1
|
Mishan MA, Choo YM, Winkler J, Hamann MT, Karan D. Manzamine A: A promising marine-derived cancer therapeutic for multi-targeted interactions with E2F8, SIX1, AR, GSK-3β, and V-ATPase - A systematic review. Eur J Pharmacol 2025; 990:177295. [PMID: 39863145 DOI: 10.1016/j.ejphar.2025.177295] [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: 08/12/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Manzamine A, a natural compound derived from various sponge genera, features a β-carboline structure and exhibits a range of biological activities, including anti-inflammatory and antimalarial effects. Its potential as an anticancer agent has been explored in several tumor models, both in vitro and in vivo, showing effects through mechanisms such as cytotoxicity, regulation of the cell cycle, inhibition of cell migration, epithelial-to-mesenchymal transition (EMT), autophagy, and apoptosis through multi-target interactions of E2F transcriptional factors, ribosomal S6 kinases, androgen receptor (AR), SIX1, GSK-3β, v-ATPase, and p53/p21/p27 cascades. This systematic review evaluates existing literature on the potential application of this marine alkaloid as a novel cancer therapy, highlighting its promising ability to inhibit cancer cell growth while causing minimal side effects.
Collapse
Affiliation(s)
- Mohammad Amir Mishan
- Department of Urology, Brown Cancer Center, 505 S Hancock Street, Louisville, KY, USA
| | - Yeun-Mun Choo
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Jeffery Winkler
- Department of Chemistry, The University of Pennsylvania, Philadelphia, PA, USA
| | - Mark T Hamann
- Department of Drug Discovery and Biomedical Sciences and Public Health, Colleges of Pharmacy and Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Dev Karan
- Department of Urology, Brown Cancer Center, 505 S Hancock Street, Louisville, KY, USA.
| |
Collapse
|
2
|
Liao EC, Law CH, Chen HY, Wei YS, Tsai YT, Lin LH, Lin MW, Wang YS, Chou HC, Chan HL. PPIA enhances cell growth and metastasis through CD147 in oral cancer. Arch Biochem Biophys 2025; 765:110328. [PMID: 39921142 DOI: 10.1016/j.abb.2025.110328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Oral cancer is a malignant tumor, and the associated death rate has significantly increased over the past few decades. Secreted fractions are involved in various physiological processes, and their analysis has become a promising approach for discovering diagnostic and prognostic biomarkers for cancer detection and monitoring metastasis. Therefore, the discovery of potential prognostic, diagnostic, and therapeutic biomarkers for oral cancer metastasis is beneficial for developing effective strategies in oral cancer therapy. In this study, we used secretomic analysis to identify the secreted proteins involved in oral cancer. One of the identified proteins, peptidylprolyl isomerase A (PPIA), was selected for further investigation. We used RNA interference to investigate the effect of PPIA secretion on invasion and migration of OC3-I5 cells. Our results showed that reducing the expression and secretion of PPIA significantly decreased invasion and migration of OC3-I5 cells. Next, we used recombinant PPIA to investigate its direct effect on OC3 cell metastasis. The results revealed that proliferation, migration, and invasion of OC3 cells were significantly increased by treatment with the recombinant PPIA. Immunohistochemical analyses revealed higher PPIA expression in tumor tissues compared to normal tissues. Concisely, PPIA activated the ERK1/2 and p38 MAPK signaling pathways and enhanced cell proliferation and metastasis through CD147. In summary, PPIA may prove to be a novel target for oral cancer therapy as well as a prognostic marker.
Collapse
Affiliation(s)
- En-Chi Liao
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsuan Law
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsin-Yi Chen
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Shan Wei
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ting Tsai
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Hsun Lin
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Meng-Wei Lin
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Shiuan Wang
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiu-Chuan Chou
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Hong-Lin Chan
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan; Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
3
|
Xie Z, Yang T, Zhou C, Xue Z, Wang J, Lu F. Integrative Bioinformatics Analysis and Experimental Study of NLRP12 Reveal Its Prognostic Value and Potential Functions in Ovarian Cancer. Mol Carcinog 2025; 64:383-398. [PMID: 39601513 DOI: 10.1002/mc.23854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
NLRP12 plays a significant role in cellular functional behavior and immune homeostasis, influencing inflammation, tumorigenesis, and prognosis. This study aimed to explore its specific effects on the tumor microenvironment (TME) and its contribution to heterogeneity in ovarian cancer (OV) through bioinformatics analysis and experimental verification. Utilizing various bioinformatics databases and clinical specimens, we investigated NLRP12 expression and its relationship with OV prognosis and immune infiltration. In vitro assays were conducted to assess the impact of NLRP12 on the proliferation and invasion of OV cells. Our findings indicate that NLRP12 is upregulated in OV, with high expression correlating with a negative prognosis. Furthermore, NLRP12 expression demonstrated a positive correlation with the infiltration of various immune cells and the expression of immune checkpoint molecules in OV. Analysis of The Cancer Immunome Atlas (TCIA) database revealed that OV patients with lower NLRP12 expression may exhibit an enhanced response to immunotherapy, particularly CTLA4 blockers, a finding validated in animal experiments. Additionally, the study emphasized the role of NLRP12 in influencing the prognosis of OV patients by promoting epithelial-mesenchymal transition (EMT) in ovarian cancer cells. Finally, we identified a potential therapeutic compound, Schisandrin B (Schi B), which decreases NLRP12 expression in ovarian cancer cells by binding to the transcription factor SPI1 associated with NLRP12. Our findings suggest that NLRP12 serves as a crucial immune-related biomarker predicting poor outcomes in OV, and targeting NLRP12 may represent a promising therapeutic approach for OV patients in the future.
Collapse
Affiliation(s)
- Zhihui Xie
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Tiantian Yang
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Chuchu Zhou
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Zixin Xue
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Jianjun Wang
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Feng Lu
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| |
Collapse
|
4
|
Roshanzadeh A, Medeiros HCD, Herrera CK, Malhado C, Tomich AW, Fisher SP, Lovera SO, Bates M, Lavallo V, Lunt RR, Lunt SY. Next-Generation Photosensitizers: Cyanine-Carborane Salts for Superior Photodynamic Therapy of Metastatic Cancer. Angew Chem Int Ed Engl 2025; 64:e202419759. [PMID: 39841576 DOI: 10.1002/anie.202419759] [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] [Revised: 01/03/2025] [Accepted: 01/22/2025] [Indexed: 01/24/2025]
Abstract
Photodynamic therapy (PDT) has emerged as a promising targeted treatment for cancer. However, current PDT is limited by low tissue penetration, insufficient phototoxicity (toxicity with light irradiation), and undesirable cytotoxicity (toxicity without light irradiation). Here, we report the discovery of cyanine-carborane salts as potent photosensitizers (PSs) that harness the near-infrared (NIR) absorbing [cyanine+] with the inertness of [carborane-]. The implementation of [cyanine+] [carborane-] salts dramatically enhance cancer targeting of the PSs and decrease cytotoxicity. We characterize the cellular uptake of the cyanine-carborane PSs, organelle localization, generation of reactive oxygen species (ROS) with the ability to cogenerate multiple ROS species, suppression of pro-metastatic pathways, and activation of apoptotic pathways. We further demonstrate the ability of optimized PSs to eliminate tumors in vivo using an orthotopic mouse model of breast cancer. These newly developed [cyanine+] [carborane-] salt PSs introduce a potent therapeutic approach against aggressive breast cancer while decreasing side effects.
Collapse
Affiliation(s)
- Amir Roshanzadeh
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States
| | - Hyllana C D Medeiros
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States
| | - Christopher K Herrera
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824, United States
| | - Carson Malhado
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824, United States
| | - Anton W Tomich
- Department of Chemistry, University of California, Riverside, CA 92521, United States
| | - Steven P Fisher
- Department of Chemistry, University of California, Riverside, CA 92521, United States
| | - Sergio O Lovera
- Department of Chemistry, University of California, Riverside, CA 92521, United States
| | - Matthew Bates
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824, United States
| | - Vincent Lavallo
- Department of Chemistry, University of California, Riverside, CA 92521, United States
| | - Richard R Lunt
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824, United States
- Department of Physics and Astronomy, Michigan State University, East Lansing, MI 48824, United States
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824, United States
| |
Collapse
|
5
|
Zhang H, Nie J, Bao Z, Shi Y, Gong J, Li H. FOXC1 promotes EMT and colorectal cancer progression by attracting M2 macrophages via the TGF-β/Smad2/3/snail pathway. Cell Signal 2025; 130:111680. [PMID: 39978609 DOI: 10.1016/j.cellsig.2025.111680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Colorectal cancer is a highly prevalent and deadly malignancy worldwide. Current treatment strategies, including surgery, chemotherapy, and targeted therapy, still face limitations due to recurrence and metastasis. By conducting a weighted gene coexpression network analysis on gene expression data from The Cancer Genome Atlas, we pinpointed critical genes linked to M2 macrophages and tumor metastasis. Among these, FOXC1 emerged as a significant prognostic indicator within our predictive model. Clinical sample analysis further confirmed that FOXC1 is upregulated in colorectal cancer tissues and associated with an unfavorable patient outcome. Both in vivo and in vitro experimental results revealed that FOXC1 promotes CRC cell migration, invasion and proliferation by regulating the expression of Snail and TGF-β/Smad2/3 pathways, thereby facilitating the epithelial-mesenchymal transition process. Additionally, FOXC1 recruits M2 macrophages to the tumor microenvironment by regulating CXCL2 expression through Snail. The TGF-β factor secreted by M2 macrophages further activates the TGF-β/Smad2/3 pathway, forming a positive feedback loop. In these processes, FOXC1 plays a critical regulatory role. In summary, this study highlights the critical significance of FOXC1 in CRC progression and indicates its viability as a therapeutic target, offering a novel theoretical foundation for the development of future CRC treatment strategies.
Collapse
Affiliation(s)
- Haoran Zhang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, PR China
| | - Jinlin Nie
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, PR China; Department of Hepatobiliary Pancreatic Hernia Surgery, the Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, PR China
| | - Zhen Bao
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, PR China
| | - Yangdong Shi
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, PR China
| | - Jin Gong
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, PR China.
| | - Hailiang Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, the Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, PR China.
| |
Collapse
|
6
|
Chiang CH, Yang JD, Liu WL, Chang FY, Yang CJ, Hsu KW, Chiang IT, Hsu FT. Mechanistic insights of lenvatinib: enhancing cisplatin sensitivity, inducing apoptosis, and suppressing metastasis in bladder cancer cells through EGFR/ERK/P38/NF-κB signaling inactivation. Cancer Cell Int 2025; 25:47. [PMID: 39955573 PMCID: PMC11829490 DOI: 10.1186/s12935-024-03597-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/03/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND The persistent activation of the epidermal growth factor receptor (EGFR) leads to the activation of downstream oncogenic kinases and transcription factors, resulting in tumor progression and an increased resistance to cisplatin in bladder cancer (BC) cells. Lenvatinib, an oral multikinase inhibitor, has the potential to offer therapeutic benefits as an adjuvant treatment for BC patients. The investigation into its application in bladder cancer treatment is a valuable endeavor. The primary goal of this study is to confirm the effectiveness and mechanism of lenvatinib in inhibiting the progression of BC and enhancing the anticancer efficacy of cisplatin. MATERIALS Three BC cell lines, namely, TSGH-8301, T24, and MB49, along with an MB49-bearing animal model, were utilized in this study. RESULTS In vitro experiments utilizing MTT assays demonstrated that lenvatinib sensitized BC cells to cisplatin, exhibiting a synergistic effect. Flow cytometry indicated apoptotic events and signaling, presenting that lenvatinib effectively induced apoptosis and triggered extrinsic/intrinsic apoptotic pathways. In vivo studies using a mouse model of BC confirmed the antitumor efficacy of lenvatinib, demonstrating significant tumor growth suppression without inducing toxicity in normal tissues. Western blotting analysis and immunohistochemistry stain revealed EGF-phosphorylated EGFR and EGFR-mediated ERK/P38/NF-κB signaling were suppressed by treatment with lenvatinib. In addition, lenvatinib also suppressed anti-apoptotic (MCL1, c-FLIP, and XIAP) and metastasis-related factors (Twist, Snail-1, ZEB-1, ZEB-2, and MMP9) and promoted epithelial markers (E-cadherin) while reducing mesenchymal markers (N-cadherin). CONCLUSION In conclusion, the induction of apoptosis and the inhibition of EGFR/ERK/P38/NF-κB signaling are correlated with lenvatinib's ability to hinder tumor progression and enhance the cytotoxic effects of cisplatin in bladder cancer. These findings underscore the potential of lenvatinib as a therapeutic option for bladder cancer, either as a standalone treatment or in combination with cisplatin.
Collapse
Grants
- YSVH111-04 Taipei Veterans General Hospital, Yuan-Shan Branch, Taiwan
- RD2021-007 National Yang Ming Chiao Tung University Hospital, Yilan, Taiwan
- SRD-108008 Show-Chwan Memorial Hospital, Changhua, Taiwan
- BRD-108027 Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
- MOST 109-2314-B-039-021-MY3 Ministry of Science and Technology (MOST), Taipei, Taiwan
Collapse
Affiliation(s)
- Chih-Hung Chiang
- Division of Urology, Department of Surgery and Department of Research and Development, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan, R.O.C
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan, R.O.C
- Department and Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Jr-Di Yang
- Division of Urology, Department of Surgery, National Yang-Ming Chiao Tung University Hospital, Yilan, Taiwan, R.O.C
| | - Wei-Lin Liu
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Fang-Yu Chang
- Department of Biological Science and Technology, China Medical University, Office: 7F, Research building, No. 100, Jingmao 1st Rd., Beitun Dist, Taichung City, 406040, Taiwan, R.O.C
| | - Che-Jui Yang
- Division of Urology, Department of Surgery, Chang Bing Show-Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Kai-Wen Hsu
- Research Center for Cancer Biology, China Medical University, Taichung, Taiwan, R.O.C..
