β-catenin signaling, while these effects of FOXN3 overexpression were reversed by the overexpression of SOX12. Mechanistically, EP300 increased SOX12 expression in CRC cells by promoting H3K27ac enrichment in the SOX12 promoter. In addition, FOXN3 transcriptionally inhibited EP300 expression in CRC cells by binding to the EP300 promoter. As expected, EP300 overexpression weakened the inhibitory effect of FOXN3 overexpression on CRC cell stemness. Collectively, FOXN3 upregulation inhibited CRC cell stemness by suppressing EP300-mediated epigenetic upregulation of SOX12.
Collapse
Affiliation(s)
- Yanjie Xu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ke Xie
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ling Li
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhong Li
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qicheng Lu
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jin Feng
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
14
|
Xu W, Xu J, Liu J, Wang N, Zhou L, Guo J. Liver Metastasis in Cancer: Molecular Mechanisms and Management. MedComm (Beijing) 2025; 6:e70119. [PMID: 40027151 PMCID: PMC11868442 DOI: 10.1002/mco2.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Liver metastasis is a leading cause of mortality from malignant tumors and significantly impairs the efficacy of therapeutic interventions. In recent years, both preclinical and clinical research have made significant progress in understanding the molecular mechanisms and therapeutic strategies of liver metastasis. Metastatic tumor cells from different primary sites undergo highly similar biological processes, ultimately achieving ectopic colonization and growth in the liver. In this review, we begin by introducing the inherent metastatic-friendly features of the liver. We then explore the panorama of liver metastasis and conclude the three continuous, yet distinct phases based on the liver's response to metastasis. This includes metastatic sensing stage, metastatic stress stage, and metastasis support stage. We discuss the intricate interactions between metastatic tumor cells and various resident and recruited cells. In addition, we emphasize the critical role of spatial remodeling of immune cells in liver metastasis. Finally, we review the recent advancements and the challenges faced in the clinical management of liver metastasis. Future precise antimetastatic treatments should fully consider individual heterogeneity and implement different targeted interventions based on stages of liver metastasis.
Collapse
Affiliation(s)
- Wenchao Xu
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jia Xu
- State Key Laboratory of Fine ChemicalsDepartment of Pharmaceutical SciencesSchool of Chemical EngineeringDalian University of TechnologyDalianChina
| | - Jianzhou Liu
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nanzhou Wang
- Department of Colorectal SurgeryState Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerGuangzhouChina
| | - Li Zhou
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junchao Guo
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
15
|
Lu J, Zhang Y, Yan C, Liu J, Qi D, Zhou Y, Wang Q, Yang J, Jiang J, Wu B, Yang M, Zhang W, Zhang X, Shi X, Zhang Y, Liu K, Liang Y, Wang C, Yang H, Gao Y, Sun Y, Ke R, Huang JH, Wu M, Wang H, Li C, Zhou S, Guo B, Wu E, Zhang G. TClC effectively suppresses the growth and metastasis of NSCLC via polypharmacology. Bioact Mater 2025; 45:567-583. [PMID: 39759535 PMCID: PMC11700266 DOI: 10.1016/j.bioactmat.2024.11.019] [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: 06/29/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 01/07/2025] Open
Abstract
Despite significant advances in targeted therapies and immunotherapies, non-small cell lung cancer (NSCLC) continues to present a global health challenge, with a modest five-year survival rate of 28 %, largely due to the emergence of treatment-resistant and metastatic tumors. In response, we synthesized a novel bioactive compound, ethyl 6-chlorocoumarin-3-carboxylyl L-theanine (TClC), which significantly inhibited NSCLC growth, epithelial mesenchymal transition (EMT), migration, and invasion in vitro and tumor growth and metastasis in vivo without inducing toxicity. TClC disrupts autocrine loops that promote tumor progression, particularly in stem-like CD133-positive NSCLC (CD133+ LC) cells, which are pivotal in tumor metastasis. Through targeted molecular assays, we identified direct binding targets of TClC, including Akt, NF-κB, β-catenin, EZH2, and PD-L1. This interaction not only suppresses the expression of oncogenic factors and cancer stem cell markers but also downregulates the expression of a multidrug resistance transporter, underscoring the compound's polypharmacological potential. These results position TClC as a promising candidate for NSCLC treatment, signaling a new era in the development of cancer therapies that directly target multiple critical cancer pathways.
Collapse
Affiliation(s)
- Jing Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
| | - Ying Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
- Shandong Yingdong Yinghao Biotechnology Inc., Yantai, Shandong, 264670, China
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, 58105, USA
| | - Chunyan Yan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
- Department of Pharmacy, Yantai Yuhuangding Hospital, Yantai, Shandong, 264000, China
| | - Jingwen Liu
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, USA
| | - Dan Qi
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX, 76502, USA
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX, 76502, USA
| | - Yue Zhou
- Department of Statistics, North Dakota State University, Fargo, ND, 58102, USA
| | - Qinwen Wang
- The Center of Non-Traumatic Treatment and Diagnosis of Tumor, Binzhou Medical College affiliated The PLA 107 Hospital, Yantai, Shandong, 264002, China
- Outpatient Department, No. 26 Rest Center for Retired Cadres, Haidian district, Beijing, 100036, China
| | - Juechen Yang
- Department of Biomedical Informatics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jing Jiang
- RemeGen, Ltd, Yantai, 264000, Shandong, China
- Department of Pharmacology, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Benhao Wu
- Shandong Yingdong Yinghao Biotechnology Inc., Yantai, Shandong, 264670, China
| | - Meiling Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
- Shandong Yingdong Yinghao Biotechnology Inc., Yantai, Shandong, 264670, China
| | - Weiwei Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
- Shandong Yingdong Yinghao Biotechnology Inc., Yantai, Shandong, 264670, China
| | - Xin Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
- Shandong Yingdong Yinghao Biotechnology Inc., Yantai, Shandong, 264670, China
| | - Xiaoyu Shi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
- Shandong Yingdong Yinghao Biotechnology Inc., Yantai, Shandong, 264670, China
| | - Yan Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
- Shandong Yingdong Yinghao Biotechnology Inc., Yantai, Shandong, 264670, China
| | - Kun Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
| | - Yongcai Liang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
| | - Chaoyang Wang
- Department of Thoracic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, 264000, China
| | - Hanyu Yang
- Shiyao Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., LTD., State Key Laboratory of New Pharmaceutical Preparations and Excipients, Shijiazhuang, 050035, China
| | - Yuqing Gao
- Shiyao Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., LTD., State Key Laboratory of New Pharmaceutical Preparations and Excipients, Shijiazhuang, 050035, China
| | - Yuping Sun
- Phase I Clinical Trial Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250013, China
| | - Ronghu Ke
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX, 76502, USA
| | - Jason H. Huang
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX, 76502, USA
- College of Medicine, Texas A&M University, College Station, TX, 77843, USA
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX, 76502, USA
| | - Min Wu
- Drug Discovery Center, Wenzhou Institute University of Chinese Academy of Sciences, Wenzhou, 325001, China
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 646000, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
| | - Chunlei Li
- Shiyao Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., LTD., State Key Laboratory of New Pharmaceutical Preparations and Excipients, Shijiazhuang, 050035, China
| | - Shuang Zhou
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, USA
| | - Bin Guo
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, USA
| | - Erxi Wu
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX, 76502, USA
- College of Medicine, Texas A&M University, College Station, TX, 77843, USA
- College of Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX, 77843, USA
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX, 76502, USA
- LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, the University of Texas at Austin, Austin, TX, 78712, USA
| | - Guoying Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, China
| |
Collapse
|
16
|
Feng JN, Chen PR, Yuan SF, Dai Q, Zheng WE, Lin HL. Pingxiao pian attenuate invasiveness and proliferation of lung adenocarcinoma through regulating miR-29b-3p/TGF-β1/Smad/EMT pathway. Discov Oncol 2025; 16:232. [PMID: 39992570 PMCID: PMC11850683 DOI: 10.1007/s12672-025-01959-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
INTRODUCTION Lung cancer is a highly prevalent and deadly disease worldwide, causing over 1.2 million deaths each year. Pingxiao Pian (PXP) tablets, a Chinese traditional medicine, have been widely applied in the treatment of lung cancer. However, the mechanism underlying therapeutic effects of PXP tablets remains undisclosed. METHODS A549 human LUAD cell line was utilized for in vitro experiments. Transfection of miR-29b mimic was performed using Lipofectamine 3000. PXP was purchased and dissolved into PBS and drinking water after carefully removing the outer coating. Dual-luciferase reporter assay was conducted to assess the regulatory effect of miR-29b on TGF-β1. The protein levels of epithelial-mesenchymal transition (EMT) markers and activation of TGF-β1 pathway were characterized using immunoblotting analysis. RESULTS PXP reduced the invasiveness and proliferation of LUAD cells by increasing miR-29b-3p expression in vitro. Overexpression of miR-29b-3p resulted in decreased cell proliferation and invasiveness, while silencing of miR-29b-3p in the A549 cells displayed the opposite effect. Moreover, PXP treatment reversed the increased cell proliferating rate triggered by miR-29b-3p silencing. Additionally, PXP was found to hamper EMT occurrence in A549 cells by regulating miR-29b-3p and reduce expression of N-cad and vimentin. Overexpression of miR-29b-3p blocked the phosphorylation of Smad2/3 and decreased TGF-β1 expression. Luciferase assay results indicated that miR-29b-3p directly regulated TGF-β1 expression. In vivo tumor formation experiments confirmed the tumor-reducing effects of PXP and the role of miR-29b in tumor progression. PXP treatment decreased tumor size and weight via regulating miR-29b-3p. CONCLUSION Our study suggests that PXP exerts anti-tumor effects in LUAD through the regulation of miR-29b and the inhibition of EMT via the TGF-β1/Smad2/3 pathway.
Collapse
Affiliation(s)
- Jie-Ni Feng
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Pei-Rui Chen
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Shao-Fei Yuan
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Qiang Dai
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Wei-E Zheng
- Department of Chemoradiation and Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Hua-Long Lin
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China.
| |
Collapse
|
17
|
Chen Y, Shi H, Dong Y, Cui W. LncRNA MSTO2P affects the proliferation, invasion and migration of non-small cell lung cancer by regulating the Wnt/β-catenin pathway. Discov Oncol 2025; 16:150. [PMID: 39928213 PMCID: PMC11811353 DOI: 10.1007/s12672-025-01920-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025] Open
Abstract
This study aimed to investigate the regulatory effects of the long non-coding RNA (lncRNA) MISATO family member 2 (MSTO2P) on non-small cell lung cancer (NSCLC) cell viability, invasion, and migration, as well as the underlying mechanism. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to analyze the expression levels of MSTO2P. The effects of MSTO2P on cell viability, invasion, and migration were assessed using cell counting kit-8 (CCK-8), Transwell invasion, and wound healing assays in A549 and H1229 cells. A human phospho-kinase array kit was employed to identify potential phosphorylated kinases or signaling nodes affected by MSTO2P. The interaction between MSTO2P and β-catenin was evaluated using RNA pull-down and RNA immunoprecipitation (RIP) assays. A xenograft tumor mouse model was established to evaluate tumor growth. The results demonstrated that MSTO2P expression was elevated in NSCLC tissues and cells compared to normal counterparts. Silencing MSTO2P inhibited the viability, invasion, and migration of A549 and H1229 cells. MSTO2P interacted with β-catenin, thereby activating the Wnt/β-catenin pathway. Overexpression of MSTO2P or β-catenin promoted the viability, invasion, and migration of A549 and H1229 cells, effects that were reversed by treatment with XAV-939. In vivo studies showed that silencing MSTO2P suppressed tumor growth. In conclusion, MSTO2P promoted NSCLC cell viability, invasion, and migration by regulating the Wnt/β-catenin pathway, suggesting that MSTO2P may be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Yunfeng Chen
- Department of Respiratory and Critial Care Medicine, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou City, 221116, Jiangsu Province, China
- Department of Respiratory and Critial Care Medicine, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Hai Shi
- Department of Respiratory and Critial Care Medicine, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou City, 221116, Jiangsu Province, China
- Department of Respiratory and Critial Care Medicine, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Yuan Dong
- Department of Respiratory and Critial Care Medicine, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou City, 221116, Jiangsu Province, China.
