1
|
Allgayer H, Mahapatra S, Mishra B, Swain B, Saha S, Khanra S, Kumari K, Panda VK, Malhotra D, Patil NS, Leupold JH, Kundu GC. Epithelial-to-mesenchymal transition (EMT) and cancer metastasis: the status quo of methods and experimental models 2025. Mol Cancer 2025; 24:167. [PMID: 40483504 PMCID: PMC12144846 DOI: 10.1186/s12943-025-02338-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/19/2025] [Indexed: 06/11/2025] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a crucial cellular process for embryogenesis, wound healing, and cancer progression. It involves a shift in cell interactions, leading to the detachment of epithelial cells and activation of gene programs promoting a mesenchymal state. EMT plays a significant role in cancer metastasis triggering tumor initiation and stemness, and activates metastatic cascades resulting in resistance to therapy. Moreover, reversal of EMT contributes to the formation of metastatic lesions. Metastasis still needs to be better understood functionally in its major but complex steps of migration, invasion, intravasation, dissemination, which contributes to the establishment of minimal residual disease (MRD), extravasation, and successful seeding and growth of metastatic lesions at microenvironmentally heterogeneous sites. Therefore, the current review article intends to present, and discuss comprehensively, the status quo of experimental models able to investigate EMT and metastasis in vitro and in vivo, for researchers planning to enter the field. We emphasize various methods to understand EMT function and the major steps of metastasis, including diverse migration, invasion and matrix degradation assays, microfluidics, 3D co-culture models, spheroids, organoids, or latest spatial and imaging methods to analyze complex compartments. In vivo models such as the chorionallantoic membrane (CAM) assay, cell line-derived and patient-derived xenografts, syngeneic, genetically modified, and humanized mice, are presented as a promising arsenal of tools to analyze intravasation, site specific metastasis, and treatment response. Furthermore, we give a brief overview on methods detecting dissemination and MRD in carcinomas, highlighting its significance in tracking the course of disease and response to treatment. Enhanced lineage tracking tools, dynamic in vivo imaging, and therapeutically useful in vivo models as powerful preclinical tools may still better reveal functional interdependencies between metastasis and EMT. Future directions are discussed in light of emerging views on the biology, diagnosis, and treatment of EMT and metastasis.
Collapse
Affiliation(s)
- Heike Allgayer
- Department of Experimental Surgery-Cancer Metastasis, Mannheim Medical Faculty, Ruprecht-Karls University of Heidelberg, Ludolf-Krehl-Str. 13-17, Mannheim, 68167, Germany.
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India
| | - Suryendu Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India
| | - Venketesh K Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India
| | - Nitin S Patil
- Department of Experimental Surgery-Cancer Metastasis, Mannheim Medical Faculty, Ruprecht-Karls University of Heidelberg, Ludolf-Krehl-Str. 13-17, Mannheim, 68167, Germany
| | - Jörg H Leupold
- Department of Experimental Surgery-Cancer Metastasis, Mannheim Medical Faculty, Ruprecht-Karls University of Heidelberg, Ludolf-Krehl-Str. 13-17, Mannheim, 68167, Germany
| | - Gopal C Kundu
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, 751024, India.
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to Be University, Bhubaneswar, 751024, India.
| |
Collapse
|
2
|
Zhang J, Yin R, Xue Y, Qin R, Wang X, Wu S, Zhu J, Li YS, Zhang C, Wei Y. Advances in the study of epithelial mesenchymal transition in cancer progression: Role of miRNAs. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:69-90. [PMID: 40185337 DOI: 10.1016/j.pbiomolbio.2025.04.001] [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: 10/06/2024] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Epithelial-mesenchymal transition (EMT) has been extensively studied for its roles in tumor metastasis, the generation and maintenance of cancer stem cells and treatment resistance. Epithelial mesenchymal plasticity allows cells to switch between various states within the epithelial-mesenchymal spectrum, resulting in a mixed epithelial/mesenchymal phenotypic profile. This plasticity underlies the acquisition of multiple malignant features during cancer progression and poses challenges for EMT in tumors. MicroRNAs (miRNAs) in the microenvironment affect numerous signaling processes through diverse mechanisms, influencing physiological activities. This paper reviews recent advances in EMT, the role of different hybrid states in tumor progression, and the important role of miRNAs in EMT. Furthermore, it explores the relationship between miRNA-based EMT therapies and their implications for clinical practice, discussing how ongoing developments may enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Jia Zhang
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Runting Yin
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China.
| | - Yongwang Xue
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Rong Qin
- Department of Medical Oncology, Jiangsu University Affiliated People's Hospital, Zhenjiang Clinical Medical College of Nanjing Medical University, Zhenjiang, China
| | - Xuequan Wang
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Shuming Wu
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Jun Zhu
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Yan-Shuang Li
- Department of Breast Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Cai Zhang
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Yuan Wei
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China.
| |
Collapse
|
3
|
Oikawa Y, Umakoshi M, Suzuki K, Kudo-Asabe Y, Miyabe K, Koyama K, Yoshida M, Tanaka M, Nanjo H, Fukuda M, Yamada T, Goto A. Prognostic significance of cancer-associated fibroblasts and tumor-associated macrophages in the tongue squamous cell carcinoma and their correlation with tumor budding. Oral Oncol 2025; 165:107295. [PMID: 40327897 DOI: 10.1016/j.oraloncology.2025.107295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/26/2025] [Accepted: 04/05/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is the most prevalent malignancy of the oral cavity and is characterized by a high propensity for invasion and a poor prognosis. Recent studies have highlighted the critical roles of cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) in tumor progression, particularly within the tumor microenvironment (TME). OBJECTIVE This study aimed to investigate the correlation between CAFs, TAMs, and tumor budding in tongue squamous cell carcinoma (TSCC), and evaluate their impact on prognostic factors. METHODS A total of 88 cases of surgically resected TSCC were analyzed. Immunohistochemical staining was performed using markers of CAFs (fibroblast activation protein, FAP) and TAMs (CD163). The correlation between CAF and TAM scores, tumor budding, and various clinicopathological factors was assessed. TAM scores were evaluated for the number of TAMs in the intratumoral areas (TAM-t) and the invasive front (TAM-fr). CAF scores were evaluated for cancer cells in the intratumoral area (cCAF-t), stromal cells in the intratumoral area (sCAF-t), stromal cells in the invasive front (sCAF-fr), and the infiltration pattern of CAF (IPC). RESULTS The IPC score was significantly associated with the tumor budding scores (p < 0.001) and poor DFS (p < 0.01). In the multivariate analysis, cCAF-t, sCAF-t, and IPC scores emerged as independent prognostic factors (p < 0.05) for early-stage TSCC. CAFs may play a pivotal role in tumor invasion. CONCLUSION These findings indicate that CAFs significantly influence the invasive characteristics of TSCC and are correlated with tumor budding and a poor prognosis. These results underscore the potential of targeting CAFs as a therapeutic strategy for improving OSCC outcome.
Collapse
Affiliation(s)
- Yuki Oikawa
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan; Department of Dentistry and Oral Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Michinobu Umakoshi
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan; Department of Pathology, Akita City Hospital, Akita, Japan.
| | - Kenichiro Suzuki
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan; Department of Dentistry and Oral Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yukitsugu Kudo-Asabe
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Ken Miyabe
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Kei Koyama
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Makoto Yoshida
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroshi Nanjo
- Department of Pathology, Akita University Hospital, Akita, Japan
| | - Masayuki Fukuda
- Department of Dentistry and Oral Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Takechiyo Yamada
- Department of Otorhinolaryngology, Head and Neck Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
4
|
Sun F, Gao X, Wang W, Zhao X, Zhang J, Zhu Y. Predictive biomarkers in the era of immunotherapy for gastric cancer: current achievements and future perspectives. Front Immunol 2025; 16:1599908. [PMID: 40438098 PMCID: PMC12116377 DOI: 10.3389/fimmu.2025.1599908] [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: 03/25/2025] [Accepted: 04/24/2025] [Indexed: 06/01/2025] Open
Abstract
Gastric cancer (GC) is one of the primary contributors to cancer-related mortality on a global scale. It holds a position within the top five most prevalent malignancies both in terms of occurrence and fatality rates. Immunotherapy, as a breakthrough cancer treatment, brings new hope for GC patients. Various biomarkers, such as the expression of programmed death ligand-1 (PD-L1), the microsatellite instability (MSI) status, tumor mutational burden (TMB), and Epstein-Barr virus (EBV) infection, demonstrate potential to predict the effectiveness of immunotherapy in treating GC. Nevertheless, each biomarker has its own limitations, which leads to a significant portion of patients continue to be unresponsive to immunotherapy. With the understanding of the tumor immune microenvironment (TIME), genome sequencing technology, and recent advances in molecular biology, new molecular markers, such as POLE/POLD1mutations, circulating tumor DNA, intestinal flora, lymphocyte activation gene 3 (LAG-3), and lipid metabolism have emerged. This review aims to consolidate clinical evidence to offer a thorough comprehension of the existing and emerging biomarkers. We discuss the mechanisms, prospects of application, and limitations of each biomarker. We anticipate that this review will open avenues for fresh perspectives in the investigation of GC immunotherapy biomarkers and promote the precise choice of treatment modalities for gastric cancer patients, thereby advancing precision immuno-oncology endeavors.
Collapse
Affiliation(s)
- Fujing Sun
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaozhuo Gao
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Wentao Wang
- Department of Gastric Surgery, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaoyan Zhao
- Department of Gynecology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Jingdong Zhang
- Department of Gastroenterology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Yanmei Zhu
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| |
Collapse
|
5
|
Muratoğlu B, Özdemir C, Eylem CC, Reçber T, Nemutlu E, Yet İ, Uçkan-Çetinkaya D. Circadian rhythm and aryl hydrocarbon receptor crosstalk in bone marrow adipose tissue and implications in leukemia. Sci Rep 2025; 15:16387. [PMID: 40350529 PMCID: PMC12066725 DOI: 10.1038/s41598-025-93169-0] [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: 04/26/2024] [Accepted: 03/05/2025] [Indexed: 05/14/2025] Open
Abstract
Leukemic cells modulate the bone marrow microenvironment to enhance their survival. Lipolysis in bone marrow adipose tissue (BMAT) has emerged as a critical factor supporting leukemic cell survival, yet understanding its primary role in leukemia development remains limited. Fanconi anemia (FA), characterized by a predisposition to acute myeloid leukemia (AML) and hypersensitivity to environmental toxins, is a transitional model for studying leukemic transformation. İntegrated multi-omics analyses were conducted on BMAT-derived mesenchymal stem/stromal cells (MSCs) from healthy donors (HD), AML, and FA patients. These analyses revealed intricate interactions among genes, metabolites, and lipids. Particularly noteworthy were the effects observed following the inhibition of aryl hydrocarbon receptor (AhR) signaling by StemRegenin1 (SR1). BMAT-MSCs showed increased expression of epithelial-mesenchymal transition (EMT) genes in FA and AML, suggesting a potential shift towards cancer-associated fibroblasts in the dysregulated marrow microenvironment. Identification of potential circadian rhythm biomarkers (NPAS2, PER2, BHLHE40, PER3, CIART) in BMAT-MSCs indicates a link between related lipid metabolism genes (e.g., PTGS1, PIK3R1) and SR1 treatment, implicating them in lipolysis processes. Dysregulation of circadian rhythm-related genes (CIART, BHLHE40, NPAS2) in AML BMAT-MSCs, along with changes in circulating lipid metabolites like palmitate suggests their role in shaping the leukemia microenvironment. Upregulation of FABP5 and CD36 suggests a novel molecular mechanism involving FABP5 in AhR-mediated circadian regulation and identifies CD36 as a potential partner for FABP5 in BMAT-MSCs. Overall, this study unveils the interplay between AhR signaling, circadian rhythm, and the leukemia microenvironment in BMAT-MSCs, offering new insights into leukemia pathogenesis and therapeutic opportunities.
Collapse
Affiliation(s)
- Bihter Muratoğlu
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Cansu Özdemir
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
| | - Cemil Can Eylem
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - İdil Yet
- Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Division of Hematology, Department of Pediatrics, Hacettepe University Faculty of Medicine, 06100, Sihhiye, Ankara, Turkey.
| |
Collapse
|
6
|
Zhou R, Wu H, You H, Wang X, Yuan X, Sun Z, Zhou D, Jiang Y, Shen Y. ESPN activates ZEB1-mediated EMT through the PI3K/AKT/mTOR axis to promote osteosarcoma metastasis. J Transl Med 2025; 23:527. [PMID: 40346630 PMCID: PMC12065200 DOI: 10.1186/s12967-025-06500-8] [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: 11/01/2024] [Accepted: 04/13/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a primary bone malignancy characterized by early metastasis and generally poor prognosis. ESPN is highly expressed and plays an important role in regulating the aggressive phenotypes of several cancer cell types. However, little is known about the molecular mechanisms underlying ESPN-mediated migration and invasion in OS cells. METHODS In this study, we first analyzed the survival of osteosarcoma patients using Kaplan-Meier analysis to assess the prognostic relevance of ESPN. To further evaluate its clinical significance, we performed immunohistochemical analysis on osteosarcoma tissue samples and benign osteochondroma (OC) tissues. The biological function of ESPN in osteosarcoma was confirmed by a series of experiments conducted both in vitro and in vivo. Additionally, we explored the underlying molecular mechanisms through Western blotting, co-immunoprecipitation, immunofluorescence, and PCR, revealing key downstream signaling pathways. RESULTS In this study, we demonstrate that ESPN, acting as an oncogene, is highly expressed in OS cell lines and tissues, promoting OS cell proliferation and metastasis. Mechanistically, ESPN promoted the phosphorylation of PI3K by direct interaction with it and active the AKT/mTOR pathway, which enhanced the expression of the transcription factor ZEB1 and initiating the epithelial-mesenchymal transition (EMT) cascade. Furthermore, we validated that mTOR-mediated activation of p70 ribosomal protein S6 kinase (p70S6K) promotes the translation of ZEB1, thereby enhancing the growth and motility of OS cells. CONCLUSIONS Our findings reveal a previously unrecognized function of ESPN in OS, closely linked with EMT and cancer metastasis progression. Targeting ESPN may represent a potential therapeutic approach for patients with OS.
