1
|
Chen J, Li M, Mao J, Wang X, Guo B, Chen X, Kalvakolanu DV, Hong J, Yang M, Liu J, Luo Q, Yang J, Sun X, Tian Y, Zhang L. Transglutaminase 2 promotes breast cancer cell autophagy by targeting p53/ mTOR axis. Biochem Pharmacol 2025; 237:116926. [PMID: 40216264 DOI: 10.1016/j.bcp.2025.116926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/22/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025]
Abstract
Breast cancer (BC) is the most common malignant tumor among women. There is a pressing demand to develop new therapeutic strategies for advanced BC. In this report, we investigated whether Transglutaminase 2 (TGM2) is involved in BC growth. Moreover, a high expression of TGM2 in BC was found to be correlated with a poor prognosis. Thus, TGM2 might be a prospective therapeutic target in BC. We examined if a small molecule inhibitor of TGM2, GK921, could suppress BC growth. GK921 inhibited TGM2 activity and induced autophagy-dependent cell death in BC cell lines. Additionally, we found that GK921 increased p53 levels, which decreased the expression of mTOR, a classical upstream regulator of autophagy. Finally, we found that down regulation of TGM2 using RNAi activated autophagy. Consistent with these findings, GK921 significantly suppressed breast tumor growth in mouse models, suggesting that TGM2 may be a potential therapeutic target for BC treatment.
Collapse
Affiliation(s)
- Jiasi Chen
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China
| | - Mengxin Li
- Department of Breast Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| | - Juanjuan Mao
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China
| | - Xuanzhong Wang
- Department of Radiation Oncology, First Hospital of Jilin University, Changchun, China
| | - Baofeng Guo
- Department of Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuyang Chen
- School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Dhan V Kalvakolanu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jinghui Hong
- Department of Breast Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| | - Mei Yang
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China
| | - Jixuan Liu
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China
| | - Qian Luo
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China
| | - Jiaying Yang
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China
| | - Xinze Sun
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China
| | - Yong Tian
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China
| | - Ling Zhang
- College of Basic Medical Sciences, the Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, Jilin University, Changchun 130021, China.
| |
Collapse
|
2
|
McCabe M, Bhattacharyya R, Sereda R, Santiago-Fernández O, Khawaja RR, Diaz A, Lindenau K, Ozturk DG, Garner TP, Sidoli S, Cuervo AM, Gavathiotis E. Small molecule disruption of RARα/NCoR1 interaction inhibits chaperone-mediated autophagy in cancer. EMBO Mol Med 2025:10.1038/s44321-025-00254-y. [PMID: 40490560 DOI: 10.1038/s44321-025-00254-y] [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: 07/24/2024] [Revised: 05/10/2025] [Accepted: 05/16/2025] [Indexed: 06/11/2025] Open
Abstract
Chaperone-mediated autophagy (CMA), a type of selective degradation of cytosolic proteins in lysosomes, is commonly upregulated in cancer cells, contributing to their survival and growth. The lack of a specific target for CMA inhibition has limited CMA blockage to genetic manipulations or global lysosomal function inhibition. Here, using genetic modulation, transcriptional analysis, and functional studies, we demonstrate a regulatory role for the interaction of the retinoic acid receptor alpha (RARα) and its corepressor, the nuclear receptor corepressor 1 (NCoR1), on CMA in non-small cell lung cancer (NSCLC). By targeting the disruption of the NCoR1/RARα complex with a structure-based screening strategy, we identified compound CIM7, a potent and selective CMA inhibitor that has no effect on macroautophagy. CIM7 preferentially inhibits CMA in NSCLC cells over normal cells, reduces tumor growth in NSCLC cells, and demonstrates efficacy in an in vivo xenograft mouse model with no observed toxicity in blood or major tissues. These findings reveal a druggable mechanism for selective CMA inhibition and a first-in-class CMA inhibitor as a potential therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Mericka McCabe
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Rajanya Bhattacharyya
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Rebecca Sereda
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Olaya Santiago-Fernández
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Rabia R Khawaja
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Kristen Lindenau
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Deniz Gulfem Ozturk
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Thomas P Garner
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Evripidis Gavathiotis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
3
|
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
|
4
|