- Department of Respiratory and Critial Care Medicine, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China.
| | - Wenjie Cui
- Department of Respiratory and Critial Care Medicine, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou City, 221116, Jiangsu Province, China.
- Department of Respiratory and Critial Care Medicine, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China.
| |
Collapse
|
18
|
Shi M, Zhang R, Lyu H, Xiao S, Guo D, Zhang Q, Chen XZ, Tang J, Zhou C. Long non-coding RNAs: Emerging regulators of invasion and metastasis in pancreatic cancer. J Adv Res 2025:S2090-1232(25)00073-6. [PMID: 39933650 DOI: 10.1016/j.jare.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND The invasion and metastasis of pancreatic cancer (PC) are key factors contributing to disease progression and poor prognosis. This process is primarily driven by EMT, which has been the focus of recent studies highlighting the role of long non-coding RNAs (lncRNAs) as crucial regulators of EMT. However, the mechanisms by which lncRNAs influence invasive metastasis are multifaceted, extending beyond EMT regulation alone. AIM OF REVIEW This review primarily aims to characterize lncRNAs affecting invasion and metastasis in pancreatic cancer. We summarize the regulatory roles of lncRNAs across multiple molecular pathways and highlight their translational potential, considering the implications for clinical applications in diagnostics and therapeutics. KEY SCIENTIFIC CONCEPTS OF REVIEW The review focuses on three principal scientific themes. First, we primarily summarize lncRNAs orchestrate various signaling pathways, such as TGF-β/Smad, Wnt/β-catenin, and Notch, to regulate molecular changes associated with EMT, thereby enhancing cellular motility and invasivenes. Second, we summarize the effects of lncRNAs on autophagy and ferroptosis and discuss the role of exosomal lncRNAs in the tumor microenvironment to regulate the behavior of neighboring cells and promote cancer cell invasion. Third, we emphasize the effects of RNA modifications (such as m6A and m5C methylation) on stabilizing lncRNAs and enhancing their capacity to mediate invasive metastasis in PC. Lastly, we discuss the translational potential of these findings, emphasizing the inherent challenges in using lncRNAs as clinical biomarkers and therapeutic targets, while proposing prospective research strategies.
Collapse
Affiliation(s)
- Mengmeng Shi
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Qi Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China.
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China.
| |
Collapse
|
19
|
Li L, Yang Y, Peng M, Wang B, Zhu L, Chen C, Fan Z, Duan X, Xue R, Lv X, Cheng M, Zhao J. Molecular Subtyping and Therapeutic Targeting of IFNG-Driven Immunogenic Cell Death in Lung Adenocarcinoma. Cancer Med 2025; 14:e70678. [PMID: 39945555 PMCID: PMC11822994 DOI: 10.1002/cam4.70678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/07/2024] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) can be triggered by various therapies to induce anti-tumor immune responses, significantly enhancing treatment effectiveness, and is widely utilized in tumor immunotherapy. METHODS LUAD data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) validated ICD-related molecular subtypes via consensus clustering. Clinical features, ICD genes, driver genes, mutations, tumor microenvironment, immune checkpoints, and drug sensitivity were compared. RT-qPCR, Western blot, immunofluorescence, ELISA, flow cytometry, and tube formation assays validated findings. RESULTS Differential expression of 33 ICD genes was observed between tumor and normal tissues. These genes were clustered into two groups via consensus clustering and validated with GEO data. Prognostic analysis indicated superior outcomes in cluster 2 across TCGA and GEO cohorts. Significant disparities in clinicopathological characteristics like stage, gender, and age were noted between subtypes. Cluster 2 exhibited heightened expression of ICD-related genes, driver genes, immune checkpoints, and immune cells. Cluster 2 also showed increased sensitivity to chemotherapy drugs. IFNG overexpression in A549 and H1299 cells induced CRT exposure, HMGB1 release, and ATP secretion, thereby promoting dendritic cell maturation and enhancing CD8+ T cell function. Additionally, IFNG boosted tumor angiogenesis via HMGB1 pathways, which could be mitigated by HMGB1 inhibition. CONCLUSION Identification of novel ICD-related molecular subtypes holds promise for guiding personalized therapies, assessing prognosis, and predicting immunotherapy efficacy in LUAD. IFNG emerges as a potential prognostic biomarker and therapeutic target, influencing both the tumor microenvironment and angiogenesis. These findings offer new insights into therapeutic strategies targeting IFNG-mediated pathways in LUAD.
Collapse
Affiliation(s)
- Lifeng Li
- National Engineering Laboratory for Internet Medical Systems and Applications, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
- Cancer Center, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
- Medical SchoolHuanghe Science and Technology UniversityZhengzhouHenanChina
- Fuwai Central China Cardiovascular HospitalInternet Medical and System Applications of National Engineering LaboratoryZhengzhouHenanChina
| | - Yaqi Yang
- National Engineering Laboratory for Internet Medical Systems and Applications, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
- Cancer Center, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
| | - Mengle Peng
- Department of Clinical LaboratoryHenan No. 3 Provincial People's HospitalZhengzhouHenanChina
- College of Public HealthZhengzhou UniversityZhengzhouChina
| | - Biyue Wang
- Department of NephrologySeventh People's Hospital of ZhengzhouZhengzhouHenanChina
| | - Lili Zhu
- National Engineering Laboratory for Internet Medical Systems and Applications, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
| | - Chengxin Chen
- National Engineering Laboratory for Internet Medical Systems and Applications, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
| | - Zhirui Fan
- Department of Integrated Traditional and Western Medicine, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
| | - Xiaoran Duan
- National Engineering Laboratory for Internet Medical Systems and Applications, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
| | - Ruyue Xue
- National Engineering Laboratory for Internet Medical Systems and Applications, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
| | - Xuefeng Lv
- Department of Clinical LaboratoryThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Ming Cheng
- Department of Medical Information, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
| | - Jie Zhao
- National Engineering Laboratory for Internet Medical Systems and Applications, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
20
|
Chen J, Zhu C, He Y, Huang L, Wang W, Huang S. FOXP3 as a prognostic marker and therapeutic target in immunogenic cell death modulation for clear cell renal cell carcinoma. Discov Oncol 2025; 16:102. [PMID: 39883234 PMCID: PMC11782763 DOI: 10.1007/s12672-025-01831-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) remains a challenging cancer type due to its resistance to standard treatments. Immunogenic cell death (ICD) has the potential to activate anti-tumor immunity, presenting a promising avenue for ccRCC therapies. METHODS We analyzed data from GSE29609, TCGA-KIRC, and GSE159115 to identify ICD-related prognostic genes in ccRCC. By applying consensus clustering, patients were categorized based on ICD modification patterns, and an ICD signature (ICDS) model was developed using a PCA approach. Functional studies were conducted with FOXP3 knockdown in ccRCC cell lines to explore its impact on cell behavior. RESULTS Eleven ICD-related genes were identified as key prognostic indicators in ccRCC, with high ICDS linked to worse survival outcomes. High ICDS also correlated with increased levels of immune-suppressive cells within the tumor microenvironment. FOXP3 was highlighted as a critical gene influencing ICD, where its knockdown significantly reduced ccRCC cell proliferation and migration, underscoring its role in tumor progression. CONCLUSIONS This study establishes FOXP3 as a pivotal factor in ICD regulation and ccRCC progression. Targeting FOXP3 and other ICD pathways could enhance treatment efficacy in ccRCC, providing a foundation for ICD-based therapeutic strategies. Evaluating ICD patterns in ccRCC may guide patient-specific interventions, paving the way for improved management of this aggressive cancer.
Collapse
Affiliation(s)
- Jian Chen
- Medical Department, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Cheng Zhu
- Medical Department, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Yan He
- Medical Department, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Liping Huang
- Medical Department, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Weizhuo Wang
- Center for Reproductive Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Shuaishuai Huang
- Department of Laboratory, Ningbo Yinzhou No.2 Hospital, No.998 Qianhe Road, Yinzhou Distrinct, Ningbo, 315100, China.
| |
Collapse
|
21
|
Zhang ZH, Yan HX, Liu MD, Niu FW, Yao K, Feng SY, Li X, Chen YH, Xie DD. Chronic NaAsO 2 exposure promotes migration and invasion of prostate cancer cells by Akt/GSK-3β/β-catenin/TCF4 axis-mediated epithelial-mesenchymal transition. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117741. [PMID: 39818140 DOI: 10.1016/j.ecoenv.2025.117741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Inorganic arsenic is a Class I human Carcinogen. However, the role of chronic inorganic arsenic exposure on prostate cancer metastasis still unclear. This study aimed to investigate the effects and mechanism of chronic NaAsO2 exposure on migration and invasion of prostate cancer cells. DU145 and PC-3 cells were exposed to NaAsO2 (2 μM) for 25 generations. Wound healing and Transwell assays showed that chronic NaAsO2 exposure promoted migration and invasion of DU145 and PC-3 cells. In addition, chronic NaAsO2 exposure induced epithelial-mesenchymal transition (EMT) of DU145 cells by promoting β-catenin/TCF4 transcriptional activity. Mechanically, NaAsO2 promoted GSK-3β inactivation in the "disruption complex" through Akt- mediated phosphorylation at serine 9, and then inhibited the phosphorylation and ubiquitination degradation of β-catenin, which led to its nuclear translocation. Ly294002, a selective phosphatidylinositol 3-kinase (PI3K)/Akt inhibitor, suppressed the β-catenin/TCF4 complex activation and EMT through blocking Akt-mediated GSK-3β inactivation in the "disruption complex" in chronic NaAsO2 exposed DU145 and PC-3 cells. Moreover, Ly294002 alleviated chronic NaAsO2-induced migration and invasion in DU145 and PC-3 cells. These findings provide evidence that chronic arsenic exposure promotes migration and invasion of prostate cancer cells via an EMT mechanism driven by the AKT/GSK-3β/β-catenin/TCF4 signaling axis. Akt is expected to be a potential therapeutic target for chronic arsenic exposure-mediated prostate cancer metastasis.