- Drug Development Center, Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan, R.O.C..
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan, R.O.C..
| | - I-Tsang Chiang
- Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Lukang, Taiwan, R.O.C
- Department of Medical Imaging and Radiologic Sciences, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C
- Medical Administrative Center, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Office: 7F, Research building, No. 100, Jingmao 1st Rd., Beitun Dist, Taichung City, 406040, Taiwan, R.O.C..
- Department of Life Sciences, National Central University, Taoyuan, Taiwan, R.O.C..
| |
Collapse
|
7
|
Kanwal R, Esposito JE, Jawed B, Zakir SK, Pulcini R, Martinotti R, Botteghi M, Gaudio F, Martinotti S, Toniato E. Exploring the Role of Epithelial-Mesenchymal Transcriptional Factors Involved in Hematological Malignancy and Solid Tumors: A Systematic Review. Cancers (Basel) 2025; 17:529. [PMID: 39941895 PMCID: PMC11817253 DOI: 10.3390/cancers17030529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/13/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND The epithelial mesenchymal transition (EMT) is a biological process in which epithelial cells lose their polarity and adhesion characteristics, and adopt a mesenchymal phenotype. While the EMT naturally occurs during tissue fibrosis, wound healing, and embryonic development, it can be exploited by cancer cells and is strongly associated with cancer stem cell formation, tissue invasiveness, apoptosis, and therapy resistance. Transcription factors (TFs) such as SNAIL, ZEB, and TWIST play a pivotal role in driving the EMT. This systematic review aims to assess the impact of EMT-TFs on hematological malignancy and solid tumors. METHODS English-language literature published between 2010 and 2024 was systematically reviewed, utilizing databases such as PubMed and Google Scholar. RESULTS A total of 3250 studies were extracted. Of these, 92 publications meeting the inclusion criteria were analyzed to elucidate the role of EMT-TFs in cancer. The results demonstrated that the EMT-TFs play a critical role in both hematological and solid tumor development and progression. They promote invasive, migratory, and metastatic properties in these tumors, and contribute to therapeutic challenges by enhancing chemoresistance. A strong correlation between EMT-TFs and poor overall survival has been identified. CONCLUSIONS Our research concluded that EMT-TFs may serve as important predictive and prognostic factors, as well as potential therapeutic targets to mitigate cancer progression.
Collapse
Affiliation(s)
- Rimsha Kanwal
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
- Unit of Clinical Pathology and Microbiology, Miulli Generale Hospital, 70021 Acquaviva delle Fonti, Italy
| | - Jessica Elisabetta Esposito
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
| | - Bilal Jawed
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
- Unit of Clinical Pathology and Microbiology, Miulli Generale Hospital, 70021 Acquaviva delle Fonti, Italy
| | - Syed Khuram Zakir
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
- Unit of Clinical Pathology and Microbiology, Miulli Generale Hospital, 70021 Acquaviva delle Fonti, Italy
| | - Riccardo Pulcini
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
| | - Riccardo Martinotti
- Residency Program in Clinical Oncology, Faculty of Medicine, Umberto I University Hospital, University of Rome “La Sapienza”, 00185 Rome, Italy;
| | - Matteo Botteghi
- Experimental Pathology Research Group, Department of Clinical and Molecular Sciences, Universita Politecnica delle Marche, 60126 Ancona, Italy;
| | - Francesco Gaudio
- Unit of Haematology, Department of Medicine and Surgeon, F. Miulli University Hospital, LUM University, Casamassima, 70010 Bari, Italy
| | - Stefano Martinotti
- Unit of Clinical Pathology, Department of Medicine and Surgeon, F. Miulli University Hospital, LUM University, Casamassima, 70010 Bari, Italy
| | - Elena Toniato
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
| |
Collapse
|
8
|
López-Collazo E, Hurtado-Navarro L. Cell fusion as a driver of metastasis: re-evaluating an old hypothesis in the age of cancer heterogeneity. Front Immunol 2025; 16:1524781. [PMID: 39967663 PMCID: PMC11832717 DOI: 10.3389/fimmu.2025.1524781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Numerous studies have investigated the molecular mechanisms and signalling pathways underlying cancer metastasis, as there is still no effective treatment for this terminal stage of the disease. However, the exact processes that enable primary cancer cells to acquire a metastatic phenotype remain unclear. Increasing attention has been focused on the fusion of cancer cells with myeloid cells, a phenomenon that may result in hybrid cells, so-called Tumour Hybrid Cells (THCs), with enhanced migratory, angiogenic, immune evasion, colonisation, and metastatic properties. This process has been shown to potentially drive tumour progression, drug resistance, and cancer recurrence. In this review, we explore the potential mechanisms that govern cancer cell fusion, the molecular mediators involved, the metastatic characteristics acquired by fusion-derived hybrids, and their clinical significance in human cancer. Additionally, we discuss emerging pharmacological strategies aimed at targeting fusogenic molecules as a means to prevent metastatic dissemination.
Collapse
Affiliation(s)
- Eduardo López-Collazo
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
- CIBER of Respiratory Diseases (CIBERES), Madrid, Spain
- UNIE University, Madrid, Spain
| | - Laura Hurtado-Navarro
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
9
|
Dou L, Yan Y, Lu E, Li F, Tian D, Deng L, Zhang X, Zhang R, Li Y, Zhang Y, Sun Y. Composition analysis and mechanism of Guizhi Fuling capsule in anti-cisplatin-resistant ovarian cancer. Transl Oncol 2025; 52:102244. [PMID: 39662450 PMCID: PMC11683237 DOI: 10.1016/j.tranon.2024.102244] [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: 08/18/2024] [Revised: 11/18/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024] Open
Abstract
OBJECTIVE Cisplatin is the main chemotherapy drug for advanced ovarian cancer, but drug resistance often occurs. The aim of this study is to explore the molecular mechanism by which Guizhi Fuling capsule inhibits cisplatin resistance in ovarian cancer. METHODS First, differences in cisplatin resistance, PA2G4 gene expression, migration, and invasion in A2780 cells and A2780/DDP cells were analyzed by qRT-PCR, scratch assay, transwell, immunofluorescence, and western blotting. Then, LC-MS/MS analysis of GFC chemical composition. qRT-PCR, scratch tests, transwell, pseudopodium formation, immunofluorescence, and western blotting were used to explore the mechanism by which GFC inhibited A2780/DDP cell migration and invasion. Finally, the anti-tumor efficacy of GFC was verified by in vivo experiments. RESULTS A2780/DDP cells had a greater ability to migrate and invade compared to their parents. Cell viability experiments showed that the migration and invasion ability of A278/DDP cells were significantly inhibited with the increase of GFC concentration. qRT-PCR results showed that compared with the blank control group, cisplatin group and GFC group, the transcription level of PA2G4 gene in the combination treatment group was significantly reduced. We also found that GFC combined with cisplatin inhibited the PI3K/AKT/GSK-3β signaling pathway by targeting PA2G4 gene expression, inhibited the epithelial-mesenchymal transition signaling pathway, decreased cell adhesion and inhibited the formation of cell pseudopodias. CONCLUSION GFC combined with cisplatin can target PA2G4 gene to regulate PI3K/AKT/GSK-3β Signaling pathway, inhibiting the invasion and migration of cisplatin resistant A2780/DDP cells in ovarian cancer.
Collapse
Affiliation(s)
- Lei Dou
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yan Yan
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Enting Lu
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Fangmei Li
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Dongli Tian
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Lei Deng
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xue Zhang
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Rongjin Zhang
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yin Li
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yi Zhang
- Department of Gynecology, the First Hospital of China Medical University, Shenyang 110001, China.
| | - Ye Sun
- Department of Pathogenic Biology, College of Basic Medical Sciences, Shenyang Medical College, Shenyang 110034, China.
| |
Collapse
|
10
|
Qi L, Wang J, Hou S, Liu S, Zhang Q, Zhu S, Liu S, Zhang S. Unraveling the tumor microenvironment of esophageal squamous cell carcinoma through single-cell sequencing: A comprehensive review. Biochim Biophys Acta Rev Cancer 2025; 1880:189264. [PMID: 39805342 DOI: 10.1016/j.bbcan.2025.189264] [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: 08/27/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly heterogeneous and aggressive malignancy. The progression, invasiveness, and metastatic potential of ESCC are shaped by a multitude of cells within the tumor microenvironment (TME), including tumor cells, immune cells, endothelial cells, as well as fibroblasts and other cell types. Recent advancements in single-cell sequencing technologies have significantly enhanced our comprehension of the diverse landscape of ESCC. Single-cell multi-omics technology, particularly single-cell transcriptome sequencing, have shed light on the expression profiles of individual cells and the molecular characteristics of distinct tumor cell populations. This review summarizes the latest literature on single-cell research in the field of ESCC, aiming to elucidate the heterogeneity of tumor cells, immune cells, and stromal cells at the single-cell level. Furthermore, it explores the impact of cellular interactions within the TME on the progression of ESCC. By compiling a comprehensive overview of single-cell omics research on ESCC, this article aims to enhance our understanding of ESCC diagnosis and treatment by elucidating the intricate interplay within the TME. It explores the cellular composition, spatial arrangement, and functional attributes of the ESCC TME, offering potential therapeutic targets and biomarkers for personalized treatment strategies.
Collapse
Affiliation(s)
- Lingyu Qi
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Jiaxin Wang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Songyuan Hou
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Siying Liu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Qian Zhang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Shengtao Zhu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Si Liu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China.
| | - Shutian Zhang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China.
| |
Collapse
|
11
|
Chen Q, Yang M, Duan X, Zhang J, Shi F, Chen R, Li Y. Linker Histone H1.4 Inhibits the Growth, Migration and EMT Process of Non-Small Cell Lung Cancer by Regulating ERK1/2 Expression. Biochem Genet 2025; 63:576-591. [PMID: 38472566 DOI: 10.1007/s10528-024-10760-2] [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: 09/21/2023] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
H1.4 is one of the 11 variants of linker histone H1, and is associated with tumorigenesis and development of various cancers. However, it is unclear for the role of histone H1.4 in non-small cell lung cancer (NSCLC). In this study, we found that overexpression of H1.4 significantly inhibited the cell viability, migration, invasion and epithelial-mesenchymal transition (EMT) processes, whereas silencing H1.4 by shRNA knockdown promoted these processes in NSCLC cell lines A549 and H1299. We further showed that H1.4 overexpression reduced ERK1/2 expression or its phosphorylation levels, while H1.4 knockdown increased ERK1/2 expression or phosphorylation levels in NSCLC. Furthermore, we demonstrated that H1.4 bound to the promoter of ERK1/2, and acted as a transcriptional suppressor to inhibit ERK1/2 expression in A549 or H1299 cells. Importantly, we found that ERK ecto-expression can largely recovered the inhibitory effects of H1.4 on cell viability, migration, invasion and EMT processes. In summary, our study reveals that the H1.4-ERK pathway is crucial for cell viability, migration, invasion and EMT of NSCLC and could be a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Qian Chen
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Mengqi Yang
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Xinyue Duan
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Jie Zhang
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Fan Shi
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Rong Chen
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Yong Li
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China.
- Center for Stem Cell and Translational Medicine, Anhui University, Hefei, Anhui Province, China.
| |
Collapse
|
12
|
Janjua D, Chaudhary A, Joshi U, Tripathi T, Bharti AC. Circulating tumor cells in solid malignancies: From advanced isolation technologies to biological understanding and clinical relevance in early diagnosis and prognosis. Biochim Biophys Acta Rev Cancer 2025; 1880:189236. [PMID: 39662757 DOI: 10.1016/j.bbcan.2024.189236] [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: 09/17/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
Circulating tumor cells (CTCs) are shed from primary tumors and travel through the body via circulation, eventually settling to form micrometastases under favorable conditions. Numerous studies have identified CTCs as a negative prognostic indicator for survival across various cancer types. CTCs mirror the current heterogeneity and genetic and biological state of tumors, making their study invaluable for understanding tumor progression, cell senescence, and cancer dormancy. However, their isolation and characterization still poses a major challenge that limits their clinical translation. A wide array of methods, each with different levels of specificity, utility, cost, and sensitivity, have been developed to isolate and characterize CTCs. Moreover, innovative techniques are emerging to address the limitations of existing methods. In this review, we provide insights into CTC biology addressing spectra of markers employed for molecular analysis and functional characterization. It also emphasizes current label-dependent and label-independent isolation procedures, addressing their strengths and limitations. SIGNIFICANCE: A comprehensive overview of CTC biology, their molecular and functional characterization, along with their current clinical utility will help in understanding the present-day extent to which the clinical potential of CTCs is getting tapped in personalized medicine.
