1
|
Zuo CJ, Tian J. Advancing the understanding of the role of apoptosis in lung cancer immunotherapy: Global research trends, key themes, and emerging frontiers. Hum Vaccin Immunother 2025; 21:2488074. [PMID: 40186454 PMCID: PMC11980473 DOI: 10.1080/21645515.2025.2488074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/12/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025] Open
Abstract
Apoptosis is vital for improving the efficacy of lung cancer (LC) immunotherapy by targeting cancer cell elimination. Despite its importance, there is a lack of comprehensive bibliometric studies analyzing global research on apoptosis in LC immunotherapy. This analysis aims to address this gap by highlighting key trends, contributors, and future directions. A total of 969 publications from 1996 to 2024 were extracted from the Web of Science Core Collection. Analysis was conducted using VOSviewer, CiteSpace, and the R package 'bibliometrix.' The study included contributions from 6,894 researchers across 1,469 institutions in 61 countries, with research published in 356 journals. The volume of publications has steadily increased, led by China and the United States, with Sichuan University as the top contributor. The journal Cancers published the most articles, while Cancer Research had the highest co-citations. Yu-Quan Wei was the leading author, and Jemal, A. was the most frequently co-cited. Key research themes include "cell death mechanisms," "immune regulation," "combination therapies," "gene and nanomedicine applications," and "traditional Chinese medicine (TCM)." Future research is likely to focus on "coordinated regulation of multiple cell death pathways," "modulation of the tumor immune microenvironment," "optimization of combination therapies," "novel strategies in gene regulation," and the "integration of TCM" for personalized treatment. This is the first bibliometric analysis on the role of apoptosis in LC immunotherapy, providing an landscape of global research patterns and emerging therapeutic strategies. The findings offer insights to guide future research and optimize treatment approaches.
Collapse
Affiliation(s)
- Chun-Jian Zuo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Wang H, Cheng G, Zhang S, Qu H, Zhao X, Yang A, Sun X, Pan H. Sevoflurane: A dual modulator of miR‑211‑5p and mitochondrial apoptosis in glioma therapy. Mol Med Rep 2025; 32:179. [PMID: 40280112 PMCID: PMC12046963 DOI: 10.3892/mmr.2025.13544] [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: 05/14/2024] [Accepted: 01/07/2025] [Indexed: 04/29/2025] Open
Abstract
The present study aimed to investigate how sevoflurane (SEV) regulated the apoptosis of glioma cells through the mitochondrial apoptosis pathway. First, an evaluation was performed on the viability, apoptosis, mitochondrial reactive oxygen species levels, mitochondrial membrane potential and apoptosis and autophagy‑related protein expression of glioma cells according to experimental groups. Next, the expression of microRNA‑211‑5p (miR‑211‑5p), silent information regulator 1 (SIRT1) and phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (AKT) signaling pathway was detected by reverse transcription‑quantitative PCR or western blotting. Dual luciferase reporter gene assay confirmed the targeting relationship between miR‑211‑5p and SIRT1. In addition, SEV suppressed the proliferation and induced the apoptosis in human glioma cell line cells via the mitochondrial apoptosis pathway. In mechanistic analysis, the miR‑211‑5p level in glioma cells was low, while following SEV treatment, it was increased. Furthermore, SEV regulated SIRT1 by upregulating miR‑211‑5p expression, thereby blocking the PI3K/AKT signaling pathway activation. Moreover, functional rescue experiments showed that downregulation of SIRT1 or miR‑211‑5p could reverse the effects of SEV on glioma cells. Collectively, SEV promoted apoptosis in glioma cells by inducing miR‑211‑5p, which regulated SIRT1/PI3K/AKT pathway, mediating mitochondria‑dependent apoptosis pathway. This finding may open new possibilities for SEV as a potential treatment for glioma in the future.
Collapse
Affiliation(s)
- Haili Wang
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Guofang Cheng
- Department of Orthopedics, Sanmenxia Orthopedic Hospital, Sanmenxia, Henan 472000, P.R. China
| | - Shuyuan Zhang
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Haibo Qu
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Xibo Zhao
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Ailing Yang
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Xuejia Sun
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Hua Pan
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| |
Collapse
|
3
|
Li CX, Xu Q, Jiang ST, Liu D, Tang C, Yang WL. Anticancer effects of salvianolic acid A through multiple signaling pathways (Review). Mol Med Rep 2025; 32:176. [PMID: 40280109 PMCID: PMC12056544 DOI: 10.3892/mmr.2025.13541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Salvia miltiorrhiza Bunge (Salvia miltiorrhiza), commonly referred to as Danshen, is a well‑known herb in traditional Chinese medicine, the active ingredients of which are mostly categorized as water soluble and lipid soluble. Salvianolic acids are the major water‑soluble phenolic acid constituents of Danshen; salvianolic acid B is the most prevalent, with salvianolic acid A (SAA) being the next most predominant form. SAA offers a wide array of pharmacological benefits, including cardiovascular protection, and anti‑inflammatory, antioxidant, antiviral and anticancer activities. SAA is currently undergoing phase III clinical trials for diabetic peripheral neuropathy and has shown protective benefits against cardiovascular illnesses; furthermore, its safety and effectiveness are encouraging. By targeting several signaling pathways, preventing cell cycle progression, tumor cell migration, invasion and metastasis, normalizing the tumor vasculature and encouraging cell apoptosis, SAA can also prevent the growth of malignancies. In addition, it enhances sensitivity to chemotherapeutic drugs, and alleviates their toxicity and side effects. However, the broad therapeutic use of SAA has been somewhat limited by its low content in Salvia miltiorrhiza Bunge and the difficulty of its extraction techniques. Therefore, the present review focuses on the potential mechanisms of SAA in tumor prevention and treatment. With the anticipation that SAA will serve a notable role in clinical applications in the future, these discoveries may offer a scientific basis for the combination of SAA with conventional chemotherapeutic drugs in the treatment of cancer, and could establish a foundation for the development of SAA as an anticancer drug.
Collapse
Affiliation(s)
- Cheng-Xia Li
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qi Xu
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Shi-Ting Jiang
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Dan Liu
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chao Tang
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wen-Li Yang
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
4
|
Peng X, Feng J, Yang H, Xia P, Pu F. Nrf2: A key regulator in chemoradiotherapy resistance of osteosarcoma. Genes Dis 2025; 12:101335. [PMID: 40242036 PMCID: PMC12000747 DOI: 10.1016/j.gendis.2024.101335] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/24/2024] [Accepted: 04/03/2024] [Indexed: 04/18/2025] Open
Abstract
Osteosarcoma (OS), frequently observed in children and adolescents, is one of the most common primary malignant tumors of the bone known to be associated with a high capacity for invasion and metastasis. The incidence of osteosarcoma in children and adolescents is growing annually, although improvements in survival remain limited. With the clinical application of neoadjuvant chemotherapy, chemotherapy combined with limb-preserving surgery has gained momentum as a major intervention. However, certain patients with OS experience treatment failure owing to chemoradiotherapy resistance or metastasis. Nuclear factor E2-related factor 2 (Nrf2), a key antioxidant factor in organisms, plays a crucial role in maintaining cellular physiological homeostasis; however, its overactivation in cancer cells restricts reactive oxygen species production, promotes DNA repair and drug efflux, and ultimately leads to chemoradiotherapy resistance. Recent studies have also identified the functions of Nrf2 beyond its antioxidative function, including the promotion of proliferation, metastasis, and regulation of metabolism. The current review describes the multiple mechanisms of chemoradiotherapy resistance in OS and the substantial role of Nrf2 in the signaling regulatory network to elucidate the function of Nrf2 in promoting OS chemoradiotherapy resistance and formulating relevant therapeutic strategies.
Collapse
Affiliation(s)
- Xianglin Peng
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| | - Jing Feng
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| | - Han Yang
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Ping Xia
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan 430030, China
| | - Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| |
Collapse
|
5
|
Wang Z, Su X, Zhan Z, Wang H, Zhou S, Mao J, Xu H, Duan S. miR-660: A novel regulator in human cancer pathogenesis and therapeutic implications. Gene 2025; 953:149434. [PMID: 40120868 DOI: 10.1016/j.gene.2025.149434] [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: 09/29/2024] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
MicroRNAs (miRNAs) are non-coding RNAs that regulate gene expression. Among these, miR-660, located on chromosome Xp11.23, is increasingly studied for its role in cancer due to its abnormal expression in various biological contexts. It is regulated by 8 competing endogenous RNAs (ceRNAs), which adds complexity to its function. miR- 660 targets 19 genes involved in 6 pathways such as PI3K/AKT/mTOR, STAT3, Wnt/β-catenin, p53, NF‑κB, and RAS, influencing cell cycle, proliferation, apoptosis, and invasion/migration. It also plays a role in resistance to chemotherapies like cisplatin, gemcitabine, and sorafenib in lung adenocarcinoma (LUAD), pancreatic ductal adenocarcinoma (PDAC), and hepatocellular carcinoma (HCC), thus highlighting its clinical importance. Additionally, leveraging liposomes as nanocarriers presents a promising avenue for enhancing cancer drug delivery. Our comprehensive study not only elucidates the aberrant expression patterns, biological functions, and regulatory networks of miR-660 and its ceRNAs but also delves into the intricate signaling pathways implicated. We envisage that our findings will furnish a robust framework and serve as a seminal reference for future investigations of miR-660, fostering advancements in cancer research and potentially catalyzing breakthroughs in cancer diagnosis and treatment paradigms.
Collapse
Affiliation(s)
- Zehua Wang
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xinming Su
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhiqing Zhan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hangxuan Wang
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shuhan Zhou
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Jiasheng Mao
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hening Xu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shiwei Duan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Kahkesh S, Hedayati N, Rahimzadeh P, Farahani N, Khoozani MF, Abedi M, Nabavi N, Naeimi B, Khoshnazar SM, Alimohammadi M, Alaei E, Mahmoodieh B. The function of circular RNAs in regulating Wnt/β-catenin signaling: An innovative therapeutic strategy for breast and gynecological cancers. Pathol Res Pract 2025; 270:155944. [PMID: 40228402 DOI: 10.1016/j.prp.2025.155944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025]
Abstract
Breast cancer (BC) and gynecological malignancies, including cervical, ovarian, and uterine cancers, are significant global health challenges due to their high prevalence, complex nature, and elevated mortality rates. Dysregulation of the Wnt/β-catenin signaling pathway is a common feature in gynecological malignancies, contributing to cancer cell growth, progression, migration, and metastasis. Recent studies have highlighted the pivotal role of non-coding RNAs (ncRNAs), particularly circular RNAs (circRNAs), in modulating the Wnt/β-catenin signaling pathway. Acting as sponges for microRNAs (miRNAs), circRNAs regulate key oncogenic and tumor-suppressive processes by influencing Wnt-related components. This research explores the role of circRNAs in breast and gynecological malignancies, focusing on their regulatory effects on the Wnt/β-catenin pathway. The findings reveal that circRNAs modulate critical cellular processes such as proliferation, apoptosis, autophagy, and metastasis, with potential implications for therapeutic interventions. Targeting circRNA-mediated dysregulation of Wnt signaling could offer novel strategies for improving diagnostic precision, treatment efficacy, and survival outcomes in breast and gynecological cancers.
Collapse
Affiliation(s)
- Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdi Farhadi Khoozani
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Bita Naeimi
- Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
7
|
Navin AK, Rejani CT, Chandrasekaran B, Tyagi A. Urolithins: Emerging natural compound targeting castration-resistant prostate cancer (CRPC). Biomed Pharmacother 2025; 187:118058. [PMID: 40253830 DOI: 10.1016/j.biopha.2025.118058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025] Open
Abstract
Castration-resistant prostate cancer (CRPC) presents a significant challenge due to its resistance to conventional androgen deprivation therapies. Urolithins, bioactive metabolites derived from ellagitannins, have recently emerged as promising therapeutic agents for CRPC. Urolithins not only inhibit androgen receptor (AR) signaling, a crucial factor in the progression of CRPC, but also play a key role in regulating oxidative stress by their antioxidant properties, thereby inhibiting increased reactive oxygen species, a common feature of the aggressive nature of CRPC. Research has shown that urolithins induce apoptosis and diminish pro-survival signaling, leading to tumor inhibition. This review delves into the intricate mechanisms through which urolithins exert their therapeutic effects, focusing on both AR-dependent and AR-independent pathways. It also explores the exciting potential of combining urolithins with androgen ablation therapy, opening new avenues for CRPC treatment.