Collapse
Affiliation(s)
- Zhi-Hui Zhang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Hai-Xin Yan
- Department of Urology, Chaohu Hospital of Anhui Medical University, Chaohu 238000, China
| | - Ming-Dong Liu
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Feng-Wen Niu
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Kai Yao
- Department of Urology, Chaohu Hospital of Anhui Medical University, Chaohu 238000, China
| | - Shi-Yao Feng
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Xi Li
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yuan-Hua Chen
- Department of Histology and Embryology, Anhui Medical University, Hefei 230032, China
| | - Dong-Dong Xie
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Department of Urology, Chaohu Hospital of Anhui Medical University, Chaohu 238000, China.
| |
Collapse
|
22
|
Ni M, Wang Y, Yang J, Ma Q, Pan W, Li Y, Xu Q, Lv H, Wang Y. IL-33 aggravates extranodal NK/T cell lymphoma aggressiveness and angiogenesis by activating the Wnt/β-catenin signaling pathway. Mol Cell Biochem 2025; 480:265-278. [PMID: 38443748 DOI: 10.1007/s11010-024-04944-y] [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: 09/15/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024]
Abstract
Extranodal NK/T cell lymphoma (ENKTCL) is an extremely aggressive form of lymphoma and lacks of specific diagnostic markers. The study intended to unearth the role of interleukin-33 (IL-33) in ENKTCL. RT-qPCR was conducted to assess mRNA levels of ENKTCL tissues and cells, while western blot assay was performed for evaluating protein levels. Plate cloning experiment and transwell assay were employed to measure aggressiveness of ENKTCL. Tube formation assay was executed to determine the angiogenesis ability. Mice ENKTCL xenograft model was designed to probe the impacts of IL-33 in vivo. IL-33 and suppression of tumorigenicity 2 receptor (ST2, receptor of IL-33) were enhanced in ENKTCL. IL-33 inhibition suppressed viability, migration, and invasion of ENKTCL cells. Moreover, IL-33 knockdown restricted angiogenesis in human umbilical vein endothelial cells (HUVECs). Furthermore, Wnt/β-catenin pathway associated proteins (β-catenin, c-myc, and cyclin D1) were downregulated by loss of IL-33. However, these impacts were overturned by Wnt/β-catenin signaling agonist lithium chloride (LiCl). Additionally, IL-33 silencing exerted anti-tumor effect via Wnt/β-catenin pathway in vivo. Silencing of IL-33 inhibited ENKTCL tumorigenesis and angiogenesis by inactivating Wnt/β-catenin signaling pathway. As such, IL-33 might be a prospective treatment target for ENKTCL.
Collapse
Affiliation(s)
- Mingli Ni
- Department of Oncology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, 450099, Henan, China
- Medical Oncology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471099, Henan, China
| | - Yuhui Wang
- Day Operating Room, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Jiezhi Yang
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Qianwen Ma
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Wei Pan
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Yulin Li
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Qian Xu
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Hongqiong Lv
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Yunlong Wang
- Department of Oncology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, 450099, Henan, China.
- Henan Bioengineering Research Center, No. 81, Zhengshang Road, Zhengzhou, 450066, Henan, China.
| |
Collapse
|
23
|
Zu F, Chen C, Geng Q, Li H, Chan B, Luo G, Wu M, Ilmer M, Renz BW, Bentum-Ennin L, Gu H, Sheng W. Smad2 Cooperating with TGIF2 Contributes to EMT and Cancer Stem Cells Properties in Pancreatic Cancer via Co-Targeting SOX2. Int J Biol Sci 2025; 21:524-543. [PMID: 39781447 PMCID: PMC11705628 DOI: 10.7150/ijbs.102381] [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: 08/15/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
The underlying mechanisms between cancer stem cells (CSC) and epithelial-mesenchymal transition (EMT) in pancreatic cancer (PC) remain unclear. In this study, we identified TGIF2 as a target gene of CSC using sncRNA and machine learning. TGIF2 is closely related to the expression of SOX2, EGFR, and E-cadherin, indicating poor prognosis. Mechanistically, TGIF2 promoted the EMT phenotype and CSC properties following the activation of SOX2, Slug, CD44, and ERGF/MAPK signaling, which were rescued by SOX2 silencing. TGIF2 silencing contributes to the opposite phenotype via SOX2. Notably, Smad2 cooperates with TGIF2 to co-regulate the SOX2 promoter, which in turn promotes EMT and CSC signaling by transactivating Slug and EGFR, respectively. The transactivation of EGFR/MAPK signaling by SOX2 promotes TGIF2 nuclear translocation, forming a positive feedback loop in vitro. Moreover, the interaction of TGIF2 and SOX2 with EGFR inhibitors promoted subcutaneous tumors and liver metastasis in vivo. Thus, the TGIF2/SOX2 axis contributes to CSC, EMT, and chemoresistance, providing a promising target for PC therapy.
Collapse
Affiliation(s)
- Fuqiang Zu
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - ChuanPing Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Qilong Geng
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Haoyu Li
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Boyuan Chan
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Mengcheng Wu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Matthias Ilmer
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernhard W Renz
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lutterodt Bentum-Ennin
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Weiwei Sheng
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| |
Collapse
|
24
|
Robinson E, Rodriguez I, Argueta V, Xie Y, Lou H, Milano R, Lee HJ, Burdett L, Mishra SK, Yeager M, Mirabello L, Dean M, Orozco R. Analysis of the progression of cervical cancer in a low-and-middle-income country: From pre-malignancy to invasive disease. Tumour Virus Res 2024; 19:200299. [PMID: 39672307 PMCID: PMC11729683 DOI: 10.1016/j.tvr.2024.200299] [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: 09/12/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024] Open
Abstract
To better understand cervical cancer progression, we analyzed RNA from 262 biopsies from women referred for colposcopy. We determined the HPV type and analyzed the expression of 51 genes. HPV31 was significantly more prevalent in precancer than stage 1 cancer and invasive cancer (p < 0.0001), and HPV16 increased in invasive disease (p < 0.0001). CCNE1, MELTF, and ULBP2 were significantly increased in HPV16-positive compared to HPV31 precancers, while NECTIN2 and HLA-E expression decreased. Markers of the innate immune system, DNA repair genes, and cell cycle genes are significantly increased during cancer progression (p = 0.0001). In contrast, the TP53 and RB1 tumor suppressor gene expression is significantly decreased in cancer cells. The T cell markers CD28 and FLT3LG expression decreased in cancer while FOXP3, IDO1, and ULBP2 expression increased. There is a significantly higher survival rate in individuals with increased expression of CD28 (p = 0.0005), FOXP3 (p = 0.0002), IDO1 (p = 0.038), FLT3LG (p = 0.026), APOBEC3B (p = 0.0011), and RUNX3 (p = 0.019), and a significantly lower survival rate in individuals with increased expression of ULBP2 (p = 0.035). These results will help us elucidate the molecular factors influencing the progression of cervical precancer to cancer. Understanding the risk of progression of specific HPV types and sublineages may aid in the triage of positive patients, and better knowledge of the immune response may aid in developing and applying immunotherapies.
Collapse
Affiliation(s)
- Emma Robinson
- HLA Immunogenetics, Basic Science Program, Frederick National Laboratory for Cancer Research, Gaithersburg, MD, USA
| | - Isabel Rodriguez
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Gaithersburg, MD, USA
| | - Victor Argueta
- Hospital General San Juan de Dios, Guatemala City, Guatemala
| | - Yi Xie
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Gaithersburg, MD, USA
| | - Hong Lou
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Frederick National Laboratory for Cancer Research, Gaithersburg, MD, USA
| | - Rose Milano
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Gaithersburg, MD, USA
| | - Hyo Jung Lee
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Frederick National Laboratory for Cancer Research, Gaithersburg, MD, USA
| | - Laurie Burdett
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Frederick National Laboratory for Cancer Research, Gaithersburg, MD, USA
| | - Sambit K Mishra
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Frederick National Laboratory for Cancer Research, Gaithersburg, MD, USA
| | - Meredith Yeager
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Frederick National Laboratory for Cancer Research, Gaithersburg, MD, USA
| | - Lisa Mirabello
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Gaithersburg, MD, USA
| | - Michael Dean
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Gaithersburg, MD, USA.
| | - Roberto Orozco
- Hospital General San Juan de Dios, Guatemala City, Guatemala
| |
Collapse
|
25
|
Gaur V, Tyagi W, Das S, Ganguly S, Bhattacharyya J. CD40 agonist engineered immunosomes modulated tumor microenvironment and showed pro-immunogenic response, reduced toxicity, and tumor free survival in mice bearing glioblastoma. Biomaterials 2024; 311:122688. [PMID: 38943821 DOI: 10.1016/j.biomaterials.2024.122688] [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: 01/29/2024] [Revised: 05/29/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
CD40 agonist antibodies (αCD40) have shown promising anti-tumor response in both preclinical and early clinical studies. However, its systemic administration is associated with immune- and hepato-toxicities which hampers its clinical usage. In addition, αCD40 showed low tumor retention and induced PD-L1 expression which makes tumor microenvironment (TME) immunosuppressive. To overcome these issues, in this study, we have developed a multifunctional Immunosome where αCD40 is conjugated on the surface and RRX-001, a small molecule immunomodulator was encapsulated inside it. Immunosomes showed higher tumor accumulation till 96 h of administration and displayed sustained release of αCD40 in vivo. Immunosomes significantly delayed tumor growth and showed tumor free survival in mice bearing GL-261 glioblastoma by increasing the population of CD45+CD8+ T cells, CD45+CD20+ B cells, CD45+CD11c+ DCs and F4/80+CD86+ cells in TME. Immunosome significantly reduced the population of T-regulatory cells, M2 macrophage, and MDSCs and lowered the PD-L1 expression. Moreover, Immunosomes significantly enhanced the levels of Th1 cytokines (IFN-γ, IL-6, IL-2) over Th2 cytokines (IL-4 and IL-10) which supported anti-tumor response. Most interestingly, Immunosomes averted the in vivo toxicities associated with free αCD40 by lowering the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), IL-6, IL-1α and reduced the degree of liver damage. In addition, Immunosomes treated long-term surviving mice showed tumor specific immune memory response which prevented tumor growth upon rechallenge. Our results suggested that this novel formulation can be further explored in clinics to improve in vivo anti-tumor efficacy of αCD40 with long-lasting tumor specific immunity while reducing the associated toxicities.
Collapse
Affiliation(s)
- Vidit Gaur
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, India; Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, India
| | - Witty Tyagi
- Molecular Oncology Laboratory, National Institute of Immunology, Delhi, India
| | - Sanjeev Das
- Molecular Oncology Laboratory, National Institute of Immunology, Delhi, India
| | - Surajit Ganguly
- Department of Molecular Medicine, Jamia Hamdard University, Delhi, India
| | - Jayanta Bhattacharyya
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, India; Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, India.
| |
Collapse
|
26
|
Lin C, Wu J, Wang Z, Xiang Y. Long non-coding RNA LNC-POTEM-4 promotes HCC progression via the LNC-POTEM-4/miR-149-5p/Wnt4 signaling axis. Cell Signal 2024; 124:111412. [PMID: 39278454 DOI: 10.1016/j.cellsig.2024.111412] [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: 06/29/2024] [Revised: 08/27/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Information on the potential role of the long non-coding RNA LNC-POTEM-4 in cancer progression is limited. Our preliminary study found that LNC-POTEM-4 was overexpressed in hepatocellular carcinoma (HCC) tissues, which led us to further investigate the biological function and molecular mechanism of LNC-POTEM-4 in HCC development. LNC-POTEM-4 expression in HCC tissues was examined using transcriptome sequencing and quantitative reverse transcription PCR. The relationships between LNC-POTEM-4 and the stage and prognosis of HCC in patient data from the TCGA database were analyzed. The effects of LNC-POTEM-4 on proliferation, invasion/migration, and epithelial-mesenchymal transition marker expression in HCC cells were evaluated in vitro using gain- and loss-of-function assays, while its effects on tumor growth and metastasis were explored through animal experiments. A LNC-POTEM-4/microRNA (miR)-149-5p/Wnt4 regulatory signaling axis was identified using bioinformatics analysis, and dual luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays. Co-transfection of LNC-POTEM-4 and Wnt4 expression plasmids was employed to confirm the new signaling pathway. We found that LNC-POTEM-4 was overexpressed in HCC tissues and was linked to poor staging and prognosis. LNC-POTEM-4 promoted proliferation, invasion, migration, and the epithelial-mesenchymal transition of HCC cells in vitro. Silencing of LNC-POTEM-4 inhibited HCC growth and distant metastasis in vivo. Mechanically, LNC-POTEM-4 was found to function as a competitive endogenous RNA, upregulating Wnt4 by sponging miR-149-5p to promote HCC progression. Wnt4 overexpression may have counteracted the tumor-inhibition effect of LNC-POTEM-4 silencing. In conclusion, LNC-POTEM-4 upregulated Wnt4 to activate the Wnt signaling pathway and stimulate the malignancy tendency of HCC by sponging miR-149-5p, providing a prospective target for the detection and therapy of HCC. However, the effects of LNC-POTEM-4 on the miR-149-5p/Wnt4 signaling axis should be further studied in animal experiments.