Collapse
Affiliation(s)
- Divya Janjua
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), New Delhi, India
| | - Apoorva Chaudhary
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), New Delhi, India
| | - Udit Joshi
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), New Delhi, India
| | - Tanya Tripathi
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), New Delhi, India
| | - Alok Chandra Bharti
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), New Delhi, India.
| |
Collapse
|
13
|
Shi Y, Min X, Li Y, Guo L, Cai Z, Li D, Jiang X, Feng N, Li X, Yang X. Yinjia pills inhibits the malignant biological behavior of HeLa cells through PKM2-medicated inhibition of JAK/STAT3 pathway. Cytotechnology 2025; 77:5. [PMID: 39575323 PMCID: PMC11579276 DOI: 10.1007/s10616-024-00668-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024] Open
Abstract
Cervical cancer is one of the most common tumors in women and is a major problem in gynecological health. Studies have shown that Yinjia pills (YJP), a traditional Chinese medicine, can effectively slow the progression of cervical cancer. Therefore, this study mainly explored the molecular mechanism by which YJP delays the progression of cervical cancer. The expression level of PKM2 in cervical cancer was evaluated by the gene expression profiling interactive analysis (GEPIA) database, and the prognostic value of the PKM2 gene was evaluated by the Kaplan‒Meier plotter database. HeLa cervical cancer cells were treated with different concentrations of YJP (2.5, 5, 10, and 20 mg/mL). The levels of the inflammatory factors were detected by ELISA. Cell proliferation activity, migration and invasion were detected by CCK-8 assay, Transwell assays and cell scratch experiment. Apoptosis was detected by flow cytometry. Western blotting was used to detect the expression of proteins. In this study, PKM2 was upregulated in both cervical squamous cell carcinoma (CESC) and endometrial adenocarcinoma tissues, and a Kaplan‒Meier analysis showed that higher PKM2 expression was associated with lower patient survival. YJP inhibited the proliferation, migration and invasion of HeLa cells in a dose-dependent manner, promoted the apoptosis of HeLa cells, and inhibited the expression of inflammatory factors. In addition, YJP inhibited the activation of the JAK/STAT3 pathway and the occurrence of EMT. Knockdown of PKM2 also inhibited the malignant biological behavior of HeLa cells, but overexpression of PKM2 weakened the inhibitory effect of YJP on the malignant biological behavior of HeLa cells. Angoline, a JAK/STAT3 pathway inhibitor, attenuated the effect of PKM2 overexpression on the efficacy of YJP. In conclusion, YJP can inhibit the activation of the JAK/STAT3 pathway by regulating PKM2, thereby inhibiting the malignant biological behavior of HeLa cells.
Collapse
Affiliation(s)
- Ying Shi
- Oncology Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, 120 Guanghua Street, Kunming, 650200 Yunnan China
| | - Xiaoli Min
- Department of Cerebrovascular Disease, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan China
| | - Yi Li
- Oncology Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, 120 Guanghua Street, Kunming, 650200 Yunnan China
| | - Lihua Guo
- Oncology Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, 120 Guanghua Street, Kunming, 650200 Yunnan China
| | - Zheng Cai
- Oncology Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, 120 Guanghua Street, Kunming, 650200 Yunnan China
| | - Dongge Li
- Oncology Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, 120 Guanghua Street, Kunming, 650200 Yunnan China
| | - Xueying Jiang
- The First Clinical Medical College, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan China
| | - Ni Feng
- Oncology Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, 120 Guanghua Street, Kunming, 650200 Yunnan China
| | - Xiaolin Li
- Oncology Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, 120 Guanghua Street, Kunming, 650200 Yunnan China
| | - Xiaoxia Yang
- Oncology Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, 120 Guanghua Street, Kunming, 650200 Yunnan China
| |
Collapse
|
14
|
Xiong L, Li D, Xiao G, Tan S, Xu L, Wang G. HSP70 Promotes Pancreatic Cancer Cell Epithelial-Mesenchymal Transformation and Growth Via the NF-κB Signaling Pathway. Pancreas 2025; 54:e89-e96. [PMID: 39352012 DOI: 10.1097/mpa.0000000000002398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
OBJECTIVE To study the effects of HSP70 on proliferation, migration, invasion, and epithelial-mesenchymal transformation (EMT) of pancreatic cancer cells and explore its underlying mechanisms. METHODS Pancreatic cancer cell models with reduced HSP70 or increased HSP70 expression were established. Reverse transcription quantitative polymerase chain reaction and Western blot assays were used to determine mRNA and protein levels of HSP70, IKK/IκBa/NF-κB signaling pathway-related genes, and EMT markers. CCK-8 and cell cloning assays were used to evaluate cell proliferation and cloning abilities. Transwell and wound healing assays were used to assess the invasive and migratory properties of cells. Electrophoresis mobility shift assay (EMSA) and luciferase reporter assays were conducted to analyze NF-κB's promoter binding and transcriptional activities. RESULTS HSP70 knockdown inhibited p-p65 nuclear translocation, the expression of p-p65, p-IKKα/β, p-IκBα, N-cadherin, Vimentin and Twist, NF-κB's promoter binding and transcriptional activities, pancreatic cancer cell proliferation, cloning, migration and invasion, while increased E-cadherin levels. HSP70 overexpression took the opposite effects. NF-κB signaling pathway modulation reversed EMT changes induced by altered HSP70 expression levels. rhHSP70 increased p-IKKα/β and p-IκBα protein levels. CONCLUSIONS HSP70 promotes EMT and enhances pancreatic cancer cell proliferation, migration, and invasion by activating NF-κB pathway.
Collapse
Affiliation(s)
- Liumei Xiong
- From the Department of Gastroenterology, Pingxing Hospital, Southern Medical University, Pingxiang, China
| | - Danming Li
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gui Xiao
- Department of International School of Nursing, Hainan Medical University, Haikou, China
| | - Sipin Tan
- Sepsis Translational Medicine, Key Lab of Hunan, Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Linfang Xu
- From the Department of Gastroenterology, Pingxing Hospital, Southern Medical University, Pingxiang, China
| | | |
Collapse
|
15
|
Gao S, Li J, Wang W, Wang Y, Shan Y, Tan H. Rabdosia rubescens (Hemsl.) H. Hara: A potent anti-tumor herbal remedy - Botany, phytochemistry, and clinical applications and insights. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119200. [PMID: 39631716 DOI: 10.1016/j.jep.2024.119200] [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: 09/14/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese herbal medicine has unique advantages as anti-cancer drugs and adjuvant therapies. Rabdosia rubescens (Hemsl.) H. Hara (R. rubescens) is a traditional medicinal plant known for its anti-inflammatory, antioxidant, antibacterial, anti-angiogenic and antitumor properties. The antitumor activity of R. rubescens is widely recognized among the folk communities in Henan Province, China. AIM OF THE STUDY This study reviews the botany, ethnopharmacology, phytochemistry, anti-tumor active ingredients, mechanisms, and clinical applications of R. rubescens, aiming to provide a comprehensive understanding for its use as an anti-cancer drug and adjuvant therapy. MATERIALS AND METHODS We systematically searched the literature in PubMed, Web of Science, and CNKI using the following keywords: "Rabdosia rubescens", "Isodon rubescens", "traditional application", "anti-tumor", "phytochemistry", "anti-tumor active compounds", "oridonin" and "clinical application". The search covered publications from 1997 to 2024. Inclusion criteria included original studies or reviews focusing on the anti-tumor properties of R. rubescens or its active components. Exclusion criteria included studies related to non-R. rubescens applications. RESULTS R. rubescens is a perennial herbaceous plant in the family Lamiaceae, mainly found in central and southern China. Historically, it has been used to treat conditions such as sore throat, cough, and excess phlegm. The plant contains various compounds, including diterpenes, triterpenes, steroids, flavonoids, phenolic acids, essential oils, amino acids, alkaloids, and polysaccharides, with diterpenes, triterpenes, flavonoids, and phenolic acids being the most active. This review identifies 50 compounds with anti-tumor properties, comprising 34 diterpenes, 2 triterpenes, 7 flavonoids, and 7 phenolic acids. Notably, besides oridonin and ponicidin, the ent-kaurane diterpenoids (20S)-11β,14β,20-trihydroxy-7α,20-epoxy-ent-kaur-16-en15-one and (20S)-11β,14β-dihydroxy-20-ethoxy7α,20-epoxy-ent-kaur-16-en-15-one demonstrate significant anti-tumor activity, attributed to their carbonyl group at C-15, hydroxyl group at C-1, and OEt group at C-20. Mechanistically, R. rubescens combats tumors by blocking the tumor cell cycle, promoting apoptosis, inhibiting cell migration and angiogenesis, inducing ferroptosis, reversing drug resistance, and enhancing radiosensitivity in tumor cells. Clinically, R. rubescens is available in various forms, including tablets, drops, syrups, capsules, and lozenges, and is primarily used for tonsillitis, pharyngitis, and stomatitis. According to the 2020 edition of the Pharmacopoeia of China, R. rubescens tablets are recognized as an adjuvant therapy for cancer. Clinical studies indicate that R. rubescens syrup, tablets, and thermal therapy can enhance cancer patient survival rates and lower tumor recurrence rates. CONCLUSIONS Given its traditional and modern uses, active anti-tumor components, and mechanisms, R. rubescens is a promising resource in traditional Chinese medicine for anti-tumor therapy. To realize its full potential, future research should explore additional active anti-tumor compounds beyond oridonin and ponicidin. For these key components, studies should focus on structural modifications to identify new active molecules and essential anti-tumor structures. Clinically, it is important to investigate how R. rubescens interacts with other Chinese herbs in anti-tumor formulations to enhance treatment efficacy and guide appropriate clinical use. Furthermore, future studies should undergo ethical review and include larger-scale randomized controlled trials to validate the efficacy of R. rubescens in treating tumors, thereby promoting its role as an anti-tumor traditional Chinese medicine.
Collapse
Affiliation(s)
- Shiyong Gao
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Jianwen Li
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Weiya Wang
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Yue Wang
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Yanmin Shan
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Huixin Tan
- Department of Pharmacy, Fourth Affiliated Hospital of Harbin Medicine University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
16
|
Xie Y, Guan S, Li Z, Cai G, Liu Y, Li G, Huang P, Lin M. Identification of a metabolic-immune signature associated with prognosis in colon cancer and exploration of potential predictive efficacy of immunotherapy response. Clin Exp Med 2025; 25:46. [PMID: 39853414 PMCID: PMC11762008 DOI: 10.1007/s10238-025-01566-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/10/2025] [Indexed: 01/26/2025]
Abstract
The role of metabolic reprogramming of the tumor immune microenvironment in cancer development and immune escape has increasingly attracted attention. However, the predictive value of differences in metabolism-immune microenvironment on the prognosis of colon cancer (CC) and the response to immunotherapy have not been elucidated. The aim of this study was to investigate changes in metabolism and immune profile of CC and to identify a reliable signature for predicting prognosis and therapeutic response. The metabolism and immune-related differential genes in CC were screened out by differential gene expression analysis. A metabolism and immune related prognostic signature was established by the least absolute shrinkage and selection operator (LASSO) Cox algorithm. The training cohort with 417 patients from The Cancer Genome Atlas (TCGA) database and the validation cohort of 232 patients from GSE17538 were used to confirm the robustness of the prognostic signature. Immunohistochemical staining scores were used to assess gene expression levels in our clinical samples. Gene ontology (GO) analysis, gene set enrichment analysis (GSEA), single nucleotide variation (SNV) analysis, immune infiltration and immune factors analysis were used to explore the characteristics of patients with different subtypes. Multiple cancer immunotherapy datasets were used to assess the response of patients with different subtypes to immune checkpoint inhibitors. We established the Metabolism and Immune-Related Prognostic Score (MIRPS) based on six genes (CD36, PCOLCE2, SCG2, CALB2, STC2, CLDN23) to predict the prognosis of CC patients. We found a correlation between MIRPS and the malignant phenotype, microsatellite subtype, mutation load, and immune escape in CC. Tumors with high MIRPS presented a higher tumor mutation load and a more prominent immunosuppressive microenvironment. This subset of patients may potentially respond well to immune checkpoint inhibitor therapy. MIRPS may be used as a novel prognostic tool for CC and have potential value for immunotherapy response prediction.
Collapse
Affiliation(s)
- Yuwen Xie
- Department of Radiation Oncology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Shenyuan Guan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhenkang Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guohao Cai
- Department of Anorectal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Yuechen Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Ping Huang
- Department of Anorectal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China.
| | - Mingdao Lin
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Department of Anorectal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China.
| |
Collapse
|
17
|
Boldu-Fernández S, Lliberos C, Simon C, Mas A. Mapping Human Uterine Disorders Through Single-Cell Transcriptomics. Cells 2025; 14:156. [PMID: 39936948 DOI: 10.3390/cells14030156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
Disruptions in uterine tissue function contribute to disorders such as endometriosis, adenomyosis, endometrial cancer, and fibroids, which all significantly impact health and fertility. Advances in transcriptomics, particularly single-cell RNA sequencing, have revolutionized uterine biological research by revealing the cellular heterogeneity and molecular mechanisms underlying disease states. Single-cell RNA sequencing and spatial transcriptomics have mapped endometrial and myometrial cellular landscapes, which helped to identify critical cell types, signaling pathways, and phase-specific dynamics. Said transcriptomic technologies also identified stromal and immune cell dysfunctions, such as fibroblast-to-myofibroblast transitions and impaired macrophage activity, which drive fibrosis, chronic inflammation, and lesion persistence in endometriosis. For endometrial cancer, scRNA-seq uncovered tumor microenvironmental complexities, identifying cancer-associated fibroblast subtypes and immune cell profiles contributing to progression and therapeutic resistance. Similarly, studies on adenomyosis highlighted disrupted signaling pathways, including Wnt and VEGF, and novel progenitor cell populations linked to tissue invasion and neuroinflammation, while single-cell approaches characterized smooth muscle and fibroblast subpopulations in uterine fibroids, elucidating their roles in extracellular matrix remodeling and signaling pathways like ERK and mTOR. Despite challenges such as scalability and reproducibility, single-cell transcriptomic approaches may have potential applications in biomarker discovery, therapeutic target identification, and personalized medicine in gynecological disorders.