Ju J, Cheng C, Cui L, Hong B, Zhan Q, Wang Q, Cui X, Su D, Huang Y, Kang C. EPIC-1042 alleviates cerebral ischemic/reperfusion injury through TAX1BP1-induced mitophagy. Free Radic Biol Med 2025; 232:367-381. [PMID: 40107570 DOI: 10.1016/j.freeradbiomed.2025.03.023] [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: 01/09/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Post ischemia-reperfusion (I/R) injury, an upregulation in Polymerase I and transcript release factor (PTRF) expression is observed. PTRF is implicated in the regulation of various cellular processes within neuronal cells, thereby exacerbating the deleterious effects of I/R injury. EPIC-1042 is a small molecule pharmacological agent that exhibits specificity in binding to PTRF. Therefore, this study aimed to explore whether EPIC-1042 could be used as a treatment for I/R injury. To achieve this goal, we observed brain injury in mice following EPIC-1042 pre-administration, and then transitioned to therapeutic administration. After observing the pre-protective and therapeutic effects of the drug, proteomic analysis revealed that the expression of TAX1BP1 continued to decline in a time-dependent manner, while EPIC-1042 was able to inhibit this decline. However, the function of TAX1BP1 in ischemic stroke is not yet fully understood. Subsequent experiments confirmed that the addition of EPIC-1042 resulted in an enhancement of mitophagy. Silencing the expression of TAX1BP1 abrogated the drug's effects, indicating that EPIC-1042 exerts a protective function by promoting mitophagy via TAX1BP1 mediation. We further investigated the synergistic effects of EPIC-1042 and edaravone by administering the two drugs in combination, observing an enhanced therapeutic efficacy compared to the administration of each drug alone. Subsequently, we optimized the administration protocol for the two drugs by utilizing liposome encapsulation for both drugs. This approach enabled us to achieve significant therapeutic outcomes while reducing both the dosage and frequency of administration, thereby demonstrating the potential for clinical translation of EPIC-1042.
Collapse
Affiliation(s)
- Jiasheng Ju
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Chunchao Cheng
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Longtao Cui
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Biao Hong
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Qi Zhan
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Qixue Wang
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Xiaoteng Cui
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Dongyuan Su
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Yanping Huang
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Chunsheng Kang
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| |
Collapse
|
5
|
Lei X, Zheng Y, Su W. RNA-binding proteins and autophagy in lung cancer: mechanistic insights and therapeutic perspectives. Discov Oncol 2025; 16:599. [PMID: 40272614 PMCID: PMC12022210 DOI: 10.1007/s12672-025-02413-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/16/2025] [Indexed: 04/27/2025] Open
Abstract
BACKGROUND Lung cancer remains a leading cause of cancer-related mortality worldwide. Its progression is intricately associated with the dynamic regulation of autophagy and RNA-binding proteins (RBPs), which play crucial roles in mRNA stability, alternative splicing, and cellular stress responses. OBJECTIVES This review aims to systematically analyze the mechanisms through which RBPs and autophagy contribute to lung cancer progression and explore potential therapeutic strategies targeting these pathways. METHODS We reviewed recent studies on the molecular mechanisms by which RBPs regulate tumor proliferation, metabolic adaptation, and their interaction with autophagy. The review also examines the dual roles of autophagy in lung cancer, highlighting its context-dependent effects on cell survival and death. RESULTS The interactions and regulatory networks between RBPs and autophagy involve multiple levels of regulation. RBPs can directly influence autophagy processes and act as microRNA (miRNA) sponges to regulate mRNA stability. The modulation of RBPs affects the expression of autophagy-related genes (ATGs) and autophagosome formation. Additionally, RBPs participate in complex regulatory interactions with non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and other proteins. CONCLUSIONS This review proposes innovative therapeutic strategies that combine RBP-targeting approaches (e.g., small molecule inhibitors, CRISPR gene editing) with autophagy modulators (e.g., mTOR inhibitors, chloroquine) to enhance treatment efficacy. Nanoparticle drug delivery systems and epigenetic regulation offer further opportunities for targeted interventions. This review lays a theoretical foundation for advancing lung cancer research and provides novel insights into synergistic therapies that target both RBPs and autophagy to improve treatment outcomes for lung cancer.
Collapse
Affiliation(s)
- Xiao Lei
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
- Zhanjiang Key Laboratory of Tumor Microenvironment and Organoid Research, Zhanjiang, 524001, China
- Department of Guangdong Medical University, Zhanjiang, 524023, China
| | - Yuexin Zheng
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
- Zhanjiang Key Laboratory of Tumor Microenvironment and Organoid Research, Zhanjiang, 524001, China
- Department of Guangdong Medical University, Zhanjiang, 524023, China
| | - Wenmei Su
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
- Zhanjiang Key Laboratory of Tumor Microenvironment and Organoid Research, Zhanjiang, 524001, China.