Collapse
Affiliation(s)
- Ruikai Zhou
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Hongyu Wu
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Hao You
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Xiaofei Wang
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Xiuchen Yuan
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Zhengyi Sun
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Dong Zhou
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China.
- Affiliated with Changzhou Children's Hospital of Nantong University, Changzhou, China.
| | - Yuqing Jiang
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China.
| | - Yifei Shen
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China.
| |
Collapse
|
7
|
Li X, Wu Y, Xie B, Xu M, Xie T, Yue W, Lin M, Lin Y, Chen Y. SPP1 Promotes NSCLC Brain Metastasis Via Sequestration of Ubiquitin Ligase RNF114 to Facilitate P85α Ubiquitination. Mol Carcinog 2025; 64:829-841. [PMID: 39918025 DOI: 10.1002/mc.23866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/04/2024] [Accepted: 12/02/2024] [Indexed: 04/12/2025]
Abstract
Brain metastasis (BM) is a significant factor contributing to the poor prognosis of patients with non-small cell lung cancer (NSCLC). Secreted phosphoprotein 1 (SPP1) is implicated in the progression and metastasis of several cancers. The role of SPP1 in NSCLC remains unclear, especially in NSCLC BM. This study aimed to identify genes associated with NSCLC BM and to investigate the involvement of SPP1 in NSCLC BM. Integrated genomic analysis was utilized to identify candidate genes in NSCLC. The expression levels of SPP1 were evaluated in NSCLC tissues and cell lines. In vitro and in vivo experiments were conducted to assess the effect of SPP1 on NSCLC cell behavior and BM. The potential mechanisms of SPP1 were demonstrated by CO-IP and liquid chromatography-mass spectrometry (LC-MS). The underlying mechanism involving the PI3K/AKT/mTOR pathway was explored. The results showed that SPP1 expression was upregulated in NSCLC tissues and cell lines. Depletion of SPP1 using shRNA inhibited cell proliferation, migration, and invasion in vitro and suppressed BM in vivo. Mechanistically, SPP1 facilitates the ubiquitination of P85α by interacting with the ubiquitin ligase RNF114, thus playing a role in regulating NSCLC BM through the PI3K/AKT/mTOR signaling pathway. Moreover, immunohistochemistry staining confirmed higher expression of SPP1 in NSCLC tissues with BM compared to those without BM. In summary, elevated SPP1 expression was associated with poor clinical outcomes in NSCLC patients. This study highlights the role of SPP1 as a regulator of cell metastasis and suggests its potential as a novel therapeutic target for BM in NSCLC.
Collapse
Affiliation(s)
- Xiaoqin Li
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| | - Yun Wu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Center for Geriatrics, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Baosong Xie
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| | - Mingxiao Xu
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai, China
| | - Tianjian Xie
- Xiapu County Hospital of Fujian Province, Ningde, China
| | - Wenxiang Yue
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Ming Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Ying Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, China
| | - Yusheng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| |
Collapse
|
8
|
Wei H, Li X, Feng P, He Z. SERPINH1 and CTSZ are Key Markers of Glioma Angiogenesis. J Mol Neurosci 2025; 75:51. [PMID: 40257572 DOI: 10.1007/s12031-025-02349-0] [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: 02/07/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Glioma, as one of the most complex and prognostically variable malignant tumors of the central nervous system, poses a significant challenge to clinical decision-making due to its molecular heterogeneity. This study aims to deepen our understanding of glioma molecular subtypes and explore key gene markers with prognostic and diagnostic value. We utilized an angiogenesis-related gene set and employed the Non-negative Matrix Factorization (NMF) algorithm to successfully identify two distinct prognostic subtypes, with subtype one exhibiting more unfavorable prognostic characteristics. To further elucidate the biological functional differences between these two subtypes, we conducted Gene Ontology (GO) functional annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and Gene Set Enrichment Analysis (GSEA). Building on this, we integrated differentially expressed genes between subtypes with core genes revealed by Weighted Gene Co-expression Network Analysis (WGCNA) through intersection analysis to pinpoint a series of key candidate genes. Subsequently, we constructed a Protein-Protein Interaction (PPI) network to identify genes occupying central nodes within the network. To screen markers with high specificity and sensitivity for prognosis and diagnosis, we adopted a dual-track strategy: on the one hand, we utilized machine learning algorithms such as Lasso regression, Support Vector Machine (SVM), and Random Forest (RF) to select core genes, identifying markers that can accurately predict the subtype with a poor prognosis; on the other hand, we employed a comprehensive evaluation system incorporating 101 machine learning ensemble algorithms to further validate and screen prognosis-related genes. Through cross-validation using these two strategies, we ultimately determined SERPINH1 and CTSZ as dual prognostic and diagnostic markers for glioma. This study not only provides a new perspective and tool for the molecular subclassification of glioma but also, through a rigorous multi-algorithm, multi-dimensional screening process, uncovers SERPINH1 and CTSZ as markers with potential clinical translational value. These findings are expected to offer more precise biomarker support for the early diagnosis and prognostic assessment of glioma, potentially paving new avenues for the development of personalized treatment strategies and improving patient outcomes. This has far-reaching implications for the clinical management of glioma in the field of neurosurgery.
Collapse
Affiliation(s)
- Haotian Wei
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xinlong Li
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Lanzhou, 730030, China
| | - Peng Feng
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Lanzhou, 730030, China
| | - Zhaohui He
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
9
|
Khalili-Tanha G, Radisky ES, Radisky DC, Shoari A. Matrix metalloproteinase-driven epithelial-mesenchymal transition: implications in health and disease. J Transl Med 2025; 23:436. [PMID: 40217300 PMCID: PMC11992850 DOI: 10.1186/s12967-025-06447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells, defined by apical-basal polarity and tight intercellular junctions, acquire migratory and invasive properties characteristic of mesenchymal cells. Under normal conditions, EMT directs essential morphogenetic events in embryogenesis and supports tissue repair. When dysregulated, EMT contributes to pathological processes such as organ fibrosis, chronic inflammation, and cancer progression and metastasis. Matrix metalloproteinases (MMPs)-a family of zinc-dependent proteases that degrade structural components of the extracellular matrix-sit at the nexus of this transition by dismantling basement membranes, activating pro-EMT signaling pathways, and cleaving adhesion molecules. When normally regulated, MMPs promote balanced ECM turnover and support the cyclical remodeling necessary for proper development, wound healing, and tissue homeostasis. When abnormally regulated, MMPs drive excessive ECM turnover, thereby promoting EMT-related pathologies, including tumor progression and fibrotic disease. This review provides an integrated overview of the molecular mechanisms by which MMPs both initiate and sustain EMT under physiological and disease conditions. It discusses how MMPs can potentiate EMT through TGF-β and Wnt/β-catenin signaling, disrupt cell-cell junction proteins, and potentiate the action of hypoxia-inducible factors in the tumor microenvironment. It discusses how these pathologic processes remodel tissues during fibrosis, and fuel cancer cell invasion, metastasis, and resistance to therapy. Finally, the review explores emerging therapeutic strategies that selectively target MMPs and EMT, ranging from CRISPR/Cas-mediated interventions to engineered tissue inhibitors of metalloproteinases (TIMPs), and demonstrates how such approaches may suppress pathological EMT without compromising its indispensable roles in normal biology.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
10
|
Castaneda M, den Hollander P, Werden S, Ramirez-Peña E, Vasaikar SV, Kuburich NA, Gould C, Soundararajan R, Mani SA. β-Catenin Drives the FOXC2-Mediated Epithelial-Mesenchymal Transition and Acquisition of Stem Cell Properties. Cancers (Basel) 2025; 17:1114. [PMID: 40227590 PMCID: PMC11987759 DOI: 10.3390/cancers17071114] [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: 11/01/2024] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Aggressive forms of breast cancer, such as triple-negative breast cancer (TNBC), are associated with an increase in cancer cells that exhibit stem cell properties. The activation of the epithelial-mesenchymal transition (EMT) program, mediated by the transcription factor FOXC2, generates these stem-like cells. FOXC2 is linked to poor prognoses across various cancer types and is notably upregulated in TNBC, where it establishes and sustains these stem-like cells within the tumor population. Methods: Here, we decode the pathways regulating FOXC2 activation using EMT-enriched cell line models. Stemness was assessed using mammosphere assays and mesenchymal markers by western blot. Expression correlations with clinical data was examined using the EMTome. Results: We demonstrate that β-catenin serves as a critical mediator of mesenchymal and stemness characteristics through FOXC2 upregulation. By disrupting β-catenin, we find that FOXC2 expression, mesenchymal properties, and stemness are reduced; however, the introduction of exogenous FOXC2 expression in β-catenin deficient cells is enough to restore the mesenchymal and stemness phenotype. These findings support the idea that FOXC2 acts as the downstream regulator of β-catenin and influences both mesenchymal and stemness properties. Moreover, there is a positive correlation between the expression of β-catenin and FOXC2 in various cancer subtypes observed in clinical patient samples. Conclusions: Our study clarifies the role of the β-catenin/FOXC2 signaling axis in maintaining stemness properties, suggesting potential targets for TNBC and other cancers driven by EMT-related mesenchymal and stemness characteristics.
Collapse
Affiliation(s)
- Maria Castaneda
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Research Center, Houston, TX 77030, USA; (M.C.); (R.S.)
| | - Petra den Hollander
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (P.d.H.); (N.A.K.); (C.G.)
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Steve Werden
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Research Center, Houston, TX 77030, USA; (M.C.); (R.S.)
| | - Esmeralda Ramirez-Peña
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Research Center, Houston, TX 77030, USA; (M.C.); (R.S.)
| | - Suhas V. Vasaikar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Research Center, Houston, TX 77030, USA; (M.C.); (R.S.)
| | - Nick A. Kuburich
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (P.d.H.); (N.A.K.); (C.G.)
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Claire Gould
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (P.d.H.); (N.A.K.); (C.G.)
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Research Center, Houston, TX 77030, USA; (M.C.); (R.S.)
| | - Sendurai A. Mani
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (P.d.H.); (N.A.K.); (C.G.)
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| |
Collapse
|
11
|
Zhu W, Sun J, Jing F, Xing Y, Luan M, Feng Z, Ma X, Wang Y, Jia Y. GLI2 inhibits cisplatin sensitivity in gastric cancer through DEC1/ZEB1 mediated EMT. Cell Death Dis 2025; 16:204. [PMID: 40133270 PMCID: PMC11937514 DOI: 10.1038/s41419-025-07564-6] [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: 07/16/2024] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Cisplatin (CDDP) based chemotherapy has emerged as the predominant therapeutic regimen for patients with advanced gastric cancer (GC). However, its efficacy is dampened by the development of chemoresistance, which results in poor prognosis of patients. GLI2, a key transcription factor in the Hedgehog (Hh) signaling pathway, is regarded as a target for cancer therapy. However, the significance of GLI2 for CDDP resistance in GC has not been well established. Here, we show that GLI2 expression was upregulated in EMT-type GC and associated with poor prognosis. GLI2 promotes proliferation, migration, and CDDP resistance of GC cells by inducing EMT. In terms of mechanism, GLI2 binds to the promoter region of DEC1 and enhances its expression, thereby co-transcriptionally regulating ZEB1 expression. Animal experiments have demonstrated that both GLI2 knockdown and GLI2 inhibitor significantly enhance CDDP sensitivity in GC. Our data not only identify a novel GLI2/DEC1/ZEB1/EMT pathway in GC CDDP resistance but also provide novel strategies to treat GC in the future.
Collapse
Affiliation(s)
- Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Jingguo Sun
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Fubo Jing
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Zhaotian Feng
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China.
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China.
| | - Yanfei Jia
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China.
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China.