Collapse
Affiliation(s)
- Ajit Kumar Navin
- Department of Pharmacology, College of Pharmacy, Texas A&M University, College Station, TX 77845, USA
| | | | - Balaji Chandrasekaran
- Department of Pharmacology, College of Pharmacy, Texas A&M University, College Station, TX 77845, USA
| | - Ashish Tyagi
- Department of Pharmacology, College of Pharmacy, Texas A&M University, College Station, TX 77845, USA.
| |
Collapse
|
8
|
Yang MH, Basappa B, Deveshegowda SN, Ravish A, Mohan A, Nagaraja O, Madegowda M, Rangappa KS, Deivasigamani A, Pandey V, Lobie PE, Hui KM, Sethi G, Ahn KS. A novel drug prejudice scaffold-imidazopyridine-conjugate can promote cell death in a colorectal cancer model by binding to β-catenin and suppressing the Wnt signaling pathway. J Adv Res 2025; 72:615-632. [PMID: 39067696 DOI: 10.1016/j.jare.2024.07.022] [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: 03/28/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
INTRODUCTION Globally, colorectal cancer (CRC) is the third most common type of cancer, and its treatment frequently includes the utilization of drugs based on antibodies and small molecules. The development of CRC has been linked to various signaling pathways, with the Wnt/β-catenin pathway identified as a key target for intervention. OBJECTIVES We have explored the impact of imidazopyridine-tethered chalcone-C (CHL-C) in CRC models. METHODS To determine the influence of CHL-C on apoptosis and autophagy, Western blot analysis, annexin V assay, cell cycle analysis, acridine orange staining, and immunocytochemistry were performed. Next, the activation of the Wnt/β-catenin signaling pathway and the anti-cancer effects of CHL-C in vivo were examined in an orthotopic HCT-116 mouse model. RESULTS We describe the synthesis and biological assessment of the CHL series as inhibitors of the viability of HCT-116, SW480, HT-29, HCT-15, and SNU-C2A CRC cell lines. Further biological evaluations showed that CHL-C induced apoptosis and autophagy in down-regulated β-catenin, Wnt3a, FZD-1, Axin-1, and p-GSK-3β (Ser9), and up-regulated p-GSK3β (Tyr216) and β-TrCP. In-depth analysis using structure-based bioinformatics showed that CHL-C strongly binds to β-catenin, with a binding affinity comparable to that of ICG-001, a well-known β-catenin inhibitor. Additionally, our in vivo research showed that CHL-C markedly inhibited tumor growth and triggered the activation of both apoptosis and autophagy in tumor tissues. CONCLUSION CHL-C is capable of inducing apoptosis and autophagy by influencing the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Min Hee Yang
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Suresha N Deveshegowda
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Akshay Ravish
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Arunkumar Mohan
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Omantheswara Nagaraja
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Mahendra Madegowda
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Kanchugarakoppal S Rangappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Amudha Deivasigamani
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, 169610, Singapore
| | - Vijay Pandey
- Shenzhen Bay Laboratory, Shenzhen 518055, China; Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Peter E Lobie
- Shenzhen Bay Laboratory, Shenzhen 518055, China; Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, 169610, Singapore.
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Jia H, Wei J, Zheng W, Li Z. The dual role of autophagy in cancer stem cells: implications for tumor progression and therapy resistance. J Transl Med 2025; 23:583. [PMID: 40414839 DOI: 10.1186/s12967-025-06595-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Cancer stem cells (CSCs) constitute a small yet crucial subgroup in tumors, known for their capacity to self-renew, differentiate, and promote tumor growth, metastasis, and resistance to therapy. These characteristics position CSCs as significant factors in tumor recurrence and unfavorable clinical results, emphasizing their role as targets for therapy. Autophagy, an evolutionarily preserved cellular mechanism for degradation and recycling, has a complex function in cancer by aiding cell survival during stress and preserving balance by eliminating damaged organelles and proteins. Although autophagy can hinder tumor growth by reducing genomic instability, it also aids tumor advancement, particularly in harsh microenvironments, highlighting its dual characteristics. Recent research has highlighted the complex interactions between autophagy and CSCs, showing that autophagy governs CSC maintenance, boosts survival, and aids in resistance to chemotherapy and radiotherapy. On the other hand, in specific situations, autophagy may restrict CSC growth by increasing differentiation or inducing cell death. These intricate interactions offer both obstacles and possibilities for therapeutic intervention. Pharmacological modulation of autophagy, via inhibitors like chloroquine or by enhancing autophagy when advantageous, has demonstrated potential in making CSCs more responsive to standard treatments. Nonetheless, applying these strategies in clinical settings necessitates a better understanding of context-dependent autophagy dynamics and the discovery of dependable biomarkers indicating autophagic activity in CSCs. Progressing in this area might unveil novel, accurate strategies to tackle therapy resistance, lessen tumor recurrence, and ultimately enhance patient outcomes.
Collapse
Affiliation(s)
- Haiqing Jia
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China
| | - Jing Wei
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China
| | - Wei Zheng
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China.
| | - Zhuo Li
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China.
| |
Collapse
|
10
|
Li L, Cui X, Lin Z, Chen Y, Zhang X, Zhu Y. Advances in the role of baicalin and baicalein in colon cancer: mechanisms and therapeutic potential. Discov Oncol 2025; 16:860. [PMID: 40404880 PMCID: PMC12098228 DOI: 10.1007/s12672-025-02719-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 05/15/2025] [Indexed: 05/24/2025] Open
Abstract
Colorectal cancer (CRC) remains a malignancy with high incidence and mortality rates worldwide, necessitating the development of more effective therapeutic strategies due to the limitations of current treatments, including drug resistance and adverse effects. Scutellaria baicalensis, a traditional Chinese medicinal herb, contains baicalin and baicalein as its primary bioactive flavonoids, which exhibit notable pharmacological properties such as antibacterial, anti-inflammatory, antioxidant, and antitumor effects. Recent studies have demonstrated that baicalin and baicalein show promising antitumor activity in CRC treatment through mechanisms such as scavenging reactive oxygen species, immune regulation, and inhibition of tumor cell proliferation, induction of apoptosis, and modulation of the gut microenvironment. This study further investigates the molecular mechanisms underlying the therapeutic effects of baicalin and baicalein in CRC, aiming to provide new research perspectives and potential clinical applications for the integration of flavonoid-based compounds from Scutellaria baicalensis in CRC treatment.
Collapse
Affiliation(s)
- Lexin Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250000, Shangdong Province, China
| | - Xuyang Cui
- Medical College, Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250000, Shangdong Province, China
| | - Zhanyu Lin
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250000, Shangdong Province, China
| | - Yiming Chen
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250000, Shangdong Province, China
| | - Xiaoyu Zhang
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250000, Shangdong Province, China.
| | - Yong Zhu
- Department of Emergency Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No.16369, Jingshi Road, Lixia District, Jinan, 250000, Shangdong Province, China.
| |
Collapse
|
11
|
Li Y, Wang F. Research Progress on Traditional Chinese Medicines Reversing Multidrug Resistance and Mechanisms in Lung Cancer. Cancer Biother Radiopharm 2025. [PMID: 40402865 DOI: 10.1089/cbr.2025.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025] Open
Abstract
Lung cancer continues to be a primary contributor to cancer-related deaths globally, and multidrug resistance (MDR) poses a significant obstacle in its management. Traditional Chinese medicines (TCMs), recognized for their comprehensive therapeutic strategies and low incidence of adverse effects, have garnered attention due to their capacity to mitigate MDR in cancer cells. Nevertheless, deciphering the precise mechanisms through which TCMs reverse MDR in lung cancer presents a substantial scientific challenge. The objective of this review is to examine prevalent manifestations of MDR in lung cancer and underscore recent advancements in understanding how TCMs might surmount this form of resistance. The review begins by investigating the unique characteristics of TCMs and their pivotal function in reversing MDR in lung cancer. Subsequently, it explores various forms of MDR in lung cancer, such as aberrant expression of cell membrane transport proteins, dysregulation of intracellular enzyme systems, disrupted apoptosis, and heightened cellular repair mechanisms, emphasizing their detrimental impact on lung cancer treatment outcomes. Central to this review is a thorough analysis of the intricate mechanisms by which TCMs counteract MDR, along with an assessment of their efficacy in lung cancer therapy. Based on this analysis, the review offers insights into potential future research directions for utilizing TCMs to overcome MDR. This review seeks to provide a thorough examination of the role of TCMs in reversing MDR in lung cancer and to stimulate additional research into their clinical applications.
Collapse
Affiliation(s)
- Yuying Li
- School of Clinic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu Shuangliu District Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Fei Wang
- School of Clinic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
12
|
Li RC, Liu C, Wang GJ, Wang Z, Li RL, Lu HT, Xie XX, Zhang QM, Feng DQ, Yun X, Luo B. ELMOD2 Overexpression Predicts Adverse Outcomes and Regulates Tumor Progression in Gliomas. Curr Med Sci 2025:10.1007/s11596-025-00057-9. [PMID: 40397299 DOI: 10.1007/s11596-025-00057-9] [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: 01/24/2025] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVE Glioma is a highly heterogeneous and malignant intracranial tumor that presents challenges for clinical treatment. ELMO domain containing 2 (ELMOD2) is a GTPase-activating protein that regulates a range of cellular biological processes. However, its specific role and prognostic value in tumorigenesis are still unknown. This study aimed to assess the prognostic relevance and signaling function of ELMOD2 in gliomas. METHODS The Chinese Glioma Genome Atlas (CGGA) and The Cancer Genome Atlas (TCGA) databases were utilized to conduct a comprehensive analysis of the expression profile of ELMOD2 in gliomas, elucidating its associations with clinicopathological parameters and patient prognosis. Single-cell analysis was performed to characterize ELMOD2 expression across distinct glioma cell subpopulations. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and Gene Set Variation Analysis (GSVA) were employed to evaluate the potential biological functions of ELMOD2 in gliomagenesis. Specific small interfering RNAs (siRNAs) were used to knock down ELMOD2 in the glioma cell lines U251 and A172 to assess their cellular behaviors and examine the levels of multiple key signaling molecules associated with the occurrence of gliomas. RESULTS ELMOD2 was overexpressed in gliomas, and this upregulation was correlated with tumor grade, isocitrate dehydrogenase mutation, and 1p/19q codeletion status. Notably, ELMOD2 expression was elevated in classical and mesenchymal subtypes, and single-cell resolution analysis revealed predominant enrichment within malignant cells. Functionally, ELMOD2 regulated cell cycle progression, and its overexpression was related to independent adverse outcomes. In vitro experiments revealed that ELMOD2 was located in the cytoplasm and nucleoplasm. Furthermore, ELMOD2 knockdown reduced proliferation, migration, and invasion and increased apoptosis in U251 and A172 cell lines. Finally, ELMOD2 knockdown significantly decreased p-Erk1/2. CONCLUSIONS ELMOD2 expression in glioma is positively correlated with tumorigenesis and is a crucial independent prognostic marker. Thus, ELMOD2 is a promising biomarker and therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Rui-Chao Li
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, 530021, China
- Department of Prenatal Genetics, Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Chang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Guo-Jian Wang
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, 530021, China
| | - Zi Wang
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, 530021, China
| | - Rong-Lin Li
- Department of Thoracic Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Hao-Tian Lu
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xiao-Xun Xie
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, 530021, China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Nanning, 530021, China
| | - Qing-Mei Zhang
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, 530021, China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Nanning, 530021, China
| | - Da-Qin Feng
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Xiang Yun
- Department of International Cooperation and Exchanges, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Bin Luo
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, 530021, China.
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Nanning, 530021, China.
| |
Collapse
|
13
|
Ahmad M, Roy NJ, Mondal D, Vijayakanth T, Lahiri M, Talukdar P. Illuminating apoptosis: a visible light-activated chloride carrier for chloride transport and cell death. J Mater Chem B 2025; 13:5957-5966. [PMID: 40314174 DOI: 10.1039/d4tb02436b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Synthetic chloride carriers are known to induce chloride-mediated apoptosis inside cancer cells. One of the main disadvantages is the unfavorable cytotoxicity towards healthy cells due to the lack of selectivity. The use of stimuli, such as light, enzymes, ligands, etc., has enabled the selective activation of these systems in cancer cells. Light, notably, is a significant stimulus that has been utilized due to its excellent spatiotemporal control, remote addressability, and low cytotoxicity. However, previously reported photoresponsive systems require UV radiation for their activation, which has low tissue penetration and can lead to phototoxic cell damage or death. Herein, we report 3-substituted indole-2-carboxamide ion carriers and their o-nitrobenzyl (ONB) linked procarriers. The incorporation of the electron-donating substituents to the ONB photocleavable group leads to a significant red shift in the absorption wavelength, and for the N,N-dimethyl-based procarrier, the absorbance peak extends up to 500 nm. Eventually, all the synthesized procarriers were photoactivated inside MCF-7 cancer cells under 400 nm electromagnetic radiation, and the N,N-dimethyl-based procarrier was also photoactivated at 450 nm. This photoactivation at a higher wavelength of electromagnetic radiation is highly desirable for its practical biological applications.
Collapse
Affiliation(s)
- Manzoor Ahmad
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| | - Naveen J Roy
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| | - Debashis Mondal
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| | - Thangavel Vijayakanth
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Mayurika Lahiri
- Department of Biology, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| |
Collapse
|
14
|
Lan Y, Sun J, Xu J, Chen X. Anti-lung cancer activity of lotusine in non-small cell lung cancer HCC827 via reducing proliferation, oxidative stress, induction of apoptosis, and G0/G1 cell cycle arrest via suppressing EGFR-Akt-ERK signalling. In Vitro Cell Dev Biol Anim 2025:10.1007/s11626-025-01048-9. [PMID: 40392483 DOI: 10.1007/s11626-025-01048-9] [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: 02/13/2025] [Accepted: 04/10/2025] [Indexed: 05/22/2025]
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related deaths worldwide, with resistance to targeted therapies and the need for novel therapeutic agents driving ongoing research. In this study, we investigated the anti-lung cancer activity of lotusine, a natural alkaloid, in the A549 (non-EGFR mutant), and EGFR-mutant HCC827 NSCLC cell line (deletion in exon 19). Our results demonstrated that lotusine significantly inhibited cell proliferation in a concentration- and time-dependent manner of HCC827 cells in comparison to A549 cells. Mechanistic analysis revealed that lotusine induced apoptosis in HCC827 cells, as evidenced by increased expression of pro-apoptotic markers (Bax and cleaved caspase-3) and decreased levels of anti-apoptotic proteins (Bcl-2). Cell cycle analysis indicated that lotusine caused G0/G1 phase arrest. Importantly, lotusine exerted its effects through the inhibition of the epidermal growth factor receptor (EGFR) EGFR-Akt-ERK signaling pathway, as evidenced by reduction of p-EGFR, p-Akt, and p-ERK in a western blot analysis in HCC827 cells. These findings suggest that lotusine exerts potent anti-cancer effects via a multifaceted mechanism, including inhibition of proliferation, apoptosis induction, and cell cycle arrest, predominantly mediated by EGFR suppression. This study highlights lotusine as a promising therapeutic candidate for the treatment of EGFR-mutant NSCLC and provides insights into its molecular mechanisms of action, paving the way for further preclinical and clinical evaluations.