- Department of Guangdong Medical University, Zhanjiang, 524023, China.
| |
Collapse
|
6
|
Walweel N, Cinar V, Mersin O, Macit S, Yildiz U, Demirel E, Tunç CU, Ulutabanca H, Hamurcu Z, Yuksel Durmaz Y, Aydin O. Enhanced In Vitro and In Vivo Autophagy Suppression via LC3 siRNA-Loaded "Smart" Nanoparticles and Doxorubicin Combination Therapy in Triple Negative Breast Cancer. ACS APPLIED BIO MATERIALS 2025; 8:2938-2953. [PMID: 40056448 DOI: 10.1021/acsabm.4c01778] [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: 03/10/2025]
Abstract
Autophagy plays a complex role in cancer progression, serving as both a tumor suppressor and a promoter, depending on the context. In triple-negative breast cancer (TNBC), a particularly aggressive subtype with limited therapeutic options, autophagy inhibition has emerged as a promising strategy to enhance the efficacy of chemotherapy. This study investigates the synergistic effects of autophagy suppression using LC3 siRNA-loaded "smart" nanoparticles (LC3siRNA-NPs) in combination with doxorubicin (DOX) to overcome chemoresistance in TNBC. We engineered a well-defined copolymer, poly[hexyl methacrylate-co-2-(dimethylamino) ethyl methacrylate-co-trimethylaminoethyl methacrylate iodide], and a PEG heteroarm beta-cyclodextrin (βCD) core star copolymer that delivers LC3 siRNA, effectively silencing the autophagy-related gene LC3. In vitro, the coadministration of LC3siRNA-NPs and DOX significantly inhibited TNBC cell proliferation, migration, and colony formation, while inducing apoptosis more effectively than either treatment alone. Mechanistically, this combination downregulated key oncogenic markers such as PARP, cyclin D1, and Src, enhancing the therapeutic outcome. In vivo, treatment with LC3siRNA-NPs and DOX in a TNBC xenograft model resulted in superior tumor growth suppression compared to that with monotherapy alone. Our findings highlight the potential of autophagy-targeting nanocarriers to improve chemotherapy outcomes and provide an effective approach to TNBC treatment by enhancing chemotherapeutic sensitivity and reducing tumor resistance.
Collapse
MESH Headings
- Doxorubicin/pharmacology
- Doxorubicin/chemistry
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/metabolism
- Humans
- Autophagy/drug effects
- Nanoparticles/chemistry
- RNA, Small Interfering/pharmacology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/chemistry
- Animals
- Mice
- Female
- Cell Proliferation/drug effects
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/metabolism
- Microtubule-Associated Proteins/antagonists & inhibitors
- Biocompatible Materials/chemistry
- Biocompatible Materials/pharmacology
- Biocompatible Materials/chemical synthesis
- Drug Screening Assays, Antitumor
- Materials Testing
- Particle Size
- Apoptosis/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Mice, Nude
- Cell Line, Tumor
- Mice, Inbred BALB C
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/chemistry
- Dose-Response Relationship, Drug
Collapse
Affiliation(s)
- Nada Walweel
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
| | - Venhar Cinar
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri 38030, Turkey
- GENKOK-Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri 38280, Turkey
| | - Osman Mersin
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Semih Macit
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Ummugulsum Yildiz
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
| | - Erhan Demirel
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Cansu Umran Tunç
- NanoThera Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, Utah 84112, United States
| | - Halil Ulutabanca
- Department of Neurosurgery, Erciyes University Medical School, Kayseri 38030, Turkey
| | - Zuhal Hamurcu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri 38030, Turkey
- GENKOK-Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri 38280, Turkey
| | - Yasemin Yuksel Durmaz
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
- Research Institute of Health Science and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Omer Aydin
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Turkey
- ERKAM-Clinical-Engineering Research and Implementation Center, Erciyes University, Kayseri 38030, Turkey
| |
Collapse
|
7
|