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, People's Republic of China.
| |
Collapse
|
12
|
Li J, Wang ZY, Jin Y, Xu J, Ya YJ, Wan TQ, Li X, Wang X. Transmembrane channel-like 5 drives hepatocellular carcinoma progression by regulating epithelial-mesenchymal transition. World J Clin Oncol 2025; 16:94091. [PMID: 40130046 PMCID: PMC11866081 DOI: 10.5306/wjco.v16.i3.94091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a difficult cancer to manage due to its highly invasive and metastatic nature. AIM To investigate the molecular function of transmembrane channel-like 5 (TMC5) in vitro and in vivo, with the objective of identifying novel diagnosis and treatment targets for HCC. METHODS The expression of TMC in cancer and normal tissues, along with its correlation with HCC prognosis, was analyzed using the GENT2, GEPIA database, and Human Protein Atlas. COX analysis was conducted to assess the relationship between TMC5 expression and overall survival in TCGA-LIHC patients. Further experiments were conducted to investigate the effect of TMC5 in cancer progression through loss- and gain-of-function assays in vitro and in vivo. RESULTS Bioinformatics revealed that TMC5 expression was generally higher in tumors than in normal tissues, and its expression was associated with poorer patient survival outcomes. TMC5 expression in HCC tissues and cells was consistent with the results of the bioinformatics analysis. Suppression of TMC5 expression reduced migration, invasion, and proliferation, while also decreasing the expression of epithelial-mesenchymal transition (EMT)-associated molecules in MHCC97-LM3 cells. Conversely, higher TMC5 expression significantly increased cell migration, invasion, proliferation, and EMT in MHCC97 L cells. TMC5 knockdown significantly decreased both the formation and spread of nodules in liver tissue, whereas TMC5 overexpression promoted them. CONCLUSION Our study provides compelling evidence that TMC5 is highly expressed in HCC and drives cancer progression through the activation of EMT-mediated invasion. TMC5 could represent a valuable molecular target for the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Jiao Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Zi-Yu Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Yan Jin
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Jing Xu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Yun-Jin Ya
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Ting-Qiu Wan
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Xi Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Xi Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| |
Collapse
|
13
|
Wei W, Dang Y, Chen G, Han C, Zhang S, Zhu Z, Bie X, Xue J. Comprehensive analysis of senescence-related genes identifies prognostic clusters with distinct characteristics in glioma. Sci Rep 2025; 15:9540. [PMID: 40108265 PMCID: PMC11923138 DOI: 10.1038/s41598-025-93482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
Cellular senescence, defined as a state of permanent arrest in cell growth, is regarded as a crucial tumor suppression mechanism. However, accumulating scientific evidence suggests that senescent cells play a detrimental role in the progression of cancer. Unfortunately, the current lack of reliable markers that specifically reflect the level of senescence in cancer greatly hinders our in-depth understanding of this important biological foundation. Therefore, the search for more specific and reliable markers to reveal the specific role of senescent cells in cancer progression is particularly urgent and important. To uncover the role of senescence in gliomas, we collected senescence-related genes for integrated analysis. Consensus clustering was used to subtype gliomas based on the senescence gene set, and we identified two robust prognostic clusters of gliomas with distinct survival outcomes, multi-omics landscapes, immune characteristics, and differential drug responses. Multiple external datasets were used to validate the stability of our subtypes. Various computational and experimental methods, including WGCNA (Weighted Gene Co-expression Network Analysis), ssGSEA (single-sample Gene Set Enrichment Analysis), and machine learning algorithms (lasso regression, support vector machines, random forests), were employed for analysis. We found that CEBPB and LMNA are associated with poor prognosis in gliomas and may mediate immunosuppression and tumor proliferation. Drug prediction indicated that dasatinib is a potential therapeutic agent. Our findings provide insights into the role of the senescence gene set in patient stratification and precision medicine.
Collapse
Affiliation(s)
- Wenyuan Wei
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Ying Dang
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
- Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Gang Chen
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Chao Han
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Siwei Zhang
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Ziqiang Zhu
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Xiaohua Bie
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| | - Jungang Xue
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
14
|
Wang M, Huang X, Zhang D, Liu Y, Liu P. The role of fructose-1,6-bisphosphatase 1 on regulating the cancer progression and drug resistance. Discov Oncol 2025; 16:346. [PMID: 40100307 PMCID: PMC11920503 DOI: 10.1007/s12672-025-02112-2] [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: 12/09/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
Fructose-1,6-bisphosphatase 1 (FBP1) is the enzyme that limits the process of gluconeogenesis as it facilitates the hydrolysis of fructose-1,6-bisphosphate(F-1,6-BP) to produce fructose-6-phosphate(F6P) and inorganic phosphate. Gluconeogenesis is the production of glucose from small carbohydrate substrates. The gluconeogenic process is typically suppressed in cancer because it inhibits glycolysis. Apart from its involvement in cellular glucose metabolism, FBP1 also plays a role in gene transcription, mRNA translation and stability regulation, and the immune microenvironment of tumors. Because of its multifaceted functions, the mechanisms by which FBP1 is involved in tumor development are complex. Moreover, FBP1 deficiency is associated with radiation and chemotherapy resistance and poor prognosis in cancer patients. Restoration of FBP1 expression in cancer cells is expected to hold promise for cancer therapy. However, up to now few reviews have systematically summarized the important functional mechanisms of FBP1 in tumorigenesis and the small molecule compounds that restore FBP1 expression. Therefore, this article addresses the question "How does FBP1 contribute to cancer progression, and can targeting FBP1 be a potential therapeutic approach?" by summarizing the effects of FBP1 on cancer development and progression as well as its mediated drug resistance and the future clinical applications of potential small molecule modulators targeting FBP1.
Collapse
Affiliation(s)
- Mengmeng Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Xiaoju Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Dan Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yisan Liu
- Department of Urology, People's Hospital of Cili, Cili, 427200, Hunan, China.
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| |
Collapse
|
15
|
Lin H, Hua J, Wang Y, Chen M, Liang Y, Yan L, Zhao W, Luo S, Hong D, Chen X, Pan X, Liu J, Liu Z. Prognostic and predictive values of a multimodal nomogram incorporating tumor and peritumor morphology with immune status in resectable lung adenocarcinoma. J Immunother Cancer 2025; 13:e010723. [PMID: 40050046 PMCID: PMC11887283 DOI: 10.1136/jitc-2024-010723] [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: 10/02/2024] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Current prognostic and predictive biomarkers for lung adenocarcinoma (LUAD) predominantly rely on unimodal approaches, limiting their characterization ability. There is an urgent need for a comprehensive and accurate biomarker to guide individualized adjuvant therapy decisions. METHODS In this retrospective study, data from patients with resectable LUAD (stage I-III) were collected from two hospitals and a publicly available dataset, forming a training dataset (n=223), a validation dataset (n=95), a testing dataset (n=449), and the non-small cell lung cancer (NSCLC) Radiogenomics dataset (n=59). Tumor and peritumor scores were constructed from preoperative CT radiomics features (shape/intensity/texture). An immune score was derived from the density of tumor-infiltrating lymphocytes (TILs) within the cancer epithelium and stroma on hematoxylin and eosin-stained whole-slide images. A clinical score was constructed based on clinicopathological risk factors. A Cox regression model was employed to integrate these scores, thereby constructing a multimodal nomogram to predict disease-free survival (DFS). The adjuvant chemotherapy benefit rate was subsequently calculated based on this nomogram. RESULTS The multimodal nomogram outperformed each of the unimodal scores in predicting DFS, with a C-index of 0.769 (vs 0.634-0.731) in the training dataset, 0.730 (vs 0.548-0.713) in the validation dataset, and 0.751 (vs 0.660-0.692) in the testing dataset. It was independently associated with DFS after adjusting for other clinicopathological risk factors (training dataset: HR=3.02, p<0.001; validation dataset: HR=2.33, p<0.001; testing dataset: HR=2.03, p=0.001). The adjuvant chemotherapy benefit rate effectively distinguished between patients benefiting from adjuvant chemotherapy and those from observation alone (interaction p<0.001). Furthermore, the high-/low-risk groups defined by the multimodal nomogram provided refined stratification of candidates for adjuvant chemotherapy identified by current guidelines (p<0.001). Gene set enrichment analyses using the NSCLC Radiogenomics dataset revealed associations between tumor/peritumor scores and pathways involved in epithelial-mesenchymal transition, angiogenesis, IL6-JAK-STAT3 signaling, and reactive oxidative species. CONCLUSION The multimodal nomogram, which incorporates tumor and peritumor morphology with anti-tumor immune response, provides superior prognostic accuracy compared with unimodal scores. Its defined adjuvant chemotherapy benefit rates can inform individualized adjuvant therapy decisions.
Collapse
Affiliation(s)
- Huan Lin
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Junjie Hua
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yumeng Wang
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, Guangxi, China
| | - Mingwei Chen
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, Guangxi, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - LiXu Yan
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Zhao
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shiwei Luo
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Deqing Hong
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xipeng Pan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, Guangxi, China
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Shi M, He Y, Zhong X, Huang H, Hua J, Wang S, Xu J, Zhao S, Liang H, Huang Y. A Smart mRNA-Initiated Theranostic Multi-shRNA Nanofactory for Precise and Efficient Cancer Gene Therapy. Adv Healthc Mater 2025; 14:e2404159. [PMID: 39790038 DOI: 10.1002/adhm.202404159] [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: 10/24/2024] [Revised: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Despite the significant potential of short hairpin RNA (shRNA)-mediated gene therapy for various diseases, the clinical success of cancer treatment remains poor, partly because of low selectivity and low efficiency. In this study, an mRNA-initiated autonomous multi-shRNA nanofactory (RNF@CM) is designed for in vivo amplification imaging and precise cancer treatment. The RNF@CM consists of a gold nanoparticle core, an interlayer of two types of three-stranded DNA/RNA hybrid probes, one of which is bound to aptamer-inhibited DNA polymerases, and an outer layer of the cancer cell membrane. After the specific delivery of RNF@CM into target cancer cells, an intracellular tumour-related mRNA target can initiate the RNF@CM with a circular strand-displacement polymerisation reaction, resulting in the release of significantly amplified fluorescence and continuous production of three types of shRNAs. The RNF@CM effectively distinguished cancer cells from normal cells, exclusively produced multiple shRNAs in response to a specific mRNA target in cancer cells, accurately diagnosed tumours in vivo, and significantly inhibited tumour growth with negligible toxicity, expanding the toolbox for on-demand gene delivery and precision theranostics.
Collapse
Affiliation(s)
- Ming Shi
- Department of Chemistry and Pharmacy, Guilin Normal College, Guilin, 541004, China
| | - Yifang He
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Xiaohong Zhong
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Huakui Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Jing Hua
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Shulong Wang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, Yulin, 537000, China
| | - Jiayao Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Shulin Zhao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Yong Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| |
Collapse
|
17
|
Wang L, Li N, Chen Y, Qiao Y, Song Y, Zhang X, Zhao H, Ran W, Li G, Xing X. GPSM1 interacts and cooperates with MMP19 to promote proliferation and EMT in colorectal cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119903. [PMID: 39855604 DOI: 10.1016/j.bbamcr.2025.119903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Among patients with colorectal cancer (CRC), metastasis accounts for the majority of deaths, and epithelial-mesenchymal transition (EMT) is important in the metastatic process. However, the mechanism underlying the correlation between the two in CRC is unknown. Here, we verified that a receptor-independent protein, G-protein signaling modulator 1 (GPSM1), was increased in CRC and had a significant positive correlation with matrix metalloproteinase 19 (MMP19). GPSM1 and MMP19 knockdown or overexpression decreased and increased proliferation, migration and invasion of CRC cells, respectively. In addition, overexpression or knockdown of GPSM1 and MMP19 upregulated and inhibited EMT, respectively. Interfering with MMP19 reversed EMT activation via GPSM1 overexpression. Apoptosis was induced by GPSM1 and MMP19 knockdown and activated the caspase3/Bcl-2/Bax signaling pathway. In conclusion, these results support the role of GPSM1 and MMP19 in CRC progression.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Na Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Chen
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yehua Qiao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaolin Song
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangyan Zhang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Han Zhao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenwen Ran
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guangqi Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoming Xing
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
18
|
Lu Y, Huang Y, Zhu C, Li Z, Zhang B, Sheng H, Li H, Liu X, Xu Z, Wen Y, Zhang J, Zhang L. Cancer brain metastasis: molecular mechanisms and therapeutic strategies. MOLECULAR BIOMEDICINE 2025; 6:12. [PMID: 39998776 PMCID: PMC11861501 DOI: 10.1186/s43556-025-00251-0] [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: 09/18/2024] [Revised: 01/06/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Brain metastases (BMs) are the most common intracranial tumors in adults and the major cause of cancer-related morbidity and mortality. The occurrence of BMs varies according to the type of primary tumors with most frequence in lung cancer, melanoma and breast cancer. Among of them, lung cancer has been reported to have a higher risk of BMs than other types of cancers with 40 ~ 50% of such patients will develop BMs during the course of disease. BMs lead to many neurological complications and result in a poor quality of life and short life span. Although the treatment strategies were improved for brain tumors in the past decades, the prognosis of BMs patients is grim. Poorly understanding of the molecular and cellular characteristics of BMs and the complicated interaction with brain microenvironment are the major reasons for the dismal prognosis of BM patients. Recent studies have enhanced understanding of the mechanisms of BMs. The newly identified potential therapeutic targets and the advanced therapeutic strategies have brought light for a better cure of BMs. In this review, we summarized the mechanisms of BMs during the metastatic course, the molecular and cellular landscapes of BMs, and the advances of novel drug delivery systems for overcoming the obstruction of blood-brain barrier (BBB). We further discussed the challenges of the emerging therapeutic strategies, such as synergistic approach of combining targeted therapy with immunotherapy, which will provide vital clues for realizing the precise and personalized medicine for BM patients in the future.