Collapse
Affiliation(s)
| | - Carolina Lliberos
- Carlos Simón Foundation, INCLIVA Health Research Institute, 46010 Valencia, Spain
| | - Carlos Simon
- Carlos Simón Foundation, INCLIVA Health Research Institute, 46010 Valencia, Spain
- Department of Obstetrics and Gynecology, Universidad de Valencia, 46010 Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215, USA
| | - Aymara Mas
- Carlos Simón Foundation, INCLIVA Health Research Institute, 46010 Valencia, Spain
| |
Collapse
|
18
|
Wang B, Zhang N, Dai L, Zhang Y, Yin S, Yang X. Nonylphenol promotes epithelial-mesenchymal transition in colorectal cancer cells by upregulating miR-151a-3p. Discov Oncol 2025; 16:63. [PMID: 39832042 PMCID: PMC11747012 DOI: 10.1007/s12672-025-01805-y] [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: 09/03/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Nonylphenol (NP) is a common environmental contaminant and endocrine disruptor. Our previous research demonstrated that NP could promote the proliferation and epithelial-mesenchymal transition (EMT) of colorectal cancer (CRC) cells; however, the specific mechanism remains unclear. miRNA sequencing revealed that NP upregulated the expression levels of microRNA(miR)-151a-3p in CRC. Analysis of The Cancer Genome Atlas (TCGA) data revealed increased expression levels of miR-151a-3p in CRC tissues. The present experiments showed that NP could activate the WNT/β-catenin signaling pathway, and promoted the migration and invasion of CRC cells by increasing the expression levels of miR-151a-3p. Through bioinformatics analysis and dual-luciferase reporter assays, Fyn-related kinase (FRK) was identified as a target gene of miR-151a-3p. Knockdown of FRK promoted NP-induced EMT in CRC cells and activated the WNT/β-catenin signaling pathway, while overexpression had the opposite effect. In summary, the present study demonstrated that NP could inhibit FRK expression via miR-151a-3p, activate the WNT/β-catenin signaling pathway, and promote EMT in CRC cells.
Collapse
Affiliation(s)
- Biao Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Nianjie Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Lin Dai
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Yuanwei Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Shuo Yin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Xuefeng Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563006, Guizhou, People's Republic of China.
| |
Collapse
|
19
|
Zhang Z, Zhao S, Lv X, Gao Y, Guo Q, Ren Y, He Y, Jin Y, Yang H, Liu S, Zhang X. CRAT downregulation promotes ovarian cancer progression by facilitating mitochondrial metabolism through decreasing the acetylation of PGC-1α. Cell Death Discov 2025; 11:15. [PMID: 39828731 PMCID: PMC11743791 DOI: 10.1038/s41420-025-02294-2] [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: 06/06/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Mitochondrial dysfunctions are closely associated with different types of disease, including cancer. Carnitine acetyltransferase (CRAT) is a mitochondrial-localized enzyme catalyzing the reversible transfer of acyl groups from an acyl-CoA thioester to carnitine and regulates the ratio of acyl-CoA/CoA. Our bioinformatics analysis using public database revealed a significant decrease of CRAT expression in ovarian cancer (OC). However, the functions of CRAT have rarely been investigated in human cancers, especially in OC. Here, we found a frequent down-regulation of CRAT in OC, which is mainly caused by up-regulation of miR-132-5p. Downregulation of CRAT was significantly associated with shorter survival time for patients with OC. Forced expression of CRAT suppressed OC growth and metastasis by inducing cell cycle arrest and epithelial to mesenchymal transition (EMT). By contrast, CRAT knockdown promoted OC growth and metastasis. Mechanistically, we found that CRAT downregulation promoted OC growth and metastasis by increasing mitochondrial biogenesis to facilitate mitochondrial metabolism through reducing the acetylation of peroxisome proliferator-activated receptor-γ coactivator (PGC-1α). In summary, CRAT functions as a critical tumor suppressor in OC progression by enhancing PGC-1α-mediated mitochondrial biogenesis and metabolism, suggesting CRAT as a potential therapeutic target in treatment of OC.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of stomatology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Shuhua Zhao
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xiaohui Lv
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yan Gao
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Qian Guo
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yanjie Ren
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yuanyuan He
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yihua Jin
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Hong Yang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Shujuan Liu
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Xiaohong Zhang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China.
| |
Collapse
|
20
|
Wen P, Jiang D, Qu F, Wang G, Zhang N, Shao Q, Huang Y, Li S, Wang L, Zeng X. PFDN5 plays a dual role in breast cancer and regulates tumor immune microenvironment: Insights from integrated bioinformatics analysis and experimental validation. Gene 2025; 933:149000. [PMID: 39396557 DOI: 10.1016/j.gene.2024.149000] [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: 05/08/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Although the prognosis for patients with breast cancer has improved, breast cancer remains the leading cause of death for women worldwide. Prefoldin 5 (PFDN5), as a subunit of the prefoldin complex, plays a vital role in aiding the correct folding of newly synthesized proteins. However, the exact impact of PFDN5 on breast cancer development and its prognostic implications remain unclear. METHODS We conducted bioinformatics analysis to investigate the correlation between PFDN5 and patient survival, as well as various clinicopathological characteristics in breast cancer. Additionally, various assays were employed to validate the biological functions of PFDN5 in breast cancer. Finally, RNA sequencing (RNA-seq) was utilized to investigate the molecular mechanisms associated with PFDN5. RESULTS Compared to normal tissues, PFDN5 exhibited lower expression levels in breast cancer tissues, and lower expression of PFDN5 is associated with poorer prognosis. PFDN5 led to G2/M phase arrest in the cell cycle and reduced proliferative potential in breast cancer cells. However, PFDN5 also promoted migration and invasion of breast cancer cells. Also, RNA-seq analysis revealed an involvement of PFDN5 in the cell cycle and TGF-β signaling pathway. Furthermore, PFDN5 had a significant impact on tumor immune microenvironment by promoting macrophage polarization towards the M1 phenotype and exhibited a positive correlation with CD8+ T cell infiltration levels. CONCLUSIONS PFDN5 plays a dual role in breast cancer and serves as a key factor in tumor immune microenvironment. Therefore, PFDN5 holds promise as a valuable biomarker for predicting both metastatic and prognosis in breast cancer.
Collapse
Affiliation(s)
- Ping Wen
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Dongping Jiang
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Fanli Qu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Guanwen Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Ningning Zhang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Qing Shao
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yuxin Huang
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Sisi Li
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Long Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China.
| | - Xiaohua Zeng
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China; Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing 400030, China.
| |
Collapse
|
21
|
Jiang L, Wang J, Liu Z, Zhang Q, Yang XL. Seryl-tRNA synthetase inhibits Wnt signaling and breast cancer progression and metastasis. FASEB J 2025; 39:e70294. [PMID: 39760229 PMCID: PMC11817322 DOI: 10.1096/fj.202401720r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/06/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Tumors require ample protein synthesis to grow, and aminoacyl-tRNA synthetases, as critical translation factors, are expected to support cancer progression. Unexpectedly, overexpression of seryl-tRNA synthetase (SerRS) suppresses primary tumor growth of breast cancer. However, the effects of SerRS on metastasis have not been studied. We observe a decrease in SerRS expression in breast cancer patient metastases compared with matched primary tumors, suggesting an inhibitory role of SerRS in metastasis. Through mouse metastasis models using breast cancer cell lines overexpressing SerRS, we show that SerRS impedes not only primary tumor growth but also establishment of metastases, and the effect of SerRS on metastasis can be independent of its impact on the primary tumor. SerRS also inhibits tumor growth with induced, post-tumor-onset overexpression, demonstrating its potential as an anticancer therapeutic. Tumor RNA-seq analysis identified Wnt signaling among the top SerRS-regulated pathways. Using cell-based studies, we confirm SerRS suppresses Wnt signaling and metastatic processes in breast cancer cells. To the best of our knowledge, this is the first study to show a component of the translation machinery can act as both a tumor and metastasis suppressor.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Justin Wang
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ze Liu
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Qian Zhang
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Xiang-Lei Yang
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
22
|
Ju M, Jin Z, Yu X, Huang C, Li Y, Gao Z, Li H, Huang H, Zheng C, Jia S, Zhang Y, Liu X, Zhou H, Zhang X, Li K. Gastric Cancer Models Developed via GelMA 3D Bioprinting Accurately Mimic Cancer Hallmarks, Tumor Microenvironment Features, and Drug Responses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2409321. [PMID: 39811968 DOI: 10.1002/smll.202409321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Current in vitro models for gastric cancer research, such as 2D cell cultures and organoid systems, often fail to replicate the complex extracellular matrix (ECM) found in vivo. For the first time, this study utilizes a gelatin methacryloyl (GelMA) hydrogel, a biomimetic ECM-like material, in 3D bioprinting to construct a physiologically relevant gastric cancer model. GelMA's tunable mechanical properties allow for the precise manipulation of cellular behavior within physiological ranges. Genetic and phenotypic analyses indicate that the 3D bioprinted GelMA (3Db) model accurately mimics the clinical tumor characteristics and reproduces key cancer hallmarks, such as cell proliferation, invasion, migration, angiogenesis, and the Warburg effect. Comparisons of gene expression and drug responses between the 3Db model and patient-derived xenograft models, both constructed from primary gastric cancer cells, validate the model's clinical relevance. The ability of the 3Db model to closely simulate in vivo conditions highlights its crucial role in identifying treatment targets and predicting patient-specific responses, showcasing its potential in high-throughput drug screening and clinical applications. This study is the first to report the pivotal role of GelMA-based 3D bioprinting in advancing gastric cancer research and regenerative medicine.
Collapse
Affiliation(s)
- Mingguang Ju
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Zhizhong Jin
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Caihao Huang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Yanshu Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, 110122, China
| | - Ziming Gao
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - He Li
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Haibo Huang
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Chen Zheng
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Shiheng Jia
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yixiao Zhang
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaofang Liu
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Heng Zhou
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xing Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Kai Li
- Department of Surgical Oncology and General Surgery Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
23
|
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
|
24
|
Wang B, Cheng H, Ji Z, Jiang Z, Wang R, Ding Y, Ni J. Synergistic Target-Attacking Tumor Cells and M2 Macrophages via a Triple-Responsive Nanoassembly for Complete Metastasis Blocking. Adv Healthc Mater 2025; 14:e2304096. [PMID: 39663738 DOI: 10.1002/adhm.202304096] [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: 11/21/2023] [Revised: 08/24/2024] [Indexed: 12/13/2024]
Abstract
Collaboration of cancerous cells and microenvironment is the root for tumor spreading, leading to difficulty in complete metastasis blockage via mono-intervention. Herein, a triple-responsive nanoassembly is designed for orienting tumor cells and migration-driving M2 tumor associated macrophages (TAMs) in microenvironment for efficient anti-metastatic therapy. Structurally, a reactive oxygen species (ROS)-responsive crosslinked short-chain polyquaternium is synthesized to bridge graphene oxide (GO) scaffold with apolipoprotein A-I crown via borate-crosslinking, electrostatic adherence, and coordinative coupling. The protein-crowning polymeric GO nanoparticles could give multimodal shielding and triple-responsive release of doxorubicin and Snail-targeted siRNA. Tailor-made apolipoprotein A-I crown fulfills nanoparticles synergistically attacking tumor cells and M2 TAMs via binding with overexpressed scavenger receptors. The findings witness the targeted accumulation and potent cytotoxicity of the hybrid nanoparticles for M2 TAMs and tumor cells; especially, elimination of M2 TAMs in tumor microenvironment holds back Snail-enhancing transforming growth factor (TGF)-β signal pathway, which collaborates with Snail silencing in tumor cells to reverse epithelial mesenchymal transition (EMT) and metastasis-promoting niche. Collectively, the synergistic targeting therapeutic platform could provide a promising solution for metastatic tumor treatment.
Collapse
Affiliation(s)
- Bei Wang
- Institute of Integration of Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| | - Hao Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhongsheng Ji
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zijun Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Rong Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiang Ni
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| |
Collapse
|
25
|
Ren X, Liu X, Zhang Q, Yang C, Xu Z. Simultaneous imaging of telomerase activity and protein tyrosine kinase 7 in living cells during epithelial-mesenchymal transformation via a near-infrared light-activatable nanoprobe. Talanta 2025; 282:126993. [PMID: 39383724 DOI: 10.1016/j.talanta.2024.126993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Exploring the relationship between key regulation molecules (such as telomerase and protein tyrosine kinase 7) during epithelial-mesenchymal transformation of cells is beneficial for studying malignant tumor metastasis. Fluorescence is usually used for real-time monitoring the distribution and expression of regulatory molecules in living cells. However, the recognition function of these classical nanoprobes is "always active" due to the absence of exogenous control, which leads to the amplification of both the background signal and the response signal, making it difficult to distinguish changes in biomolecule expression levels. To improve the fluorescence ratio between tumor and normal cells, we constructed near-infrared light-activatable nanoprobes by engineering the functional units of catalytic hairpin assembly and integrating upconversion luminescence nanoparticles. Under near-infrared light irradiation, the nanoparticles, serving as a near-infrared-to-ultraviolet light transducer, induced the photolysis of the photo-cleavable linkers sealed in hairpins. The recognition function of the nanoprobes can be controlled by near-infrared light, preventing them from recognizing the targets in non-irradiated regions. By employing the nanoprobes, we realized simultaneous imaging of two regulatory molecules in living cells and observed an increase in telomerase activity and a decrease in protein tyrosine kinase 7 expression during drug-induced epithelial-mesenchymal transformation. This work provides a promising method for revealing changes and relationships of regulatory molecules during tumor metastasis.