Wang B, Liu D, Shi D, Li X, Li Y. The role and machine learning analysis of mitochondrial autophagy-related gene expression in lung adenocarcinoma. Front Immunol 2025; 16:1509315. [PMID: 40313958 PMCID: PMC12043613 DOI: 10.3389/fimmu.2025.1509315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Objective Lung adenocarcinoma (LUAD) continues to be a primary cause of cancer-related mortality globally, highlighting the urgent need for novel insights finto its molecular mechanisms. This study aims to investigate the relationship between gene expression and mitophagy in LUAD, with an emphasis on identifying key biomarkers and elucidating their roles in tumorigenesis and immune cell infiltration. Methods We utilized datasets GSE151101 and GSE203609 from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) associated with lung cancer and mitophagy. DEGs were identified using GEO2R, filtered based on criteria of P < 0.05 and log2 fold change ≥ 1. Subsequently, Weighted Gene Co-expression Network Analysis (WGCNA) was conducted to classify DEGs into modules. Functional annotation of these modules was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Gene Set Enrichment Analysis (GSEA) was applied to the most relevant module, designated as the greenyellow module. To identify critical biomarkers, machine learning algorithms including Random Forest, Least Absolute Shrinkage and Selection Operator (LASSO) regression, and Support Vector Machine (SVM) were employed. Validation of the findings was conducted using The Cancer Genome Atlas (TCGA) database, Human Protein Atlas (HPA), quantitative PCR (qPCR), and immune cell infiltration analysis via CIBERSORTx. Results Our analysis identified 11,012 overlapping DEGs between the two datasets. WGCNA revealed 11 modules, with the green-yellow module exhibiting the highest correlation. Functional enrichment analysis highlighted significant associations with FOXM1 signaling pathways and retinoblastoma in cancer. Machine learning algorithms identified COASY, FTSJ1, and MOGS as pivotal genes. These findings were validated using TCGA data, qPCR experiments, which demonstrated high expression levels in LUAD samples. Immunohistochemistry from HPA confirmed consistency between protein levels and RNA-seq data. Furthermore, pan-cancer analysis indicated that these genes are highly expressed across various cancer types. Immune infiltration analysis suggested significant correlations between these genes and specific immune cell populations. Conclusion COASY, FTSJ1 and MOGS have emerged as critical biomarkers in LUAD, potentially influencing tumorigenesis through mitophagy-related mechanisms and immune modulation. These findings provide promising avenues for future research into targeted therapies and diagnostic tools, thereby enhancing LUAD management.
Collapse
Affiliation(s)
- Binyu Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Huzhou University, The First People’s Hospital of Huzhou City, Huzhou, Zhejiang, China
| | - Di Liu
- Department of Clinical Laboratory, Huzhou Maternity & Child Health Care Hospital, Huzhou, Zhejiang, China
| | - Danfei Shi
- Department of Pathology, The First Affiliated Hospital of Huzhou University, The First People’s Hospital of Huzhou City, Huzhou, Zhejiang, China
| | - Xinmin Li
- Department of Clinical Laboratory, Chongqing Hospital of Traditional Chinese Medicine, ChongQing, China
| | - Yong Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Huzhou University, The First People’s Hospital of Huzhou City, Huzhou, Zhejiang, China
| |
Collapse
|
8
|
Jiang T, Fei L. cGAS-STING signaling in melanoma: regulation and therapeutic targeting. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04141-8. [PMID: 40223035 DOI: 10.1007/s00210-025-04141-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
Melanocytes are the source of the skin cancer known as melanoma. It usually affects the viscera, mucous membranes, and skin. Even so, melanoma only makes for 7% of all skin cancer occurrences. By triggering the generation of type I interferons (IFN-I) and inflammatory cytokines upon identifying microbial DNA, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway promotes anti-microbial innate immunity. A growing body of research indicates that antitumor immunity depends on the cGAS-STING axis being activated. The cGAS-STING-regulated downstream cytokines, particularly IFN-I, act as linkages between adaptive and innate immunity. As a result, an increasing amount of research has concentrated on the synthesis and screening of agonists of the STING pathway. As a result, an increasing amount of research has concentrated on the synthesis and screening of agonists of the STING pathway. The many implications of the cGAS-STING pathway in the pathophysiology and therapy of melanoma are thoroughly examined in this study. Our research highlights the significance of the cGAS-STING pathway in melanoma and identifies it as a key target for boosting immunity against tumors.