Collapse
Affiliation(s)
- Yuanmin Lan
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People's Hospital of Shenzhen, Guangdong, Guangdong, 518172, China
| | - Jing Sun
- Department Of Oncology, The Fifth People's Hospital Of Dalian, Dalian Liaoning, 116021, China
| | - Jiqing Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People's Hospital of Shenzhen, Guangdong, Guangdong, 518172, China
| | - Xiaoying Chen
- Department of Respiratory and Critical Care Medicine, Lishui Second People's Hospital, Lishui Zhejiang, 323000, China.
| |
Collapse
|
15
|
Chen X, Ma X, Hu X, Wang C, Zhang X, Yan C. Mechanisms and potential therapeutic strategies of withaferin A in breast cancer. Pharmacol Rep 2025:10.1007/s43440-025-00736-3. [PMID: 40392517 DOI: 10.1007/s43440-025-00736-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/22/2025]
Abstract
Breast cancer (BC) is one of the most common malignant tumors in women worldwide, and its treatment faces numerous challenges. Despite the effectiveness of modern treatment methods such as surgery, radiotherapy, chemotherapy, and targeted therapy, issues like recurrence, metastasis, and drug resistance still significantly affect patient prognosis and survival rates. This is particularly true for triple-negative breast cancer (TNBC) and HER2-positive BC, for which treatment outcomes are relatively poor. Withaferin A (WA), a natural plant-derived compound, has shown significant anti-cancer effects in the treatment of BC. WA inhibits the progression of BC through multiple mechanisms, including suppressing cell migration and invasion, inducing tumor cell apoptosis, regulating autophagy and metabolic pathways, and modulating miRNA expression. In combination therapy, WA exhibits a good synergistic effect when used with other anti-cancer drugs such as phenethyl isothiocyanate (PEITC), cisplatin, and sulforaphane, significantly enhancing therapeutic efficacy and reducing drug resistance. This review summarizes the research progress on the mechanisms of WA in combating BC, aiming to provide a foundation for the scientific development and clinical application of WA in BC treatment.
Collapse
Affiliation(s)
- Xin Chen
- Shandong Electric Power Central Hospital, Jinan, Shangdong Province, China
| | - Xijun Ma
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Jinan, Shangdong Province, China
| | - Xiaofei Hu
- Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province, China
| | - Cihang Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province, China
| | - Xiaoyu Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province, China.
| | - Chunchun Yan
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Jinan, Shangdong Province, China.
| |
Collapse
|
16
|
Ruban M, Pozhidaeva E, Bolotina L, Kaprin A. The Role of Diet and Nutrition in Cancer Development and Management: From Molecular Mechanisms to Personalized Interventions. Foods 2025; 14:1788. [PMID: 40428567 DOI: 10.3390/foods14101788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/08/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Diet plays a crucial role in cancer development and progression, beyond traditional risk factors. This review aims to summarize current evidence on the role of diet and specific nutrients in cancer development and progression, focusing on molecular mechanisms. We also discuss the potential of personalized dietary interventions, based on tumor and patient characteristics, in enhancing cancer prevention and treatment strategies. The review covers the impact of calories, protein, sugar, and other dietary components on signaling pathways and growth factors involved in carcinogenesis. We examine the influence of obesity, insulin resistance, and other metabolic factors on cancer risk and outcomes. The article also explores current dietary strategies, including calorie restriction, ketogenic diets, and the role of the gut microbiome in modulating response to anticancer therapies. Finally, we highlight the need for further research to develop targeted, personalized dietary recommendations based on an individual's tumor profile, stage of disease, and other clinical factors. Integrating such personalized dietary approaches into cancer prevention and treatment holds promise for improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Maxim Ruban
- P. Hertsen Moscow Oncology Research Institute-Branch of the National Medical Research Radiological Centre, 105425 Moscow, Russia
| | - Elizaveta Pozhidaeva
- P. Hertsen Moscow Oncology Research Institute-Branch of the National Medical Research Radiological Centre, 105425 Moscow, Russia
| | - Larisa Bolotina
- P. Hertsen Moscow Oncology Research Institute-Branch of the National Medical Research Radiological Centre, 105425 Moscow, Russia
| | - Andrey Kaprin
- P. Hertsen Moscow Oncology Research Institute-Branch of the National Medical Research Radiological Centre, 105425 Moscow, Russia
- Department of Urology and Operative Nephrology, Peoples' Friendship University of Russia (RUDN University), Mikluho-Maklaya St., 6, 117198 Moscow, Russia
| |
Collapse
|
17
|
Sun C, Gui J, Sheng Y, Huang L, Zhu X, Huang K. Specific signaling pathways mediated programmed cell death in tumor microenvironment and target therapies. Discov Oncol 2025; 16:776. [PMID: 40377777 PMCID: PMC12084487 DOI: 10.1007/s12672-025-02592-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Increasing evidence has shown that programmed cell death (PCD) plays a crucial role in tumorigenesis and cancer progression. The components of PCD are complex and include various mechanisms such as apoptosis, necroptosis, alkaliptosis, oxeiptosis, and anoikis, all of which are interrelated in their functions and regulatory pathways. Given the significance of these processes, it is essential to conduct a comprehensive study on PCD to elucidate its multifaceted nature. Key signaling pathways, particularly the caspase signaling pathway, the RIPK1/RIPK3/MLKL pathway, and the mTOR signaling pathway, are pivotal in regulating PCD and influencing tumor progression. In this review, we briefly describe the generation mechanisms of different PCD components and focus on the regulatory mechanisms of these three major signaling pathways within the context of global PCD. Furthermore, we discuss various tumor therapeutic compounds that target different signaling axes of these pathways, which may provide novel strategies for effective tumor therapy and help improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Chengpeng Sun
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Jiawei Gui
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Yilei Sheng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Le Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China
| | - Xingen Zhu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Kai Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
18
|
Li X, Wang L, Ni B, Wang J, Sun Y. Research Progress of Natural Compounds from Chinese Herbal Medicine in the Treatment of Melanoma. Curr Treat Options Oncol 2025:10.1007/s11864-025-01322-8. [PMID: 40372659 DOI: 10.1007/s11864-025-01322-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/16/2025]
Abstract
OPINION STATEMENT Melanoma is a malignant tumor that originates from activated or genetically altered epidermal melanocytes, resulting from the interplay of genetic, somatic, and environmental factors. It is the fastest-growing malignancy among the Caucasian population and has a high mortality rate, second only to lung cancer. Current mainstream treatments have led to unavoidable drug resistance and toxic side effects despite improvements in efficacy and prognosis. Traditional Chinese Medicine is a significant component of complementary and alternative medicine, playing a vital role in cancer treatment. Natural compounds derived from Chinese herbal medicines offer notable advantages owing to their multimolecular, multitarget, and multipathway characteristics. These compounds exert anti-melanoma effects through various mechanisms, including antiproliferation, promotion of apoptosis, inhibition of metastasis, suppression of angiogenesis, modulation of autophagy, and enhancement of the immune response. Furthermore, combining natural compounds with mainstream antagonistic medicine not only enhances treatment efficacy but also significantly reverses multidrug resistance. This article discusses the specific mechanisms by which natural compounds combat melanoma and reviews the recent research advancements in this field. It also addresses the challenges faced in the widespread clinical application of these natural compounds in melanoma treatment and outlines the future directions for their development.
Collapse
Affiliation(s)
- Xin Li
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Lankang Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Baoyi Ni
- Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Wang
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Yifeng Sun
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China.
| |
Collapse
|
19
|
Majeed NS, Mohammed MH, Hatem ZA, El-Sehrawy AAMA, Ganesan S, Singh A, Akoul MA, Sudan P, Singh R, Hamad HA. Interplay between NETosis and the lncRNA-microRNA regulatory axis in the immunopathogenesis of cancer. J Physiol Biochem 2025:10.1007/s13105-025-01082-x. [PMID: 40358898 DOI: 10.1007/s13105-025-01082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025]
Abstract
Neutrophil extracellular traps (NETs), web-like complex structures secreted by neutrophils, have emerged as key players in the modulation of immune responses and the immunopathogenesis of immune disorders. Initially described for their antimicrobial function, NETs now play a part in the fundamental processes of cancer biology, including cancer initiation, metastatic dissemination, and immune evasion strategies. NETs hijack anti-tumor immunity by entrapping circulating cancer cells, fostering the growth of tumors, and reorganizing the tumor microenvironment such that it is pro-malignancy. Emerging evidence emphasizes the role of NETosis coupled with non-coding RNAs-long non-coding RNAs (lncRNAs) and microRNAs (miRNAs)-as key regulators of gene expression and controllers of processes vital for cancer growth, such as immune response and programmed cell death processes like apoptosis, necroptosis, pyroptosis, and ferroptosis. Aberrantly expressed non-coding RNAs have been attributed to immune dysregulation and excessive NET production, promoting tumor growth. NETs are also associated with a myriad of pathological conditions, such as autoimmune disorders, cystic fibrosis, sepsis, and thrombotic disorders. New therapeutic approaches-such as DNase therapy and PAD4 inhibitors-target NET production and their degradation to modify immune function and the efficiency of immunotherapies. Further clarification of the intricate interactions of NETosis, lncRNAs, and miRNAs has the potential to establish new strategies for the suppression of the growth of tumors and preventing immune evasion. This review seeks to elucidate the interactions between NETosis and the regulatory networks involving non-coding RNAs that significantly contribute to the immunopathogenesis of cancer.
Collapse
Affiliation(s)
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques department, College of Health and medical technology, Al-Maarif University, Anbar, Iraq.
| | - Zainab Amer Hatem
- College of Science, Biotechnology Department, Diyala University, Diyala, Iraq
| | | | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Marwa Azeez Akoul
- Biotechnology Department, College of Applied Science, Fallujah University, Anbar, Iraq
| | - Puneet Sudan
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Roshni Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Hamad Ali Hamad
- Department of Pathological Analysis, Collage of Applied Sciences, University of Fallujah, Fallujah, Iraq
| |
Collapse
|
20
|
Phamornnak C, Klaharn N, Suwanno T, Bunjongpru W, Jeamsaksiri W, Oonkhanond B, Srinives S. Modified ISFET for Real-Time Calcium Ion Sensing in MDA-MB-231 Breast Cancer Cells. Chem Asian J 2025:e00268. [PMID: 40354185 DOI: 10.1002/asia.202500268] [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: 02/14/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Transfer of extracellular and intracellular calcium ions (Ca2⁺) plays a crucial role in programmed cell death, with varied Ca2⁺ concentrations associated with cell growth, proliferation, apoptosis, and so on. Real-time monitoring of extracellular Ca2⁺ concentrations ([Ca2+]e) can provide valuable quantitative insights into programmed cell death, which may greatly benefit cancer therapy and other biological applications. This study used a modified ion-sensitive field-effect transistor (ISFET) as a platform for Ca2⁺ sensing. The ISFET device was functionalized with a Ca2⁺-selective membrane containing polyurethane (PU) and calcium ionophore II. The Ca2⁺-FET device exhibited a sensitivity of 35 ± 3 mV/pCa (pCa 0-5) toward Ca2⁺ with insignificant cross-sensitivity to other ions, such as sodium (Na⁺) and potassium (K⁺). The device was applied to monitor [Ca2+]e concentrations in the MDA-MB-231 breast cancer cell culture, showing sufficient reliance in detecting Ca2⁺ concentrations (0.1 mM to 1 M (pCa 0-5)) correlated with cellular activities. This technique offers a noninvasive and label-free approach for real-time Ca2⁺ monitoring in cell culture, with potential applications in cancer research and therapeutic development.