Collapse
Affiliation(s)
- Xiuyan Ren
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Xiaopeng Liu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Qi Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Chunguang Yang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China.
| |
Collapse
|
26
|
Guo L, Zhao Y, Bai X, Wang X, Tuoheti K, Cao Y, Zuo Y, Zhang X, Liu T. RRM2 Is a Putative Biomarker and Promotes Bladder Cancer Progression via PI3K/AKT/mTOR Pathway. J Cell Physiol 2025; 240:e31501. [PMID: 39676643 DOI: 10.1002/jcp.31501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/24/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Bladder cancer (BLCA) is the tenth most common cancer worldwide, characterized by its high recurrence and progression rates. Thus, identifying prognostic biomarkers and understanding its underlying mechanisms are imperative to enhance patient outcomes. In this study, we aimed to investigate the prognostic significance, expression, functional activity, and underlying mechanisms of RRM2 in BLCA. RRM2 expression and its association with pathological grading and survival were investigated in samples from TCGA dataset and BLCA tissue microarray. CCK8 assays, colony formation assays, wound healing, and transwell assays were performed to assess the role of RRM2 in BLCA cell proliferation and migration. Western blot was employed to investigate alterations in markers associated with epithelial-to-mesenchymal transition (EMT), apoptosis, and cell cycle regulation. Gene set enrichment analysis was performed to investigate the biological processes associated with RRM2, which were subsequently validated. The expression of RRM2 was significantly elevated in both BLCA tissues and cells. Our results also indicated a positive correlation between RRM2 expression and high tumor stage, high tumor grade, and poor survival. Knockdown of RRM2 inhibited cell proliferation and migration of BLCA. RRM2 knockdown significantly induced apoptosis and arrested the cell cycle at the G0/G1 phase. Opposite results were observed in the RRM2 overexpression cells. Additionally, our study demonstrates that RRM2 promotes BLCA progression by activating the PI3K/AKT/mTOR pathway. RRM2 is remarkably correlated with poor prognosis in BLCA and facilitate its progression via PI3K/AKT/mTOR pathway. It is suggested that RRM2 might become an effective prognostic biomarker and potential therapeutic target for BLCA.
Collapse
Affiliation(s)
- Linfa Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yiqiao Zhao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaojie Bai
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaolong Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kuerban Tuoheti
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuanfei Cao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingtong Zuo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xinhua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tongzu Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
27
|
Gong H, Yang X, An L, Zhang W, Liu X, Shu L, Yang L. PCSK5 downregulation promotes the inhibitory effect of andrographolide on glioblastoma through regulating STAT3. Mol Cell Biochem 2025; 480:521-533. [PMID: 38553549 DOI: 10.1007/s11010-024-04977-3] [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: 01/07/2024] [Accepted: 02/24/2024] [Indexed: 01/03/2025]
Abstract
Proprotein convertase subtilisin/kexin type 5 (PCSK5) is a member of the proprotein convertase (PC) family, which processes immature proteins into functional proteins and plays an important role in the process of cell migration and transformation. Andrographolide is a non-peptide compound with PC inhibition and antitumor activity. Our research aimed to investigate the functional role of PCSK5 downregulation combined with Andro on GBM progression. Results from the cancer genome atlas (TCGA) and clinical samples revealed a significant upregulation of PCSK5 in GBM tissues than in non-tumor brain tissues. Higher expression of PCSK5 was correlated with advanced GBM stages and worse patient prognosis. PCSK5 knockdown attenuated the epithelial-mesenchymal transition (EMT)-like properties of GBM cells induced by IL-6. PCSK5 knockdown in combination with Andro treatment significantly inhibited the proliferation and invasion of GBM cells in vitro, as well as tumor growth in vivo. Mechanistically, PCSK5 downregulation reduced the expression of p-STAT3 and Matrix metalloproteinases (MMPs), which could be rescued by the p-STAT3 agonist. STAT3 silencing downregulated the expression of MMPs without affecting PCSK5. Furthermore, Andro in combination with PCSK5 silencing significantly inhibited STAT3/MMPs axis. These observations provided evidence that PCSK5 functioned as a potential tumor promoter by regulating p-STAT3/MMPs and the combination of Andro with PCSK5 silencing might be a good strategy to prevent GBM progression.
Collapse
Affiliation(s)
- Huiyuan Gong
- Department of Immunology, Basic Medical College, Guizhou Medical University, No.6, Ankang Road, Guian New District, Guiyang, 550004, Guizhou, People's Republic of China
| | - Xiaomin Yang
- Department of Immunology, Basic Medical College, Guizhou Medical University, No.6, Ankang Road, Guian New District, Guiyang, 550004, Guizhou, People's Republic of China
| | - Lijun An
- Department of Immunology, Basic Medical College, Guizhou Medical University, No.6, Ankang Road, Guian New District, Guiyang, 550004, Guizhou, People's Republic of China
| | - Wangming Zhang
- Department of Immunology, Basic Medical College, Guizhou Medical University, No.6, Ankang Road, Guian New District, Guiyang, 550004, Guizhou, People's Republic of China
| | - Xiaohua Liu
- Department of Immunology, Basic Medical College, Guizhou Medical University, No.6, Ankang Road, Guian New District, Guiyang, 550004, Guizhou, People's Republic of China
| | - Liping Shu
- Department of Immunology, Basic Medical College, Guizhou Medical University, No.6, Ankang Road, Guian New District, Guiyang, 550004, Guizhou, People's Republic of China
| | - Liuqi Yang
- Department of Immunology, Basic Medical College, Guizhou Medical University, No.6, Ankang Road, Guian New District, Guiyang, 550004, Guizhou, People's Republic of China.
| |
Collapse
|
28
|
Xu Y, Xue G, Zhou L, Wu G, Hu L, Ma S, Zhang J, Li X. KIF4A promotes epithelial-mesenchymal transition by activating the TGF-β/SMAD signaling pathway in glioma cells. Mol Cell Biochem 2025; 480:217-230. [PMID: 38411896 DOI: 10.1007/s11010-024-04943-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: 09/09/2023] [Accepted: 01/14/2024] [Indexed: 02/28/2024]
Abstract
Gliomas are the most prevalent type of primary brain tumor, with poor prognosis reported in patients with high-grade glioma. Kinesin family member 4 A (KIF4A) stimulates the proliferation, migration, and invasion of tumor cells. However, its function in gliomas has not been clearly established. Therefore, this study aimed to investigate the effects of KIF4A on the epithelial-mesenchymal transition and invasion of glioma cells. We searched The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases to identify KIF4A-related signaling pathways and downstream genes. We further validated them using western blotting, transwell migration and invasion, wound-healing scratch, and dual-luciferase reporter assays in U251 and U87 human glioblastoma cells. Our analysis of the Cancer Genome Atlas and Chinese Glioma Genome Atlas data showed elevated KIF4A expression in patients with gliomas and was associated with clinical grade. Here, KIF4A overexpression promoted the migration, invasion, and proliferation of glioma cells, whereas KIF4A knockdown showed contrasting results. Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) analyses demonstrated that KIF4A positively controls TGF-β/SMAD signaling in glioma cells. Additionally, genetic correlation analysis revealed that KIF4A transcriptionally controls benzimidazoles-1 expression in glioma cells. KIF4A promotes the epithelial-mesenchymal transition by regulating the TGF-β/SMAD signaling pathway via benzimidazoles-1 in glioma cells.
Collapse
Affiliation(s)
- Yao Xu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guangren Xue
- Department of Neurosurgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Lei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gaotian Wu
- Laboratory of Cancer Molecular Genetics, Soochow University, Medical College of Soochow University, Suzhou, China
| | - Lingji Hu
- Laboratory of Cancer Molecular Genetics, Soochow University, Medical College of Soochow University, Suzhou, China
| | - Shuchen Ma
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Xiangdong Li
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
29
|
Li Q, Zhang H, He Y, Zhang H, Han C. Inhibition of Colorectal Cancer Metastasis by Total Flavones of Abelmoschus manihot via LncRNA AL137782-mediated STAT3/EMT Pathway Regulation. Curr Pharm Des 2025; 31:219-232. [PMID: 39289944 DOI: 10.2174/0113816128298998240828060306] [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: 05/27/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) ranks among the most lethal malignancies globally, particularly following metastasis which results in poor prognosis. In recent years, CRC incidence in China has persistently increased. Total flavonoids (TFA) from Abelmoschus manihot, a natural compound, are recognized for their anti-inflammatory, analgesic, and antioxidant properties. However, despite extensive research into the therapeutic potential of TFA, coverage of its role in cancer treatment is notably lacking. To address this research void, our study aims to unveil the role and potential mechanisms of TFA in treating CRC. METHODS We conducted a series of experiments to assess the impact of TFA on CRC cells. Two specific CRC cell lines, DLD-1 and HCT116, were employed in cell proliferation, colony formation, flow cytometry, and cell migration assays. Additionally, to test the in vivo effects of TFA, we developed a nude mouse xenograft tumor model to assess TFA's impact on tumor growth and liver metastasis. Furthermore, we meticulously analyzed the gene expression differences between CRC cells pretreated with TGF-β and those treated with TFA using RNA-seq technology. We also examined the molecular mechanisms of TFA and assessed the expression of proteins related to the STAT3/EMT signaling pathway through Western blotting and siRNA technology. RESULTS Our research findings reveal for the first time the effect of TFA on CRC cells. Result shows that TFA could suppress cell proliferation, migration, and induce apoptosis. In vivo results showed that TFA inhibited tumor growth and liver metastasis. Molecular mechanism studies have shown that TFA exerts these effects by upregulating the expression of non-coding RNA AL137782, inhibiting the EMT/STAT3 signaling pathway. These results suggest that TFA is a potential candidate for mitigating CRC metastasis. CONCLUSION However, further research is needed to comprehensively evaluate the efficacy and safety of TFA in animal models and clinical settings. These findings bring great hope for the development of innovative CRC treatment methods.
Collapse
MESH Headings
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/antagonists & inhibitors
- STAT3 Transcription Factor/genetics
- Humans
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/metabolism
- Abelmoschus/chemistry
- Animals
- Mice
- Flavones/pharmacology
- Flavones/chemistry
- Flavones/isolation & purification
- Cell Proliferation/drug effects
- Mice, Nude
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Mice, Inbred BALB C
- Cell Movement/drug effects
- Drug Screening Assays, Antitumor
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/chemistry
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Qian Li
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, PR China
- Department of Proctology, Xinyi Hospital of Traditional Chinese Medicine, Xinyi, Jiangsu 221400, PR China
| | - Hui Zhang
- Department of Ultrasound, Xu Zhou Hospital of Traditional Chinese Medicine, Zhongshan South Road 169, Yunlong District, Xu Zhou, Jiangsu 221000, PR China
| | - Yongshan He
- Department of Colorectal Surgery, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Kongjiang Road 1665, Yangpu District, Shanghai 200092, PR China
| | - Hao Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Daxue Road 4655, Changqing District, Jinan, Shandong, PR China
| | - Conghui Han
- Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, 221000, PR China
| |
Collapse
|
30
|
Famta P, Shah S, Dey B, Kumar KC, Bagasariya D, Vambhurkar G, Pandey G, Sharma A, Srinivasarao DA, Kumar R, Guru SK, Raghuvanshi RS, Srivastava S. Despicable role of epithelial-mesenchymal transition in breast cancer metastasis: Exhibiting de novo restorative regimens. CANCER PATHOGENESIS AND THERAPY 2025; 3:30-47. [PMID: 39872366 PMCID: PMC11764040 DOI: 10.1016/j.cpt.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/03/2024] [Accepted: 01/10/2024] [Indexed: 01/30/2025]
Abstract
Breast cancer (BC) is the most prevalent cancer in women globally. Anti-cancer advancements have enabled the killing of BC cells through various therapies; however, cancer relapse is still a major limitation and decreases patient survival and quality of life. Epithelial-to-mesenchymal transition (EMT) is responsible for tumor relapse in several cancers. This highly regulated event causes phenotypic, genetic, and epigenetic changes in the tumor microenvironment (TME). This review summarizes the recent advancements regarding EMT using de-differentiation and partial EMT theories. We extensively review the mechanistic pathways, TME components, and various anti-cancer adjuvant and neo-adjuvant therapies responsible for triggering EMT in BC tumors. Information regarding essential clinical studies and trials is also discussed. Furthermore, we also highlight the recent strategies targeting various EMT pathways. This review provides a holistic picture of BC biology, molecular pathways, and recent advances in therapeutic strategies.