Collapse
Affiliation(s)
- Ting Jiang
- Cancer Center, The First Bethune Hospital of Jilin University, Changchun, 130000, China
| | - Lixue Fei
- Cancer Center, The First Bethune Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
9
|
Jalali P, Shahmoradi A, Samii A, Mazloomnejad R, Hatamnejad MR, Saeed A, Namdar A, Salehi Z. The role of autophagy in cancer: from molecular mechanism to therapeutic window. Front Immunol 2025; 16:1528230. [PMID: 40248706 PMCID: PMC12003146 DOI: 10.3389/fimmu.2025.1528230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
Autophagy is a cellular degradation process that plays a crucial role in maintaining metabolic homeostasis under conditions of stress or nutrient deprivation. This process involves sequestering, breaking down, and recycling intracellular components such as proteins, organelles, and cytoplasmic materials. Autophagy also serves as a mechanism for eliminating pathogens and engulfing apoptotic cells. In the absence of stress, baseline autophagy activity is essential for degrading damaged cellular components and recycling nutrients to maintain cellular vitality. The relationship between autophagy and cancer is well-established; however, the biphasic nature of autophagy, acting as either a tumor growth inhibitor or promoter, has raised concerns regarding the regulation of tumorigenesis without inadvertently activating harmful aspects of autophagy. Consequently, elucidating the mechanisms by which autophagy contributes to cancer pathogenesis and the factors determining its pro- or anti-tumor effects is vital for devising effective therapeutic strategies. Furthermore, precision medicine approaches that tailor interventions to individual patients may enhance the efficacy of autophagy-related cancer treatments. To this end, interventions aimed at modulating the fate of tumor cells by controlling or inducing autophagy substrates necessitate meticulous monitoring of these mediators' functions within the tumor microenvironment to make informed decisions regarding their activation or inactivation. This review provides an updated perspective on the roles of autophagy in cancer, and discusses the potential challenges associated with autophagy-related cancer treatment. The article also highlights currently available strategies and identifies questions that require further investigation in the future.
Collapse
Affiliation(s)
- Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arvin Shahmoradi
- Department of Laboratory Medicine, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amir Samii
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Radman Mazloomnejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hatamnejad
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Afshin Namdar
- Program in Cell Biology, The Hospital for Sick Children Peter Gilgan Centre for Research and Learning, Toronto, ON, United States
| | - Zahra Salehi
- Department of Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Hasan MM, Goto S, Sekiya R, Hayashi T, Li TS, Kawabata T. Sustained induction of autophagy enhances survival during prolonged starvation in newt cells. Life Sci Alliance 2025; 8:e202402772. [PMID: 39904566 PMCID: PMC11794943 DOI: 10.26508/lsa.202402772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025] Open
Abstract
Salamanders demonstrate exceptional resistance to starvation, allowing them to endure extended periods without food in their natural habitats. Although autophagy, a process involving evolutionarily conserved proteins, promotes survival during food scarcity, the specific mechanism by which it contributes to the extreme starvation resistance in newt cells remains unexplored. Our study, using the newt species Pleurodeles waltl, reveals that newt primary fibroblasts maintain constant autophagy activation during prolonged cellular starvation. Unlike normal mammalian fibroblasts, where autophagosome formation increases during acute starvation but returns to baseline levels after extended periods, newt cells maintain elevated autophagosome numbers even 4 d after autophagy initiation, surpassing levels observed in nutrient-rich conditions. Unique P. waltl mTOR orthologs show reduced lysosomal localization compared with mammalian cells in both nutrient-rich and starved states. However, newt cells exhibit dephosphorylation of mTOR substrates under starvation conditions, similar to mammalian cells. These observations suggest that newts may have evolved a distinctive system to balance seemingly conflicting factors: high regenerative capacity and autophagy-mediated survival during starvation.
Collapse
Affiliation(s)
- Md Mahmudul Hasan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Reiko Sekiya
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Toshinori Hayashi
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tsuyoshi Kawabata
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
11
|
Zhai X, Zhang Z, Chen Y, Wu Y, Zhen C, Liu Y, Lin Y, Chen C. Current and future therapies for small cell lung carcinoma. J Hematol Oncol 2025; 18:37. [PMID: 40170056 PMCID: PMC11959764 DOI: 10.1186/s13045-025-01690-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/14/2025] [Indexed: 04/03/2025] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy characterized by rapid proliferation and high metastatic potential. It is characterized by universal inactivation of and RB1, overexpression of the MYC family and dysregulation of multiple oncogenic signaling pathways. Among different patients, SCLCs are similar at the genetic level but exhibit significant heterogeneity at the molecular level. The classification of SCLC has evolved from a simple neuroendocrine (NE)/non-neuroendocrine (non-NE) classification system to a transcription factor-based molecular subtype system; lineage plasticity adds further complexity and poses challenges for therapeutic development. While SCLC is initially sensitive to platinum-based chemotherapy, resistance develops rapidly, leading to a dismal prognosis. Various antibodies, including PD-1/PD-L1 inhibitors and antibody‒drug conjugates, have been introduced into clinical practice or are being evaluated in clinical trials. However, their therapeutic benefits for SCLC patients remain limited. This review summarizes SCLC carcinogenic mechanisms, tumor heterogeneity, and the immune microenvironment of SCLC, with a focus on recent advances in metastasis and resistance mechanisms. Additionally, the corresponding clinical progress in tackling these challenges is discussed.