Collapse
Affiliation(s)
- Yu Lu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunhang Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chenyan Zhu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhidan Li
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Bin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui Sheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haotai Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xixi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongwen Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Wen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liguo Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Haynes J, Manogaran P. Mechanisms and Strategies to Overcome Drug Resistance in Colorectal Cancer. Int J Mol Sci 2025; 26:1988. [PMID: 40076613 PMCID: PMC11901061 DOI: 10.3390/ijms26051988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related mortality worldwide, with a significant impact on public health. Current treatment options include surgery, chemotherapy, radiotherapy, molecular-targeted therapy, and immunotherapy. Despite advancements in these therapeutic modalities, resistance remains a significant challenge, often leading to treatment failure, poor progression-free survival, and cancer recurrence. Mechanisms of resistance in CRC are multifaceted, involving genetic mutations, epigenetic alterations, tumor heterogeneity, and the tumor microenvironment. Understanding these mechanisms at the molecular level is crucial for identifying novel therapeutic targets and developing strategies to overcome resistance. This review provides an overview of the diverse mechanisms driving drug resistance in sporadic CRC and discusses strategies currently under investigation to counteract this resistance. Several promising strategies are being explored, including targeting drug transport, key signaling pathways, DNA damage response, cell death pathways, epigenetic modifications, cancer stem cells, and the tumor microenvironment. The integration of emerging therapeutic approaches that target resistance mechanisms aims to enhance the efficacy of current CRC treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Jennifer Haynes
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive, Huntington, WV 25701, USA;
| | | |
Collapse
|
20
|
Wang JF, Wang MC, Jiang LL, Lin NM. The neuroscience in breast cancer: Current insights and clinical opportunities. Heliyon 2025; 11:e42293. [PMID: 39975839 PMCID: PMC11835589 DOI: 10.1016/j.heliyon.2025.e42293] [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: 08/06/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/21/2025] Open
Abstract
The involvement of nerves in the development of breast cancer has emerged as a significant factor. Interaction between the nervous system and breast cancer can influence tumor initiation, growth, invasion, metastasis, reverse resistance to drugs, promote inflammation in tumors, and impair the immune system's ability to combat cancer. This review examined the intricate relationship linking the nervous system with breast cancer, emphasizing both central and peripheral aspects of the nervous system. Moreover, we reviewed neural cell factors and their impact on breast cancer progression, alongside the interactions between nerves and immunology, microbiota in breast cancer. Furthermore, the study discussed the potential of nerves as biomarkers for diagnosing and prognosticating breast cancer, and evaluated prospects for improving chemotherapy and immunotherapy therapeutic outcomes in breast cancer treatment. We hope to provide a deeper understanding of the neurobiological underpinnings of breast cancer and pave the way for the discovery of innovative therapeutic targets and prognostic markers.
Collapse
Affiliation(s)
- Jia-feng Wang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Meng-chuan Wang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, China
| | - Lei-lei Jiang
- The First Affiliated Hospital of Anhui University of Chinese Medicine,Hefei, 230031, China
| | - Neng-ming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, 310024, China
| |
Collapse
|
21
|
Yang S, Seo J, Choi J, Kim SH, Kuk Y, Park KC, Kang M, Byun S, Joo JY. Towards understanding cancer dormancy over strategic hitching up mechanisms to technologies. Mol Cancer 2025; 24:47. [PMID: 39953555 PMCID: PMC11829473 DOI: 10.1186/s12943-025-02250-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025] Open
Abstract
Delving into cancer dormancy has been an inherent task that may drive the lethal recurrence of cancer after primary tumor relief. Cells in quiescence can survive for a short or long term in silence, may undergo genetic or epigenetic changes, and can initiate relapse through certain contextual cues. The state of dormancy can be induced by multiple conditions including cancer drug treatment, in turn, undergoes a life cycle that generally occurs through dissemination, invasion, intravasation, circulation, immune evasion, extravasation, and colonization. Throughout this cascade, a cellular machinery governs the fate of individual cells, largely affected by gene regulation. Despite its significance, a precise view of cancer dormancy is yet hampered. Revolutionizing advanced single cell and long read sequencing through analysis methodologies and artificial intelligence, the most recent stage in the research tool progress, is expected to provide a holistic view of the diverse aspects of cancer dormancy.
Collapse
Affiliation(s)
- Sumin Yang
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Jieun Seo
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Jeonghyeon Choi
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Sung-Hyun Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Yunmin Kuk
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Kyung Chan Park
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Mingon Kang
- Department of Computer Science, University of Nevada, Las Vegas, NV, 89154, USA
| | - Sangwon Byun
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea.
| | - Jae-Yeol Joo
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea.
- Department of Pharmacy, College of Pharmacy, Hanyang University, Rm 407, Bldg.42, 55 Hanyangdaehak-ro, Sangnok-gu Ansan, Gyeonggi-do, 15588, Republic of Korea.
| |
Collapse
|
22
|
Zhou W, Cheng H, Fan C, Zhou X, Chen W, Xie C, Hu Y, Chen Y, Wang X, Wu J. LAMP3-mediated epithelial-mesenchymal transition promotes the invasion and excessive proliferation of fibroblast-like synoviocytes in rheumatoid arthritis. J Autoimmun 2025; 151:103359. [PMID: 39799877 DOI: 10.1016/j.jaut.2025.103359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/27/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
OBJECTIVE The aim of this study was to explore the functional role of LAMP3-mediated epithelial-mesenchymal transition (EMT) in fibroblast-like synoviocytes (FLSs) in rheumatoid arthritis (RA) patients and to evaluate its potential as a therapeutic target. METHODOLOGY Changes in EMT and LAMP3 were investigated in the synovial tissue and FLSs of RA patients. In vitro experiments were performed using the EMT inhibitor C19, siRNA, and lentivirus to examine the impact of EMT and LAMP3 on RA-FLSs and the underlying mechanisms involved. Finally, C19 was administered to mice with collagen-induced arthritis (CIA) to validate the therapeutic efficacy of C19 in treating arthritis. RESULTS Compared with patients with osteoarthritis (OA), RA patients exhibited increased EMT and increased expression of LAMP3 in the synovium. The results from the in vitro experiments demonstrated that inhibiting EMT effectively reduced the excessive proliferation, anti-senescent properties, migration, and invasive behavior of RA-FLSs, as well as the secretion of MMP1, MMP3, and MMP13. Additionally, regulating the expression of LAMP3 not only affected the EMT pathway but also impacted the excessive proliferation and invasive behavior of RA-FLSs. In the CIA model, administration of the EMT inhibitor C19 significantly alleviated the progression of arthritis. CONCLUSION These findings demonstrate the inhibitory impact of EMT on arthritis and suggest that inhibiting EMT or LAMP3 may be a promising novel therapeutic approach for treating RA.
Collapse
Affiliation(s)
- Wenxian Zhou
- Institute of Genomic Medicine, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Hui Cheng
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Chenghu Fan
- Institute of Genomic Medicine, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Xin Zhou
- The Second School of Medicine, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Wenyu Chen
- Institute of Genomic Medicine, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Chenglong Xie
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Yuezheng Hu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Yue Chen
- Institute of Genomic Medicine, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Xiaobing Wang
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China; Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China.
| | - Jinyu Wu
- Institute of Genomic Medicine, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China.
| |
Collapse
|
23
|
König J, Rokavec M, Öner-Ziegler MG, Fei Y, Hermeking H. Myeloid Mir34a suppresses colitis-associated colon cancer: characterization of mediators by single-cell RNA sequencing. Cell Death Differ 2025; 32:225-241. [PMID: 39425000 PMCID: PMC11802797 DOI: 10.1038/s41418-024-01380-9] [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: 04/24/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 10/21/2024] Open
Abstract
We have previously shown that general deletion of the gene encoding the p53-inducible Mir34a microRNA enhances the number and invasion of colitis-associated colorectal cancers (CACs) in mice. Since the p53-pathway has been implicated in tumor-suppression mediated by cells in the tumor microenvironment (TME) we deleted Mir34a in myeloid cells and characterized CACs in these with scRNA-Seq (single cell RNA sequencing). This revealed an increase in specific macrophage subtypes, such as Cdk8+ macrophages and Mrc1+, M2-like macrophages. The latter displayed elevated expression of 21 known Mir34a target mRNAs, including Csf1r, Axl, Foxp1, Ccr1, Nampt, and Tgfbr2, and 32 predicted Mir34a target mRNAs. Furthermore, Mir34a-deficient BMDMs showed enhanced migration, elevated expression of Csf1r and a shift towards M2-like polarization when compared to Mir34a-proficient BMDMs. Concomitant deletion of Csf1r or treatment with a Csf1r inhibitor reduced the CAC burden and invasion in these mice. Notably, loss of myeloid Mir34a function resulted in a prominent, inflammatory CAC cell subtype, which displayed epithelial and macrophage markers. These cells displayed high levels of the EMT transcription factor Zeb2 and may therefore enhance the invasiveness of CACs. Taken together, our results provide in vivo evidence for a tumor suppressive role of myeloid Mir34a in CACs which is, at least in part, mediated by maintaining macrophages in an M1-like state via repression of Mir34a targets, such as Csf1r. Collectively, these findings may serve to identify new therapeutic targets and approaches for treatment of CAC.
Collapse
Affiliation(s)
- Janine König
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany
| | - Matjaz Rokavec
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany
| | - Meryem Gülfem Öner-Ziegler
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany
| | - Ye Fei
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany
| | - Heiko Hermeking
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany.
- German Cancer Consortium (DKTK), Partner site Munich, D-80336, Munich, Germany.
- German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany.
| |
Collapse
|
24
|
Keeling J, Falchook G. Oncology Clinical Trials Targeting Members of the Cadherin Superfamily: A Review. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2025; 8:23-33. [PMID: 39811419 PMCID: PMC11728384 DOI: 10.36401/jipo-24-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/31/2024] [Accepted: 09/09/2024] [Indexed: 01/16/2025]
Abstract
The cadherin superfamily of proteins is critical for cell-cell interactions and demonstrates tissue-specific expression profiles. In cancers, disruption of cell-cell adhesion is frequently associated with oncogenesis and metastasis. As such, these proteins have been the targets of multiple attempts to develop novel therapeutics in malignancy. This review article discusses prior and current clinical trials targeting the cadherin proteins.
Collapse
Affiliation(s)
- Jacob Keeling
- Sky Ridge Internal Medicine Residency Program, Lone Tree, CO, USA
| | - Gerald Falchook
- Sarah Cannon Research Institute (SCRI) at HealthONE, Denver, CO, USA
| |
Collapse
|
25
|
Liao YY, Tsai CL, Huang HP. Optimizing Osimertinib for NSCLC: Targeting Resistance and Exploring Combination Therapeutics. Cancers (Basel) 2025; 17:459. [PMID: 39941826 PMCID: PMC11815769 DOI: 10.3390/cancers17030459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide, with epidermal growth factor receptor (EGFR) mutations present in a substantial proportion of patients. Third-generation EGFR tyrosine kinase inhibitors (EGFR TKI), exemplified by osimertinib, have dramatically improved outcomes by effectively targeting the T790M mutation-a primary driver of acquired resistance to earlier-generation EGFR TKI. Despite these successes, resistance to third-generation EGFR TKIs inevitably emerges. Mechanisms include on-target mutations such as C797S, activation of alternative pathways like MET amplification, histologic transformations, and intricate tumor microenvironment (TME) alterations. These resistance pathways are compounded by challenges in tolerability, adverse events, and tumor heterogeneity. In light of these hurdles, this review examines the evolving landscape of combination therapies designed to enhance or prolong the effectiveness of third-generation EGFR TKIs. We explore key strategies that pair osimertinib with radiotherapy, anti-angiogenic agents, immune checkpoint inhibitors, and other molecularly targeted drugs, and we discuss the biological rationale, preclinical evidence, and clinical trial data supporting these approaches. Emphasis is placed on how these combinations may circumvent diverse resistance mechanisms, improve survival, and maintain a favorable safety profile. By integrating the latest findings, this review aims to guide clinicians and researchers toward more individualized and durable treatment options, ultimately enhancing both survival and quality of life for patients with EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Yan-You Liao
- Department of Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| | - Chia-Luen Tsai
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| |
Collapse
|
26
|
Guo H, Zhang N, Huang T, Shen N. MicroRNA-200c in Cancer Generation, Invasion, and Metastasis. Int J Mol Sci 2025; 26:710. [PMID: 39859424 PMCID: PMC11766322 DOI: 10.3390/ijms26020710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
MicroRNA-200c (miR-200c) is increasingly recognized as a crucial small RNA molecule that plays a significant and multifaceted role in the complex processes of tumor development, invasion, and metastasis across various types of cancers. Recent studies have compellingly demonstrated that miR-200c exerts its influence on tumor biology by meticulously regulating a range of critical processes, including cell proliferation, apoptosis, epithelial-mesenchymal transition (EMT), and cell migration, all of which are essential for the progression and aggressiveness of tumors. This comprehensive review aims to summarize the expression characteristics and functional implications of miR-200c across a diverse array of tumor types, delving into its potential utility as both a biomarker for early detection and a therapeutic target in the realm of cancer treatment. By synthesizing current research findings and insights, we aspire to provide valuable information that could significantly enhance early diagnostic capabilities and inform the strategic development of targeted therapy approaches in oncology.