Collapse
Affiliation(s)
- Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Biswajit Dey
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Kondasingh Charan Kumar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Deepkumar Bagasariya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Dadi A. Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | | | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
31
|
Chen P, Liang K, Mao X, Wu Q, Chen Z, Jin Y, Lin K, He T, Yang S, Huang H, Ye G, Gao J, Zhou D, Zeng Z. Single-cell transcriptomes of dissecting the intra-tumoral heterogeneity of breast cancer microenvironment. J Cancer Res Clin Oncol 2024; 151:17. [PMID: 39724260 PMCID: PMC11671554 DOI: 10.1007/s00432-024-06015-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 10/23/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE To investigate the mechanism by which heterogeneity in breast cancer developed and acted in single-cell transcriptomes. METHODS The composition of breast cancer based on the single-cell transcriptomes of 54,055 high-quality cells from clinical specimens of 4 malignant and 4 non-malignant patients were investigated. RESULTS We identified six common expression programs and six subtype-specific expression programs form malignant epithelial cells. The expression program of malignant epithelial cells exhibited activated EMT (Epithelial Mesenchymal Transition) in TME, which might indicate EMT intervention have a general therapeutic effect on various subtypes. Gene set enrichment analysis (GSEA) based on the top 50 highly NMF (non-negative matrix factorization) score genes in each program depicted the distinct function of each program in breast cancer progression. Moreover, we revealed the profound cellular heterogeneity of myeloid cell lineages in breast cancer. In macrophages, two mainly tumor associated macrophages (TAMs), TAM1 and TAM2, were also detected and the highly variable genes in TAM2 were strongly enriched in IFN-α and IFN-γ. The changes of lipid metabolism pathways in macrophages are closely related to the microenvironment of breast cancer. CONCLUSION We constructed a comprehensive single-cell transcriptome atlas of 54,055 cells from 4 malignant and 4 nonmalignant patients, providing insights into the mechanisms underlying breast cancer progression and the development of potential therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Peixian Chen
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, Guangdong Province, China
| | - Kaifeng Liang
- Department of Breast Surgery, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), #1, Jiazi Road, Lunjiao, Shunde District, Foshan, 528308, Guangdong Province, China
| | - Xiaofan Mao
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Qiuyuan Wu
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, Guangdong Province, China
- Guangdong Medical University, Zhanjiang, 524000, China
| | - Zhiyan Chen
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, Guangdong Province, China
- Guangdong Medical University, Zhanjiang, 524000, China
| | - Yabin Jin
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Kairong Lin
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Tiancheng He
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, Guangdong Province, China
| | - Shuqing Yang
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, Guangdong Province, China
| | - Huiqi Huang
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, Guangdong Province, China
| | - Guolin Ye
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, Guangdong Province, China
| | - Juntao Gao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
| | - Dan Zhou
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, Guangdong Province, China
- Guangdong Medical University, Zhanjiang, 524000, China
| | - Zhihao Zeng
- Department of Breast Surgery, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), #1, Jiazi Road, Lunjiao, Shunde District, Foshan, 528308, Guangdong Province, China.
| |
Collapse
|
32
|
Sun D, Song Y, Gao W, Lin B, Wang B, Yang X, Li S, Jin Y, Zhang J. DNA-templated nanosheets for enhanced chemodynamic therapy and gene therapy to inhibit tumor recurrence and metastasis. Int J Pharm 2024; 667:124910. [PMID: 39500474 DOI: 10.1016/j.ijpharm.2024.124910] [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: 06/23/2024] [Revised: 10/13/2024] [Accepted: 11/01/2024] [Indexed: 11/10/2024]
Abstract
Recurrence and metastasis stand as the primary contributors to mortality among patients with triple-negative breast cancer post-surgery, presenting a formidable clinical obstacle. Chemodynamic therapy (CDT), leveraging metal-ion-mediated Fenton-like reactions within the tumor microenvironment (TME), emerges as a promising avenue for addressing cancer metastasis. Despite recent progress, challenges such as tumor cell antioxidant defenses and epithelial-mesenchymal transition (EMT) impede the efficacy of CDT. Here, we introduce a novel approach using DNA-templated nanosheets (Dz-MnO2) that combine the functions of Mn2+-mediated CDT and DNAzyme-mediated gene therapy to suppress tumor growth and metastasis. The Dz-MnO2 nanosheets respond effectively to the TME, releasing Mn2+ and DNAzyme. The DNAzyme exhibits mRNA cleavage activity, specifically targeting oncogenic transcripts to reduce tumor progression. Mn2+ not only facilitates a Fenton-like reaction, enhancing the chemodynamic treatment effect, but also serves as a cofactor for DNAzyme, improving its catalytic efficiency. Concurrently, the nanosheets robustly silence the Twist1 gene, mitigating the EMT process and reinforcing CDT efficacy by suppressing apoptosis resistance. Results indicate that Dz-MnO2 nanosheets efficiently polarize M2-tumor-associated macrophages (TAMs) into M1-TAMs by locally mitigating tumor hypoxia via catalyzing the decomposition of H2O2 into O2. This collaborative strategy presents a promising approach to enhance CDT, effectively inhibiting tumor recurrence and metastasis.
Collapse
Affiliation(s)
- Danna Sun
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Yuwei Song
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China
| | - Wenyan Gao
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China
| | - Boyang Lin
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China
| | - Bei Wang
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China
| | - Xinjian Yang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Shaochun Li
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China
| | - Yi Jin
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China.
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China.
| |
Collapse
|
33
|
Yan Z, Liu Y, Yuan Y. The plasticity of epithelial cells and its potential in the induced differentiation of gastric cancer. Cell Death Discov 2024; 10:512. [PMID: 39719478 DOI: 10.1038/s41420-024-02275-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Cell plasticity refers to the deviation of cells from normal terminal differentiation states when faced with environmental and genetic toxic stresses, resulting in the phenomenon of transforming into other cell or tissue phenotypes. Unlocking phenotype plasticity has been defined as a hallmark of malignant tumors. The stomach is one of the organs in the body with the highest degree of self-renewal and exhibits significant cell plasticity. In this paper, based on the review of the characteristics of normal differentiation of gastric epithelial cells and their markers, the four main phenotypes of gastric epithelial cell remodeling and their relationship with gastric cancer (GC) are drawn. Furthermore, we summarize the regulatory factors and mechanisms that affect gastric epithelial cell plasticity and outline the current status of research and future prospection for the treatment targeting gastric epithelial cell plasticity. This study has important theoretical reference value for the in-depth exploration of epithelial cell plasticity and the tumor heterogeneity caused by it, as well as for the precise treatment of GC.
Collapse
Affiliation(s)
- Ziwei Yan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yingnan Liu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
34
|
Chen L, Chen N, Xie Z, Xiao Y, Jiang H. Prognostic and immunological role of LASP2 in clear cell renal cell carcinoma. Genes Genomics 2024:10.1007/s13258-024-01612-9. [PMID: 39714590 DOI: 10.1007/s13258-024-01612-9] [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: 09/03/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) represents a common renal carcinoma subtype influenced by the immune microenvironment. LIM and SH3 Protein 2 (LASP2), an actin-binding protein within the nebulin family, contributes to cellular immunity and adhesion mechanisms. OBJECTIVE This study aimed to clarify the immunological and prognostic relevance of LASP2 in ccRCC. METHODS Using clinical and expression data from TCGA, LASP2 expression levels were analyzed alongside clinicopathological features in ccRCC patients. Validation was conducted through real-world samples and tissue microarrays. Comprehensive analysis using online databases examined genetic mutations, DNA methylation patterns, and immune microenvironment characteristics. Gene set enrichment analysis (GSEA) provided insights into LASP2's potential mechanisms in ccRCC. RESULTS LASP2 expression was notably reduced and correlated with adverse clinicopathological features and prognosis in ccRCC patients. Prognostic associations were identified across multiple CpG DNA methylation sites. LASP2 levels showed significant correlations with immune cell infiltration and checkpoint genes, including PDCD1 and CTLA4. GSEA findings highlighted LASP2's enrichment within metabolic pathways and signaling networks, such as fatty acid metabolism, TGF-β signaling, and epithelial-mesenchymal transition. CONCLUSION LASP2 emerged as an immune-associated biomarker linked to poorer survival outcomes in ccRCC, suggesting its potential as a novel anti-cancer target and prognostic indicator in ccRCC.
Collapse
Affiliation(s)
- Libo Chen
- Department of Urology, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, No. 63, Huang Tang Road, Meizhou, 514031, Guangdong Province, People's Republic of China
| | - Nanhui Chen
- Department of Urology, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, No. 63, Huang Tang Road, Meizhou, 514031, Guangdong Province, People's Republic of China
| | - Zhouzhou Xie
- Meizhou Clinical Institute of Shantou University Medical College, Meizhou, People's Republic of China
| | - Yuchen Xiao
- Shantou University Medical College, Shantou, People's Republic of China
| | - Huiming Jiang
- Department of Urology, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, No. 63, Huang Tang Road, Meizhou, 514031, Guangdong Province, People's Republic of China.
| |
Collapse
|
35
|
He J, Zhang HP. Research progress and treatment status of malignant ascites. Front Oncol 2024; 14:1390426. [PMID: 39737405 PMCID: PMC11682990 DOI: 10.3389/fonc.2024.1390426] [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: 02/23/2024] [Accepted: 11/20/2024] [Indexed: 01/01/2025] Open
Abstract
Malignant ascites (MA), a common and serious complication of various cancers in the abdominal cavity, originates from the extensive infiltration, metastasis, and growth of cancer cells in or on the abdominal cavity, leading to abnormal accumulation of fluid in the abdominal cavity and the formation of MA. MA seriously reduces the quality of life of cancer patients, shortens their survival period, and generally has a poor prognosis. Modern medicine has developed various strategies for the treatment of MA, including targeted supportive treatment, diuretic treatment, abdominal paracentesis, surgical intervention, and intraperitoneal administration therapy. Among them, chemotherapy, as one of the important treatment methods, includes both systemic chemotherapy and intraperitoneal chemotherapy, especially pressurized intraperitoneal aerosol chemotherapy (PIPAC), hyperthermic intraperitoneal chemotherapy (HIPEC), and foam-based intraperitoneal chemotherapy (FBIC), providing a new choice for the treatment of MA. In addition, innovative treatment methods such as gas-based intra-abdominal hyperthermia (GIH) combined with dehydration therapy have also shown promising application prospects. This article delves into multiple aspects of MA, including its concept, mechanism of occurrence, clinical manifestations, differential diagnostic methods, and current treatment status and research progress. This comprehensive review aims to provide valuable references for effectively controlling MA, improving cancer patients' quality of life, and prolonging the survival cycle of cancer patients in clinical practice. Malignant ascites (MA) is a common complication of cancer, which originates from the extensive infiltration, metastasis, and growth of cancer cells in the abdominal cavity or peritoneum, leading to abnormal accumulation of peritoneal fluid. It is a common clinical manifestation in the late stage of cancer. Its symptoms are stubborn and recurrent, which can lead to abdominal pain, bloating, poor appetite, fatigue, breathing difficulties, and even multiple organ failure. The median survival time for cancer patients with MA is generally 5 to 6 months. The prognosis is poor, and it is imperative to seek more active and effective treatment plans. This article reviews the research and treatment status of MA, aiming to provide certain value for controlling MA and improving the quality of life of patients.
Collapse
Affiliation(s)
- Jing He
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Dali University, Dali, China
| | - Hui-ping Zhang
- Department of Oncology, Guang’anmen Hospital Jinan Hospital (Jinan Hospital of Traditional Chinese Medicine), Jinan, China
| |
Collapse
|
36
|
Zhou H, Xiao J, Cheng Q, Wang W, Peng H, Lin X, Chen J, Wang X. Metformin inhibits migration and epithelial-to-mesenchymal transition in non-small cell lung cancer cells through AMPK-mediated GDF15 induction. Eur J Pharmacol 2024; 985:177127. [PMID: 39528101 DOI: 10.1016/j.ejphar.2024.177127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
The growth differentiation factor 15 (GDF15) may serve as a biomarker of metformin, which mediates the bodyweight lowering effect of metformin. However, whether GDF15 also serves as a molecular target of metformin to inhibit carcinogenesis remains largely unknown. This study examined the role and molecular mechanisms of GDF15 in the anticancer effects of metformin in non-small cell lung cancer (NSCLC) cells, which has never been reported before. We found that metformin significantly inhibited the migration of NSCLC A549 and NCI-H460 cells and reduced the expression of epithelial-to-mesenchymal transition (EMT)-related molecules, including neuro-cadherin (N-cadherin), matrix metalloproteinase 2 (MMP2), and the zinc finger transcription factor Snail, but increased epithelial cadherin (E-cadherin) expression. Furthermore, metformin increased GDF15 and its upstream transcription factors activated transcription factor 4 (ATF4) and C/EBP-homologous protein (CHOP) expressions and increased AMP-activated protein kinase (AMPK) phosphorylation in NSCLC cells. GDF15 siRNA partially reverses the inhibitory effect of metformin on NSCLC cell migration. Moreover, metformin-induced increases in GDF15, CHOP, and ATF4 expression and the inhibition of migration were partially reversed by treatment with Compound C, a specific AMPK inhibitor. Meanwhile, metformin significantly inhibited NCI-H460 xenograft tumor growth in nude mice, increased GDF15 expression, and regulated EMT- and migration-related protein expression in xenograft tumors. In conclusion, our results provide novel insights into revealing that GDF15 can serve as a potential molecular target of metformin owing to its anti-cancer effect in NSCLC, which is mediated by AMPK activation.