Collapse
Affiliation(s)
- Chinnawich Phamornnak
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, 25/25 Phutthamonthon 4 Rd., Salaya, Nakhon Pathom, 73170, Thailand
- Cell Technology and Tissue Engineering Research Team, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Natakorn Klaharn
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, 25/25 Phutthamonthon 4 Rd., Salaya, Nakhon Pathom, 73170, Thailand
| | - Theetat Suwanno
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, 25/25 Phutthamonthon 4 Rd., Salaya, Nakhon Pathom, 73170, Thailand
| | - Win Bunjongpru
- Thai Microelectronics Center, 51/4 Suwintawong Rd., Wangtakien, Chachoengsao, 24000, Thailand
| | - Wutthinan Jeamsaksiri
- Thai Microelectronics Center, 51/4 Suwintawong Rd., Wangtakien, Chachoengsao, 24000, Thailand
| | - Bovornlak Oonkhanond
- Department of Chemical Engineering, Faculty of Engineering, Mahidol University, 25/25 Phutthamonthon 4 Rd., Salaya, Nakhon Pathom, 73170, Thailand
- Globrick Refractories Co., Ltd., Bangkhen, Muang, Nonthaburi, 11000, Thailand
| | - Sira Srinives
- Department of Chemical Engineering, Faculty of Engineering, Mahidol University, 25/25 Phutthamonthon 4 Rd., Salaya, Nakhon Pathom, 73170, Thailand
| |
Collapse
|
21
|
Ma Y, Wang Y, Wang S, Wang H, Zhao Y, Peng C, Liu X, Yang J. Regulatory roles of non-coding RNAs in programmed cell death pathways and drug resistance in gastrointestinal stromal tumors. Clin Exp Med 2025; 25:150. [PMID: 40347390 PMCID: PMC12065685 DOI: 10.1007/s10238-025-01667-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/02/2025] [Indexed: 05/12/2025]
Abstract
Gastrointestinal stromal tumors (GIST) are the most common mesenchymal tumors of the gastrointestinal tract, primarily driven by KIT or PDGFRA mutations. Programmed cell death (PCD), including apoptosis, autophagy, and ferroptosis, plays a crucial role in GIST pathogenesis, progression, and treatment response. Non-coding RNAs (ncRNAs) have emerged as key regulators of PCD pathways, influencing GIST proliferation, metastasis, and drug resistance, particularly in response to tyrosine kinase inhibitors (TKIs) such as imatinib. Apoptosis suppression is strongly associated with poor prognosis, while autophagy contributes to tumor dormancy and TKI resistance. Ferroptosis, a novel iron-dependent cell death pathway, represents a promising therapeutic target. Recent evidence suggests that ncRNAs modulate these PCD pathways through interactions with key molecular regulators such as miR-494, miR-30a, and lncRNAs, which affect signaling networks including PI3K/AKT, MAPK, and mTOR. Furthermore, ncRNAs have mediated secondary resistance to imatinib by promoting autophagic flux and altering ferroptosis sensitivity. Understanding the molecular interplay between ncRNAs and PCD in GIST provides novel insights into disease mechanisms and offers potential therapeutic strategies to overcome drug resistance. Targeting ncRNA-mediated regulation of apoptosis, autophagy, and ferroptosis may enhance treatment efficacy and improve patient outcomes. Future research should focus on elucidating the mechanistic roles of ncRNAs in PCD pathways to develop innovative diagnostic and therapeutic approaches for GIST.
Collapse
Affiliation(s)
- Yuxuan Ma
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 127, Changlexi Road, Xi'an, 710032, Shaanxi Province, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuhao Wang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 127, Changlexi Road, Xi'an, 710032, Shaanxi Province, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Shu Wang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 127, Changlexi Road, Xi'an, 710032, Shaanxi Province, China
| | - Haoyuan Wang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 127, Changlexi Road, Xi'an, 710032, Shaanxi Province, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Zhao
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 127, Changlexi Road, Xi'an, 710032, Shaanxi Province, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Chaosheng Peng
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 127, Changlexi Road, Xi'an, 710032, Shaanxi Province, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Xin Liu
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 127, Changlexi Road, Xi'an, 710032, Shaanxi Province, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Jianjun Yang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 127, Changlexi Road, Xi'an, 710032, Shaanxi Province, China.
| |
Collapse
|
22
|
Leandri R, Power K, Martano M, De Vico G. Immunohistochemical Detection of Iron-Related Proteins in Sertoli Cell-Only Patterns in Canine Testicular Lesions. Animals (Basel) 2025; 15:1377. [PMID: 40427255 DOI: 10.3390/ani15101377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Sertoli cell-only (SCO) tubules are a histologic pattern characterized by the absence of germ cells within seminiferous tubules, leading to infertility in both humans and dogs. While its association with testicular tumors has been documented, the role of iron metabolism in SCO tubules remains unclear. This study investigates the immunolabeling of key iron-related proteins (Transferrin Receptor 1, Transferrin Receptor 2, and Ferritin Heavy chain 1) and Proliferating Cell Nuclear Antigen (PCNA) in canine SCO tubules within distinct microenvironments: seminomas, Sertoli cell tumors, and isolated. We confirm the presence and distribution of iron-related proteins in Sertoli cells as a part of a Sertoli cell-only pattern across different microenvironments. Our findings suggest a potential increase in iron uptake in association with tumors, and the cytoplasmic PCNA immunolabeling suggests a preferential activation of cell survival rather than proliferation, potentially facilitating neoplastic transformation. In contrast, Sertoli cells in the isolated Sertoli cell-only pattern exhibit nuclear PCNA immunolabeling, possibly correlated to the state of immaturity of Sertoli cells. These findings highlight the role of iron homeostasis and apoptosis in testicular tumorigenesis. Immunohistochemistry revealed that Sertoli cells in SCO tubules actively uptake iron in all conditions, yet their capacity to utilize it for proliferation appears restricted. Interestingly, PCNA labeling exhibits a pattern dependent on the microenvironment: in tumor-associated SCO tubules, it showed cytoplasmic localization, characteristic of an anti-apoptotic function, whereas isolated SCO tubules showed nuclear PCNA labeling, suggesting a potential role in DNA synthesis and repair. These findings highlight the interplay between iron homeostasis and cellular survival mechanisms, offering novel perspectives on its pathophysiology and implications for testicular cancer development.
Collapse
Affiliation(s)
- Rebecca Leandri
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Karen Power
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Manuela Martano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy
| | - Gionata De Vico
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| |
Collapse
|
23
|
Sun X, Chen Z, Yang H, Yu J, Lin H, Zhang L. Targeting ferroptosis for precision medicine in cervical cancer. Apoptosis 2025:10.1007/s10495-025-02120-1. [PMID: 40335818 DOI: 10.1007/s10495-025-02120-1] [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] [Accepted: 04/17/2025] [Indexed: 05/09/2025]
Abstract
Cervical cancer (CC) is a prevalent malignant tumor in the female reproductive system, with rising incidence rates among younger women posing a significant public health challenge. Human papillomavirus (HPV) infection is the primary cause, driving carcinogenesis by promoting abnormal proliferation of tumor cells. Ferroptosis is a form of regulated necrosis that is caused by an iron-dependent accumulation of lipid peroxides with rupture of the plasma membrane. Targeting ferroptosis-related molecules and pathways can selectively induce cervical cancer cell death, while alterations in the expression of ferroptosis-related genes provide promising biomarkers for prognostic assessment. Advances in research on biomarkers and molecular targets are improving predictions of therapeutic outcomes, overcoming drug resistance, and optimizing immunotherapy strategies, thereby opening new avenues for precision medicine. This review focuses on the molecular mechanisms underlying ferroptosis in cervical cancer, discusses its potential applications in early diagnosis and prognosis evaluation, and summarizes the latest advancements in targeted therapy, aiming to provide a novel perspective for the clinical management of cervical cancer.
Collapse
Affiliation(s)
- Xin Sun
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, China
| | - Zhuoxi Chen
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, China
| | - Hui Yang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, China
| | - Jianing Yu
- The Second School of Clinical Medicine, Binzhou Medical University, Yantai, 264100, China.
| | - Haiyan Lin
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, China.
| | - Leiming Zhang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
24
|
Feng H, Deng D, Zhu F, Chen S, Geng J, Jiang S, Zhang K, Jiang J, Yin S, Zhang C. Acute exposure to glufosinate-ammonium induces hepatopancreas toxicity in juvenile Chinese mitten crab (Eriocheir sinensis). JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137487. [PMID: 39914334 DOI: 10.1016/j.jhazmat.2025.137487] [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: 08/30/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 03/19/2025]
Abstract
Glufosinate-ammonium (GLA) is a widely used organophosphorus herbicide, which poses a potential threat to non-target aquatic species. This study aimed to evaluate the toxic effects of acute exposure to GLA on the hepatopancreas of juvenile Eriocheir sinensis, and to preliminarily reveal the toxicity mechanism. The results showed that the 96h-LC50 of GLA on juvenile E. sinensis was 386.61 mg/L. The acute test showed that GLA exposure caused hepatopancreas histological lesions, DNA damage and a higher apoptosis rate. The activities of aspartate aminotransferase and alanine aminotransferase in serum increased significantly and had a concentration-dependent effect. Moreover, GLA exposure resulted in a significant increase in malondialdehyde content, which subsequently activated the antioxidant system and detoxification system, and the related enzyme activities and gene expression levels were significantly increased. In addition, the RNA-Seq analysis showed that the toxic effects of GLA exposure on juvenile crabs may mainly involve physiological pathways such as energy metabolism, protein synthesis and nervous system function. This study highlights the hepatotoxic effects of GLA on aquatic crustaceans and preliminarily reveals the key pathways of action. The results of this study will helpful to provide new insights into the toxic effects and risk assessment of herbicides on non-target organisms.
Collapse
Affiliation(s)
- Huixia Feng
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Dunqian Deng
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Fei Zhu
- Jiangsu Marine Fisheries Research Institute, Nantong 226007, China
| | - Shuyin Chen
- Jiangsu Marine Fisheries Research Institute, Nantong 226007, China
| | - Jiayin Geng
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Su Jiang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Kai Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Jianbin Jiang
- Nantong Tongzhou District Aquatic Technology Guidance Station, Nantong 226399, China
| | - Shaowu Yin
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China.
| | - Cong Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China.
| |
Collapse
|
25
|
Naidoo DB, Phulukdaree A, Chuturgoon AA, Sewram V. Centella Asiatica Fraction-3 Enhances Antioxidant Capacity and Apoptotic Cell Death in HEK293 Kidney Cells. J Med Food 2025; 28:455-464. [PMID: 40160106 DOI: 10.1089/jmf.2022.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
The traditional medicinal plant Centella asiatica is commonly used in Chinese and Ayurvedic medicine due to its vast range of therapeutic properties. Previously, the ethanolic C. asiatica leaf extract was subjected to silica column fractionation, and the C3 fraction was obtained. We investigated the antioxidant and anti-proliferative effects of C3 in human embryonic kidney (HEK293) cells. In HEK293 cells, C3 cytotoxicity was assessed (viability assay; 24 h; [0.2-3 mg/mL]), and a half maximal inhibitory concentration (IC50) was determined. Malondialdehyde (MDA), lactate dehydrogenase (LDH) (spectrophotometry), mitochondrial depolarization (Δψm), intracellular reactive oxygen species (flow cytometry), glutathione (GSH), oxidized glutathione (GSSG) concentrations, caspase activities, ATP levels (luminometry), and fragmentation of DNA (SCGE assay) were evaluated. Protein expressions were assessed by western blotting. Gene expressions were quantified by qPCR. Cell viability in HEK293 cells was decreased in a dose-dependent manner by C3. MDA, Δψm, LDH, caspase activities, and DNA fragmentation (P < .0004) were significantly increased by C3. Nuclear factor erythroid 2-related factor 2 (Nrf-2) protein expression, GSH, and GSSG concentrations were increased, whereas antioxidant (Nrf-2, GPx, SOD, and CAT) gene expression was significantly decreased by C3 (P < .001). C3 decreased both Bax and Bcl-2 protein expression (P < .03). Gene expression of c-myc was significantly increased, whereas OGG-1 was significantly reduced by C3 (P < .05). C3 reduced antioxidant gene expression, increased antioxidant levels, and elevated anti-proliferative effects in HEK293 cells, suggesting that high concentrations of C3 are potentially toxic to kidney cells, thus rendering cause for concern with its human use.
Collapse
Affiliation(s)
- Dhaneshree B Naidoo
- Faculty of Health Sciences, Discipline of Medical Biochemistry and Chemical Pathology, Howard College, University of KwaZulu-Natal, Durban, South Africa
| | - Alisa Phulukdaree
- Faculty of Health Sciences, Discipline of Medical Biochemistry and Chemical Pathology, Howard College, University of KwaZulu-Natal, Durban, South Africa
| | - Anil Armichund Chuturgoon
- Faculty of Health Sciences, Discipline of Medical Biochemistry and Chemical Pathology, Howard College, University of KwaZulu-Natal, Durban, South Africa
| | - Vikash Sewram
- Division of Health Systems and Public Health, Faculty of Medicine and Health Sciences, African Cancer Institute, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
26
|
Wen Q, Liu J, Hu J, Kou KI, Li H, Zhang J, Zhang R, Zhong S, Huang R. Molecular mechanisms underlying the anti-Colon Cancer effects of Caulerpa lentillifera polysaccharides (CLP). Int J Biol Macromol 2025; 308:142594. [PMID: 40157667 DOI: 10.1016/j.ijbiomac.2025.142594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/15/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Colon cancer (CC) ranks is the second leading cause of cancer-related deaths globally. Despite chemotherapy being a primary treatment its effectiveness significantly declines in advanced in stage. Emerging evidence suggests that dietary components particularly polysaccharides, play a role in CC progression. This study employed multi-omics and network pharmacology to elucidate the mechanisms underlying the apoptotic effects of Caulerpa lentillifera polysaccharide (CLP) in CC, validated through in vitro and in vivo experiments. Transcriptomics and network pharmacology analysis identified the p53/Bax/Caspase-3 pathway as a key regulatory axis. Further targeted analysis of amino acid metabolism revealed that CLP significantly decreased intracellular aspartate (Asp) levels. Additionally, reactive oxygen species (ROS) accumulation was detected in cells. CLP treatment reduced Asp content, leading to ROS accumulation, which activated the p53/Bax/Caspase-3 pathway, triggering apoptosis. In vivo, CLP effectively inhibited tumor growth in BALB/c mice bearing CT26 colon cancer cells. These findings suggest that CLP exerts anti-colon cancer effects by modulating amino acid metabolism and inducing apoptosis via the p53/Bax/Caspase-3 axis, providing a promising therapeutic strategy for CC.