Collapse
Affiliation(s)
| | | | | | - Na Shen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China; (H.G.); (N.Z.); (T.H.)
| |
Collapse
|
27
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
28
|
Błaszczak E, Miziak P, Odrzywolski A, Baran M, Gumbarewicz E, Stepulak A. Triple-Negative Breast Cancer Progression and Drug Resistance in the Context of Epithelial-Mesenchymal Transition. Cancers (Basel) 2025; 17:228. [PMID: 39858010 PMCID: PMC11764116 DOI: 10.3390/cancers17020228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most difficult subtypes of breast cancer to treat due to its distinct clinical and molecular characteristics. Patients with TNBC face a high recurrence rate, an increased risk of metastasis, and lower overall survival compared to other breast cancer subtypes. Despite advancements in targeted therapies, traditional chemotherapy (primarily using platinum compounds and taxanes) continues to be the standard treatment for TNBC, often with limited long-term efficacy. TNBC tumors are heterogeneous, displaying a diverse mutation profile and considerable chromosomal instability, which complicates therapeutic interventions. The development of chemoresistance in TNBC is frequently associated with the process of epithelial-mesenchymal transition (EMT), during which epithelial tumor cells acquire a mesenchymal-like phenotype. This shift enhances metastatic potential, while simultaneously reducing the effectiveness of standard chemotherapeutics. It has also been suggested that EMT plays a central role in the development of cancer stem cells. Hence, there is growing interest in exploring small-molecule inhibitors that target the EMT process as a future strategy for overcoming resistance and improving outcomes for patients with TNBC. This review focuses on the progression and drug resistance of TNBC with an emphasis on the role of EMT in these processes. We present TNBC-specific and EMT-related molecular features, key EMT protein markers, and various signaling pathways involved. We also discuss other important mechanisms and factors related to chemoresistance in TNBC within the context of EMT, highlighting treatment advancements to improve patients' outcomes.
Collapse
Affiliation(s)
- Ewa Błaszczak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland
| | | | | | | | | | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland
| |
Collapse
|
29
|
Hou Y, Wang Z, Wu Y, Deng T, Yang S, Ma S, Zhang Y, Luo D, Chen Y, Dai X. Sophaline B inhibits non-small cell lung cancer by activating NLRP3/caspase-1/GSDMD-dependent pyroptosis and PI3K/AKT/mTOR-mediated autophagy. Nat Prod Res 2025:1-13. [PMID: 39797590 DOI: 10.1080/14786419.2024.2448839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 12/01/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025]
Abstract
Sophaline B (SPB), extracted from the seeds of Sophora alopecuroides L., is a natural bioactive compound that effectively exerts antiviral activities against the hepatitis B virus. This is the first study to demonstrate that SPB exerts anti-tumor effects on NSCLC by inducing pyroptosis and autophagy. SPB promotes NSCLC cell death by increasing the reactive oxygen species (ROS) production to induce pyroptosis via activating the NLRP3/Caspase-1/GSDMD signalling pathway. Meanwhile, SPB inhibits cancer cell proliferation by activating autophagy via blocking the PI3K/AKT/mTOR signalling pathway. Further investigation indicates that SPB inhibits NSCLC cell migration and invasion by increasing E-cadherin while decreasing N-cadherin, vimentin, and snail at the protein level. In addition, our results show that SPB inhibits cancer cell colony formation and human umbilical vein endothelial cell angiogenesis. Our study revealed the mechanisms of SPB triggering pyroptosis and autophagy in NSCLC, thus providing evidence and information on the SPB as an anti-cancer agent in NSCLC.
Collapse
Affiliation(s)
- Yibo Hou
- Institute of Biopharmaceutical and Health Engineering, State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Gene and Antibody Therapy, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China
| | - Zhongfu Wang
- Department of Interventional, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Yang Wu
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Ting Deng
- Institute of Biopharmaceutical and Health Engineering, State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Gene and Antibody Therapy, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China
| | - Siyu Yang
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Gene and Antibody Therapy, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China
| | - Yubo Zhang
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Ding Luo
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yanjun Chen
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoyong Dai
- Institute of Biopharmaceutical and Health Engineering, State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Gene and Antibody Therapy, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China
| |
Collapse
|
30
|
Sokoli L, Takáč P, Budovská M, Michalková R, Kello M, Nosálová N, Balážová Ľ, Salanci Š, Mojžiš J. The Proapoptotic Effect of MB-653 Is Associated with the Modulation of Metastasis and Invasiveness-Related Signalling Pathways in Human Colorectal Cancer Cells. Biomolecules 2025; 15:72. [PMID: 39858466 PMCID: PMC11762530 DOI: 10.3390/biom15010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Colorectal cancer is one of the most common cancers worldwide and has a high mortality rate. In this study, we investigated the cytotoxic, proapoptotic, and anti-invasive effects of the synthetic indole phytoalexin MB-653. The antiproliferative effect was determined using an MTT assay, showing IC50 values of 5.8 ± 0.3 μmol/L for HCT116 cells and 6.1 ± 2.1 μmol/L for Caco2 cells. Flow cytometry and Western blot analysis were employed to investigate the molecular mechanisms underlying cytotoxicity, proapoptotic action, and anti-invasion effects. The proapoptotic activity was evidenced by the activation of caspases 3 and 7, mitochondrial dysfunction, and an increased number of apoptotic cells, confirmed by annexin V/PI and AO/PI staining. Additionally, MB-653 induces dose-dependent G2/M phase cell cycle arrest, the cause of which could be cyclin B1/CDC2 complex dysfunction and/or a decrease in α-tubulin protein expression. Another important observation was that MB-653 modulated several signalling pathways associated with various cellular activities, including survival, proliferation, tumour invasiveness, metastasis, and epithelial-mesenchymal transition (EMT). We further demonstrated its safety for topical and parenteral application. To sum up, our results indicate the real potential of MB-653 in treating colorectal cancer.
Collapse
Affiliation(s)
- Libor Sokoli
- Department of Pharmacology and Toxicology, University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (R.M.); (M.K.)
| | - Peter Takáč
- Department of Pharmacology and Toxicology, University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
| | - Mariana Budovská
- Department of Organic Chemistry, Institute of Chemistry, Faculty of Science, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Radka Michalková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (R.M.); (M.K.)
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (R.M.); (M.K.)
| | - Natália Nosálová
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
| | - Ľudmila Balážová
- Department of Pharmaceutical Technology, Pharmacognosy and Botany, University of Veterinary Medicine and Pharmacy, 041 81 Košice, Slovakia;
| | - Šimon Salanci
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (R.M.); (M.K.)
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (R.M.); (M.K.)
| |
Collapse
|
31
|
Ye J, Wang J, Liu R, Chen C, Wang W. The prognostic significance and potential mechanism of PFDN4 in hepatocellular carcinoma. Int Immunopharmacol 2025; 145:113761. [PMID: 39644788 DOI: 10.1016/j.intimp.2024.113761] [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: 09/13/2024] [Revised: 11/15/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
PFDN4, a subunit of the prefoldin complex, has been previously shown to be upregulated in breast and colorectal cancers, where its expression correlates with poor clinical outcomes. This study investigates PFDN4 expression across various cancer types, with a specific focus on its role in hepatocellular carcinoma (HCC) development and progression. Analysis of TCGA data revealed that PFDN4 is highly expressed in several cancers and is associated with poor prognosis. Further validation through multiple databases, tissue microarrays, and clinical samples confirmed that PFDN4 protein levels are significantly elevated in HCC tissues. Meanwhile, multiple database multivariate and univariate Cox regression analyses suggest that PFDN4 is an independent prognostic marker for HCC. To evaluate the functional effects of PFDN4, we established stable HCC cell lines with PFDN4 knockdown and overexpression. Using CCK-8, EdU, wound healing, and Transwell assays, we found that PFDN4 knockdown significantly suppressed cell proliferation, migration, and invasion, while its overexpression enhanced these behaviors. These findings were further validated in vivo. Mechanistically, transcriptome sequencing suggested that PFDN4 modulates HCC cell behavior through the MAPK/ERK signaling pathway, a result confirmed by Western blot and the use of the MAPK/ERK inhibitor SCH772984. Additionally, single-cell RNA sequencing data revealed that PFDN4 is primarily expressed in several immune cell types, including B cells, CD8 + Tex, DC, ILC, mast cells, macrophages, Tprolif, and Treg. In conclusion, our study demonstrates that PFDN4 is upregulated in HCC and drives tumor progression via the MAPK/ERK pathway, highlighting its potential as both a prognostic marker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Jing Ye
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China; Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China
| | - Jianguo Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China; Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China; Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Rongqiang Liu
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China; Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China; Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China; Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei 430060, China.
| |
Collapse
|
32
|
Zuo Q, Kang Y. Metabolic Reprogramming and Adaption in Breast Cancer Progression and Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:347-370. [PMID: 39821033 DOI: 10.1007/978-3-031-70875-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Recent evidence has revealed that cancer is not solely driven by genetic abnormalities but also by significant metabolic dysregulation. Cancer cells exhibit altered metabolic demands and rewiring of cellular metabolism to sustain their malignant characteristics. Metabolic reprogramming has emerged as a hallmark of cancer, playing a complex role in breast cancer initiation, progression, and metastasis. The different molecular subtypes of breast cancer exhibit distinct metabolic genotypes and phenotypes, offering opportunities for subtype-specific therapeutic approaches. Cancer-associated metabolic phenotypes encompass dysregulated nutrient uptake, opportunistic nutrient acquisition strategies, altered utilization of glycolysis and TCA cycle intermediates, increased nitrogen demand, metabolite-driven gene regulation, and metabolic interactions with the microenvironment. The tumor microenvironment, consisting of stromal cells, immune cells, blood vessels, and extracellular matrix components, influences metabolic adaptations through modulating nutrient availability, oxygen levels, and signaling pathways. Metastasis, the process of cancer spread, involves intricate steps that present unique metabolic challenges at each stage. Successful metastasis requires cancer cells to navigate varying nutrient and oxygen availability, endure oxidative stress, and adapt their metabolic processes accordingly. The metabolic reprogramming observed in breast cancer is regulated by oncogenes, tumor suppressor genes, and signaling pathways that integrate cellular signaling with metabolic processes. Understanding the metabolic adaptations associated with metastasis holds promise for identifying therapeutic targets to disrupt the metastatic process and improve patient outcomes. This chapter explores the metabolic alterations linked to breast cancer metastasis and highlights the potential for targeted interventions in this context.
Collapse
Affiliation(s)
- Qianying Zuo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
33
|
Keerthiga R, Xie Y, Pei DS, Fu A. The multifaceted modulation of mitochondrial metabolism in tumorigenesis. Mitochondrion 2025; 80:101977. [PMID: 39505244 DOI: 10.1016/j.mito.2024.101977] [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: 11/24/2023] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Changes in mitochondrial metabolism produce a malignant transformation from normal cells to tumor cells. Mitochondrial metabolism, comprising bioenergetic metabolism, biosynthetic process, biomolecular decomposition, and metabolic signal conversion, obviously forms a unique sign in the process of tumorigenesis. Several oncometabolites produced by mitochondrial metabolism maintain tumor phenotype, which are recognized as tumor indicators. The mitochondrial metabolism synchronizes the metabolic and genetic outcome to the potent tumor microenvironmental signals, thereby further promoting tumor initiation. Moreover, the bioenergetic and biosynthetic metabolism within tumor mitochondria orchestrates dynamic contributions toward cancer progression and invasion. In this review, we describe the contribution of mitochondrial metabolism in tumorigenesis through shaping several hallmarks such as microenvironment modulation, plasticity, mitochondrial calcium, mitochondrial dynamics, and epithelial-mesenchymal transition. The review will provide a new insight into the abnormal mitochondrial metabolism in tumorigenesis, which will be conducive to tumor prevention and therapy through targeting tumor mitochondria.