Collapse
Affiliation(s)
- Xiaoqian Zhai
- Department of Medical Oncology, State Key Laboratory of Biotherapy and Cancer Center and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 1, Keyuan 4th Road, Gaopeng Avenue, Chengdu, 610041, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhengkun Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- College of Life Sciences, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuxin Chen
- West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yanmou Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- College of Life Sciences, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Cheng Zhen
- West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Liu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 1, Keyuan 4th Road, Gaopeng Avenue, Chengdu, 610041, Sichuan, China.
| | - Yiyun Lin
- Department of Medicine, Weill Cornell Medicine, East 69th Street, New York, NY, 10021, USA.
| | - Chong Chen
- Department of Medical Oncology, State Key Laboratory of Biotherapy and Cancer Center and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 1, Keyuan 4th Road, Gaopeng Avenue, Chengdu, 610041, Sichuan, China.
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Al Amin M, Bouhenni H, Zehravi M, Sweilam SH, Durgawale TP, Qureshi MS, Durgapal S, Haque MA, Vodeti R, Urs D, Shatu MM, Rab SO, Doukani K, Emran TB. Natural compounds and programmed necrosis: pioneering a new frontier in cancer treatments. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04050-w. [PMID: 40137962 DOI: 10.1007/s00210-025-04050-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025]
Abstract
Programmed necrosis, a controlled cell death method that bypasses resistance mechanisms that render apoptosis ineffective, is a potential cancer treatment target. Due to their diverse biological activities and low side effects, natural products are being explored as modulators of programmed necrosis pathways. This review highlights the potential of natural compounds to target cancer cells while preserving healthy tissues and their interaction with essential programmed necrosis mechanisms like ferroptosis and necroptosis. Recent developments have identified various types of programmable necrosis, including necroptosis, ferroptosis, pyroptosis, proptosis, mitochondrial permeability transition-driven necrosis, and oncosis. Natural compounds are increasingly being utilized as a primary source of anti-cancer medications, providing new cancer treatments. This review demonstrates the molecular mechanisms behind lipid peroxidation, mixed lineage kinase domain-like protein, and receptor-interacting protein kinases (RIPK1 and RIPK3) inducing cell death. Recent research has identified natural compounds like polyphenols, alkaloids, and terpenoids that can modulate pathways and benefit preclinical cancer models. The review underscores the potential of natural compounds in developing innovative cancer treatments by integrating pharmacology and cellular signaling knowledge. Integrating natural compound studies and programmed necrosis research presents a promising avenue for oncologists to overcome treatment resistance. Natural compounds have shown potential in developing programmed necrosis as a novel cancer treatment approach, enhancing therapeutic effectiveness and minimizing side effects through preclinical research, pharmacology, and molecular biology.
Collapse
Affiliation(s)
- Md Al Amin
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1216, Bangladesh.
| | - Hasna Bouhenni
- Laboratory of Agrobiotechnology and Nutrition in Semi-Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Trupti Pratik Durgawale
- Department of Pharmaceutical Chemistry, Krishna Institute of Pharmacy Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, Maharashtra, India
| | - Mohammad Shamim Qureshi
- Department of Pharmacognosy & Phytochemistry, Anwarul Uloom College of Pharmacy, New Mallepally, Hyderabad, 500001, India
| | - Sumit Durgapal
- Department of Pharmaceutics, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Premnagar, Dehradun, Uttarakhand, 248007, India
| | - M Akiful Haque
- School of Pharmacy, Anurag University, Venkatapur, Hyderabad, Telangana , 500088, India
| | - Rajeshwar Vodeti
- Deportment of Pharmaceutics, School of Pharmacy, Anurag University, Hyderabad, India
| | - Deepadarshan Urs
- Inflammation Research Laboratory, Department of Studies & Research in Biochemistry, Mangalore University, Jnana Kaveri Post Graduate Centre, Kodagu, Karnataka, 571232, India
| | - Mst Maharunnasa Shatu
- Department of Botany, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Koula Doukani
- Laboratory of Agrobiotechnology and Nutrition in Semi-Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
- Laboratory of Animal Production Sciences and Techniques, Faculty of Nature and Life Sciences, University of Abdelhamid Ibn Badis, Mostaganem, Algeria
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1216, Bangladesh.