Collapse
Affiliation(s)
- Hongyu Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Jun Xiao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Qi Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Wen Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China.
| |
Collapse
|
37
|
Guo Y, Miao S, Jin Y, Li Q, Wang Y, Zhang X, Li J. Tumor-associated macrophages contribute to cholangiocarcinoma progression and chemoresistance through activation of ID1. Ann Hepatol 2024; 30:101773. [PMID: 39674368 DOI: 10.1016/j.aohep.2024.101773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/25/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
INTRODUCTION AND OBJECTIVES Tumor-associated macrophages (TAM) can influence both cancer growth and chemoresistance, but the specific mechanisms involved in these processes in cholangiocarcinoma (CCA) are unclear. MATERIALS AND METHODS We explored the distribution of TAM in CCA samples by multiplex immunofluorescence staining and tested the effects of TAM on CCA in vitro and in vivo. We then investigated the mechanisms underlying these effects using the Luminex assay, RNA sequencing, western blotting, flow cytometry, and co-immunoprecipitation. RESULTS The infiltration of TAM was strongly increased in the cholangiocarcinoma tumor microenvironment. Oncostain M (OSM) secreted by TAM increased the proliferation and chemotherapeutic resistance of CCA cells both in vitro and in vivo. The results of transcriptome sequencing analysis, Western blot analysis, and immunofluorescence staining confirmed that OSM can promote Yap nuclear translocation and its subsequent formation of complexes with SMADs to upregulate the expression of inhibitor of DNA binding 1 (ID1). CONCLUSIONS TAM promotes CCA progression and chemoresistance through activating OSM-Yap-ID1.
Collapse
Affiliation(s)
- Yinghao Guo
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Shuangda Miao
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Yun Jin
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Qi Li
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Yihang Wang
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Xiaoxiao Zhang
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Jiangtao Li
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China.
| |
Collapse
|
38
|
Li D, Bao L, Liu S, Ji K, Xu X, Yuan J, Xia G. Identification and Validation of Molecular Features of the Anoikis Gene-Related Hub Genes in Nasopharyngeal Carcinoma. Appl Biochem Biotechnol 2024:10.1007/s12010-024-05130-y. [PMID: 39666232 DOI: 10.1007/s12010-024-05130-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 12/13/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor originating from nasopharyngeal mucosa. Anoikis, a form of programmed cell death induced by detachment from the extracellular matrix, normally prevents metastasis. Resistance to anoikis in cancer cells can enhance their metastatic potential. This study identifies anoikis-related genes (ARGs) associated with NPC to elucidate tumorigenesis mechanisms. Analysis of the GSE12452 dataset from GEO revealed 77 differentially expressed ARGs in NPC tissues. GO and KEGG analyses highlighted significant enrichment in apoptosis-related pathways. A PPI network identified MYC, FN1, BRCA1, and FGF2 as Hub genes. Correlation analysis showed MYC positively correlated with activated dendritic cells (p < 0.01) but negatively with naive CD4 T cells (p < 0.001). FN1 was positively correlated with activated dendritic cells (p < 0.01) and negatively with M1 macrophages (p < 0.05). FGF2 negatively correlated with naive CD4 T cells (p < 0.001), while BRCA1 was positively correlated with eosinophils (p < 0.01). GSVA and GSEA indicated that MYC, FN1, BRCA1, and FGF2 were significantly enriched in cell cycle and DNA replication pathways. Immunohistochemistry and qPCR of 50 NPC samples confirmed the overexpression of these genes. Knockdown of MYC, FN1, BRCA1, and FGF2 led to increased tumor cell malignancy, with statistical significance (p < 0.05). This study identifies MYC, FN1, BRCA1, and FGF2 as anoikis-related genes (ARGs) with significant regulatory roles in nasopharyngeal carcinoma (NPC). These ARGs are found to be involved in the development and progression of NPC, suggesting their potential as therapeutic targets for this cancer.
Collapse
Affiliation(s)
- Dong Li
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Lihao Bao
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Shaosheng Liu
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Ke Ji
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Xujiu Xu
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Jie Yuan
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Guihua Xia
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China.
| |
Collapse
|
39
|
Zhang F, Cui X, Yang K, Guo R, Zhu L, Zhao W, Liu Z, Liu B. Activin A inhibits the migration of human lung adenocarcinoma A549 cells induced by EGF. Int Immunopharmacol 2024; 142:113170. [PMID: 39288626 DOI: 10.1016/j.intimp.2024.113170] [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: 03/11/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Activin A, a member of the transforming growth factor β (TGF-β) superfamily, is involved in tumorigenesis and tumor progression. However, it remains unclear whether activin A can affect the migration of lung adenocarcinoma (LUAD) cells. In this study, the results of differentially expressed genes (DEGs) identification revealed that lung adenocarcinoma tissues exhibited lower expression of activin βA mRNA, but higher expression of epidermal growth factor (EGF) and MMP9 mRNA compared to nontumor tissues. Moreover, we found that activin A inhibited human LUAD A549 cell proliferation promoted by EGF. Additionally, EGF induced A549 cell migration in microfluidic device, while activin A attenuated EGF actions. Simultaneously, EGF increased the levels of migration-related proteins, but activin A played the opposite role. Furthermore, the study revealed that EGF upregulated the ratio of p-ERK/ERK in A549 cells, which was weakened by activin A, and A549 cell migration regulated by activin A was not related to calcium signaling. In addition, the inhibitory effect of activin A on EGF-induced A549 cell migration was attenuated by the ERK inhibitor FR180204. These findings demonstrate that activin A effectively hinders the migration of A549 cells induced by EGF through ERK1/2 signaling, suggesting that targeting activin A may hold promise in the treatment of EGF-dependent LUAD growth and metastasis.
Collapse
Affiliation(s)
- Fenglin Zhang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China
| | - Xueling Cui
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China; Key Laboratory of Neuroimmunology and Clinical Immunology in Jilin Province, Jilin Province 130021, China
| | - Ke Yang
- Institute of Applied Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui Province 230031, China
| | - Rui Guo
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China
| | - Linjing Zhu
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China
| | - Wei Zhao
- Key Laboratory of Neuroimmunology and Clinical Immunology in Jilin Province, Jilin Province 130021, China; Department of Internal Medicine, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Zhonghui Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China; Key Laboratory of Neuroimmunology and Clinical Immunology in Jilin Province, Jilin Province 130021, China
| | - Boyang Liu
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China; Department of Scientific Research, Jilin Jianzhu University, Changchun, Jilin Province 130118, China.
| |
Collapse
|
40
|
Lin L, Xiao X, Guo X, Zhong C, Zhuang M, Xu J, Wang Y, Chen F. AKR1C3 mediates gastric cancer cell invasion and metastasis via the AKT and JNK/p-NF-κB signaling pathways. Sci Rep 2024; 14:30263. [PMID: 39632995 PMCID: PMC11618362 DOI: 10.1038/s41598-024-82039-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: 04/19/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Gastric cancer (GC) is globally recognized as the fifth most common cancer and the third leading cause of cancer-related mortality. Early metastasis in GC significantly contributes to its high mortality and unfavorable prognosis. However, the underlying mechanisms of this phenomenon remain largely unexplored. Among the various factors involved, AKR1C3 has emerged as a crucial component in the pathways of tumorigenesis and metastasis across multiple cancer types. Yet, the precise significance of AKR1C3 in GC patients' prognosis and its role in GC progression remain elusive. This study illuminated the significant downregulation of AKR1C3 in GC tissues, linking it to an aggressive phenotype and poor prognosis. Interestingly, while AKR1C3 overexpression did not affect the proliferation of GC cells, it significantly inhibited their ability to invade and metastasize. The underlying mechanism appears to involve AKR1C3's inhibition of the p-JNK pathway, which leads to reduced phosphorylation of IKKα/β and IKBα, lowering p-NF-κB levels and hindering its movement into the nucleus, thereby stifling the epithelial-mesenchymal transition (EMT) process in GC cells. These insights reveal AKR1C3's tumor-suppressive effects in GC and suggest its potential as a diagnostic and prognostic biomarker, offering new avenues for targeted therapies in gastric cancer management.
Collapse
Affiliation(s)
- Liying Lin
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Xinzhu Xiao
- Department of Infectious disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
- Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Xiaoxiong Guo
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Canmei Zhong
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Mingkai Zhuang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Jie Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Yin Wang
- Department of Gastroenterology, Tongan Ward of the First Affiliated Hospital of Xiamen University, Xiamen, 361026, Fujian, China
| | - Fenglin Chen
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
41
|
Feng Q, Shen Z, Wang F, Shi C. Mediation of circ_0007142 on miR-128-3p/S100A14 pathway to stimulate the progression of cervical cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9919-9933. [PMID: 38951152 DOI: 10.1007/s00210-024-03250-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/27/2023] [Indexed: 07/03/2024]
Abstract
A previous study has confirmed the upregulation of circ_0007142 expression in CC. Here, we aimed to investigate the effect and mechanism of circ_0007142 in CC progression. The expression of circ_0007142, microRNA-128-3p (miR-128-3p), S100 calcium-binding protein A14 (S100A14), and epithelial mesenchymal transition (EMT)-related markers was measured by qRT-PCR and Western blot. Cell proliferative, migratory, and invasion abilities were evaluated using cell counting Kit-8, cell colony formation, 5-ethynyl-2'-deoxyuridine, and transwell assays, respectively. The interaction among circ_0007142, miR-128-3p and S100A14 was identified by dual-luciferase reporter and RNA immunoprecipitation assays. In vivo experiment was implemented to investigate the effect of circ_0007142 on tumor growth. CC tissues and cells displayed high expression of circ_0007142 and S100A14, and low expression of miR-128-3p in comparison to the controls. Knockdown of circ_0007142 resulted in the inhibition of cell proliferation, migration invasion, and EMT in vitro. In support, circ_0007142 deficiency hindered tumor growth and EMT in vivo. In rescue experiments, downregulation of miR-128-3p relieved circ_0007142 absence-mediated anticancer impacts. MiR-128-3p overexpression-induced inhibitory effects on cell growth and metastasis were attenuated by S100A14 overexpression. Importantly, circ_0007142 regulated S100A14 expression by sponging miR-128-3p. Circ_0007142 knockdown suppressed CC cell malignant behaviors by miR-128-3p/S100A14 pathway, providing a possible circRNA-targeted therapy for CC.
Collapse
Affiliation(s)
- Qinqin Feng
- Department of Obstetrics, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, No. 80, Guilin South Road, Xialu District, Huangshi, 435000, China
| | - Zhangzhou Shen
- Medical School, Hubei Polytechnic University, Huangshi, 435003, China
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Huangshi, 435003, China
| | - Fen Wang
- Department of Obstetrics, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, No. 80, Guilin South Road, Xialu District, Huangshi, 435000, China
| | - Cheng Shi
- Department of Obstetrics, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, No. 80, Guilin South Road, Xialu District, Huangshi, 435000, China.
| |
Collapse
|
42
|
Li Y, Tang X, Wang B, Chen M, Zheng J, Chang K. Current landscape of exosomal non-coding RNAs in prostate cancer: Modulators and biomarkers. Noncoding RNA Res 2024; 9:1351-1362. [PMID: 39247145 PMCID: PMC11380467 DOI: 10.1016/j.ncrna.2024.07.003] [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: 02/29/2024] [Revised: 06/12/2024] [Accepted: 07/18/2024] [Indexed: 09/10/2024] Open
Abstract
Prostate cancer (PCa) has the highest frequency of diagnosis among solid tumors and ranks second as the primary cause of cancer-related deaths. Non-coding RNAs (ncRNAs), such as microRNAs, long non-coding RNAs and circular RNAs, frequently exhibit dysregulation and substantially impact the biological behavior of PCa. Compared with circulating ncRNAs, ncRNAs loaded into exosomes are more stable because of protection by the lipid bilayer. Furthermore, exosomal ncRNAs facilitate the intercellular transfer of molecules and information. Increasing evidence suggests that exosomal ncRNAs hold promising potential in the progression, diagnosis and prognosis of PCa. This review aims to discuss the functions of exosomal ncRNAs in PCa, evaluate their possible applications as clinical biomarkers and therapeutic targets, and provide a comprehensive overview of the ncRNAs regulatory network in PCa. We also identified ncRNAs that can be utilized as biomarkers for diagnosis, staging, grading and prognosis assessment in PCa. This review offers researchers a fresh perspective on the functions of exosomal ncRNAs in PCa and provides additional options for its diagnosis, progression monitoring, and prognostic prediction.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, PR China
- School of Medicine, Chongqing University, Chongqing, 400030, PR China
| | - Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Binpan Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, PR China
- School of Medicine, Chongqing University, Chongqing, 400030, PR China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| |
Collapse
|
43
|
Liu Z, Ding X, Zhang B, Pang Y, Wang Y, Xu D, Wang H. Endosulfan promotes cell growth, migration and invasion via CCL5/CCR5 axis in MCF-7 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117344. [PMID: 39549571 DOI: 10.1016/j.ecoenv.2024.117344] [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: 06/14/2024] [Revised: 11/03/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
Endosulfan, recognized as an endocrine disruptor, has emerged as an important risk factor for human breast cancer. The chemokine ligand 5 (CCL5) and its receptor CCR5 constitute a biological axis, that is implicated in tumorigenesis and cancer progression. However, the role of the CCL5/CCR5 axis in breast cancer when exposure to endosulfan remains unclear. The present study aimed to determine the significance of the CCL5/CCR5 axis in the carcinogenic effects of endosulfan in human breast cancer MCF-7 cells. The results showed that endosulfan significantly promoted cell proliferation, increased the rate of colony formation, and enhanced cell migration ability in a dose-dependent manner by activating the PI3K/AKT signaling pathway, which were rescued by the specific inhibitor (LY-294002) for PI3K/AKT signaling pathway. We utilized Cytoscape software to construct protein-protein interaction (PPI) network when exposure to endosulfan, and identified 47 highly connected genes in the network diagram centered on CCL5. Endosulfan significantly increased the secretion of CCL5 and the expression levels of CCL5/CCR5, which were reversed by CCR5 inhibitor (HY-13004). HY-13004 significantly counteracted the effects of endosulfan on colony formation, cell migration and the activation of PI3K/AKT signaling pathway. Endosulfan markedly altered the expression levels of epithelial-mesenchymal transition (EMT) biomarkers and enhanced transwell migration and invasion capabilities of MCF-7 cells, which were inhibited by HY-13004, similar to the effects observed with LY-294002. Collectively, our findings suggest that endosulfan activates the PI3K/AKT signaling pathway to promote cell growth, and induces EMT, thereby enhancing cell migration and invasion via the CCL5/CCR5 axis in MCF-7 cells.