Collapse
Affiliation(s)
- Rajendiran Keerthiga
- College of Pharmaceutical Science, Southwest University, Chongqing, 400716, China; Department of Computational Biology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Thandalam, Chennai 602105, Tamil Nadu, India
| | - Yafang Xie
- College of Pharmaceutical Science, Southwest University, Chongqing, 400716, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Ailing Fu
- College of Pharmaceutical Science, Southwest University, Chongqing, 400716, China.
| |
Collapse
|
34
|
Liu GL, Luo H, Liang DD, Zhong L, Dai N, Lan WH. Comprehensive Analysis of Prognostic Alternative Splicing Signatures in Tumor Immune Infiltration in Bladder Cancer. Recent Pat Anticancer Drug Discov 2025; 20:185-199. [PMID: 39473202 DOI: 10.2174/0115748928329276241020184935] [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: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Bladder cancer exhibits substantial heterogeneity encompassing genetic expressions and histological features. This heterogeneity is predominantly attributed to alternative splicing (AS) and AS-regulated splicing factors (SFs), which, in turn, influence bladder cancer development, progression, and response to treatment. OBJECTIVE This study aimed to explore the immune landscape of aberrant AS in bladder cancer and establish the prognostic signatures for survival prediction. METHODS Bladder cancer-related RNA-Seq, transcriptome, and corresponding clinical information were downloaded from The Cancer Genome Atlas (TCGA). Gene set enrichment analysis (GSEA) was used to identify significantly enriched pathways of cancer-related AS events. The underlying interactions among differentially expressed genes (DEGs) and cancer-related AS events were assessed by a protein-protein interaction network. Univariate and multivariate Cox regression analyses were performed to identify crucial prognostic DEGs that co-occurred with cancer-related AS events (DEGAS) for overall survival. The area under the curve (AUC) of receiver operating characteristic (ROC) curves was used to assess the efficiency of the prognostic signatures. The CIBERSORT algorithm was used to explore the abundance of immune infiltrating cells. RESULTS A total of 3755 cancer-related AS events and 3110 DEGs in bladder cancer were identified. Among them, 379 DEGs co-occurred with cancer-related AS events (DEGAS), of which 102 DEGAS were associated with 14 dysregulated SFs. GSEA and KEGG analysis showed that cancer-related AS events were predominantly enriched in pathways related to immunity, tumorigenesis, and treatment difficulties of bladder cancer. Multivariate Cox regression analysis identified 8 DEGAS (CABP1, KCNN2, TNFRSF13B, PCDH7, SNRPA1, APOLD1, CX3CL1, and DENND5A) significantly associated with OS, and they were further integrated into the prediction model with good AUCs at 3-year, 5-year and 7-year ROC curves (all>0.7). Immune infiltration analysis revealed the significant enrichment of three immune cell types (B cells naïve, dendritic cells resting, and dendritic cell activated) in high-risk bladder cancer patients. CONCLUSION This study not only unveiled comprehensive prognostic signatures of AS events in bladder cancer but also established a robust prognostic model based on survival-related DEGAS. These aberrant AS events, dysregulated SFs, and the identified 8 DEGAS may have significant clinical potential as therapeutic targets for bladder cancer.
Collapse
Affiliation(s)
- Gao-Lei Liu
- Department of Urology, Army Medical Center, Chongqing, 400042, China
| | - Hao Luo
- Department of Oncology, Army Medical Center, Chongqing, 400042, China
| | - Dan-Dan Liang
- People's Hospital of Chong Qing Liang Jiang New Area, Chongqing, 400042, China
| | - Li Zhong
- Department of Oncology, Army Medical Center, Chongqing, 400042, China
| | - Nan Dai
- Department of Oncology, Army Medical Center, Chongqing, 400042, China
| | - Wei-Hua Lan
- Department of Urology, Army Medical Center, Chongqing, 400042, China
| |
Collapse
|
35
|
Shang H, Sun J, Zheng Z, Sun S, Yan X. Study on the Effect of Quinoa Saponins on Human Colon Cancer HT-29 Cells. Food Sci Nutr 2025; 13:e4669. [PMID: 39803233 PMCID: PMC11717042 DOI: 10.1002/fsn3.4669] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/27/2024] [Accepted: 11/30/2024] [Indexed: 01/16/2025] Open
Abstract
Quinoa saponins can inhibit the survival of specific cancer cells. However, there is still a lack of systematic research on the effects of quinoa saponins on colon cancer cells. This experiment confirmed that quinoa saponins prevented human colon cancer HT-29 cells from growing in vitro. The MTT experiment revealed that quinoa saponins significantly decreased the proliferative vitality of HT-29 cells. In comparison to the control group, the proportion of cell number in the G0/G1 phase increased by 22.97% and the rate of apoptosis increased by 22.55% after treating cells with quinoa saponins (40 μg/mL). By regulating the expression of Cyclin D1 and p21, it caused the cell cycle to be blocked in the G0/G1 phase. It also promoted the expression of Caspase3 and Bax while suppressing the expression of Bcl-2, which led to the apoptosis of HT-29 cells. In addition, quinoa saponins caused cells to undergo autophagy by upregulating the expression of LC-3II and Beclin1, while the addition of autophagy inhibitors significantly reduced the inhibitory effect on cell proliferation. Finally, the migration of HT-29 cells was also inhibited by quinoa saponins. After treating cells with quinoa saponins (40 μg/mL), compared with that in the control group, the wound healing rate of cells decreased by 38.21% and the migration ability decreased by 69.48%. The potential mechanism could be connected to increasing E-cadherin expression while decreasing N-cadherin expression. Importantly, all of these changes induced by quinoa saponins were dose dependent. Overall, these findings give a scientific basis for the anticancer mechanism of quinoa saponins.
Collapse
Affiliation(s)
- Haijun Shang
- College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
- Anhui Business and Technology CollegeHefeiChina
| | - Jinwei Sun
- Anhui Business and Technology CollegeHefeiChina
| | - Zhi Zheng
- College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | | | | |
Collapse
|
36
|
Sun Q, Xu J, Yuan F, Liu Y, Chen Q, Guo L, Dong H, Liu B. RND1 inhibits epithelial-mesenchymal transition and temozolomide resistance of glioblastoma via AKT/GSK3-β pathway. Cancer Biol Ther 2024; 25:2321770. [PMID: 38444223 PMCID: PMC10936657 DOI: 10.1080/15384047.2024.2321770] [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: 06/25/2023] [Accepted: 02/18/2024] [Indexed: 03/07/2024] Open
Abstract
GBM is one of the most malignant tumor in central nervous system. The resistance to temozolomide (TMZ) is inevitable in GBM and the characterization of TMZ resistance seriously hinders clinical treatment. It is worthwhile exploring the underlying mechanism of aggressive invasion and TMZ resistance in GBM treatment. Bioinformatic analysis was used to analyze the association between RND1 and a series of EMT-related genes. Colony formation assay and cell viability assay were used to assess the growth of U87 and U251 cells. The cell invasion status was evaluated based on transwell and wound-healing assays. Western blot was used to detect the protein expression in GBM cells. Treatment targeted RND1 combined with TMZ therapy was conducted in nude mice to evaluate the potential application of RND1 as a clinical target for GBM. The overexpression of RND1 suppressed the progression and migration of U87 and U251 cells. RND1 knockdown facilitated the growth and invasion of GBM cells. RND1 regulated the EMT of GBM cells via inhibiting the phosphorylation of AKT and GSK3-β. The promoted effects of RND1 on TMZ sensitivity was identified both in vitro and in vivo. This research demonstrated that the overexpression of RND1 suppressed the migration and EMT status by downregulating AKT/GSK3-β pathway in GBM. RND1 enhanced the TMZ sensitivity of GBM cells both in vitro and in vivo. Our findings may contribute to the targeted therapy for GBM and the understanding of mechanisms of TMZ resistance in GBM.
Collapse
Affiliation(s)
- Qian Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junjie Xu
- Office of director, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Fan’en Yuan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lirui Guo
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huimin Dong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
37
|
Chen D, Jiang J, Zhang W, Li X, Ge Q, Liu X, Li X. Tripartite motif-containing protein 50 suppresses triple-negative breast cancer progression by regulating the epithelial-mesenchymal transition. Cancer Biol Ther 2024; 25:2427410. [PMID: 39538371 PMCID: PMC11572070 DOI: 10.1080/15384047.2024.2427410] [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: 05/31/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Tripartite motif-containing protein 50 (TRIM50) is a recently discovered E3 ubiquitin ligase that participates in tumor progression. TRIM50 is overexpressed in many cancers, although few studies focused on TRIM50's role in breast cancer. METHODS We overexpressed TRIM50 in triple-negative breast cancer cell lines using plasmid and found that TRIM50 upregulation markedly reduced breast cancer cell proliferation, clone formation, and migration, as well as promoted breast cancer cell apoptosis. Western blotting revealed that accumulated TRIM50 resulted in both mRNA and protein depletion of SNAI1, and partially attenuated the epithelial-mesenchymal transition (EMT) induced by SNAI1. RESULTS In this study, we demonstrate that TRIM50 is downregulated in human breast cancer and that its overexpression closely correlates with diminished invasion capacity in breast cancer, suggesting that TRIM50 may serve as a diagnostic marker and therapeutic target. CONCLUSION TRIM50 plays a key role in breast cancer proliferation and potentially serves as a prognostic and therapeutic target.
Collapse
Affiliation(s)
- Danxiang Chen
- Department of Oncology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, PR China
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Jing Jiang
- Department of Oncology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, PR China
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Wei Zhang
- Department of Oncology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, PR China
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Xinlin Li
- Department of Oncology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, PR China
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Qidong Ge
- Department of Oncology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, PR China
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Xia Liu
- Department of Anesthesiology, Ningbo 1st Hospital, Ningbo, Zhejiang, China
| | - Xujun Li
- Department of Oncology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, PR China
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
38
|
Zhou L, Yang Y, Qiao Q, Mi Y, Gan Y, Zheng Y, Wang Y, Liu M, Zhou Y. AKT1-Mediated NOTCH1 phosphorylation promotes gastric cancer progression via targeted regulation of IRS-1 transcription. J Cancer Res Clin Oncol 2024; 151:15. [PMID: 39724412 PMCID: PMC11671552 DOI: 10.1007/s00432-024-06039-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE This study aimed to investigate that AKT1-Mediated NOTCH1 phosphorylation promotes gastric cancer (GC) progression via targeted regulation of IRS-1 transcription. METHODS The study utilized databases such as PhosphositePlus, TRANSFAC, CHEA, GPS 5.0, and TCGA, along with experimental techniques including Western Blot, co-IP, in vitro kinase assay, construction of lentiviral overexpression and silencing vectors, immunoprecipitation, modified proteomics, immunofluorescence, ChIP-PCR, EdU assay, Transwell assay, and scratch assay to investigate the effects of AKT1-induced Notch1 phosphorylation on cell proliferation, invasion and migration in vitro, as well as growth and epithelial-mesenchymal transition (EMT) in vivo. RESULTS AKT1 was found to induce phosphorylation of Notch1 at the S2183 site in GC, subsequently altering the subcellular localization of Notch1-IC and promoting its nuclear translocation. The transcription factor RBPJ that binds to Notch1 transcriptionally regulated IRS-1, CDH5, TNL1, ASCL2, and LRP6. Experimental validation revealed that Notch1-IC can regulate the expression of IRS-1. Overexpression of Notch1-IC was shown to promote the proliferation, invasion, and metastasis of GC cells, while knockdown of IRS-1 partially inhibited the aforementioned effects induced by Notch1-IC overexpression. Further experiments in vitro and vivo confirmed that AKT1-induced Notch1 phosphorylation can regulate the expression of IRS-1 and promote the malignant behavior of GC, including proliferation, invasion, metastasis, and EMT, with knockdown of IRS-1 partially reversing these effects. CONCLUSION AKT1 induces the Notch1 phosphorylation and promotes the activation and nuclear translocation of Notch1-IC by targeting the regulation of IRS-1, thereby advancing the progression of GC.
Collapse
Affiliation(s)
- Lingshan Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Department of Geriatrics Ward 2, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yuan Yang
- Department of Gastroenterology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Qian Qiao
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yingying Mi
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yuling Gan
- The 1nd Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730000, China
| | - Ya Zheng
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yuping Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Min Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China.
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Yongning Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China.
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
39
|
Anvari S, Nikbakht M, Vaezi M, Amini-Kafiabad S, Ahmadvand M. Immune checkpoints and ncRNAs: pioneering immunotherapy approaches for hematological malignancies. Cancer Cell Int 2024; 24:410. [PMID: 39702293 DOI: 10.1186/s12935-024-03596-8] [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: 03/19/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
Hematological malignancies are typically treated with chemotherapy and radiotherapy as the first-line conventional therapies. However, non-coding RNAs (ncRNAs) are a rapidly expanding field of study in cancer biology that influences the growth, differentiation, and proliferation of tumors by targeting immunological checkpoints. This study reviews the results of studies (from 2012 to 2024) that consider the immune checkpoints and ncRNAs in relation to hematological malignancies receiving immunotherapy. This article provides a summary of the latest advancements in immunotherapy for treating hematological malignancies, focusing on the role of immune checkpoints and ncRNAs in the immune response and their capacity for innovative strategies. The paper also discusses the function of immune checkpoints in maintaining immune homeostasis and how their dysregulation can contribute to developing leukemia and lymphoma. Finally, this research concludes with a discussion on the obstacles and future directions in this rapidly evolving field, emphasizing the need for continued research to fully harness the capacity of immune checkpoints and ncRNAs in immunotherapy for hematological malignancies.