| |
Collapse
|
13
|
Liu Q, Hu J, Li X, Gao H, Kong D, Jin M. Glutamine transporter inhibitor enhances the sensitivity of NSCLC to trametinib through GSDME-dependent pyroptosis. Biochem Pharmacol 2025; 233:116796. [PMID: 39923858 DOI: 10.1016/j.bcp.2025.116796] [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: 11/01/2024] [Revised: 12/27/2024] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Trametinib, an inhibitor of mitogen-activated extracellular signal-regulated kinases 1/2 (MEK1/2), is used to treat BRAFV600E/K melanoma and non-small-cell lung cancer (NSCLC). Mutant Kirsten rat sarcoma viral oncogene homolog (KRAS) promotes glutamine utilization, therefore, in the present study we investigated the anti-cancer effects of trametinib in combination with V-9302, a glutamine transporter inhibitor, in NSCLC with KRAS mutations. Trametinib in combination with V-9302 exhibited a potent synergistic antitumor effect, inducing cell cycle arrest and pyroptosis. Mechanistically, combination treatment triggered caspase-3 activation and gasdermin E (GSDME) cleavage, as well as elevated lactate dehydrogenase (LDH) and IL-1β levels. Meanwhile, combination treatment reduced cyclin D1 and p-Rb levels and increased p27 expression. Moreover, this combination increased forkhead box class O3a (FOXO3a) levels and decreased forkhead box M1 (FOXM1) expression by regulating the phosphorylation of ERK, Akt, AMPK, and c-Jun N-terminal kinase (JNK). Trametinib in combination with V-9302 increased reactive oxygen species (ROS) generation and reduced glutathione (GSH) synthesis and ATP levels. Furthermore, V-9302 in combination with trametinib inhibited the trametinib-induced autophagy, thereby enhancing pyroptosis in cancer cells. In vivo, the co-administration of trametinib and V-9302 remarkably inhibited tumor growth in a xenograft mouse model compared to each drug alone. Taken together, the combination of trametinib and V-9302 resulted in increased pyroptosis and cell cycle arrest compared to each single agent through regulation of the FOXO3a/FOXM1 axis and autophagy and significantly enhanced antitumor efficacy in vivo. Our results suggest a potential new therapeutic strategy for KRAS-mutant NSCLC using trametinib in combination with glutamine restriction.
Collapse
Affiliation(s)
- Qingxia Liu
- Tianjin Key Laboratory On Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, International Joint Laboratory of Ocular Diseases (Ministry of Education), Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Jinxia Hu
- Tianjin Key Laboratory On Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, International Joint Laboratory of Ocular Diseases (Ministry of Education), Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Xinzhen Li
- Tianjin Key Laboratory On Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, International Joint Laboratory of Ocular Diseases (Ministry of Education), Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Haiwang Gao
- Tianjin Key Laboratory On Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, International Joint Laboratory of Ocular Diseases (Ministry of Education), Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Dexin Kong
- Tianjin Key Laboratory On Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, International Joint Laboratory of Ocular Diseases (Ministry of Education), Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China; Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Meihua Jin
- Tianjin Key Laboratory On Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, International Joint Laboratory of Ocular Diseases (Ministry of Education), Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
14
|
Pei S, Zhang D, Li Z, Liu J, Li Z, Chen J, Xie Z. The Role of the Fox Gene in Breast Cancer Progression. Int J Mol Sci 2025; 26:1415. [PMID: 40003882 PMCID: PMC11855465 DOI: 10.3390/ijms26041415] [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: 11/26/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Forkhead box (FOX) genes are a family of transcription factors that participate in many biological activities, from early embryogenesis to the formation of organs, and from regulation of glucose metabolism to regulation of longevity. Given the extensive influence in the multicellular process, FOX family proteins are responsible for the progression of many types of cancers, especially lung cancer, breast cancer, prostate cancer, and other cancers. Breast cancer is the most common cancer among women, and 2.3 million women were diagnosed in 2020. So, various drugs targeting the FOX signaling pathway have been developed to inhibit breast cancer progression. While the role of the FOX family gene in cancer development has not received enough attention, discovering more potential drugs targeting the FOX signaling pathway is urgently demanded. Here, we review the main members in the FOX gene family and summarize their signaling pathway, including the regulation of the FOX genes and their effects on breast cancer progression. We hope this review will emphasize the understanding of the role of the FOX gene in breast cancer and inspire the discovery of effective anti-breast cancer medicines targeting the FOX gene in the future.