Collapse
Affiliation(s)
- Zeming Liu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China
| | - Xiaolin Ding
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China
| | - Boxiang Zhang
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China
| | - Yue Pang
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China
| | - Yuhui Wang
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China
| | - Dan Xu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China.
| | - Hailong Wang
- Department of Clinical Epidemiology and Evidence-based Medicine, First Hospital of China Medical University, Shenyang 110016, PR China.
| |
Collapse
|
44
|
Chatterjee P, Ghosh D, Chowdhury SR, Roy SS. ETS1 drives EGF-induced glycolytic shift and metastasis of epithelial ovarian cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119805. [PMID: 39159682 DOI: 10.1016/j.bbamcr.2024.119805] [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: 01/23/2024] [Revised: 06/10/2024] [Accepted: 07/20/2024] [Indexed: 08/21/2024]
Abstract
Epithelial ovarian cancer (EOC), a leading cause of gynecological cancer-related morbidity and mortality and the most common type of ovarian cancer (OC), is widely characterized by alterations in the Epidermal Growth Factor (EGF) signaling pathways. The phenomenon of metastasis is largely held accountable for the majority of EOC-associated deaths. Existing literature reports substantiate evidence on the indispensable role of metabolic reprogramming, particularly the phenomenon of the 'Warburg effect' or aerobic glycolysis in priming the cancer cells towards Epithelial to Mesenchymal transition (EMT), subsequently facilitating EMT. Considering the diverse roles of growth factor signaling across different stages of oncogenesis, our prime emphasis was laid on unraveling mechanistic details of EGF-induced 'Warburg effect' and resultant metastasis in EOC cells. Our study puts forth Ets1, an established oncoprotein and key player in OC progression, as the prime metabolic sensor to EGF-induced cues from the tumor microenvironment (TME). EGF treatment has been found to induce Ets1 expression in OC cells predominantly through the Extracellular Signal-Regulated Kinase1/2 (ERK1/2) pathway activation. This subsequently results in pronounced glycolysis, characterized by an enhanced lactate production through transcriptional up-regulation of key determinant genes of the central carbon metabolism namely, hexokinase 2 (HK2) and monocarboxylate transporter 4 (MCT4). Furthermore, this study reports an unforeseen combinatorial blockage of HK2 and MCT4 as an effective approach to mitigate cellular metastasis in OC. Collectively, our work proposes a novel mechanistic insight into EGF-induced glycolytic bias in OC cells and also sheds light on an effective therapeutic intervention approach exploiting these insights.
Collapse
Affiliation(s)
- Priti Chatterjee
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Deepshikha Ghosh
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | | | - Sib Sankar Roy
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
45
|
Du T, Dong X, Tan J, Chen X, Liu J, Wen T, Ru X. Targeting Tn Antigen Suppresses Aberrant O-Glycosylation-Elicited Metastasis in Breast Cancer. J Cell Mol Med 2024; 28:e70279. [PMID: 39654023 PMCID: PMC11628356 DOI: 10.1111/jcmm.70279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024] Open
Abstract
The Tn antigen, a truncated O-glycan representing aberrant mucin-type O-glycosylation, is frequently observed in human breast cancer. However, the functional role of Tn antigen in breast cancer metastasis remains insufficiently investigated. This study aimed to elucidate the expression profile of Tn antigen in breast cancer and its potential as a therapeutic target for inhibiting metastasis. Immunohistochemical staining was performed to determine the levels of Tn antigen expression in breast cancer tissues and its clinical relevance was analyzed accordingly. Tn-positive breast cancer cell lines were generated through disruption of the Cosmc gene. The functional roles of Tn antigen in breast cancer metastasis were studied in both in vitro and in vivo models. Western blotting and immunofluorescence staining were employed to investigate the molecular mechanisms by which Tn antigen promotes breast cancer metastasis. Our findings revealed that Tn antigen was prevalent in breast carcinomas, particularly within metastatic lesions. Tn antigen expression was positively correlated with lymph node metastasis and poorer patient survival. Tn antigen-expressing breast cancer cells exhibited enhanced invasiveness and metastasis, along with significant activation of EMT and FAK signaling pathways. Targeting Tn-positive cells with HPA (Helix pomatia agglutinin) demonstrated the suppression of invasive and metastatic capabilities, EMT program, and FAK signaling in vitro, as well as reduced pulmonary metastasis in a xenotransplant mouse model. This study reveals that Tn antigen-mediated aberrant O-glycosylation plays a contributing role in breast cancer metastasis, which may serve as a potential therapeutic target in clinical practice.
Collapse
Affiliation(s)
- Tan Du
- Medical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Xichen Dong
- Medical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Jingyu Tan
- Medical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Xiangyu Chen
- Medical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Jian Liu
- Medical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Tao Wen
- Medical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Xiaoli Ru
- Department of Gynecology and Obstetrics, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
46
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
47
|
Zheng L, Rajamanickam V, Wang M, Zhang H, Fang S, Linnebacher M, Abd El-Aty AM, Zhang X, Zhang Y, Wang J, Chen M, Zhao Z, Ji J. Fangchinoline inhibits metastasis and reduces inflammation-induced epithelial-mesenchymal transition by targeting the FOXM1-ADAM17 axis in hepatocellular carcinoma. Cell Signal 2024; 124:111467. [PMID: 39393566 DOI: 10.1016/j.cellsig.2024.111467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/10/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide. Efforts have been focused on developing new anti-HCC agents and understanding their pharmacology. However, few agents have been able to effectively combat tumor growth and invasiveness due to the rapid progression of HCC. In this study, we discovered that fangchinoline (FAN), a bisbenzylisoquinoline alkaloid derived from Stephania tetrandra S. Moore, effectively inhibited the migration, invasion, and epithelial-mesenchymal transition (EMT) of HCC cells. FAN treatment also led to the suppression of IL6 and IL1β release, as well as the expression of inflammation-related proteins such as COX-2 and iNOS, and the activation of the NF-κB pathway, thereby reducing inflammation-related EMT. Additionally, FAN directly bound to forkhead box protein M1 (FOXM1), resulting in decreased levels of FOXM1 proteins and disruption of the FOXM1-ADAM17 axis. Our in vivo findings confirmed that FAN effectively hindered the growth and lung metastasis of HCCLM3-xenograft tumors. Importantly, the upregulation of FOXM1 in HCC tissue suggested that targeting FOXM1 inhibition with FAN or its inhibitors could be a promising therapeutic approach for HCC. Overall, this study elucidated the anti-tumor effects and potential pharmacological mechanisms of FAN, and proposed that targeting FOXM1 inhibition may be an effective therapeutic strategy for HCC with potential clinical applications.
Collapse
Affiliation(s)
- Liyun Zheng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Shenzhen University General Hospital-Lishui Hospital Joint Research Center, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Vinothkumar Rajamanickam
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Mengyuan Wang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Huajun Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Shenzhen University General Hospital-Lishui Hospital Joint Research Center, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, Rostock 18059, Germany
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
| | - Xinbin Zhang
- Shenzhen University General Hospital-Lishui Hospital Joint Research Center, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Yeyu Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jianbo Wang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Shenzhen University General Hospital-Lishui Hospital Joint Research Center, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Shenzhen University General Hospital-Lishui Hospital Joint Research Center, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China.
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Shenzhen University General Hospital-Lishui Hospital Joint Research Center, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| |
Collapse
|
48
|
Xie A, Wang H, Zhao J, Wang Z, Xu J, Xu Y. scPAS: single-cell phenotype-associated subpopulation identifier. Brief Bioinform 2024; 26:bbae655. [PMID: 39681325 DOI: 10.1093/bib/bbae655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/13/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Despite significant advancements in single-cell sequencing analysis for characterizing tissue sample heterogeneity, identifying the associations between cell subpopulations and disease phenotypes remains a challenging task. Here, we introduce scPAS, a new bioinformatics tool designed to integrate bulk data to identify phenotype-associated cell subpopulations within single-cell data. scPAS employs a network-regularized sparse regression model to quantify the association between each cell in single-cell data and a phenotype. Additionally, it estimates the significance of these associations through a permutation test, thereby identifying phenotype-associated cell subpopulations. Utilizing simulated data and various single-cell datasets from breast carcinoma, ovarian cancer, and atherosclerosis, as well as spatial transcriptomics data from multiple cancers, we demonstrated the accuracy, flexibility, and broad applicability of scPAS. Evaluations on large datasets revealed that scPAS exhibits superior operational efficiency compared to other methods. The open-source scPAS R package is available at GitHub website: https://github.com/aiminXie/scPAS.
Collapse
Affiliation(s)
- Aimin Xie
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 157 Baojian Road, Heilongjiang 150081, China
| | - Hao Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 157 Baojian Road, Heilongjiang 150081, China
| | - Jiaxu Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 157 Baojian Road, Heilongjiang 150081, China
| | - Zhaoyang Wang
- Genetron Health (Beijing) Co. Ltd, 1-2/F, Building 11, Zone 1, 8 Life Science Parkway, Changping District, Beijing 102208, China
| | - Jinyuan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 157 Baojian Road, Heilongjiang 150081, China
| | - Yan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 157 Baojian Road, Heilongjiang 150081, China
| |
Collapse
|
49
|
Ning X, Du N, Zhang X, Wang S, Zhi Y, Li Z, Ren Z, Ku T, Li G, Sang N. Metastatic effects of hydroxy-polycyclic aromatic hydrocarbons on liver cancer cells mediated by estrogen receptor α. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 952:175878. [PMID: 39222821 DOI: 10.1016/j.scitotenv.2024.175878] [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: 06/21/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Hydroxy-polycyclic aromatic hydrocarbons (OH-PAHs) are a growing worldwide concern because of their persistence, ubiquity, and toxicity. Nonetheless, research on the toxicological mechanisms of OH-PAHs remains sparse, particularly concerning the risk of liver cancer. This study evaluated the effects of OH-PAHs on disrupting estrogen receptor α (ERα) and subsequently facilitating hepatocellular invasion and metastasis. Results revealed that all six OH-PAHs exhibited ERα agonistic activities at noncytotoxic levels, which were partially validated using molecular docking (MD) and molecular dynamics simulations (MDS). Furthermore, OH-PAHs with ERα agonistic properties stimulated a concentration-dependent increase in the migration and invasion of HepG2 cells. In addition, they disturbed the expression of target genes associated with epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM), and the invasion effects were significantly reversed by adding an ERα antagonist. Our results suggest an essential role of ERα in the metastasis of liver cancer cells induced by OH-PAHs and emphasize their potential ecological and health hazards.
Collapse
Affiliation(s)
- Xia Ning
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Du
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Xiaofeng Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Shuo Wang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Yan Zhi
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Zhaoli Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Zhihua Ren
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| |
Collapse
|
50
|
Zhang Y, Zhang Y, Gong R, Liu X, Zhang Y, Sun L, Ma Q, Wang J, Lei K, Ren L, Zhao C, Zheng X, Xu J, Ren H. Label-Free Prediction of Tumor Metastatic Potential via Ramanome. SMALL METHODS 2024:e2400861. [PMID: 39558758 DOI: 10.1002/smtd.202400861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/02/2024] [Indexed: 11/20/2024]
Abstract
Assessing metastatic potential is crucial for cancer treatment strategies. However, current methods are time-consuming, labor-intensive, and have limited sample accessibility. Therefore, this study aims to investigate the urgent need for rapid and accurate approaches by proposing a Ramanome-based metastasis index (RMI) using machine learning of single-cell Raman spectra to rapidly and accurately assess tumor cell metastatic potential. Validation with various cultured tumor cells and a mouse orthotopic model of pancreatic ductal adenocarcinoma show a Kendall rank correlation coefficient of 1 compared to Transwell experiments and histopathological assessments. Significantly, lipid-related Raman peaks are most influential in determining RMI. The lipidomic analysis confirmed strong correlations between metastatic potential and phosphatidylcholine, phosphatidylethanolamine, cholesteryl ester, ceramide, and bis(monoacylglycero)phosphate, crucial in cell membrane composition or signal transduction. Therefore, RMI is a valuable tool for predicting tumor metastatic potential and providing insights into metastasis mechanisms.
Collapse
Affiliation(s)
- Yuxing Zhang
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yanmei Zhang
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Ruining Gong
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Xiaolan Liu
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yu Zhang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Luyang Sun
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Qingyue Ma
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Jia Wang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Ke Lei
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Linlin Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Chenyang Zhao
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Xiaoshan Zheng
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Jian Xu
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - He Ren
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| |
Collapse
|