Collapse
Affiliation(s)
- Qinghua Wen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jun Liu
- Laboratory of Pathogenic Biology, Guangdong Medical University, Zhanjiang 524023, China
| | - Jiaheng Hu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Kit Ian Kou
- Department of Mathematics, Faculty of Science and Technology, University of Macau, Macao
| | - Haichou Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaojiao Zhang
- College of Food and Health, Zhejiang A& F University, Hangzhou 311300, China
| | - Rongxin Zhang
- Department of Colorectal Surgery, Sun Yatsen University Cancer center, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, Guangzhou 510060, China
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; Shenzhen Research Institute, Guangdong Ocean University, Shenzhen 518108, China.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
27
|
Fiser O, Muller P. Role of HSF1 in cell division, tumorigenesis and therapy: a literature review. Cell Div 2025; 20:11. [PMID: 40287736 PMCID: PMC12034185 DOI: 10.1186/s13008-025-00153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Heat shock factor 1 (HSF1) is the master orchestrator of the heat shock response (HSR), a critical process for maintaining cellular health and protein homeostasis. These effects are achieved through rapid expression of molecular chaperones, the heat shock proteins (HSPs), which ensure correct protein folding, repair, degradation and stabilization of multiprotein complexes. In addition to its role in the HSR, HSF1 influences the cell cycle, including processes such as S phase progression and regulation of the p53 pathway, highlighting its importance in cellular protein synthesis and division. While HSF1 activity offers neuroprotective benefits in neurodegenerative diseases, its proteome-stabilizing function may also reinforce tumorigenic transformation. HSF1 overexpression in many types of cancer reportedly enhances cell growth enables survival, alters metabolism, weakens immune response and promotes angiogenesis or epithelial-mesenchymal transition (EMT) as these cells enter a form of "HSF1 addiction". Furthermore, the client proteins of HSF1-regulated chaperones, particularly Hsp90, include numerous key players in classical tumorigenic pathways. HSF1 thus presents a promising therapeutic target for cancer treatment, potentially in combination with HSP inhibitors to alleviate typical initiation of HSR upon their use.
Collapse
Affiliation(s)
- Otakar Fiser
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petr Muller
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic.
| |
Collapse
|
28
|
Gupta G, Samuel VP, M RM, Rani B, Sasikumar Y, Nayak PP, Sudan P, Goyal K, Oliver BG, Chakraborty A, Dua K. Caspase-independent cell death in lung cancer: from mechanisms to clinical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04149-0. [PMID: 40257494 DOI: 10.1007/s00210-025-04149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/05/2025] [Indexed: 04/22/2025]
Abstract
Caspase-independent cell death (CICD) has recently become a very important mechanism in lung cancer, in particular, to overcome a critical failure in apoptotic cell death that is common to disease progression and treatment failures. The pathways involved in CICD span from necroptosis, ferroptosis, mitochondrial dysfunction, and autophagy-mediated cell death. Its potential therapeutic applications have been recently highlighted. Glutathione peroxidase 4 (GPX4) inhibition-driven ferroptosis has overcome drug resistance in non-small cell lung cancer (NSCLC). In addition, necroptosis involving RIPK1 and RIPK3 causes tumor cell death and modulation of immune responses in the tumor microenvironment (TME). Mitochondrial pathways are critical for CICD through modulation of metabolic and redox homeostasis. Ferroptosis is amplified by mitochondrial reactive oxygen species (ROS) and lipid peroxidation in lung cancer cells, and mitochondrial depolarization induces oxidative stress and leads to cell death. In addition, mitochondria-mediated autophagy, or mitophagy, results in the clearance of damaged organelles under stress conditions, while this function is also linked to CICD when dysregulated. The role of cell death through autophagy regulated by ATG proteins and PI3K/AKT/mTOR pathway is dual: to suppress tumor and to sensitize cells to therapy. A promising approach to enhancing therapeutic outcomes involves targeting mechanisms of CICD, including inducing ferroptosis by SLC7A11 inhibition, modulating mitochondrial ROS generation, or combining inhibition of autophagy with chemotherapy. Here, we review the molecular underpinnings of CICD, particularly on mitochondrial pathways and their potential to transform lung cancer treatment.
Collapse
Affiliation(s)
- Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Y Sasikumar
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Priya Priyadarshini Nayak
- Department of Medical Oncology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Puneet Sudan
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Brian G Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Amlan Chakraborty
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Kamal Dua
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia.
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
29
|
Ghogare SS, Pathan EK. Intratumor fungi specific mechanisms to influence cell death pathways and trigger tumor cell apoptosis. Cell Death Discov 2025; 11:188. [PMID: 40258837 PMCID: PMC12012188 DOI: 10.1038/s41420-025-02483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer, uncontrolled cell growth due to the loss of cell cycle regulation, is often found to be associated with viral infections and, as recent studies show, with bacterial infections as well. Emerging reports also suggest a strong link between fungi and cancer. The crucial virulence trait of fungi, the switch from yeast (Y) to hyphal (H) form, is found to be associated with carcinogenesis. The physicochemical properties and signal transduction pathways involved in the switch to the hyphal form overlap with those of tumor cell formation. Inhibiting differentiation causes apoptosis in fungi, whereas preventing apoptosis leads to cancer in multicellular organisms. Literature on the fungi-cancer linkage, though limited, is increasing rapidly. This review examines cancer-specific fungal communities, the impact of fungal microbiome on cancer cell progression, similarities between fungal differentiation and cells turning cancerous at biochemical and molecular levels, including the overlaps in signal transduction pathways between fungi and cancer. Based on the available evidence, we suggest that molecules inhibiting the yeast-hyphal transition in fungi can be combined with those targeting tumor cell apoptosis for effective cancer treatment. The review points out fertile research areas where mycologists and cancer researchers can collaborate to unravel common molecular mechanisms. Moreover, antibodies targeting fungal-specific chitin and glucan can be used for the selective neutralization of tumor cells. These new combinations of potential therapies are expected to facilitate the development of target-specific, less harmful and commercially feasible anticancer therapies. We bring together available evidence to argue that fungal infections could either trigger cancer or have a significant role in the development and progression of cancer. Hence, cancer-associated fungal populations could be utilized as a target for a combination therapy involving the integration of anticancer and antifungal drugs as well as inhibitors of fungal morphogenesis to develop more effective anticancer therapies.
Collapse
Affiliation(s)
- Simran S Ghogare
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) Lavale, Pune, 412115, Maharashtra, India
| | - Ejaj K Pathan
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
30
|
Yao J, Zhu F, Feng Y, Gu C, Wang T, Li X, Yang H, Hu X, Bonnet PA, Meng X. Research Progress on the Structure-activity Relationship and Mechanism of Flavonoid Derivatives in the Treatment of Lung Cancer. Molecules 2025; 30:1827. [PMID: 40333837 PMCID: PMC12029265 DOI: 10.3390/molecules30081827] [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: 03/19/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide. The difficulty in early diagnosis, combined with the tendency for tumor invasion and metastasis, creates significant challenges for current therapeutic approaches. Additionally, the pharmaceutical agents currently used to treat NSCLC often come with severe side effects and can lead to drug resistance. As a result, there is an urgent need to develop new therapeutic agents with fewer side effects that can effectively overcome resistance mechanisms. Flavonoids, a prominent class of natural compounds, have shown promise in preventing and treating various cancers. By structurally optimizing flavonoids, it is possible to enhance their anticancer activity and improve their pharmacokinetic properties. This article reviews the different mechanisms of action and structure-activity relationships (SARs) of flavonoid derivatives in treating NSCLC, aiming to provide a scientific foundation for developing new therapeutic agents.
Collapse
Affiliation(s)
- Jiacheng Yao
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Y.); (Y.F.); (C.G.)
- Faculty of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; (T.W.); (X.L.); (H.Y.)
| | - Feng Zhu
- Yangzijiang Pharmaceutical Group Shanghai Haini Pharmaceutical Co., Ltd., Shanghai 201318, China;
| | - Yikun Feng
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Y.); (Y.F.); (C.G.)
- Faculty of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; (T.W.); (X.L.); (H.Y.)
| | - Chen Gu
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Y.); (Y.F.); (C.G.)
- Faculty of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; (T.W.); (X.L.); (H.Y.)
| | - Tianyu Wang
- Faculty of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; (T.W.); (X.L.); (H.Y.)
| | - Xinyu Li
- Faculty of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; (T.W.); (X.L.); (H.Y.)
| | - Hao Yang
- Faculty of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; (T.W.); (X.L.); (H.Y.)
| | - Xiamin Hu
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Y.); (Y.F.); (C.G.)
- Faculty of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; (T.W.); (X.L.); (H.Y.)
| | - Pierre-Antoine Bonnet
- IBMM, Faculty of Pharmacy, Montpellier University, CNRS, ENSCM, 34093 Montpellier, France
| | - Xiangguo Meng
- Faculty of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; (T.W.); (X.L.); (H.Y.)
| |
Collapse
|
31
|
Prata RBDS, Pinheiro RO. Cell Death Mechanisms in Mycobacterium abscessus Infection: A Double-Edged Sword. Pathogens 2025; 14:391. [PMID: 40333197 PMCID: PMC12030298 DOI: 10.3390/pathogens14040391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025] Open
Abstract
Infections caused by non-tuberculous mycobacteria (NTM), such as Mycobacterium abscessus, elicit diverse cell death mechanisms including apoptosis, necrosis, and pyroptosis, which play key roles in immunopathogenesis. NTM can manipulate these cell death pathways to evade host immune responses, ensuring their intracellular survival and persistence. Apoptosis may aid in antigen presentation and immune activation, while necrosis and pyroptosis trigger excessive inflammation, leading to tissue damage. Autophagy, a crucial cellular defense mechanism, is often induced in response to NTM infection; however, M. abscessus has evolved mechanisms to inhibit autophagic processes, enhancing its ability to survive within host cells. This manipulation of cell death pathways, particularly the dysregulation of autophagy and ferroptosis, contributes to chronic infection, immune evasion, and tissue damage, complicating disease management. Understanding these mechanisms offers potential therapeutic targets for improving treatment strategies against M. abscessus infections.
Collapse
Affiliation(s)
| | - Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro 21040-360, Brazil;
| |
Collapse
|
32
|
An RF, Wu KT, Pan J, Zhang WJ, Qin HY, Li XR, Liu W, Huang XF. Design, synthesis and cytotoxic activity of novel lipophilic cationic derivatives of diosgenin and sarsasapogenin. Bioorg Med Chem Lett 2025; 119:130094. [PMID: 39778752 DOI: 10.1016/j.bmcl.2025.130094] [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: 07/27/2024] [Revised: 12/27/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Novel lipophilic cationic derivatives including quaternary ammonium salt and triphenylphosphine series were designed and synthesized using diosgenin (1) and sarsasapogenin (2) as substrates to improve the cytotoxicity and selectivity. Most of the derivatives showed higher cytotoxicity against all cancer cell lines tested, compound 13 exhibited the most superior activity against A549 cells with an IC50 value of 0.95 μM, which was 34-fold of diosgenin. Preliminary cellular mechanism studies elucidated that compound 13 might arrest cell cycle at G0/G1 phase, trigger apoptosis via up-regulating the expression of Bax, down-regulating the expression of Bcl-2 and caspase-3, and induce an increase in the generation of intracellular reactive oxygen species (ROS) in A549 cells. In addition, molecular docking analysis revealed that compound 13 could occupy the active site of p38α-MAPK well and interact to the surrounding amino acids by salt bridge and conjugation. These results suggested that compound 13 had the potential to serve as an antitumor lead agent, probably exert antitumor effect through mitochondrial pathway and p38α MAPK pathway.
Collapse
Affiliation(s)
- Ren-Feng An
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Kai-Tian Wu
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Jie Pan
- Department of Pharmacology, Nanjing Medical University, Nanjing 210029 China
| | - Wen-Jin Zhang
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Hui-Ying Qin
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Xiao-Rui Li
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Wei Liu
- Department of Pharmacology, Nanjing Medical University, Nanjing 210029 China.
| | - Xue-Feng Huang
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009 China.
| |
Collapse
|
33
|
Xiao ZW, Zeng YC, Ji LT, Yuan JT, Li L. Nitric oxide synthase 1 inhibits the progression of esophageal cancer through interacting with nitric oxide synthase 1 adaptor protein. World J Gastrointest Oncol 2025; 17:103843. [PMID: 40235872 PMCID: PMC11995332 DOI: 10.4251/wjgo.v17.i4.103843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Esophageal cancer (ESCA) is among the most prevalent and lethal tumors globally. While nitric oxide synthase 1 (NOS1) is recognized for its important involvement in various cancers, its specific function in ESCA remains unclear. AIM To explore the potential role and underlying mechanisms of NOS1 in ESCA. METHODS Survival rates were analyzed using GeneCards and Gene Expression Profiling Interactive Analysis. The effects and mechanisms of NOS1 on ESCA cells were evaluated via the Cell Counting Kit-8 assay, scratch assay, Transwell assay, flow cytometry, quantitative polymerase chain reaction, western blotting, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling staining. The protein interaction network was used to screen the interacting proteins of NOS1 and validate these interactions through co-immunoprecipitation and dual luciferase assays. Additionally, a nude mouse xenograft model was established to evaluate the effect of NOS1 in vivo. RESULTS The survival rate of patients with ESCA with high NOS1 expression was higher than that of patients with low NOS1 expression. NOS1 expression in ESCA cell lines was lower than that in normal esophageal epithelial cells. Overexpression of NOS1 (oe-NOS1) inhibited proliferation, invasion, and migration abilities in ESCA cell lines, resulting in decreased autophagy levels and increased apoptosis, pyroptosis, and ferroptosis. Protein interaction studies confirmed the interaction between NOS1 and NOS1 adaptor protein (NOS1AP). Following oe-NOS1 and the silencing of NOS1AP, levels of P62 and microtubule-associated protein 1 light chain 3 beta increased both in vitro and in vivo. Furthermore, the expression levels of E-cadherin, along with the activation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (AKT), were inhibited in ESCA cell lines. CONCLUSION NOS1 and NOS1 proteins interact to suppress autophagy, activate the PI3K/AKT pathway, and exert anti-cancer effects in ESCA.