Collapse
Affiliation(s)
- Chao Lin
- Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jiacheng Wu
- Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - Zhixuan Wang
- Intensive Care Medicine, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Yien Xiang
- Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
27
|
Zang Y, Lu Y, Yu J, Dong Q, Shi Y, Ying G, Liang Z. FOXP3 inhibits proliferation and migration by competitively inhibiting YAP1 in nasopharyngeal carcinoma. Oral Oncol 2024; 159:107066. [PMID: 39413576 DOI: 10.1016/j.oraloncology.2024.107066] [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: 06/30/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/18/2024]
Abstract
Hippo signalling is involved in the coordination of extracellular signals that control tissue homeostasis and organ size. Yes-associated protein 1 (YAP1) is regulated primarily by Hippo signalling through coactivation of transcription factors with GATA domains called TEADs. However, small-molecule orthosteric inhibitors of YAP1 are difficult to develop due to its tight binding to TEAD4 via a flat interface. Previous studies have shown that chlorpromazine (CPZ) can inhibit YAP1 expression. MTT, colony formation, wound healing, Transwell migration and Western blot assays were performed to explore how CPZ affects nasopharyngeal carcinoma (NPC) cells through FOXP3. In addition, immunofluorescence and live-cell imaging were used to detect YAP1 intracellular localization after CPZ administration. Through the HDOCK website, we predicted protein binding regions between FOXP3 and TEAD4. Western blot and co-IP experiments were used to verify the relationship between FOXP3 and YAP1. The UCSC Xena database, LinkedOmics database and KM plotter website were used to assess the prognostic value of FOXP3 in head and neck squamous cell carcinoma (HNSCC). Age, sex, pathological tumour-node-metastasis (pTMN) stage, grade, smoking status and FOXP3 expression were included in an overall survival nomogram model. Our findings revealed that FOXP3 has the ability to competitively interacts competitively with TEAD4 to inhibit YAP1 expression. By increasing FOXP3 expression, CPZ induces YAP1 nuclear export and phosphorylation, consequently suppressing NPC cell proliferation and migration. Collectively, our findings indicate that FOXP3 competitively binds TEAD4 to regulate YAP1 localization in the nucleus and cytoplasm to suppress NPC progression. Consequently, FOXP3 may be a prognostic indicator for HNSCC.
Collapse
Affiliation(s)
- Yiqing Zang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Yi Lu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Jiaxi Yu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Qiuping Dong
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, PR China
| | - Yue Shi
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, PR China
| | - Guoguang Ying
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, PR China.
| | - Zheng Liang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, PR China.
| |
Collapse
|
28
|
Bao Y, Teng S, Zhai H, Zhang Y, Xu Y, Li C, Chen Z, Ren F, Wang Y. SE-lncRNAs in Cancer: Classification, Subcellular Localisation, Function and Corresponding TFs. J Cell Mol Med 2024; 28:e70296. [PMID: 39690143 DOI: 10.1111/jcmm.70296] [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: 10/06/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
Emerging evidence highlights certain long noncoding RNAs (lncRNAs) transcribed from or interacting with super-enhancer (SE) regulatory elements. These lncRNAs, known as SE-lncRNAs, are strongly linked to cancer and regulate cancer progression through multiple interactions with downstream targets. The expression of SE-lncRNAs is controlled by various transcription factors (TFs), and dysregulation of these TFs can contribute to cancer development. In this review, we discuss the characteristics, classification and subcellular distribution of SE-lncRNAs and summarise the role of key TFs in the transcription and regulation of SE-lncRNAs. Moreover, we examine the distinct functions and potential mechanisms of SE-lncRNAs in cancer progression.
Collapse
Affiliation(s)
- Yuxin Bao
- Fourth Department of Orthopaedic Surgery, Central Hospital Affiliated To Shenyang Medical College, Shenyang, Liaoning, P. R. China
| | - Songling Teng
- Department of Hand Surgery, Central Hospital Affiliated To Shenyang Medical College, Shenyang, Liaoning, P. R. China
| | - Hanjie Zhai
- Fourth Department of Orthopaedic Surgery, Central Hospital Affiliated To Shenyang Medical College, Shenyang, Liaoning, P. R. China
| | - Yuanzhuang Zhang
- Fourth Department of Orthopaedic Surgery, Central Hospital Affiliated To Shenyang Medical College, Shenyang, Liaoning, P. R. China
| | - Yeqiu Xu
- Fourth Department of Orthopaedic Surgery, Central Hospital Affiliated To Shenyang Medical College, Shenyang, Liaoning, P. R. China
| | - Chenghao Li
- Fourth Department of Orthopaedic Surgery, Central Hospital Affiliated To Shenyang Medical College, Shenyang, Liaoning, P. R. China
| | - Zhenjun Chen
- Department of Neurosurgery, Central Hospital Affiliated To Shenyang Medical College, Shenyang, Liaoning, P. R. China
| | - Fu Ren
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, P. R. China
| | - Yong Wang
- Fourth Department of Orthopaedic Surgery, Central Hospital Affiliated To Shenyang Medical College, Shenyang, Liaoning, P. R. China
| |
Collapse
|
29
|
Mei H, Luo Q, Weng J, Hao J, Cai J, Zhou R, Bian C, Ye Y, Luo S, Wen Y. The miR-1269a/PCDHGA9/CXCR4/β-catenin pathway promotes colorectal cancer invasion and metastasis. Cell Mol Biol Lett 2024; 29:144. [PMID: 39587482 PMCID: PMC11590219 DOI: 10.1186/s11658-024-00656-9] [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: 12/18/2023] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer worldwide and the second leading cause of cancer-related death. This research focuses on investigating the impact and underlying molecular mechanisms of protocadherin gamma subfamily A, 9 (PCDHGA9) on the invasion and metastasis of CRC, aiming to identify more precise molecular markers for the diagnosis and prognosis of CRC. METHODS PCDHGA9 expression was detected using quantitative real-time quantitative polymerase chain reaction (RT-qPCR) in 63 pairs of colorectal cancer tissues. Differential gene expression from high-throughput sequencing was analyzed using ingenuity pathway analysis (IPA) to explore the biological functions of PCDHGA9 and its potential regulated genes. Bioinformatics tools were employed to explore potential upstream regulatory microRNAs of PCDHGA9. Dual-luciferase assays were performed to demonstrate the regulation between PCDHGA9 and miR-1269a. Protein mass spectrometry suggested an interaction between PCDHGA9 and HOXA1. JASPAR predicted that HOXA1 may act as a transcription factor of CXCR4. Coimmunoprecipitation, dual-luciferase assays, and nuclear-cytoplasmic fractionation experiments confirmed the molecular mechanism involving PCDHGA9, CXCR4, HOXA1, and β-catenin. Transwell, wound healing, and western blot assays were conducted to confirm the impact of PCDHGA9, miR-1269a, and CXCR4 on the invasion, metastasis, and epithelial-mesenchymal transition (EMT) functions of CRC cells in in vitro experiments. A whole-body fluorescence imaging system was used to evaluate the combined impact of miR-1269a and PCDHGA9 on the invasion and metastasis of CRC in in vivo experiments. RESULTS The expression of PCDHGA9 was found to be lower in CRC tissues compared with their corresponding adjacent tissues. Low expression of PCDHGA9 potentially correlated with worse prognosis and increased chances of invasion and metastasis in CRC. miR-1269a was highly expressed in CRC tissues and acted as a negative regulator for PCDHGA9, promoting invasion, migration, and EMT of CRC cells. PCDHGA9's interaction with HOXA1 downregulated CXCR4, a transcription factor, leading to accumulation of β-catenin and further promoting invasion, migration, and EMT of CRC cells. CONCLUSIONS PCDHGA9, acting as a tumor suppressor, is downregulated by miR-1269a. The low level of PCDHGA9 activates the Wnt/β-catenin pathway by releasing its interaction with HOXA1, promoting the expression of CXCR4, and causing invasion, migration, and EMT in CRC.
Collapse
Affiliation(s)
- Haitao Mei
- Department of Gastrointestinal Surgery, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, 200071, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
- Department of Colorectal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Qingshan Luo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
| | - Junyong Weng
- Department of Colorectal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China
| | - Jialing Hao
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
| | - Jinfeng Cai
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
| | - Runkai Zhou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
| | - Ce Bian
- Department of Colorectal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Yingzi Ye
- Department of Infectious Diseases, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| | - Shengzheng Luo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China.
| | - Yugang Wen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China.
| |
Collapse
|
30
|
Li J, Xie B, Wang H, Wang Q, Wu Y. Investigating MATN3 and ASPN as novel drivers of gastric cancer progression via EMT pathways. Hum Mol Genet 2024; 33:2035-2050. [PMID: 39301785 DOI: 10.1093/hmg/ddae129] [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: 06/24/2024] [Revised: 08/09/2024] [Accepted: 08/24/2024] [Indexed: 09/22/2024] Open
Abstract
Gastric cancer (GC) is a leading cause of cancer-related deaths globally, necessitating the identification of novel therapeutic targets. This study investigates the roles of MATN3 and ASPN in GC progression via the epithelial-mesenchymal transition (EMT) pathway. Analysis of the Cancer Genome Atlas-Stomach Adenocarcinoma (TCGA-STAD) dataset revealed that both MATN3 and ASPN are significantly upregulated in GC tissues and correlate with poor patient survival. Protein-protein interaction and co-expression analyses confirmed a direct interaction between MATN3 and ASPN, suggesting their synergistic role in EMT activation. Functional assays demonstrated that MATN3 promotes GC cell proliferation, migration, and invasion, while its knockdown inhibits these malignant behaviors and induces apoptosis. ASPN overexpression further amplified these oncogenic effects. In vivo, studies in a mouse model corroborated that co-overexpression of MATN3 and ASPN enhances tumor growth and metastasis. These findings highlight the MATN3-ASPN axis as a potential therapeutic target in GC, offering new insights into the molecular mechanisms driving GC progression.