Collapse
Affiliation(s)
- Samira Anvari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohsen Nikbakht
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vaezi
- Hematology, Oncology, and Stem Cell Transplantation Research Center Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Amini-Kafiabad
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.
| | - Mohammad Ahmadvand
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Sun Y, Zhou X, Hu X. Constructing a doxycycline-inducible system for an epithelial-to-mesenchymal transition model in MCF10A cells. Biol Open 2024; 13:bio061790. [PMID: 39648980 PMCID: PMC11655024 DOI: 10.1242/bio.061790] [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: 05/23/2024] [Accepted: 11/18/2024] [Indexed: 12/10/2024] Open
Abstract
Epithelial to mesenchymal transition (EMT) has been shown to play an essential role in the early stages of cancer cell invasion and metastasis. Inducible EMT models can initiate EMT in a controlled manner, thereby providing the opportunity to determine whether a cancer-associated gene influences cancer metastasis by triggering EMT. Moreover, different inducible EMT models enable the investigation of specific mechanisms of EMT modulation by various genes, facilitating a more precise understanding of how these genes influence cancer metastasis through the induction of EMT. Unfortunately, current inducible EMT models still present unmet needs. Therefore, we aimed to establish an inducible EMT model in MCF10A cells, a spontaneously immortalized human fibrocystic mammary cell line, by manipulating the expression of mouse Twist1 (mTwist1). In this study, we first compared the EMT induction capacity between human TWIST1 (hTWIST1) and mTwist1, and selected mTwist1 for further investigation. By monitoring the changes in epithelial and mesenchymal markers at different induction time points, we examined the EMT process in both polyclonal and monoclonal MCF10A cells that express doxycycline (DOX)-inducible mTwist1. Furthermore, our results showed that doxycycline-induced mTwist1 expression triggered EMT at a similar rate to TGFβ1-induced EMT in MCF10A cells. Additionally, this process was reversible upon DOX withdrawal. Thus, we have established a robust inducible EMT model in MCF10A cells, which can be used to further study cancer metastasis-driving genes.
Collapse
Affiliation(s)
- Yaxuan Sun
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xun Zhou
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xiaohui Hu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
41
|
Gao HW, Chang KF, Huang XF, Lee MC, Tsai NM, Chen TH. Cedrus atlantica extract inhibits melanoma progression by suppressing epithelial-mesenchymal transition and inducing mitochondria-mediated apoptosis. Med Oncol 2024; 42:22. [PMID: 39643823 DOI: 10.1007/s12032-024-02573-5] [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: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
Melanoma has a low incidence, accounting for less than 5% of skin cancers; however, it is the most lethal cancer, primarily because of its high potential for metastasis and resistance to different treatments. Natural products can sensitize melanoma to chemotherapy and overcome drug resistance. Previous studies have reported Cedrus atlantica extract has various pharmacological benefits such as anti-inflammatory, antioxidant, antibacterial, and analgesic properties. This study aimed to explore the effects of C. atlantica extract (CAt) against melanoma in vitro and in vivo. The effects of CAt on B16F10 cell viability, proliferation, migration, invasion, and apoptosis were detected using MTT, colony formation, wound-healing, Boyden chamber, and TUNEL assays. Semi-quantitative RT-PCR and western blotting were used to measure mRNA and protein expression, respectively. Results revealed that CAt selectively decreased the viability of B16F10 cells and inhibited colony formation in a dose-dependent manner. CAt reduces cell migration and invasion by regulating epithelial-mesenchymal transition-associated proteins (Snail, E-cadherin, and vimentin). Moreover, CAt enhanced the Bax/Bcl-2 ratio and the expression of cleaved-caspase-9, caspase-3, and PARP1, resulting in the activation of mitochondria-mediated apoptosis. In an in vivo study, CAt significantly inhibited tumor growth and prolonged the lifespan of mice at a well-tolerated dose. Importantly, the combination of CAt and 5-fluorouracil (5-FU) exhibited synergistic growth suppression and attenuated the development of 5-FU resistance. Overall, the findings suggest that CAt holds promise as a potential drug or adjuvant to improve melanoma treatment.
Collapse
Affiliation(s)
- Hong-Wei Gao
- Department of Pathology, Tungs' Taichung MetroHarbor Hospital, Taichung, 43503, Taiwan, R.O.C
| | - Kai-Fu Chang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., Taichung, 40201, Taiwan, R.O.C
| | - Xiao-Fan Huang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., Taichung, 40201, Taiwan, R.O.C
| | - Meng-Chiao Lee
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., Taichung, 40201, Taiwan, R.O.C
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., Taichung, 40201, Taiwan, R.O.C..
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, R.O.C..
- Department of Life-and-Death Studies, Nanhua University, Chiayi, 62249, Taiwan, R.O.C..
| | - Tze-Ho Chen
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, No. 135, Nanhsiao Street, Changhua, 50006, Taiwan, R.O.C..
| |
Collapse
|
42
|
Mathew Thomas V, Sayegh N, Chigarira B, Gebrael G, Tripathi N, Nussenzveig R, Jo Y, Dal E, Galarza Fortuna G, Li H, Sahu KK, Srivastava A, Maughan BL, Agarwal N, Swami U. Differences in Tumor Gene Expression Profiles Between De Novo Metastatic Castration-sensitive Prostate Cancer and Metastatic Relapse After Prior Localized Therapy. Eur Urol Oncol 2024; 7:1462-1468. [PMID: 38735779 DOI: 10.1016/j.euo.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND AND OBJECTIVE It has been reported that patients with de novo metastatic castration-sensitive prostate cancer (dn-mCSPC) have worse prognosis and outcomes than those whose cancer relapses after prior local therapy (PLT-mCSPC). Our aim was to interrogate and validate underlying differences in tumor gene expression profiles between dn-mCSPC and PLT-mCSPC. METHODS The inclusion criteria were histologically confirmed prostate adenocarcinoma and the availability of RNA sequencing data for treatment-naïve primary prostate tissue. RNA sequencing was performed by Tempus or Caris Life Sciences, both of which have Clinical Laboratory Improvement Amendments certification. The Tempus cohort was used for interrogation, while the Caris cohort was used for validation. Differential gene expression analysis between the cohorts was conducted using the DEseq2 pipeline. The resulting gene expression profiles were further analyzed using Gene Set Enrichment software to identify pathways with enrichment in each cohort. KEY FINDINGS AND LIMITATIONS Overall, 128 patients were eligible, of whom 78 were in the Tempus cohort (dn-mCSPC 37, PLT-mCSPC 41) and 50 were in the Caris cohort (dn-mCSPC 30, PLT-mCSPC 20). Tumor tissues from patients with dn-mCSPC had higher expression of genes associated with inflammation pathways, while tissues from patients with PLT-mCSPC had higher expression of genes involved in oxidative phosphorylation, fatty acid metabolism, and androgen response pathways. CONCLUSIONS AND CLINICAL IMPLICATIONS Our study revealed upregulation of distinct genomic pathways in dn-mCSPC in comparison to PLT-mCSPC. These hypothesis-generating data could guide personalized therapy for men with prostate cancer and explain different survival outcomes for dn-mCSPC and PLT-mCSPC. PATIENT SUMMARY We measured gene expression levels in tumors from patients with metastatic castration-sensitive prostate cancer. In patients with metastatic disease at first diagnosis, inflammatory pathways were upregulated. In patients whose metastasis occurred on relapse after treatment, androgen response pathways were upregulated. These findings could help in personalizing therapy for prostate cancer and explaining differences in survival.
Collapse
Affiliation(s)
- Vinay Mathew Thomas
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicolas Sayegh
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beverly Chigarira
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nishita Tripathi
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Internal Medicine, Detroit Medical Center Sinai Grace Hospital, Detroit, MI, USA
| | - Roberto Nussenzveig
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; DDx Foundation, Lehi, UT, USA
| | - Yeonjung Jo
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Emre Dal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Gliceida Galarza Fortuna
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Haoran Li
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, Westwood, KS, USA
| | - Kamal Kant Sahu
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Ayana Srivastava
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Benjamin L Maughan
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
43
|
Luan H, Wang T, Li F, Sun S, Wang Z, Zhao X, Kong F, Hu T, Liu Y, Zhang J, Liu X, Wang H, Meng X, Li C, Zhang J, Ji S, Hui L, Nie S, Wang Y, Li Z. IGSF9 promotes tumor invasion and metastasis through GSK-3β/β-catenin mediated EMT in lung cancer. Neoplasia 2024; 58:101067. [PMID: 39383800 PMCID: PMC11492623 DOI: 10.1016/j.neo.2024.101067] [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: 02/12/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
We previously reported that immunoglobulin superfamily member 9 (IGSF9) as a tumor specific immune checkpoint promoted the tumor immune escape, however, as an adhesion molecule, whether IGSF9 promotes tumor invasion and metastasis has not been reported. Here, the full length, the intracellular domain (ID) not extracellular domain (ECD) of IGSF9 could alter tumor cell morphology from a flat and polygonal shape to elongated strips, suggesting that IGSF9 signal pathway has the potential to mediate epithelial-to-mesenchymal transition (EMT). Real-time PCR and western blotting also showed that the mesenchymal markers were significantly up-regulated, and the epithelial markers were significantly down-regulated in IGSF9 and IGSF9-ID groups. Meanwhile, immunofluorescence showed that β-catenin was clearly translocated into the nucleus in IGSF9 and IGSF9-ID groups. The in vitro and in vivo data showed that IGSF9, IGSF9-ID and ECD could promote tumor invasion and metastasis. Mechanistically, IGSF9-ID could recruit GSK-3β to result in the accumulation and nuclear translocation of β-catenin to trigger EMT. Anti-IGSF9 could significantly inhibit the invasion and metastasis, and IGSF9 is an effective candidate for lung cancer therapy.
Collapse
Affiliation(s)
- Huiwen Luan
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Ting Wang
- Department of Pathology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264099, PR China
| | - Fangmin Li
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Shuang Sun
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China; Department of Laboratory Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264099, PR China
| | - Zhenbo Wang
- Department of Binzhou Medical University Hospital, Binzhou, Shandong 256600, PR China
| | - Xinyu Zhao
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Feng Kong
- Shandong Institute of Clinical Medicine, Shandong Provincial Hospital, Jinan, Shandong 250021, PR China
| | - Tao Hu
- Department of Thoracic Surgery, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264099, PR China
| | - Yifan Liu
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Juan Zhang
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Xiaoli Liu
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Hongying Wang
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Xianhui Meng
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Chunling Li
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Jiashen Zhang
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China; Department of Biochemistry and Molecular Biology, School of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Shuhao Ji
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Lijun Hui
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Siman Nie
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Yaopeng Wang
- Department of Thoracic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong 266011, PR China.
| | - Zunling Li
- Shandong Key Lab of Complex Medical Intelligence and Aging, Shandong Medicine and Health Key Lab of Respiratory Infection and Tumor Immunity, Department of Biochemistry and Molecular Biology, Shandong Tumor Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong 264003, PR China.
| |
Collapse
|
44
|
Cui A, Liu H, Liu X, Zhang M, Xiao B, Wang B, Yang J. Steroidal saponins: Natural compounds with the potential to reverse tumor drug resistance (Review). Oncol Lett 2024; 28:585. [PMID: 39421314 PMCID: PMC11484340 DOI: 10.3892/ol.2024.14719] [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: 05/13/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Steroidal saponins are a type of natural product that have been widely used in Chinese herbal medicine, with a variety of pharmacological activities, such as antitumor, anti-inflammatory and anti-bacterial effects. Cancer has become a growing global health problem, and drug therapy is currently the most important clinical antitumor treatment. However, drug resistance is a major obstacle to the effectiveness of chemotherapy, resulting in >90% of deaths of patients with cancer receiving conventional chemotherapy. It has been found that steroidal saponins may exert an effect on the reversal of drug resistance in tumor cells by regulating apoptosis, autophagy, epithelial-mesenchymal transition and drug efflux through multiple related signaling pathways. The present study reviews the role and mechanism of steroidal saponins in the treatment of tumor drug resistance, aiming to provide a scientific basis and research ideas for the future development and clinical application of natural steroidal saponins.
Collapse
Affiliation(s)
- Aiping Cui
- The Clinical Medicine Research Center of The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- Ganzhou Key Laboratory of Antitumor Effects of Natural Products, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Hai Liu
- The Clinical Medicine Research Center of The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Xiaoxuan Liu
- The Clinical Medicine Research Center of The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- Ganzhou Key Laboratory of Antitumor Effects of Natural Products, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Minhong Zhang
- The Clinical Medicine Research Center of The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Bang Xiao
- The Clinical Medicine Research Center of The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- Ganzhou Key Laboratory of Antitumor Effects of Natural Products, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Biao Wang
- The Clinical Medicine Research Center of The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Jianqiong Yang
- The Clinical Medicine Research Center of The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- Ganzhou Key Laboratory of Antitumor Effects of Natural Products, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
- Ganzhou Key Laboratory of Osteoporosis Research, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
45
|
Guo W, Liu M, Luo W, Peng J, Liu F, Ma X, Wang L, Yang S. FERMT1 promotes epithelial-mesenchymal transition of hepatocellular carcinoma by activating EGFR/AKT/β-catenin and EGFR/ERK pathways. Transl Oncol 2024; 50:102144. [PMID: 39353234 PMCID: PMC11472111 DOI: 10.1016/j.tranon.2024.102144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 07/10/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the effects of fermitin family member 1 (FERMT1) on epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma (HCC) via the EGFR/AKT/β-catenin and EGFR/ERK pathways. METHODS The expression of FERMT1 encoding protein kindlin-1 in HCC tissues was determined by immunohistochemistry, and FERMT1 mRNA expression in HCC tissues and cell lines was analyzed by qRT-PCR. After the FERMT1 expression of SNU182 and SNU387 interfered with siRNA, the cell viability, invasion, migration, and EMT were tested by CCK-8, transwell invasion, scratching, immunofluorescence/WB, respectively. Similarly, the effects of FERMT1 on the viability and metastasis of HCC were investigated in transplanted tumor and lung metastasis mouse models. The protein expressions of EGFR/AKT/β-catenin and EGFR/ERK pathways were analyzed by WB. In addition, the relationship between FERMT1 and EGFR was further determined by immunofluorescence double staining and Co-IP. RESULTS FERMT1 was significantly upregulated in HCC, and silencing FERMT1 inhibited the viability, invasion, migration, and EMT of HCC. Silencing FERMT1 also inhibited the activation of EGFR/AKT/β-catenin and EGFR/ERK pathways. In addition, inhibition of EGFR, AKT, or ERK confirmed that EGFR/AKT/β-catenin and EGFR/ERK pathways were involved in the promoting effects of FERMT1 on HCC. Co-IP and immunofluorescence experiments confirmed the targeting relationship between FERMT1 and EGFR. CONCLUSION FERMT1 was highly expressed in HCC and promoted viability, invasion, migration, and EMT of HCC by targeting EGFR to activate the EGFR/AKT/β-catenin and EGFR/ERK pathways. Our study revealed the role of FERMT1 in HCC and suggested that FERMT1 exerts biological effects through activating the EGFR/AKT/β-catenin and EGFR/ERK pathways.