Collapse
Affiliation(s)
- Zi-Wei Xiao
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Ying-Chao Zeng
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Lin-Tao Ji
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Jia-Tao Yuan
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Lin Li
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| |
Collapse
|
34
|
Cui Y, Mei J, Zhao S, Zhu B, Lu J, Li H, Bai B, Sun W, Jin W, Zhu X, Rao S, Yi Y. Identification of a PANoptosis-related long noncoding rna risk signature for prognosis and immunology in colon adenocarcinoma. BMC Cancer 2025; 25:662. [PMID: 40211224 PMCID: PMC11987197 DOI: 10.1186/s12885-025-14021-2] [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: 06/06/2024] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND PANoptosis, a complex programmed cell death (PCD) pathway that includes apoptosis, pyroptosis and necroptosis, is significantly involved in the progression of cancers. Long noncoding RNAs (lncRNAs) play crucial roles in PCD. However, the predictive value of PANoptosis-related lncRNAs (PRlncRNAs) for colon adenocarcinoma (COAD) has not been established. METHODS Gene expression data and clinical characteristics of patients with COAD were obtained from The Cancer Genome Atlas database. Differential expression analysis and Pearson correlation analysis were used to identify PRlncRNAs. In addition to least absolute shrinkage and selection operator, univariate and multivariate Cox regression analyses were employed to obtain PRlncRNAs for constructing a risk signature. Patients with COAD in the training set, testing set and entire set were stratified into high- and low-risk groups for further comparison of survival prognosis, using the median risk score as the cut-off point. Time-dependent receiver operating characteristic curves, a nomogram and multivariate Cox regression analysis were conducted to validate the risk signature in the testing set and the entire set. In addition, critical pathways, immune infiltration cells, immune checkpoint-related genes, Tumor Immune Dysfunction and Exclusion (TIDE) scores and antitumour drugs were compared between the two risk groups in the entire set. Correlations between ferroptosis, cuproptosis, disulfidptosis and the PRlncRNA risk score were evaluated. Finally, a competitive endogenous RNA (ceRNA) network was established, and enrichment analysis of the predicted mRNAs was performed using Gene Ontology (GO) analysis. The Kaplan-Meier plotter database was used as an external database to confirm the accuracy of the risk signature in predicting patient prognosis. Additionally, small interfering RNA (siRNA), a cell counting kit- 8 assay, a cell colony formation assay, quantitative polymerase chain reaction (qPCR) and an apoptosis assay were further employed to investigate the roles of AP003555.1 in colon cancer. RESULTS A risk signature comprising four PRlncRNAs (LINC01133, FOXD3-AS1, AP001066.1, and AP003555.1) was developed to predict the prognosis of patients with COAD. Kaplan‒Meier curves demonstrated significant differences in prognosis between the high- and low-risk groups across the three sets. Multivariate Cox regression analysis confirmed that the risk signature was an independent prognostic factor across the three sets. A nomogram, receiver operating characteristic curves and calibration curves indicated strong confidence in the risk signature. Using the CIBERSORT algorithm and gene set enrichment analysis, variations in infiltrating immune cells and immune processes were observed between the two risk groups. Furthermore, TIDE algorithm suggested that the high-risk group exhibited a lower risk of immunotherapy escape and better immunotherapy outcomes than the low-risk group. Distinct responses to various antitumour drugs were observed between the two risk groups. Additionally, we constructed a ceRNA network based on PRlncRNAs, and GO enrichment analysis of the predicted mRNAs revealed different functions. In addition, the results of the Kaplan‒Meier plotter database revealed that patients who exhibited high levels of LINC01133 and FOXD3-AS1 experienced significantly shorter overall survival than those with low levels of these lncRNAs. Specifically, in terms of functionality, AP003555.1 was found to be highly expressed in colon cancer tissue and promoted viability and proliferation while suppressing the apoptosis of colon cancer cells. CONCLUSION We identified a novel risk signature consisting of four PRlncRNAs, which is an independent prognostic indicator for patients with COAD. This PRlncRNA risk signature is potentially relevant for immunotherapy and could serve as a therapeutic target for COAD.
Collapse
Affiliation(s)
- Yuekai Cui
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Mei
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shengsheng Zhao
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bingzi Zhu
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianhua Lu
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongzheng Li
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Binglong Bai
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weijian Sun
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenyu Jin
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueqiong Zhu
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China.
- Wenzhou Medical University, Wenzhou, China.
| | - Shangrui Rao
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Yongdong Yi
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
35
|
Zou JX, Chang MR, Kuznetsov NA, Kee JX, Babak MV, Ang WH. Metal-based immunogenic cell death inducers for cancer immunotherapy. Chem Sci 2025; 16:6160-6187. [PMID: 40160356 PMCID: PMC11949249 DOI: 10.1039/d4sc08495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunogenic cell death (ICD) has attracted enormous attention over the past decade due to its unique characteristics in cancer cell death and its role in activating innate and adaptive immune responses against tumours. Many efforts have been dedicated to screening, identifying and discovering ICD inducers, resulting in the validation of several based on metal complexes. In this review, we provide a comprehensive summary of current metal-based ICD inducers, their molecular mechanisms for triggering ICD initiation and subsequent protective antitumour immune responses, along with considerations for validating ICD both in vitro and in vivo. We also aim to offer insights into the future development of metal complexes with enhanced ICD-inducing properties and their applications in potentiating antitumour immunity.
Collapse
Affiliation(s)
- Jiao Xia Zou
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Meng Rui Chang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Nikita A Kuznetsov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Jia Xuan Kee
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- NUS Graduate School - Integrative Science and Engineering Programme (ISEP), National University of Singapore 21 Lower Kent Ridge Rd Singapore 119077 Singapore
| |
Collapse
|
36
|
Singh S, Singh V, Singh R, Gouri V, Koch B, Samant M. Synergistic combination of doxorubicin with fisetin for the treatment of lymphoma. Eur J Pharmacol 2025; 992:177361. [PMID: 39929420 DOI: 10.1016/j.ejphar.2025.177361] [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: 09/18/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/25/2025]
Abstract
Lymphoma is a common cancer of the lymphatic system, and its treatment presents considerable clinical difficulties due to the constraints of existing medicines. Anticancer drug such as Doxorubicin (DOX) is an effective chemotherapeutic drug that is frequently used to treat lymphoma and other cancers; however, it is linked with considerable toxicities. Fisetin, a naturally occurring flavonoid, exhibits anticancer properties and has the potential to augment the therapeutic effects of DOX. This study explores the synergistic effects of combining DOX with fisetin in the treatment of lymphoma. The combination of DOX and fisetin significantly inhibits cell viability, induced membrane blabbing, chromatin condensation, and promoted apoptosis compared to monotherapies. The study also showed that the synergistic effect of fisetin along with DOX significantly promotes apoptosis in DL cells through intracellular ROS generation, mitochondrial aggregation at the periphery of the nucleus and, increased p53, Bax, cytochrome c, caspase 3, caspase 9, and cleaved caspase 9 expression. Additionally, combination therapy not only increased the mean survival of the treated group animals but also reduced the tumor burden. While histopathological parameters have shown overall improvement in combination therapy. This study proposes a novel combinational therapy for the treatment of lymphoma and requires further clinical investigation.
Collapse
Affiliation(s)
- Sumeet Singh
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Virendra Singh
- Genotoxicology and Cancer Biology Laboratory, Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Ranjeet Singh
- Department of Zoology, Soban Singh Jeena University (Bageshwar Campus), Almora, Uttarakhand, India; Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Vinita Gouri
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India; Department of Zoology, Kumaun University, Nainital, Uttarakhand, India
| | - Biplob Koch
- Genotoxicology and Cancer Biology Laboratory, Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Mukesh Samant
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India.
| |
Collapse
|
37
|
Bhattacharya R, Avdieiev SS, Bukkuri A, Whelan CJ, Gatenby RA, Tsai KY, Brown JS. The Hallmarks of Cancer as Eco-Evolutionary Processes. Cancer Discov 2025; 15:685-701. [PMID: 40170539 DOI: 10.1158/2159-8290.cd-24-0861] [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: 06/20/2024] [Revised: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/03/2025]
Abstract
SIGNIFICANCE Viewing the hallmarks as a sequence of adaptations captures the "why" behind the "how" of the molecular changes driving cancer. This eco-evolutionary view distils the complexity of cancer progression into logical steps, providing a framework for understanding all existing and emerging hallmarks of cancer and developing therapeutic interventions.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Cancer Biology, University of South Florida, Tampa, Florida
| | - Stanislav S Avdieiev
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anuraag Bukkuri
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J Whelan
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joel S Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
38
|
Christian Y, Redkar AS, Kumar N, Jancy SV, Chandrasekharan A, Retnabai Santhoshkumar T, Ramakrishnan V. Structural regression modelling of peptide based drug delivery vectors for targeted anti-cancer therapy. Drug Deliv Transl Res 2025; 15:1284-1298. [PMID: 39117921 DOI: 10.1007/s13346-024-01674-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2024] [Indexed: 08/10/2024]
Abstract
Drug resistance in cancer poses a serious challenge in finding an effective remedy for cancer patients, because of the multitude of contributing factors influencing this complex phenomenon. One way to counter this problem is using a more targeted and dose-limiting approach for drug delivery, rather than relying on conventional therapies that exhibit multiple pernicious side-effects. Stability and specificity have traditionally been the core issues of peptide-based delivery vectors. In this study, we employed a structural regression modelling approach in the design, synthesis and characterization of a series of peptides that belong to approximately same topological cluster, yet with different electrostatic signatures encoded as a result of their differential positioning of amino acids in a given sequence. The peptides tagged with the fluorophore 5(6)-carboxyfluorescein, showed higher uptake in cancer cells with some of them colocalizing in the lysosomes. The peptides tagged with the anti-cancer drug methotrexate have displayed enhanced cytotoxicity and inducing apoptosis in triple-negative breast cancer cells. They also showed comparable uptake in side-population cells of lung cancer with stem-cell like properties. The most-optimized peptide showed accumulation in the tumor resulting in significant reduction of tumor size, compared to the untreated mice in in-vivo studies. Our results point to the following directives; (i) peptides can be design engineered for targeted delivery (ii) stereochemical engineering of peptide main chain can resist proteolytic enzymes and (iii) cellular penetration of peptides into cancer cells can be modulated by varying their electrostatic signatures.
Collapse
Affiliation(s)
- Yvonne Christian
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Amay Sanjay Redkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Naveen Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Shine Varghese Jancy
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | - Aneesh Chandrasekharan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | | | - Vibin Ramakrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
- Mehta Family School of Data Science & Artificial Intelligence, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
39
|
Xie Q, Sun T, Zhang L, Gong M, Zhang W, Liu X, Zhao Y, Wang M, Yang X, Zhang Z, Liu G, Zhou C, Zhang D. Responsive plasmonic hybrid nanorods enables metabolism reprogramming via cuproptosis-photothermal combined cancer therapy. Biomaterials 2025; 315:122971. [PMID: 39577035 DOI: 10.1016/j.biomaterials.2024.122971] [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: 08/08/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Abnormal tumor metabolism leads to tumor growth, metastasis, and recurrence, reprogramming tumor metabolism and activating potent anti-tumor immune response have been demonstrated to have good therapeutic effects on tumor elimination. Copper-based nanomaterials involved in cuproptosis show great prospects in these two aspects, but their efficiency is restricted by Cu homeostasis and the toxicity of the chelator. Here, the pH-responsive AuNRs@Cu2O core-shell plasmonic hybrid nanorods (ACNRs) have been successfully fabricated to realize microenvironment-controlled release at the tumor site for the combined therapy of cuproptosis and photothermal treatment. The AuNRs core exhibited excellent NIR-II photothermal property, which boost the intracellular concentration of copper to trigger severe cuproptosis and induce immunogenic cell death of tumor cells. In vivo studies demonstrated the ACNR exhibited efficient tumor therapy for primary, metastatic, and recurrent tumors. ACNRs-induced cuproptosis and PTT were capable of reprogramming energy metabolism, leading to a decreased production of lactic acid. This potential of metabolic reprogramming assisted in reshaping the immunosuppressive tumor microenvironment to facilitate the infiltration of immune cells and boost the immune responses triggered by PTT. The therapeutic mechanism was further verified by metabolomics analysis, which indicated that ACNRs + PTT treatment led to the inhibition of the Pentose Phosphate Pathway and Glycolysis pathways in tumor cells. The suppression of glycolytic reduced ATP synthesis, thereby hindering energy-dependent copper efflux, which in turn promoted cuproptosis. Taken together, this study offers promising insights for cuproptosis-based cancer treatment and sheds new light on nanomedicine-mediated metabolic modulation for future tumor therapy.