Collapse
Affiliation(s)
- Jing Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou 215004, Jiangsu Province, China
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - Bo Xie
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - Hu Wang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - QingKang Wang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - YongYou Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou 215004, Jiangsu Province, China
| |
Collapse
|
31
|
Zhou J, Zhou D, Zhang Q, Zhang X, Liu X, Ding L, Wen J, Xu X, Cheng Z. DCLK1 mediated cooperative acceleration of EMT by avian leukosis virus subgroup J and Marek's disease virus via the Wnt/β-catenin pathway promotes tumor metastasis. J Virol 2024; 98:e0111224. [PMID: 39445786 PMCID: PMC11575233 DOI: 10.1128/jvi.01112-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Co-infection with oncogenic retrovirus and herpesvirus significantly facilitates tumor metastasis in human and animals. Co-infection with avian leukosis virus subgroup J (ALV-J) and Marek's disease virus (MDV), which are typical oncogenic retrovirus and herpesvirus, respectively, leads to enhanced oncogenicity and accelerated tumor formation, resulting in increased mortality of affected chickens. Previously, we found that ALV-J and MDV cooperatively promoted tumor metastasis. However, the molecular mechanism remains elusive. Here, we found that doublecortin-like kinase 1 (DCLK1) mediated cooperative acceleration of epithelial-mesenchymal transition (EMT) by ALV-J and MDV promoted tumor metastasis. Mechanistically, DCLK1 induced EMT via activating Wnt/β-catenin pathway by interacting with β-catenin, thereby cooperatively promoting tumor metastasis. Initially, we screened and found that DCLK1 was a potential mediator for the cooperative activation of EMT by ALV-J and MDV, and enhanced cell proliferation, migration, and invasion. Subsequently, we revealed that DCLK1 physically interacted with β-catenin to promote the formation of the β-catenin-TCF4 complex, inducing transcription of the Wnt target gene, c-Myc, promoting EMT by increasing the expression of N-cadherin, Vimentin, and Snail, and decreasing the expression of E-cadherin. Taken together, we discovered that jointly activated DCLK1 by ALV-J and MDV accelerated cell proliferation, migration and invasion, and ultimately activated EMT, paving the way for tumor metastasis. This study elucidated the molecular mechanism underlying cooperative metastasis induced by co-infection with retrovirus and herpesvirus. IMPORTANCE Tumor metastasis, a complex phenomenon in which tumor cells spread to new organs, is one of the greatest challenges in cancer research and is the leading cause of cancer-induced death. Numerous studies have shown that oncoviruses and their encoded proteins significantly affect metastasis, especially the EMT process. ALV-J and MDV are classic tumorigenic retrovirus and herpesvirus, respectively. We found that ALV-J and MDV synergistically promoted EMT. Further, we identified the tumor stem cell marker DCLK1 in ALV-J and MDV co-infected cells. DCLK1 directly interacted with β-catenin, promoting the formation of the β-catenin-TCF4 complex. This interaction activated the Wnt/β-catenin pathway, thereby inducing EMT and paving the way for synergistic tumor metastasis. Exploring the molecular mechanisms by which ALV-J and MDV cooperate during EMT will contribute to our understanding of tumor progression and metastasis. This study provides new insights into the cooperative induced tumor metastasis by retroviruses and herpesviruses.
Collapse
Affiliation(s)
- Jing Zhou
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Defang Zhou
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Qian Zhang
- Department of Neurology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Xinyue Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Xiaoyang Liu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Longying Ding
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Jing Wen
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Xiaoyu Xu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Ziqiang Cheng
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| |
Collapse
|
32
|
Ferreira JM, Gonçalves CS, Costa BM. Emerging roles and biomarker potential of WNT6 in human cancers. Cell Commun Signal 2024; 22:538. [PMID: 39529066 PMCID: PMC11552340 DOI: 10.1186/s12964-024-01892-4] [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: 06/24/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
The WNT6 ligand is a well-known activator of the WNT signaling pathway, considered a vital player in several important physiologic processes during embryonic development and maintaining homeostasis throughout life, regulating the proliferation and differentiation of multiple stem/progenitor cell types. More recently, as it is the case for many key molecular regulators of embryonic development, dysregulation of WNT6 has been implicated in cancer development and progression in multiple studies. In this review, we overview the most significant recent findings regarding WNT6 in the context of human malignancies, exploring its influence on multiple dimensions of tumor pathophysiology and highlighting the putative underlying WNT6-associated molecular mechanisms. We also discuss the potential clinical implications of WNT6 as a prognostic and therapeutic biomarker. This critical review highlights the emerging relevance of WNT6 in multiple human cancers, and its potential as a clinically-useful biomarker, addressing key unanswered questions that could lead to new opportunities in patient diagnosis, stratification, and the development of rationally-designed precision therapies.
Collapse
Affiliation(s)
- Joana M Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Céline S Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bruno M Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
33
|
Wu F, Zhang K, Song Z, Zhou Q, Sun H, Tan Z, Huang Z, Wang F, Wang Z, Yang R, Huang Y. Reduced Proline-Rich Tyrosine Kinase 2 Promotes Tumor Metastasis by Activating Epithelial-Mesenchymal Transition in Colorectal Cancer. Dig Dis Sci 2024; 69:4098-4107. [PMID: 39414740 DOI: 10.1007/s10620-024-08643-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/05/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND Proline-rich tyrosine kinase 2 (PYK2) is involved in the occurrence, proliferation, migration, and invasion of various tumors. However, few studies have reported the role of PYK2 in colorectal cancer (CRC). AIM To explore the effects of PYK2 on CRC metastasis and elucidate the detailed molecular mechanisms involved. METHODS The expression and prognosis value of PYK2 in CRC prognosis were analyzed using data from The Cancer Genome Atlas (TCGA). PYK2 was knocked down or overexpressed in human CRC cell line, HCT116. Cell proliferation, migration, invasion, and cycle changes were analyzed using CCK-8, Transwell, and flow cytometry assays. Western blotting and quantitative real-time PCR were performed to detect the mRNA and protein levels of cell proliferation and epithelial-mesenchymal transition (EMT) indicators. Fluorescence staining was performed to examine the cytoskeleton. RESULTS Lower expression of PYK2 was observed in CRC tissues and associated with poor prognosis and metastasis in patients with CRC in TCGA database. PYK2 knockdown significantly induced the migration and invasion of CRC cells but did not affect cell proliferation or cycle. Immunofluorescence staining of phalloidin showed that the downregulation of PYK2 increased the cytoskeleton in CRC cells. Moreover, low expression of PYK2 induced the downregulation of E-cadherin and upregulation of snail and vimentin by activating Wnt/β-catenin signaling, thus promoting EMT in CRC cells. CONCLUSIONS Low PYK2 expression was found in tumor tissues, especially metastases, and significantly correlated with patient prognosis. Moreover, decreased PYK2 induces EMT by activating Wnt/β-catenin signaling, which is the potential mechanism of CRC metastasis. Regulating the expression of PYK2 to suppress tumor cell metastasis may represent a promising therapeutic strategy for metastatic CRC.
Collapse
Affiliation(s)
- Fangquan Wu
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Ke Zhang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhengyang Song
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Qishuo Zhou
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hongxia Sun
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zenglin Tan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhenxuan Huang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Fangyan Wang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhonglin Wang
- Department of Anorectal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Riwei Yang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yingpeng Huang
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
34
|
Goyal A, Murkute SL, Bhowmik S, Prasad CP, Mohapatra P. Belling the "cat": Wnt/β-catenin signaling and its significance in future cancer therapies. Biochim Biophys Acta Rev Cancer 2024; 1879:189195. [PMID: 39413855 DOI: 10.1016/j.bbcan.2024.189195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
The WNT/β-catenin is among one of the most extensively studied cellular signaling pathways involved in the initiation and progression of several deadly cancers. It is now understood that the WNT/β-catenin signaling, during tumor progression operates in a very complex fashion beyond the earlier assumed simple WNT 'On' or 'Off' mode as it recruits numerous WNT ligands, receptors, transcriptional factors and also cross-talks with other signaling molecules including the noncanonical WNT regulators. WNT/β-catenin signaling molecules are often mutated in different cancers which makes them very challenging to inhibit and sometimes ranks them among the undruggable targets. Furthermore, due to the evolutionary conservation of this pathway, inhibiting WNT/β-catenin has caused significant toxicity in normal cells. These challenges are reflected in clinical trial data, where the use of WNT/β-catenin inhibitors as standalone treatments remains limited. In this review, we have highlighted the crucial functional associations of diverse WNT/β-catenin signaling regulators with cancer progression and the phenotypic switching of tumor cells. Next, we have shed light on the roles of WNT/β-catenin signaling in drug resistance, clonal evolution, tumor heterogeneity, and immune evasion. The present review also focuses on various classes of routine and novel WNT/β-catenin therapeutic regimes while addressing the challenges associated with targeting the regulators of this complex pathway. In the light of multiple case studies on WNT/β-catenin inhibitors, we also highlighted the challenges and opportunities for future clinical trial strategies involving these treatments. Additionally, we have proposed strategies for future WNT/β-catenin-based drug discovery trials, emphasizing the potential of combination therapies and AI/ML-driven prediction approaches. Overall, here we showcased the opportunities, possibilities, and potentialities of WNT/β-catenin signaling modulatory therapeutic regimes as promising precision cancer medicines for the future.
Collapse
Affiliation(s)
- Akansha Goyal
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Satyajit Laxman Murkute
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Sujoy Bhowmik
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology Lab, DR BRA-IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Purusottam Mohapatra
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India.
| |
Collapse
|
35
|
Li J, Zhu X, Ye S, Dong Q, Hou J, Liu J, She W. Tanshinone IIA potentiates the therapeutic efficacy of glucocorticoids in lipopolysaccharide-treated HEI-OC1 cells through modulation of the FOXP3/Nrf2 signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 57:727-737. [PMID: 39483046 DOI: 10.3724/abbs.2024194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
Glucocorticoids (GCs) are commonly used to treat sudden sensorineural hearing loss (SSNHL), although some patients are resistant to this therapeutic approach. Clinical studies have demonstrated the efficacy of tanshinone IIA (TA) in combination with GC for managing various human ailments. However, it remains unclear whether TA can mitigate GC resistance in SSNHL. Our aim is to elucidate the role of NRF2-induced transcriptional regulation of HDAC2 in influencing GC resistance and investigate the involvement of TA-related molecular pathways in GC resistance. Here, HEI-OC1 cells are treated with lipopolysaccharide (LPS) to establish an in vitro model for SSNHL. The cells are subsequently treated with dexamethasone (DXE) or DXE + TA. RT-qPCR and western blot analysis are used to measure the mRNA and protein levels of Forkhead box P3 (FOXP3), nuclear factor erythroid 2-related factor 2 (NRF2), and histone deacetylase 2 (HDAC2). Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assays are carried out to assess cell proliferation. Flow cytometry analysis is performed to evaluate apoptosis. Mechanistic studies involve chromatin immunoprecipitation (ChIP), luciferase reporter, and DNA pull-down assays. Our results show that treatment with TA + DEX significantly increases proliferation and suppresses apoptosis in LPS-treated HEI-treated OC1 cells. TA upregulates HDAC2 expression by activating NRF2-mediated transcription of HDAC2, with the NRF2-HDAC2 binding site located at bases 419-429 (ATGACACTCCA) in the promoter sequence of HDAC2. Furthermore, TA upregulates FOXP3 expression to activate NRF2 transcription, with the predicted FOXP3-binding site located at bases 864-870 (GCAAACA) in the promoter sequence of NRF2. In summary, these findings suggest that TA enhances the therapeutic effects of GC on the proliferation and apoptosis of HEI OC1 cells by increasing FOXP3/Nrf2 expression. These results indicate that TA may be promising for ameliorating GC resistance in patients with SSNHL.