Collapse
Affiliation(s)
- Wubin Guo
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China; Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China; National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China; The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
| | - Mengnan Liu
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China; National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China; Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China
| | - Wei Luo
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jing Peng
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Fei Liu
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xin Ma
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Li Wang
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China; National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
| | - Sijin Yang
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China; National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
46
|
Yeh CH, Chen RY, Wu TH, Chang SY, Hsieh TY, Shih YL, Lin YW. Promoter hypermethylation-mediated downregulation of PAX6 promotes tumor growth and metastasis during the progression of liver cancer. Clin Epigenetics 2024; 16:174. [PMID: 39614377 DOI: 10.1186/s13148-024-01789-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND The progression of liver cancer is a complicated process that involves genetic and epigenetic changes. Paired box 6 (PAX6) is a critical transcription factor for embryonic development. PAX6 is abnormally methylated in human cancer. The role of the PAX6 gene in the pathogenesis of hepatocellular carcinoma (HCC) is still unclear. METHODS Transcriptional silencing of PAX6 mediated by promoter methylation was confirmed using quantitative methylation-specific polymerase chain reaction (PCR) and reverse-transcription (RT)-PCR. Then we conducted gain-and-loss of function approaches to evaluate the function of PAX6 in HCC progression in vitro. Moreover, we designed xenograft mouse models to assess the effect of PAX6 on tumor growth and metastasis. Finally, we used RNA sequencing (RNA-seq) strategy and phenotypic rescue experiments to identify potential targets of PAX6 performing tumor-suppressive function. RESULTS Constitutive expression of PAX6 suppressed anchorage-independent growth and cell invasion in vitro as well as tumor growth and metastasis in xenograft mouse models. In contrast, the inhibition of PAX6 using knockout and knockdown strategies increased tumor growth both in vitro and in vivo. Downregulation of PAX6 by doxycycline depletion partially reversed the malignant phenotypes of HCC cells induced by PAX6. Moreover, we identified E-cadherin (CDH1) and thrombospondin-1 (THBS1) as targets of PAX6. Ultimately, we demonstrated that the knockdown of CDH1 and overexpression of THBS1 in PAX6-expressing HCC cells partly reversed the tumor-suppressive effect. CONCLUSION PAX6 functions as a tumor suppressor partly through upregulation of CDH1 and downregulation of THBS1. Promoter hypermethylation-mediated suppression of PAX6 reduces the tumor suppressor function in the progression of liver cancer.
Collapse
Affiliation(s)
- Ching-Hua Yeh
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Rou-Yu Chen
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Ti-Hui Wu
- Division of Thoracic Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shan-Yueh Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tsai-Yuan Hsieh
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Lueng Shih
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Ya-Wen Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, 11490, Taiwan.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
47
|
Mussa A, Ismail NH, Hamid M, Al-Hatamleh MAI, Bragoli A, Hajissa K, Mokhtar NF, Mohamud R, Uskoković V, Hassan R. Understanding the role of TNFR2 signaling in the tumor microenvironment of breast cancer. J Exp Clin Cancer Res 2024; 43:312. [PMID: 39609700 PMCID: PMC11603874 DOI: 10.1186/s13046-024-03218-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy among women. It is characterized by a high level of heterogeneity that emerges from the interaction of several cellular and soluble components in the tumor microenvironment (TME), such as cytokines, tumor cells and tumor-associated immune cells. Tumor necrosis factor (TNF) receptor 2 (TNFR2) appears to play a significant role in microenvironmental regulation, tumor progression, immune evasion, drug resistance, and metastasis of many types of cancer, including BC. However, the significance of TNFR2 in BC biology is not fully understood. This review provides an overview of TNFR2 biology, detailing its activation and its interactions with important signaling pathways in the TME (e.g., NF-κB, MAPK, and PI3K/Akt pathways). We discuss potential therapeutic strategies targeting TNFR2, with the aim of enhancing the antitumor immune response to BC. This review provides insights into role of TNFR2 as a major immune checkpoint for the future treatment of patients with BC.
Collapse
Affiliation(s)
- Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Nor Hayati Ismail
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Mahasin Hamid
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Hunan Province, Changsha, 410013, China
- Department of Zoology, Faculty of Sciences and Information Technology, University of Nyala, Nyala, 63311, Sudan
| | - Mohammad A I Al-Hatamleh
- Division of Hematology and Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anthony Bragoli
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Khalid Hajissa
- Department of Zoology, Faculty of Science and Technology, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine (iNFORMM), Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| | - Vuk Uskoković
- TardigradeNano LLC, Irvine, CA, 92604, USA
- Division of Natural Sciences, Fullerton College, Fullerton, CA, 92832, USA
| | - Rosline Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| |
Collapse
|
48
|
Xu M, Ai H, Wang D, Wang X. Gene clusters-based pathway enrichment analysis identifies four pan-cancer subtypes with distinct molecular and clinical features. Funct Integr Genomics 2024; 24:224. [PMID: 39607532 DOI: 10.1007/s10142-024-01501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Pathways-based clustering methods have been proposed to explore tumor heterogeneity. However, such methods are currently disadvantageous in that specific pathways need to be explicitly claimed. We developed the PathClustNet algorithm, a pathway-based clustering method designed to identify cancer subtypes. This method first detects gene clusters and identifies overrepresented pathways associated with them. Based on the pathway enrichment scores, it reveals cancer subtypes by clustering analysis. We applied the method to TCGA pan-cancer data and identified four pan-cancer subtypes, termed C1, C2, C3 and C4. C1 exhibited high metabolic activity, favorable survival, and the lowest TP53 mutation rate. C2 had high immune, developmental, and stromal pathway activities, the lowest tumor purity, and intratumor heterogeneity. C3, which overexpressed cell cycle and DNA repair pathways, was the most genomically unstable and had the highest TP53 mutation rate. C4 overrepresented neuronal pathways, with the lowest response rate to chemotherapy, but the highest tumor purity and genomic stability. Furthermore, age showed positive correlations with most pathways but a negative correlation with neuronal pathways. Smoking, viral infections, and alcohol use were found to affect the activities of neuron, cell cycle, immune, stromal, developmental, and metabolic pathway in varying degrees. The PathClustNet algorithm unveils a novel classification of pan-cancer based on metabolic, immune, stromal, developmental, cell cycle, and neuronal pathways. These subtypes display different molecular and clinical features to warrant the investigation of precision oncology.
Collapse
Affiliation(s)
- Mengli Xu
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, 211198, Nanjing, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, 211198, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, 211198, Nanjing, China
| | - Hongjing Ai
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, 211198, Nanjing, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, 211198, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, 211198, Nanjing, China
| | - Danni Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, 211198, Nanjing, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, 211198, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, 211198, Nanjing, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, 211198, Nanjing, China.
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, 211198, Nanjing, China.
- Big Data Research Institute, China Pharmaceutical University, 211198, Nanjing, China.
| |
Collapse
|
49
|
He C, Chen Y, Zhang X, Feng H, Rao Y, Ji T, Wang W. Down-regulation of ESRP2 inhibits breast cancer cell proliferation via inhibiting cyclinD1. Sci Rep 2024; 14:28475. [PMID: 39557898 PMCID: PMC11574003 DOI: 10.1038/s41598-024-77980-9] [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: 06/15/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024] Open
Abstract
Epithelial splicing regulatory protein 2 (ESRP2),an important alternative splicing protein of mRNA, is reported to have a dual role in tumors, which can promote or inhibit the occurrence and development of tumors. However, the function and mechanism of ESRP2 in breast cancer (BC) remain unclear. The distribution of ESRP2 expression in breast cancer and the correlation between ESRP2 expression and the overall survival rate were detected by The Cancer Genome Atlas (TCGA) database. Gene Ontology(GO)analysis, containing biological process, cellular components, and molecular function, was utilized to evaluate the potential mechanism of ESRP2 in breast cancer. The ESRP2 expression in breast cancer cell lines was detected by real-time quantitative PCR analysis (RT-qPCR) and western blotting. Cell clone was performed to examine the proliferation of ESRP2 knockdown in MCF-7 cells. The cell cycle was measured by flow cytometry assays. The role of ESRP2 knockdown in synergistic effect with chemotherapeutic agents was also determined by MTT assay. Bioinformatics analysis demonstrated that the ESRP2 gene was elevated in breast cancer cells and its overexpression was strongly correlated with shorter overall survival. GO analysis revealed that ESRP2 expression was related to cell proliferation. ESRP2 mRNA and protein expression were elevated in breast cancer cell lines, compared to the normal human breast cell line MCF-10 A. Dwon-regulation of ESRP2 inhibited cell proliferation and promoted the sensitivity of chemotherapy drug, Cisplatin(DDP) and Paclitaxel (TAXOL), in MCF-7 cells.Additionally, ESRP2 knockdown obstructed the cell cycle at the G1 phase and caused a decrease in cyclinD1 protein expression. These findings reveal that ESRP2 is highly expressed in breast cancer and is correlated with poor prognosis in breast cancer patients. ESRP2 knockdown can inhibit MCF-7 cell proliferation by arresting the cell cycle at the G1 phase and promoting the sensitivity of chemotherapy drugs (DDP and TAXOL)in MCF-7 cells. ESRP2 may be required for the regulation of breast cancer progression, as well as a critical target for the clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Caiping He
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuting Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Ximin Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangzhou Civil Aviation College, Guangzhou, Guangdong, China
| | - Huancun Feng
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yuzhen Rao
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Tangyang Ji
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenya Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
50
|
Verstappe J, Skrypek N, De Coninck J, Soen B, Taminau J, Tatari M, Bruneel K, Loret N, De Clercq K, Van den Broecke C, Van De Vijver K, Van Dorpe J, Haigh J, De Craene B, Goossens S, Vandamme N, Berx G. ZEB2 drives intra-tumor heterogeneity and skin squamous cell carcinoma formation with distinct EMP transition states. iScience 2024; 27:111169. [PMID: 39555401 PMCID: PMC11567922 DOI: 10.1016/j.isci.2024.111169] [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: 04/25/2024] [Revised: 08/23/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024] Open
Abstract
About 5% of patients with cutaneous squamous cell carcinoma (cSCC) have a poor prognosis which is associated with a loss of tumor differentiation, invasion and metastasis, all of which are linked to the process of epithelial-to-mesenchymal plasticity (EMP). Here, we showed that the EMP-associated transcription factor ZEB2 drives cSCC heterogeneity which resembles biphasic carcinosarcoma-like tumors. Single cell RNA sequencing revealed distinct subpopulations ranging from fully epithelial (E) to intermediate (EM) to fully mesenchymal (M), associated with the gradual loss of cell surface markers EPCAM, CDH1, ITGB4, and CD200. Mesenchymal features were associated with a higher metastatic capacity and anoikis resistance, yet this comes with a sensitivity toward TNF-induced cell death. Altogether we provide insights in cSCC heterogeneity and modes to target mesenchymal-metastasis inducing cells.
Collapse
Affiliation(s)
- Jeroen Verstappe
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Nicolas Skrypek
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jordy De Coninck
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- VIB Single Cell Core, VIB, Ghent-Leuven, Belgium
| | - Bieke Soen
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Joachim Taminau
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Marianthi Tatari
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Kenneth Bruneel
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Nele Loret
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Kato De Clercq
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Caroline Van den Broecke
- Department of Pathology, Ghent Unviversity Hospital, Ghent, Belgium
- AZ Sint-Lucas Gent, Ghent, Belgium
| | - Koen Van De Vijver
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, Ghent Unviversity Hospital, Ghent, Belgium
- Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Pathology, Ghent Unviversity Hospital, Ghent, Belgium
| | - Jody Haigh
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
| | - Bram De Craene
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Niels Vandamme
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- VIB Single Cell Core, VIB, Ghent-Leuven, Belgium
- VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium
| | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|