Collapse
Affiliation(s)
- Qian Xie
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Tao Sun
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China; Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Mingfu Gong
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Wansu Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xu Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yue Zhao
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Miaomiao Wang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xiaofeng Yang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Zhipeng Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chunyu Zhou
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - Dong Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
40
|
Wang Y, Sun X, Ren M, Ma F, Zhao R, Zhu X, Xu Y, Cao N, Chen Y, Pan Y, Zhao A. Integrative network pharmacology, transcriptomics, and proteomics reveal the material basis and mechanism of the Shen Qing Weichang Formula against gastric cancer. Chin Med 2025; 20:42. [PMID: 40155922 PMCID: PMC11954191 DOI: 10.1186/s13020-025-01091-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common malignancy with poor prognosis and lack of efficient therapeutic methods. Shen Qing Weichang Formula (SQWCF) is a patented traditional herbal prescription for GC, but its efficacy and underlying mechanism remains to be clarified. PURPOSE To explore the efficacy and potential mechanism of SQWCF in treating GC. METHODS A subcutaneous transplantation tumor model of human GC was established for assessing SQWCF's efficacy and safety. A comprehensive strategy integrating mass spectrometry, network pharmacology, omics analysis, and bioinformatic methods was adopted to explore the core components, key targets, and potential mechanism of SQWCF in treating GC. Molecular docking, immunohistochemistry, quantitative real-time PCR, and western blot were applied to validation. RESULTS In the mouse model of GC, SQWCF effectively suppressed the GC growth without evident toxicity and enhanced the therapeutic efficacy of paclitaxel. Network pharmacology and molecular docking based on mass spectrometry showed that key targets (CASP3, TP53, Bcl-2, and AKT1) and core active components (Calycosin, Glycitein, Liquiritigenin, Hesperetin, and Eriodictyol) involved in the anti-GC effect of SQWCF had stable binding affinity, of which AKT1 ranked the top in the affinity. Validation based on network pharmacology and omics analysis confirmed that PI3K-AKT and MAPK signaling pathways, as well as downstream apoptosis pathway, explained the therapeutic effects of SQWCF on GC. In addition, family with sequence similarity 81 member A (FAM81A) was identified as a novel biomarker of GC that was aberrantly highly expressed in GC and associated with poor prognosis by bioinformatic analysis, and was an effector target of SQWCF at both mRNA and protein levels. CONCLUSION This study uncovers a synergistic multi-component, multi-target, and multi-pathway regulatory mechanism of SQWCF in treating GC comprehensively, emphasizing its potential for therapeutic use and providing new insights into GC treatment.
Collapse
Affiliation(s)
- Yi Wang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Xiaoyu Sun
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Mingming Ren
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Fangqi Ma
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Ruohan Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Xiaohong Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yan Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Nida Cao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yuanyuan Chen
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yongfu Pan
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China.
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China.
| |
Collapse
|
41
|
Wang Y, Yang X, Yang Z, Chen Z, Jiang H, Wang Y, Shen D, Su G. RBM39 Functions as a Potential Oncogene Through the NF-κB Signaling Pathway in Colorectal Cancer Cells. J Cancer 2025; 16:2233-2249. [PMID: 40302803 PMCID: PMC12036102 DOI: 10.7150/jca.105120] [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: 10/13/2024] [Accepted: 03/02/2025] [Indexed: 05/02/2025] Open
Abstract
Colorectal cancer (CRC) ranks as the third most frequently diagnosed cancer and is the second leading cause of cancer-related deaths globally. Recently, RNA-binding protein 39(RBM39), a critical factor in tumor-targeted mRNA and protein expression, has played a vital role in tumorigenesis and has broad development prospects in clinical treatment and drug research. However, the functional roles of RBM39 in the progression of CRC remain largely unexplored. This study found that RBM39 is notably overexpressed at both the mRNA and protein levels in CRC tissues compared with normal adjacent tissues. RBM39 was identified as a potential therapeutic target for colorectal cancer. Elevated RBM39 mRNA levels in CRC patients indicated worse survival probabilities. We show that RBM39 enhances the proliferation, migration, and invasion ability of CRC cells. Furthermore, we have made an innovative discovery that increased RBM39 inhibits apoptosis in CRC cells. Mechanistically, RNA-seq analysis indicated that RBM39 activates the NF-κB pathway, which plays a pivotal role in driving the malignant biological behaviors of colorectal cancer. Notably, these findings represent a novel contribution to our understanding of the mechanistic underpinnings of CRC, as they have not been previously documented in the literature. In the in vivo nude mouse xenograft model, our study demonstrates that the targeted knockdown of RBM39 markedly suppresses tumor formation, highlighting a novel therapeutic strategy for combating colorectal cancer. In conclusion, RBM39 emerges as a promising candidate for clinical diagnosis and targeted treatment of colorectal cancer, with implications for future research in tumor biology and therapeutic strategies.
Collapse
Affiliation(s)
- YaTao Wang
- Department of Colorectal Tumor Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen,361003, Fujian Province, China
- The Sixth People's Hospital of Luoyang, 471003, Luoyang, Henan Province, China
| | - XueSi Yang
- Department of Colorectal Tumor Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen,361003, Fujian Province, China
| | - ZhangQuan Yang
- Department of Colorectal Tumor Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen,361003, Fujian Province, China
| | - ZiRui Chen
- Department of Colorectal Tumor Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen,361003, Fujian Province, China
| | - HaiFeng Jiang
- Department of Critical Care Medicine, Second People's Hospital of Yibin City, Yibin, 644000, Sichuan Province, China
| | - YiCong Wang
- Gastrointestinal Oncology, The Affiliated Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - DongYan Shen
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian Province, China
| | - GuoQiang Su
- Department of Colorectal Tumor Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen,361003, Fujian Province, China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, 350122, China
| |
Collapse
|
42
|
Fan H, Liu J, Hu X, Cai J, Su B, Jiang J. The critical role of X-linked inhibitor of apoptosis protein (XIAP) in tumor development. Apoptosis 2025:10.1007/s10495-025-02101-4. [PMID: 40146486 DOI: 10.1007/s10495-025-02101-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2025] [Indexed: 03/28/2025]
Abstract
X-linked inhibitor of apoptosis protein (XIAP) is the most potent endogenous member of the inhibitor of apoptosis protein family. XIAP exerts its anti-apoptotic effects by inhibiting both the death receptor pathway and mitochondrial pathway of apoptosis through various mechanisms such as directly binding to caspases, activating the nuclear factor kappa B (NF-κB) pathway, and other signaling pathways. These processes are closely related to tumor development and progression, making XIAP a therapeutic target for various types of cancer. This article will first review the structural characteristics and biological functions of XIAP, followed by its effects on tumors and an overview of XIAP-targeted inhibitors.
Collapse
Affiliation(s)
- Hui Fan
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiyuan Liu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiangyan Hu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiye Cai
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Bo Su
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jinhuan Jiang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
43
|
Ying L, Chen R, Guo R, Liang Y, Hao M, Chen X, Zhang W, Yu C, Yang Z. Paeonol Suppresses Bladder Cancer Progression via Apoptotic Pathways: Insights from In Vitro and In Vivo Studies. Pharmaceuticals (Basel) 2025; 18:472. [PMID: 40283909 PMCID: PMC12030738 DOI: 10.3390/ph18040472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Bladder cancer (BC), a highly heterogeneous and mutation-prone malignancy, remains a significant therapeutic challenge due to its propensity for recurrence, metastasis, and drug resistance. Natural products, particularly paeonol, a bioactive compound derived from Moutan Cortex in traditional Chinese medicine, have shown promising potential in cancer therapy. This study aims to evaluate the anti-BC effects of paeonol and elucidate its underlying molecular mechanisms. Methods: In vitro experiments were conducted using T24 and J82 BC cell lines to assess paeonol's effects on cell viability, migration, apoptosis, and cell cycle progression via CCK-8, scratch, flow cytometry, RT-qPCR, and Western blot analyses. In vivo efficacy was evaluated using a xenograft mouse model, with tumor growth monitored and histopathological analysis performed. Results: Paeonol significantly inhibited BC cell proliferation and migration in a dose- and time-dependent manner, with IC50 values of 225 μg/mL (T24) and 124 μg/mL (J82) at 48 h. It induced apoptosis and arrested the cell cycle at the G1 phase, accompanied by upregulation of pro-apoptotic proteins (BID, BAX, BIM, and p53). In vivo, paeonol reduced tumor volume and weight without histopathological abnormalities in vital organs. Conclusions: Paeonol exhibits potent anti-BC activity by apoptotic pathways and by arresting the cell cycle at the G1 phase and inhibiting tumor growth. Its favorable safety profile and multi-target mechanisms highlight its potential as a promising therapeutic candidate for BC. These findings provide a foundation for further clinical development of paeonol-based therapies.
Collapse
Affiliation(s)
- Lu Ying
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
- College of Life Science and Technology, State Key Laboratory Incubation Base for Conservation and Utilization of Bio-Resource in Tarim Basin, Tarim University, Alar 843300, China
| | - Ruolan Chen
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
| | - Rui Guo
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
| | - Youfeng Liang
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
| | - Mingxuan Hao
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
| | - Xiaoyang Chen
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
| | - Wenjing Zhang
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
| | - Changyuan Yu
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
| | - Zhao Yang
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China; (L.Y.); (R.C.); (R.G.); (Y.L.); (M.H.); (X.C.); (W.Z.)
- College of Life Science and Technology, State Key Laboratory Incubation Base for Conservation and Utilization of Bio-Resource in Tarim Basin, Tarim University, Alar 843300, China
| |
Collapse
|
44
|
Fujimoto S, Endo M, Tonomura S, Tsuji F, Haraguchi H, Hasegawa K, Numao T, Izumi A, Tourtas T, Schlötzer-Schrehardt U, Kruse F, Oyama Y, Ikawa M, Jun AS, Koizumi N, Okumura N. Therapeutic Potential of Emricasan, a Pan-Caspase Inhibitor, in Reducing Cell Death and Extracellular Matrix Accumulation in Fuchs Endothelial Corneal Dystrophy. Cells 2025; 14:498. [PMID: 40214452 PMCID: PMC11988121 DOI: 10.3390/cells14070498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Fuchs endothelial corneal dystrophy (FECD) is a progressive disorder characterized by endothelial cell loss and excessive extracellular matrix (ECM) accumulation leading to corneal dysfunction. Emricasan, a pan-caspase inhibitor, was investigated for its therapeutic potential in suppressing these pathological changes. Patient-derived FECD cells and stress-induced cell models were treated with emricasan to assess its effects on apoptosis and ECM production. Caspase-specific knockdown experiments were performed to identify key mediators. Col8a2Q455K/Q455K mice, model mice of early-onset FECD, received twice-daily administration of 0.1% emricasan eye drops from 8 to 28 weeks of age. Endothelial cell density, hexagonality, cell size variation, and guttae area were evaluated by contact specular microscopy, while transcriptomic changes were analyzed via RNA sequencing. Emricasan effectively reduced apoptosis and ECM production in vitro by selectively inhibiting caspase-7 without affecting canonical TGF-β signaling. In vivo, emricasan-treated mice exhibited significantly higher endothelial cell density, improved hexagonality, and reduced variation in cell size compared with controls. Transcriptome analysis revealed distinct gene expression changes in the corneal endothelium following emricasan treatment. These findings suggest that emricasan exerts dual protective effects by inhibiting caspase-7-mediated ECM accumulation and broadly suppressing apoptosis, highlighting its potential as a pharmacological therapy for FECD.
Collapse
Affiliation(s)
- Sohya Fujimoto
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| | - Mako Endo
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| | - Shigehito Tonomura
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| | - Fuuga Tsuji
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| | - Hirotaka Haraguchi
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| | - Kanna Hasegawa
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| | - Taisuke Numao
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| | | | - Theofilos Tourtas
- Department of Ophthalmology, University of Erlangen-Nürnberg, 430074 Erlangen, Germany
| | | | - Friedrich Kruse
- Department of Ophthalmology, University of Erlangen-Nürnberg, 430074 Erlangen, Germany
| | - Yuki Oyama
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita 565-0871, Japan
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Masahito Ikawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita 565-0871, Japan
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Albert S. Jun
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Noriko Koizumi
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| | - Naoki Okumura
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 602-8580, Japan
| |
Collapse
|
45
|
Abid J, Al-Rawi MBA, Mahmood A, Li A, Jiang T. Identification and functional characterization of key biomarkers in diffuse large B-cell lymphoma: emphasis on STYX as a prognostic marker and therapeutic target. Hereditas 2025; 162:45. [PMID: 40128844 PMCID: PMC11931869 DOI: 10.1186/s41065-025-00411-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBC) is the most common subtype of non-Hodgkin lymphoma, characterized by its aggressive nature and poor prognosis in advanced stages. Despite advances in treatment, the molecular mechanisms driving DLBC progression remain incompletely understood, necessitating the identification of novel biomarkers for diagnosis and prognosis. In this study, we analyzed two publicly available datasets (GSE32018 and GSE56315) from the Gene Expression Omnibus database (GEO) to identify overlapping differentially expressed genes (DEGs). Later on, a comprehensive in silico and in vitro methodology was adopted to decipher the role of identify DEGs in DLBC. DEGs analysis of GSE32018 and GSE56315 datasets identified five overlapping gene: SP3, CSNK1A1, STYX, SIRT5, and MGEA5. Expression validation using the GEPIA2 database confirmed the upregulation of SP3, CSNK1A1, STYX, and SIRT5, and the downregulation of MGEA5 in DLBC tissues compared to normal controls. Furthermore, mutational analysis revealed that CSNK1A1 was the only gene among these DEGs to exhibit mutations, with a 2.7% mutation frequency in DLBC patients. Methylation analysis highlighted a negative correlation between DEGs methylation levels and mRNA expression, while survival analysis identified high STYX expression as significantly associated with poorer overall survival in DLBC patients. Functional assays demonstrated that STYX knockdown in U2932 cells led to reduced cell proliferation, colony formation, and enhanced wound healing, indicating STYX's pivotal role in DLBC cell survival and migration. Additionally, gene enrichment analysis revealed the involvement of these DEGs in key biological processes, including intracellular trafficking and myeloid progenitor cell differentiation. These findings emphasize the potential of SP3, CSNK1A1, STYX, SIRT5, and MGEA5 as biomarkers and therapeutic targets in DLBC, particularly highlighting STYX as a promising prognostic marker and potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Junaid Abid
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300222, China
| | - Mahmood Basil A Al-Rawi
- Department of Optometry, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Ahmad Mahmood
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - An Li
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Urumqi, 830054, China.