Collapse
Affiliation(s)
- Jie Li
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
- Department of Otolaryngology, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, Nantong 226000, China
| | - Xiaoyan Zhu
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Shiming Ye
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Qi Dong
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- Otorhinolaryngology Research Institute of Nanjing Drum Tower Hospital, Nanjing 210008, China
| | - Jie Hou
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- Otorhinolaryngology Research Institute of Nanjing Drum Tower Hospital, Nanjing 210008, China
| | - Jing Liu
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- Otorhinolaryngology Research Institute of Nanjing Drum Tower Hospital, Nanjing 210008, China
| | - Wandong She
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- Otorhinolaryngology Research Institute of Nanjing Drum Tower Hospital, Nanjing 210008, China
| |
Collapse
|
36
|
Zhang Y, Liu J, Yang G, Zou J, Tan Y, Xi E, Geng Q, Wang Z. Asiaticoside Inhibits Growth and Metastasis in Non-Small Cell Lung Cancer by Disrupting EMT via Wnt/β-Catenin Pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:4859-4870. [PMID: 38888371 DOI: 10.1002/tox.24359] [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: 06/08/2023] [Revised: 04/07/2024] [Accepted: 05/11/2024] [Indexed: 06/20/2024]
Abstract
Non-small cell lung cancer (NSCLC) is the primary inducer of cancer-related death worldwide. Asiaticoside (ATS) is a triterpenoid saponin that has been indicated to possess an antitumor activity in several malignancies. Nonetheless, its detailed functions in NSCLC remain unclarified. In this study, NSCLC cells were exposed to various doses of ATS. Functional experiments were employed to estimate the ATS effect on NSCLC cell behaviors. Western blotting was implemented for protein expression evaluation. A xenograft mouse model was established to assess the ATS effect on NSCLC in vivo. The results showed that ATS restrained NSCLC cell proliferation, cell cycle progression, migration, and invasiveness. ATS reversed TGF-β-induced promotion in epithelial-mesenchymal transition (EMT). Mechanistically, ATS inhibited Wnt/β-catenin signaling in NSCLC. Upregulating β-catenin restored ATS-mediated suppression of NSCLC cell aggressiveness. Moreover, ATS administration repressed tumorigenesis in tumor-bearing mice. In conclusion, ATS represses growth and metastasis in NSCLC by blocking EMT via the inhibition of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yanan Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiangyong Liu
- Department of Radiography, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Gang Yang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Jiani Zou
- Department of Radiography, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Yan Tan
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Erping Xi
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| |
Collapse
|
37
|
Wang P, Jiang N, Zhong J, Chen Q, Huang R, Liu C, Xu P. IFI27 enhances bladder cancer immunotherapy response by modulating regulatory T cell enrichment. J Cancer 2024; 15:6616-6630. [PMID: 39668835 PMCID: PMC11632990 DOI: 10.7150/jca.99014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/12/2024] [Indexed: 12/14/2024] Open
Abstract
Bladder cancer (BCa) is the 10th most prevalent cancer globally. Neoadjuvant therapy has become the standard treatment for muscle-invasive bladder cancer, yet the pathologic complete response rate for patients is only approximately 35%. However, the mechanisms underlying neoadjuvant therapy resistance in bladder cancer patients remain unclear. We collected two sets of paired bladder cancer specimens before and after neoadjuvant therapy, and performed RNA sequencing. The findings revealed a significant decrease in IFI27 expression levels in the post-neoadjuvant therapy group compared to samples collected before treatment, suggesting that IFI27 may play a role in resistance to neoadjuvant combination therapy. IFI27, a member of the interferon-alpha (IFN-α) inducible gene family, influences the efficacy of immune checkpoint blockade therapy. Further analysis demonstrated that IFI27 is predominantly expressed in the cytoplasm of bladder cancer cells and exhibited low expression levels in bladder cancer tissues and cell lines. Subsequently, we investigated the inhibitory effects of IFI27 on bladder cancer proliferation, migration, epithelial-mesenchymal transition, and lymph node metastasis. Additionally, in a mouse model, PD-1Ab immunotherapy was found to upregulate IFI27 while downregulating the protein level of FOXP3, a key transcription factor for regulatory T cells. Flow cytometric analysis further demonstrated that IFI27 inhibits bladder cancer progression by suppressing regulatory T cell infiltration and enhancing anti-tumor immune responses. In conclusion, these findings establish IFI27 as a promising molecular marker for improving the efficacy of immunotherapy in bladder cancer and offer valuable insights into strategies for enhancing immunotherapy sensitivity.
Collapse
Affiliation(s)
- Peng Wang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Jiang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianye Zhong
- Department of Urology, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
| | - Qiwei Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Renliang Huang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Chen X, Dai L. WTAP Promotes the Excessive Proliferation of Airway Smooth Muscle Cells in Asthma by Enhancing AXIN1 Levels Through the Recognition of YTHDF2. Biochem Genet 2024:10.1007/s10528-024-10947-7. [PMID: 39453546 DOI: 10.1007/s10528-024-10947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Asthma is a common chronic respiratory disease in children, the incidence rate of which has increased in recent years. Wilms tumour 1-associated protein (WTAP) is an N6-methyladenosine (m6A) methyltransferase. The purpose of this study was to explore the specific mechanism of WTAP in asthma progression, and clarify the intricate interplay between m6A modifications, WTAP, AXIN1, and their collective impact on airway smooth muscle cells (ASMCs) proliferation in asthma. Platelet-derived growth factor-BB (PDGF-BB)-treated ASMCs were used to establish an asthma model in vitro. The cell phenotype was tested using CCK-8, transwell, and wound healing assays. The expression of the Wnt signalling pathway was detected by western blotting. In addition, the relationship between WTAP/YTDHF2 and AXIN1 was assessed by a double luciferase reporter assay. Actinomycin D treatment and RT‒qPCR assays were performed to determine the mRNA stability of AXIN1. We found that WTAP was significantly increased in PDGF-BB-treated ASMCs. Knockdown of WTAP inhibited the excessive cell viability and migration of ASMCs induced by PDGF-BB. Furthermore, WTAP knockdown increased AXIN1 levels and inhibited the Wnt signalling pathway. Furthermore, WTAP knockdown decreased the m6A levels and enhanced the mRNA stability of AXIN1. WTAP overexpression showed the opposite effect. In addition, YTHDF2 was demonstrated to be the reader that recognizes the WTAP-mediated m6A modification of AXIN1. YTHDF2 knockdown enhanced the mRNA stability of AXIN1 and reversed the effect of WTAP overexpression on PDGF-BB-treated ASMCs. WTAP knockdown inhibited the excessive cell viability and migration of ASMCs by enhancing the m6A levels of AXIN1, which was further recognized by YTHDF2. The upregulation of AXIN1 mediated by the WTAP/YTHDF2 axis further inhibited the Wnt signalling pathway. Our study provides a new method for the treatment of asthma. This work not only deepens our understanding of the molecular underpinnings of asthma but also identifies potential therapeutic targets for the development of novel treatments aimed at inhibiting ASMC proliferation and alleviating asthma symptoms.
Collapse
Affiliation(s)
- Xueli Chen
- Pediatric department, Maternal and Child Health of Hubei Province, NO.745 Wuluo Road, Hongshan District, Wuhan, 430070, Hubei, People's Republic of China
| | - Li Dai
- Pediatric department, Maternal and Child Health of Hubei Province, NO.745 Wuluo Road, Hongshan District, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
39
|
Tan C, Xu J, Zhang S, Liu S, Yang X, Wu D, Yu B, Huang Y. Transcription Factor Forkhead Box Protein 3 (FOXP3) as a Prognostic Indicator for Postoperative Outcomes in Patients with Breast Cancer: Establishment of a Prognostic Nomogram. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:705-723. [PMID: 39464238 PMCID: PMC11505482 DOI: 10.2147/bctt.s484055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
Purpose The current investigation is to assess FOXP3 expression in breast cancer patients and evaluate the predictive significance of FOXP3. Patients and Methods A cohort of 313 cases between January 2015 and November 2015 were enrolled this research. Immunohistochemistry (IHC) assay was utilized to detect the expression levels of FOXP3 in primary breast carcinoma specimens. These patients were separated into two groups by semiquantitative scoring approach. Chi-square test and Fisher's exact test were conducted to investigate the correlations between FOXP3 expression in tumors and clinicopathological variables. Kaplan-Meier method and Log rank test were utilized to generate survival curves for disease-free survival (DFS) and overall survival (OS). The independent factors were examined using Cox regression analysis. Nomogram models were created for assessing DFS and OS rates. Results Depending on the levels of FOXP3 expression in tumors, these patients were categorized into two groups: low FOXP3 expression (174 cases) and high FOXP3 expression (139 cases). The patients exhibiting low levels of FOXP3 expression in tumors demonstrated a longer survival duration contrasted with those with high expression (DFS: 88.75 vs 65.87 months, χ2=36.1100, P<0.0001; OS: 89.70 vs 78.37 months, χ2=32.4900, P<0.0001). Multivariate analysis revealed that FOXP3 was a significant prognostic factor [DFS: hazard ratio (HR): 2.822, 95% CI: 1.595-4.992, P<0.0001; OS: HR: 3.232, 95% CI: 1.812-5.763, P<0.0001]. The good predictive clinical utility of FOXP3-based nomograms within the threshold probability range for different survival rates was demonstrated by calibration curve and decision curve analyses. Conclusion FOXP3 expression serves as a crucial prognostic indicator in breast cancer patients, and may aid preoperative evaluation in clinical practice.
Collapse
Affiliation(s)
- Chunlei Tan
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Jinling Xu
- Endoscope Department, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Shiyuan Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Shuqiang Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Xiaotian Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Danping Wu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Boqian Yu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yuanxi Huang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
40
|
杨 玉, 刘 雪, 刘 伟, 周 星, 张 振, 胡 妍, 刘 培, 李 娴, 刘 浩, 李 姗. [Aumolertinib combined with anlotinib inhibits proliferation of non-small cell lung cancer cells by down-regulating the PI3K/AKT pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1965-1975. [PMID: 39523097 PMCID: PMC11526449 DOI: 10.12122/j.issn.1673-4254.2024.10.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE To investigate the inhibitory effect of aumolertinib combined with anlotinib on proliferation of non-small cell lung cancer (NSCLC) cells. METHODS CCK-8 assay, colony formation assay, and flow cytometry were used to assess the effect of different concentrations of aumolertinib or anlotinib on proliferation, survival, and apoptosis of PC-9 and HCC827 cells, and their synergistic effect was evaluated using the SynergyFinder model. In PC-9 and HCC827 cells treated with aumolertinib combined with anlotinib, the changes in cell invasion and migration abilities were assessed with Transwell assay, and the expressions of apoptosis- and invasion/migration-related proteins (Bax, Bcl-2, E-cadherin, vimentin, MMP2, and MMP9) and the key PI3K-Akt pathway proteins were detected using Western blotting. RESULTS In PC-9 cells, the IC50 of aumolertinib and anlotinib was 1.701 μmol/L and 4.979 μmol/L, respectively, with a synergy score (ZIP) of 19.112; in HCC827 cells, their IC50 was 2.961 μmol/L and 7.934 μmol/L, respectively, with a ZIP of 12.325. Compared with aumolertinib and anlotinib used alone, their combined treatment more strongly inhibited the proliferation and survival, enhanced apoptosis and suppressed invasion and migration abilities of PC-9 and HCC827 cells. Western blotting showed that in both PC-9 and HCC827 cells, the combined treatment significantly upregulated the expressions of E-cadherin and Bax proteins, downregulated the expressions of Bcl-2, vimentin, MMP2, and MMP9 proteins, and reduced phosphorylation levels of PI3K and Akt. CONCLUSION Aumolertinib combined with anlotinib can effectively inhibit NSCLC cell proliferation by downregulating the PI3K-Akt pathway, suggesting a potentially new option for NSCLC treatment.