| | - Tiemin Jiang
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Urumqi, 830054, China.
| |
Collapse
|
46
|
Ouyang Y, Cao L, Zhao Q, Yang W, Lin C. Biodegradable Mg-1%Ca alloy inhibits the growth of cervical cancer. Biomed Mater 2025; 20:035002. [PMID: 39908673 DOI: 10.1088/1748-605x/adb2cc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/05/2025] [Indexed: 02/07/2025]
Abstract
The traditional treatment for cervical cancer involves aggressive surgery combined with radiotherapy and chemotherapy. Nevertheless, these treatments have certain limitations and side effects, thus breakthroughs and advances are required in cervical cancer therapy. Magnesium alloy is a promising antitumor biomaterial with excellent biocompatibility and biodegradability. However, the potential effects of magnesium alloy on cervical tumors have not been extensively explored. Recent studies have demonstrated that adding a small amount of calcium to the magnesium matrix can reduce grain size and corrosion rate while providing good biocompatibility. We conductedin vivoandin vitroexperiments to test the antitumor properties of Mg-1%Ca alloys. The results indicated that the Mg-1%Ca alloy released Mg2+and OH-more slowly, inhibited the proliferation of SiHa and HeLa cells, induced apoptosis in tumor cells, disrupted the cytoskeleton, and inhibited cell migration and invasion. At the molecular level, Mg-1%Ca alloy significantly activated the mitochondrial apoptosis pathway and inhibited the MAPK/ERK signaling pathway. In the future, Mg-1%Ca may be employed in the treatment of cervical cancer as a novel adjuvant therapeutic material with anticancer function to prevent the occurrence and progression of cancer proliferation and metastasis.
Collapse
Affiliation(s)
- Yunshan Ouyang
- Department of Pathology, School of Basic Medical Sciences
- Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang Medical University, Urumqi 830017, People's Republic of China
| | - Lingling Cao
- Department of Pathology, School of Basic Medical Sciences
- Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang Medical University, Urumqi 830017, People's Republic of China
| | - Qian Zhao
- Department of Pathology, School of Basic Medical Sciences
- Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang Medical University, Urumqi 830017, People's Republic of China
| | - Wang Yang
- Department of Pathology, School of Basic Medical Sciences
- Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang Medical University, Urumqi 830017, People's Republic of China
| | - Chen Lin
- Department of Pathology, School of Basic Medical Sciences
| |
Collapse
|
47
|
Chen J, Ji Z, Wu D, Wei S, Zhu W, Peng G, Hu M, Zhao Y, Wu H. MYBL2 promotes cell proliferation and inhibits cell apoptosis via PI3K/AKT and BCL2/BAX/Cleaved-caspase-3 signaling pathway in gastric cancer cells. Sci Rep 2025; 15:9148. [PMID: 40097530 PMCID: PMC11914465 DOI: 10.1038/s41598-025-93022-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
The transcription factor MYB proto-oncogene like 2 (MYBL2) has been reported to be involved in the occurrence and development of various tumors, however, its role in gastric cancer (GC) remains to be elucidated. In this study, the Kaplan-Meier plotter was used to evaluate the prognostic value of different MYBL2 expression levels in GC patients. The UALCAN database were applied to analyze the relationships between MYBL2 and clinicopathological characteristics of GC. GC cell proliferation, cell cycle and apoptosis were determined by CCK-8 and flow cytometry assays, and proteins were examined by Western blot analysis. Next, signaling pathway enrichment analysis of MYBL2-related genes and protein expression were analyzed by Gene Set Enrichment Analysis (GSEA) and Western blot assays. The results found that MYBL2 expression was significantly upregulated in GC compared with adjacent non-malignant tissues and associated with poor patient survival, tumor, stages and lymph node metastasis. Forced expression of MYBL2 could promote cell proliferation, resulting in an accelerated S phase progression and inhibiting cell apoptosis in GC cells. Conversely, MYBL2 silencing inhibited cell proliferation, induced G2/M phase arrest and promoted cell apoptosis in GC cells. Mechanistically, Western blot analysis showed that MYBL2 silencing decreased the expression of BCL2 and upregulated the expression of Cleaved-caspase-3 and BAX in HGC-27 cells. Conversely, MYBL2 overexpression in AGS cells resulted in the opposite effects. Furthermore, enforced expression of MYBL2 activated the PI3K/AKT signaling pathway, especially AKT phosphorylation. Additionally, the AKT inhibitor MK2206 significantly reversed the proliferation capacity of GC cells induced by MYBL2 overexpression. Therefore, these results suggest that upregulated expression of MYBL2 contributes to GC cell growth and inhibits cell apoptosis by regulating the PI3K/AKT and BCL2/BAX/Cleaved-caspase-3 signaling pathways in GC cells indicating that MYBL2 may be a new therapeutic target and prognostic marker for GC.
Collapse
Affiliation(s)
- Jingya Chen
- School of Public Health, Bengbu Medical University, Bengbu, China
| | - Zhenglei Ji
- School of Public Health, Bengbu Medical University, Bengbu, China
| | - Di Wu
- School of Life Science, Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, China
| | - Siyang Wei
- School of Public Health, Bengbu Medical University, Bengbu, China
| | - Wanjing Zhu
- School of Life Science, Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, China
| | - Guisen Peng
- School of Life Science, Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, China
| | - Mingjie Hu
- School of Life Science, Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, China.
| | - Yunli Zhao
- School of Public Health, Bengbu Medical University, Bengbu, China.
| | - Huazhang Wu
- School of Life Science, Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
48
|
An Y, Sun JX, Ma SY, Xu MY, Xu JZ, Liu CQ, Wang SG, Xia QD. From Plant Based Therapy to Plant-Derived Vesicle-Like Nanoparticles for Cancer Treatment: Past, Present and Future. Int J Nanomedicine 2025; 20:3471-3491. [PMID: 40125436 PMCID: PMC11927496 DOI: 10.2147/ijn.s499893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer stands as a formidable malady profoundly impacting human health. Throughout history, plant-based therapies have remained pivotal in the arsenal against cancer, evolving alongside the epochs. Presently, challenges such as the arduous extraction of active components and potential safety concerns impede the progression of plant-based anticancer therapies. The isolation of plant-derived vesicle-like nanoparticles (PDVLNs), a kind of lipid bilayer capsules isolated from plants, has brought plant-based anticancer therapy into a novel realm and has led to decades of research on PDVLNs. Accumulating evidence indicates that PDVLNs can deliver plant-derived active substances to human cells and regulate cellular functions. Regulating immunity, inducing cell cycle arrest, and promoting apoptosis in cancer cells are the most commonly reported mechanisms of PDVLNs in tumor suppression. Low immunogenicity and lack of tumorigenicity make PDVLNs a good platform for drug delivery. The molecules within the PDVLNs are all from source plants, so the selection of source plants is crucial. In recent years, there has been a clear trend that the source plants have changed from vegetables or fruits to medicinal plants. This review highlights the mechanisms of medicinal plant-based cancer therapies to identify candidate source plants. More importantly, the current research on PDVLN-based cancer therapy and the applications of PDVLNs for drug delivery are systematically discussed.
Collapse
Affiliation(s)
- Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Si-Yang Ma
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
49
|
Deng YR, Wu QZ, Zhang W, Jiang HP, Xu CQ, Chen SC, Fan J, Guo SQ, Chen XJ. Apoptotic cell-derived extracellular vesicles-MTA1 confer radioresistance in cervical cancer by inducing cellular dormancy. J Transl Med 2025; 23:328. [PMID: 40087679 PMCID: PMC11908104 DOI: 10.1186/s12967-025-06350-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Radioresistance presents a major challenge in the treatment of cervical cancer (CC). Apoptotic tumor cells can create an "onco-regenerative niche," contributing to radioresistance. However, the intercellular signaling mechanisms mediating the transfer of radioresistance from apoptotic to surviving cancer cells remain unclear. METHODS The role of apoptotic tumor cell-derived extracellular vesicles (apoEVs) in mediating radioresistance was investigated through integrated bioinformatics and experimental approaches. The GSE236738 dataset was analyzed to identify potential regulators, with subsequent validation of apoEV-MTA1 function using in vitro and in vivo models. Mechanistic studies focused on caspase-3 activation, p-STAT1 signaling pathway, and dormancy-associated protein networks. Furthermore, therapeutic strategies targeting MTA1 and its downstream signaling were evaluated for radiosensitization potential. RESULTS MTA1 was identified as a critical factor enriched in and transferred by apoEVs from apoptotic tumor cells to neighboring CC cells. Caspase-3 activation facilitated the nuclear export and encapsulation of MTA1 in apoEVs. Transferred MTA1 retained transcriptional activity, activated the p-STAT1 signaling pathway, and induced cellular dormancy via NR2F1, a key dormancy regulator, resulting in increased radioresistance. Knockdown of MTA1 in apoEVs or inhibition of p-STAT1 in recipient cells enhanced radiosensitivity. Furthermore, apoEV-MTA1 promoted tumor radioresistance and reduced survival rates in irradiated cervical cancer mouse model. CONCLUSIONS This study demonstrates that apoEV-MTA1 confers radioresistance in CC by promoting cellular dormancy via the p-STAT1/NR2F1 signaling axis. Targeting this pathway could improve radiosensitivity and provide a promising therapeutic strategy for CC patients.
Collapse
Affiliation(s)
- Yuan-Run Deng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, P.R. China
| | - Qiao-Zhi Wu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, P.R. China
| | - Wan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Radiation Oncology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523059, China
| | - Hui-Ping Jiang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, P.R. China
| | - Cai-Qiu Xu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, P.R. China
| | - Shao-Cheng Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, P.R. China
| | - Jing Fan
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, P.R. China
| | - Sui-Qun Guo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, P.R. China.
| | - Xiao-Jing Chen
- Department of Gynecology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, P.R. China.
| |
Collapse
|
50
|
Yang J, Wang F, Hu Z, Liu X, Zhang W, Li C, Wang W, Reaila J, Zhang X, Zhu G, Tian F, Chen B, Zhu X. Huachansu Injection induces ferroptosis in multiple myeloma through NRF2/HO-1 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119454. [PMID: 39922329 DOI: 10.1016/j.jep.2025.119454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Multiple myeloma (MM) is a common hematologic malignancy. Huachansu (HCS) is extracted from the skin of Bufo bufo gargarizans, known for its well-established and multi-target anti-tumor effect. It has been reported to be effective in treating patients with multiple myeloma but its underlying mechanism remains unclear. AIM OF THE STUDY This study aims to investigate the cellular and molecular mechanisms in which HCS induces cell death of MM. MATERIALS AND METHODS Cell viability was assessed using the CCK-8 assay. The effect of HCS on the gene expression of MM were screened by transcriptome sequencing and validated by quantitative real-time PCR, Western blot, and immunofluorescence. The ferroptosis phenotype were evaluated by measuring iron ion concentration, lipid peroxidation degree in terms of malondialdehyde (MDA), and reduced glutathione (GSH) level. Flow cytometry was adopted to measure intracellular ROS and PGSK levels. The ability of ferroptosis inhibitors to reverse these effects was also assessed. The treatment effect and ferroptosis induction of HCS on MM in vivo were explored on a xenograft nude mice model, with mitochondrial damage observed by transmission electron microscopy. RESULTS HCS modulated the NRF2/HO-1 pathway, upregulating PRP and ZIP8, leading to Fe2+ accumulation and PGSK elevation, while increasing ROS and MDA levels and reducing GSH content. These effects were significantly reversed by the ferroptosis inhibitor Ferrostin-1. HCS induced MM cell ferroptosis through the NRF2/HO-1 pathway in vivo, inhibiting MM progression similarly to the positive control drug bortezomib. CONCLUSION These results indicate that HCS can induce ferroptosis in MM cells via the NRF2/HO-1 pathway, thereby controlling MM progression. Our study provides a solid theoretical basis for the clinical use of HCS in treating MM. Additionally, it suggests an innovative treatment alternative based on natural medicine, proposing the combined use of HCS and chemotherapy drugs as a new therapeutic avenue for MM.
Collapse
Affiliation(s)
- Jing Yang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Fengnan Wang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Zhongxiao Hu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; Lianshui Hospital of Traditional Chinese Medicine, Huaian, Jiangsu, 223400, China
| | - Xixi Liu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Weiguang Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Chencheng Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; Wisdom Lake Academy of Pharmacy, Xi' an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
| | - Wanxia Wang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Jianati Reaila
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Xiaoli Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Guangrong Zhu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Fang Tian
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Biqing Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Xuejun Zhu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|