Collapse
|
41
|
Monleón-Guinot I, Bravo-Baranda L, Milián L, Sancho-Tello M, Llop-Miguel M, Galbis JM, Cremades A, Carda C, Mata M. Cancer Epithelial Cells Participate in the Self-Organization of Lung Tumor Spheroids: A Morphological Approach. Cells Tissues Organs 2024:1-23. [PMID: 39383853 DOI: 10.1159/000541524] [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: 06/14/2024] [Accepted: 09/06/2024] [Indexed: 10/11/2024] Open
Abstract
INTRODUCTION/AIMS The tumor microenvironment is known to play an important role in tumor progression. However, the specific mechanisms underlying this process are still not known in detail and more research is needed on the elements that control tumor progression in lung cancer. In this work, we aimed to investigate the involvement of epithelial and stromal cancer cells in growth, cell migration, and epithelial-to-mesenchymal transition (EMT) in a 3D in vitro model consisting of cell spheroids cultured in a type I collagen scaffold. METHODS Spheroids were manufactured using different combinations of epithelial cells, particularly H460 and H1792 cell lines, with cancer-associated fibroblasts and normal fibroblasts, both isolated from adenocarcinoma patients. We evaluated the morphology of the spheroids by analysis of F-actin and pankeratin with confocal microscopy. We determined the ultrastructure of cells in the spheroids by transmission electron microscopy and the expression of CDH1, CDH2, and VIM by RT-PCR. RESULTS We observed that, on the one hand, the type of epithelial cell influences the morphology of spheroids. Stromal cells stimulated spheroid growth and cell dissemination through the collagen matrix, either alone or organized in branches with a nucleus of epithelial cells preceded by fibroblast cells. They also induced the appearance of new cell groups in the scaffold and the presence of EMT markers. CONCLUSION The results presented here indicate the participation of both epithelial and stromal cells in the control of spheroid self-organization. The experimental model proposed here, although preliminary, is useful for the study of some aspects related to tumor progression in lung cancer.
Collapse
Affiliation(s)
- Irene Monleón-Guinot
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Lucía Bravo-Baranda
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Lara Milián
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - María Sancho-Tello
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Mauro Llop-Miguel
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, Valencia, Spain
| | | | | | - Carmen Carda
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Manuel Mata
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicina (CIBER-BBN), Madrid, Spain
| |
Collapse
|
42
|
Yan Q, Li S, He L, Chen N. Prognostic implications of tumor-infiltrating lymphocytes in non-small cell lung cancer: a systematic review and meta-analysis. Front Immunol 2024; 15:1476365. [PMID: 39372398 PMCID: PMC11449740 DOI: 10.3389/fimmu.2024.1476365] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
Background Tumor-infiltrating lymphocytes (TILs) have demonstrated potential as prognostic biomarkers across various cancer types. However, their prognostic implications in non-small cell lung cancer (NSCLC) remain ambiguous. Methods An exhaustive electronic search was executed across the Pubmed, EMBASE, Web of Science, and Cochrane Library databases to locate relevant studies published up until December 19, 2023. Studies were eligible if they assessed the association between TILs and overall survival (OS) and disease-free survival (DFS) in NSCLC patients. The OS and DFS were subsequently extracted for analysis. The prognostic significance of TILs was evaluated by calculating the Pooled Hazard Ratios (HRs) and their corresponding 95% Confidence Intervals (CIs). Results The meta-analysis incorporated 60 studies, which collectively included 15829 NSCLC patients. The collective analysis indicated that NSCLC patients exhibiting TILs infiltration demonstrated a significantly improved OS(HR: 0.67; 95%CI: 0.55-0.81). Subgroup analyses, based on TIL subtypes (CD8+, CD3+ and CD4+), consistently revealed a favorable prognostic impact on OS. However, it was observed that FOXP3+ was correlated with a poor OS (HR: 1.35; 95% CI: 0.87-2.11). Conclusion This comprehensive systematic review and meta-analysis substantiate the prognostic significance of TILs in patients diagnosed with NSCLC. Notably, elevated TILs infiltration correlates with a favorable prognosis, particularly among CD8+, CD3+ and CD4+ subtypes. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023468089 PROSPERO, identifier CRD42023468089.
Collapse
Affiliation(s)
- Qin Yan
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Cancer Prevention and Treatment Institute of Chengdu, Department of Oncology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Shuai Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lang He
- Cancer Prevention and Treatment Institute of Chengdu, Department of Oncology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Nianyong Chen
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Khambholja K, Gehani M, Kothari R, Marulkar S. Prognostic value of tumour-associated regulatory T-cells as a biomarker in non-small cell lung cancer: a systematic review and meta-analysis. Syst Rev 2024; 13:233. [PMID: 39272135 PMCID: PMC11401299 DOI: 10.1186/s13643-024-02642-w] [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: 02/11/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Tumour, nodes, and metastases (TNM) staging has been deficient in prognosticating in patients suffering from non-small cell lung cancer (NSCLC). To supplement TNM staging, this systematic review and meta-analysis aimed to evaluate the prognostic value of the regulatory T cells (Treg). METHODS A keyword search was conducted in MEDLINE and EMBASE for full-text original human studies from any region published in English during the last 12 years. Eligible for inclusion were studies evaluating the prognostic value of the number of Treg cells in NSCLC except case studies, case series, systematic reviews, and meta-analyses. Two reviewers (one reviewer used an automation tool) independently screened the studies and assessed risk-of-bias using the Quality in Prognosis Studies (QUIPS) tool. Meta-analysis was done for studies reporting significant multivariate hazard ratio (HR). RESULTS Out of 809 retrievals, 24 studies were included in the final review. The low number of Treg cells was found significantly associated with improved overall survival (pooled log OR, 1.646; 95% CI, 1.349, 1.944; p (2-tailed) < .001; SE, 0.1217), improved recurrence-free survival (HR, 1.99; 95% CI, 1.15, 3.46; p = .01), improved progression-free survival (pooled log OR, 2.231; 95% CI, 0.424, 4.038; p (2-tailed) .034; SE, 0.4200), and worse disease-free survival (pooled log OR, 0.992; 95% CI, 0.820, 1.163; p (2-tailed) .009; SE, 0.0135), especially when identified by forkhead box P3 (FOXP3), in any stage or non-metastatic NSCLC. CONCLUSION A low number of Treg cells indicated better survival, suggesting its potential use as a prognostic biomarker in NSCLC. SYSTEMATIC REVIEW REGISTRATION The protocol of this review was prospectively registered on PROSPERO on August 28, 2021, and was assigned the registration number CRD42021270598. The protocol can be accessed from PROSPERO website.
Collapse
Affiliation(s)
- Kapil Khambholja
- Department of Medical Writing, Catalyst Clinical Research, 2528 Independence Blvd, Suite 100, Wilmington, NC, 28412, USA
| | - Manish Gehani
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani-Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal, Hyderabad, Telangana, 500078, India.
| | - Rushabh Kothari
- Medical Oncology Department, Narayana Multispecialty Hospital, Opposite Police Station, Near Chakudiya Mahadev, Rakhial, Ahmedabad, Gujarat, 380023, India
| | - Sachin Marulkar
- Catalyst Clinical Research, 2528 Independence Blvd, Suite 100, Wilmington, NC, 28412, USA
| |
Collapse
|
44
|
Li S, Guo Y, Zhu G, Sun L, Zhou F. Identify BCAT1 plays an oncogenic role and promotes EMT in KIRC via single cell RNA-seq and experiment. Front Oncol 2024; 14:1446324. [PMID: 39324007 PMCID: PMC11422235 DOI: 10.3389/fonc.2024.1446324] [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: 06/09/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Background Kidney renal clear cell carcinoma (KIRC) is a major subtype of renal cell carcinoma with poor prognosis due to its invasive and metastatic nature. Despite advances in understanding the molecular underpinnings of various cancers, the role of branched-chain amino acid transferase 1 (BCAT1) in KIRC remains underexplored. This study aims to fill this gap by investigating the oncogenic role of BCAT1 in KIRC using single-cell RNA-seq data and experimental validation. Methods Single-cell transcriptomic data GSE159115 was utilized to investigate potential biomarkers in KIRC. After screening, we used BCAT1 as a target gene and investigated its function and mechanism in KIRC through databases such as TCGA-GTEx, using genome enrichment analysis (GSEA), genome variation analysis (GSVA), gene ontology (GO) and Kyoto Encyclopedia of the Genome (KEGG). BCAT1 expression was detected in clinical tissue samples using Western Blotting (WB) and immunohistochemical (IHC) staining techniques. We established cell lines stably overexpressing and knocking down BCAT1 and performed WB, qRT-PCR, cell scratch assay and transwell assay. Results BCAT1 was highly expressed in KIRC and was associated with disease prognosis and TME. Patients with mutations in the BCAT1 gene had shorter overall survival (OS) and disease-free survival (DFS). patients with high BCAT1 expression had shorter OS, progression-free interval (PFI), and disease-specific survival (DSS). GSEA showed that BCAT1 was significantly enriched in epithelial mesenchymal transition (EMT). Bioinformatics analysis and WB and IHC staining showed that BCAT1 expression was higher in KIRC than in paracancerous tissues. In vitro experiments confirmed that BCAT1 in KIRC cells may promote EMT affecting its invasion, migration. We constructed a protein interaction network (PPI) to hypothesize proteins that may interact with BCAT1. Single-sample gene set enrichment analysis (ssGSEA) revealed the immune infiltration environment of BCAT1. Furthermore, hypomethylation of the BCAT1 promoter region in KIRC may contribute to disease progression by promoting BCAT1 expression. Conclusion BCAT1 promotes KIRC invasion and metastasis through EMT and has prognostic predictive value and potential as a biomarker. It may become a novel biomarker.
Collapse
Affiliation(s)
- Shiqing Li
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yinsheng Guo
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guanhua Zhu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lu Sun
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
45
|
Ge J, Yin X, Chen L. Regulatory T cells: masterminds of immune equilibrium and future therapeutic innovations. Front Immunol 2024; 15:1457189. [PMID: 39290699 PMCID: PMC11405253 DOI: 10.3389/fimmu.2024.1457189] [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: 06/30/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Regulatory T cells (Tregs), a subset of CD4+T cells marked by the expression of the transcription factor forkhead box protein 3 (Foxp3), are pivotal in maintaining immune equilibrium and preventing autoimmunity. In our review, we addressed the functional distinctions between Foxp3+Tregs and other T cells, highlighting their roles in autoimmune diseases and cancer. We uncovered the dual nature of Tregs: they prevented autoimmune diseases by maintaining self-tolerance while contributing to tumor evasion by suppressing anti-tumor immunity. This study underscored the potential for targeted therapeutic strategies, such as enhancing Treg activity to restore balance in autoimmune diseases or depleting Foxp3+Tregs to augment anti-tumor immune responses in cancer. These insights laid the groundwork for future research and clinical applications, emphasizing the critical role of Foxp3+Tregs in immune regulation and the advancement of next-generation immunotherapies.
Collapse
Affiliation(s)
- Junwei Ge
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xuan Yin
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
46
|
Sundar R, Chia DKA, Zhao JJ, Lee ARYB, Kim G, Tan HL, Pang A, Shabbir A, Willaert W, Ma H, Huang KK, Hagihara T, Tan ALK, Ong CAJ, Wong JSM, Seo CJ, Walsh R, Chan G, Cheo SW, Soh CCC, Callebout E, Geboes K, Ng MCH, Lum JHY, Leow WQ, Selvarajan S, Hoorens A, Ang WH, Pang H, Tan P, Yong WP, Chia CSL, Ceelen W, So JBY. Phase I PIANO trial-PIPAC-oxaliplatin and systemic nivolumab combination for gastric cancer peritoneal metastases: clinical and translational outcomes. ESMO Open 2024; 9:103681. [PMID: 39288528 PMCID: PMC11421236 DOI: 10.1016/j.esmoop.2024.103681] [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: 06/12/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/19/2024] Open
Abstract
INTRODUCTION Pressurized intraperitoneal aerosol chemotherapy-oxaliplatin (PIPAC-OX) induces direct DNA damage and immunogenic cell death in patients with gastric cancer peritoneal metastases (GCPM). Combining PIPAC-OX with immune checkpoint inhibition remains untested. We conducted a phase I first-in-human trial evaluating the safety and efficacy of PIPAC-OX combined with systemic nivolumab (NCT03172416). METHODS Patients with GCPM who experienced disease progression on at least first-line systemic therapy were recruited across three centers in Singapore and Belgium. Patients received PIPAC-OX at 90 mg/m2 every 6 weeks and i.v. nivolumab 240 mg every 2 weeks. Translational studies were carried out on GCPM samples acquired during PIPAC-OX procedures. RESULTS In total, 18 patients with GCPM were prospectively recruited. The PIPAC-OX and nivolumab combination was well tolerated with manageable treatment-related adverse events, although one patient suffered from grade 4 vomiting. At second and third PIPAC-OX, respectively, the median decrease in peritoneal cancer index (PCI) was -5 (interquartile range: -12 to +1) and -7 (interquartile range: -6 to -20) and peritoneal regression grade 1 or 2 was observed in 66.7% (6/9) and 100% (3/3). Translational analyses of 43 GCPM samples revealed enrichment of immune/stromal infiltration and inflammatory signatures in peritoneal tumors after PIPAC-OX and nivolumab. M2 macrophages were reduced in treated peritoneal tumor samples while memory CD4+, CD8+ central memory and naive CD8+ T-cells were increased. CONCLUSIONS The first-in-human trial combining PIPAC-OX and nivolumab demonstrated safety and tolerability, coupled with enhanced T-cell infiltration within peritoneal tumors. This trial sets the stage for future combinations of systemic immunotherapy with locoregional intraperitoneal treatments.
Collapse
Affiliation(s)
- R Sundar
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore; Singapore Gastric Cancer Consortium, Singapore.
| | - D K A Chia
- Division of Upper Gastrointestinal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore
| | - J J Zhao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore; Department of Medicine, National University Hospital, Singapore, Singapore
| | - A R Y B Lee
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - G Kim
- Division of Upper Gastrointestinal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore
| | - H L Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - A Pang
- Division of Upper Gastrointestinal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore
| | - A Shabbir
- Division of Upper Gastrointestinal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore
| | - W Willaert
- Department of Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - H Ma
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - K K Huang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - T Hagihara
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - A L K Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - C-A J Ong
- Singapore Gastric Cancer Consortium, Singapore; Department of Sarcoma, Peritoneal and Rare Tumors (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - J S M Wong
- Department of Sarcoma, Peritoneal and Rare Tumors (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - C J Seo
- Department of Sarcoma, Peritoneal and Rare Tumors (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - R Walsh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - G Chan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - S W Cheo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - C C C Soh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - E Callebout
- Department of Digestive Oncology, Gastroenterology, Ghent University Hospital, Ghent, Belgium
| | - K Geboes
- Department of Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - M C H Ng
- Division of Medical Oncology, National Cancer Centre, Singapore; Duke NUS Medical School, Singapore
| | - J H Y Lum
- Department of Pathology, National University Hospital, Singapore
| | - W Q Leow
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - S Selvarajan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - A Hoorens
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - W H Ang
- Department of Chemistry, National University of Singapore, Singapore
| | - H Pang
- Department of Chemistry, National University of Singapore, Singapore
| | - P Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore; Singapore Gastric Cancer Consortium, Singapore
| | - W P Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore; Singapore Gastric Cancer Consortium, Singapore
| | - C S L Chia
- Department of Sarcoma, Peritoneal and Rare Tumors (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - W Ceelen
- Department of Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - J B Y So
- Singapore Gastric Cancer Consortium, Singapore; Division of Upper Gastrointestinal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Surgical Oncology, National University Cancer Institute of Singapore (NCIS), Singapore, Singapore.
| |
Collapse
|
47
|
Alqithami SM, Machwe A, Orren DK. Cigarette Smoke-Induced Epithelial-to-Mesenchymal Transition: Insights into Cellular Mechanisms and Signaling Pathways. Cells 2024; 13:1453. [PMID: 39273025 PMCID: PMC11394110 DOI: 10.3390/cells13171453] [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: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
This review delves into the molecular complexities underpinning the epithelial-to-mesenchymal transition (EMT) induced by cigarette smoke (CS) in human bronchial epithelial cells (HBECs). The complex interplay of pathways, including those related to WNT//β-catenin, TGF-β/SMAD, hypoxia, oxidative stress, PI3K/Akt, and NF-κB, plays a central role in mediating this transition. While these findings significantly broaden our understanding of CS-induced EMT, the research reviewed herein leans heavily on 2D cell cultures, highlighting a research gap. Furthermore, the review identifies a stark omission of genetic and epigenetic factors in recent studies. Despite these shortcomings, the findings furnish a consolidated foundation not only for the academic community but also for the broader scientific and industrial sectors, including large tobacco companies and manufacturers of related products, both highlighting areas of current understanding and identifying areas for deeper exploration. The synthesis herein aims to propel further research, hoping to unravel the complexities of the EMT in the context of CS exposure. This review not only expands our understanding of CS-induced EMT but also reveals critical limitations in current methodologies, primarily the reliance on 2D cell cultures, which may not adequately simulate more complex biological interactions. Additionally, it highlights a significant gap in the literature concerning the genetic and epigenetic factors involved in CS-induced EMT, suggesting an urgent need for comprehensive studies that incorporate these types of experiments.
Collapse
Affiliation(s)
- Sarah Mohammed Alqithami
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | | |
Collapse
|
48
|
Wang G, Hiramoto K, Ma N, Ohnishi S, Morita A, Xu Y, Yoshikawa N, Chinzei Y, Murata M, Kawanishi S. Immunohistochemical analyses reveal FoxP3 expressions in spleen and colorectal cancer in mice treated with AOM/DSS, and their suppression by glycyrrhizin. PLoS One 2024; 19:e0307038. [PMID: 39150932 PMCID: PMC11329161 DOI: 10.1371/journal.pone.0307038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/27/2024] [Indexed: 08/18/2024] Open
Abstract
We previously demonstrated that glycyrrhizin (GL) suppressed inflammation and carcinogenesis in an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced murine model of colorectal cancer (CC). In this study, we found an accumulation of regulatory T cells (Tregs) in the spleen and suppression by GL in model mice. ICR mice were divided into four groups: Control, GL, CC, and GL-treated CC (CC+GL), and were sacrificed 20 weeks after AOM/DSS treatment. We measured spleen weight, areas of white and red pulp, and CD8+ T cells (cytotoxic T lymphocytes, CTL), and CD11c-positive cells (dendritic cells) in splenic tissues and forkhead box protein 3 (FoxP3)-positive cells (Tregs) in colorectal and splenic tissues. In all cases, the CC group showed a significant increase compared with those in Control group, and GL administration significantly attenuated this increase. These results indicate that Tregs accumulated in the spleen may participate in inflammation-related carcinogenesis by suppressing CTL. We also suggest that GL which binds to high-mobility group box 1 (HMGB1), suppresses carcinogenesis with decreasing Tregs in the spleen. Furthermore, there was an expression of FoxP3 in cancer cells, indicating that it may be involved in the malignant transformation of cancer cells.
Collapse
Affiliation(s)
- Guifeng Wang
- Department of Acupuncture and Moxibustion Medical Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Keiichi Hiramoto
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Ning Ma
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
- Institute of Traditional Chinese Medicine, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Shiho Ohnishi
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Akihiro Morita
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Yifei Xu
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | | | - Yasuo Chinzei
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Shosuke Kawanishi
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| |
Collapse
|
49
|
Meyiah A, Elkord E. What is the relevance of FoxP3 in the tumor microenvironment and cancer outcomes? Expert Rev Clin Immunol 2024; 20:803-809. [PMID: 38512803 DOI: 10.1080/1744666x.2024.2334258] [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: 01/13/2024] [Accepted: 03/20/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Forkhead box P3 (FoxP3) transcription factor plays critical roles in controlling immune responses and cancer progression in different cancers. FoxP3 expression within the tumor microenvironment (TME) may influence clinical outcomes negatively or positively, and it could play dual roles in cancer, either by promoting or inhibiting tumor development and progression. Some studies reported that high levels of FoxP3 could be associated with tumor progression and worse prognosis, while others reported contradictory results. AREAS COVERED In this special report, we present a brief account on the role and function of FoxP3 in the TME, and its contribution to the clinical outcomes of cancer patients. Importantly, we give insights on the potential factors that could contribute to different clinical outcomes in cancer patients. EXPERT OPINION Different studies showed that FoxP3 expression can be associated with bad prognoses in cancer patients. However, FoxP3 could have opposing roles by enhancing cancer progression or regression. Location and expression of FoxP3 in T cells or tumor cells can have different impacts on cancer prognoses. Different factors should be considered to establish FoxP3 as a more robust prognostic biomarker and a potential therapeutic target for enhancing anti-tumor immunity and improving clinical outcomes of cancer patients.
Collapse
Affiliation(s)
- Abdo Meyiah
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Eyad Elkord
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, UK
| |
Collapse
|
50
|
Zhang H, Ly A, Chou E, Wang L, Zhang P, Prado K, Gu Y, Pellegrini M, Chin AI. Role of Forkhead Box P3 in IFNγ-Mediated PD-L1 Expression and Bladder Cancer Epithelial-to-Mesenchymal Transition. CANCER RESEARCH COMMUNICATIONS 2024; 4:2228-2241. [PMID: 39099201 PMCID: PMC11345674 DOI: 10.1158/2767-9764.crc-23-0493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 06/26/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Antagonism of the PD-1/PD-L1 axis is a critical therapeutic strategy for patients with advanced bladder cancer. IFNγ functions as a key regulator of PD-L1 in both immune as well as cancer cells. Forkhead box P3 (FOXP3) is a transcription factor synonymous in T regulatory cell function but with increasingly described functions in cancer cells. Here, we investigated the relationship between FOXP3 and PD-L1 in bladder cancer. We showed that FOXP3 is critical in the ability for IFNγ to activate PD-L1 in bladder cancer cells. FOXP3 can bind to the PD-L1 promoter and induces a gene program that leads to regulation of multiple immune-related genes and genes involved in epithelial-to-mesenchymal transition (EMT). Using in vitro and in vivo human and murine models, we showed that FOXP3 can influence bladder cancer EMT as well as promote cancer metastases. Furthermore, FOXP3 may be a convergent factor for multiple activators of PD-L1, including the chemotherapeutic drug cisplatin. SIGNIFICANCE Historically a key transcription factor driving T regulatory cell function, FOXP3 has an increasingly recognized role in cancer cells. In bladder cancer, we defined a novel mechanism whereby FOXP3 mediates the activation of the immune checkpoint PD-L1 by the cytokine IFNγ. We also showed that FOXP3 induces other immune checkpoints as well as genes involved in EMT, promoting immune resistance and cancer metastases.
Collapse
Affiliation(s)
- Hanwei Zhang
- Department of Urology, University of California, Los Angeles, California.
| | - Ann Ly
- Department of Urology, University of California, Los Angeles, California.
| | - Emily Chou
- Department of Urology, University of California, Los Angeles, California.
| | - Liang Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California.
| | - Paul Zhang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California.
| | - Kris Prado
- Department of Urology, University of California, Los Angeles, California.
| | - Yiqian Gu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California.
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California.
- UCLA Broad Stem Cell Research Center, Los Angeles, California.
| | - Arnold I. Chin
- Department of Urology, University of California, Los Angeles, California.
- UCLA Broad Stem Cell Research Center, Los Angeles, California.
| |
Collapse
|