Collapse
Affiliation(s)
- Shaoxuan Pei
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Dechun Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhuohan Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jinkai Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Ziyi Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
| |
Collapse
|
15
|
Zhang Q, Guo S, Ge H, Wang H. The protective role of baicalin regulation of autophagy in cancers. Cytotechnology 2025; 77:33. [PMID: 39760060 PMCID: PMC11699138 DOI: 10.1007/s10616-024-00689-0] [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: 11/25/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025] Open
Abstract
Autophagy is a conservative process of self degradation, in which abnormal organelles, proteins and other macromolecules are encapsulated and transferred to lysosomes for subsequent degradation. It maintains the intracellular balance, and responds to cellular conditions such as hunger or stress. To date, there are mainly three types of autophagy: macroautophagy, microautophagy and chaperone-mediated autophagy. Autophagy plays a key role in regulating multiple physiological and pathological processes, such as cell metabolism, development, energy homeostasis, cell death and hunger adaptation, and so on. Increasing evidence indicates that autophagy dysfunction participates in many kinds of cancers, such as liver cancer, pancreatic cancer, prostate cancer, and so on. However, the relevant mechanisms are not yet fully understood. Baicalin is a natural flavonoid compound extracted from the traditional Chinese medicine Scutellaria baicalensis. The research has shown that after oral or intravenous administration of baicalin, it is delivered to various organs through the systemic circulation, with the highest volume in the kidneys and lungs. More and more evidence suggests that baicalin has antioxidant, anticancer, anti-inflammatory, anti-apoptotic, immunomodulatory and antiviral effects. Therefore, baicalin plays an important role in various diseases, such as cancers, lung diseases, liver diseases, cardiovascular diseases, ans so on. However, the relevant mechanisms have not yet been fully clear. Recently, increasing evidence indicates that baicalin participates in different cancer by regulating autophagy. Herein, we reviewed the current knowledge about the role and mechanism of baicalin regulation of autophagy in multiple types of cancers to lay the theoretical foundation for future related researches.
Collapse
Affiliation(s)
- Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Hangwei Ge
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| |
Collapse
|
16
|
Li X, Chaouhan HS, Yu S, Wang I, Yu T, Chuang Y, Chen K, Lin F, Chen MY, Hsu C, Sun K, Li C. Hypoxia-Induced Metabolic and Functional Changes in Oral CSCs: Implications for Stemness and Viability Modulation Through BNIP3-Driven Mitophagy. J Cell Mol Med 2025; 29:e70400. [PMID: 39945227 PMCID: PMC11822456 DOI: 10.1111/jcmm.70400] [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: 06/09/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Oral squamous cell carcinomas (OSCCs), like several solid tumours, contain heterogeneous subpopulations of a small subset of cancer cells, termed cancer stem cells (CSCs), that are highly relevant to cancer metastasis and invasive properties. CSCs have also shown a high capacity to survive against various stressful environments, such as hypoxia. However, the molecular underpinnings behind the high potential of CSCs to survive under this stress remain unclear. The current study aimed to investigate the significance of autophagy systems in oral CSC maintenance and survival under stress conditions. Human OSCC cell lines OECM-1 and OECM-1 CSCs were cultured in different hypoxic time periods for proliferation and cytotoxicity analyses. The stemness property of CSCs is evaluated by sphere formation, transwell and wound healing assays protein expression of stemness, and epithelial-to-mesenchymal transition markers. Mitochondrial functions, including mitochondrial ROS generation, mitochondria dynamics, mitophagy, and mitochondrial metabolism (glycolysis and oxidative phosphorylation [OXPHOS]) were examined by western blotting, immunohistochemistry, and XF-seahorse assays, respectively. Under hypoxia, oral CSCs showed a higher proliferation rate with increased invasion/migration/EMT properties than OECM-1 cells. Further, hypoxia-induced BNIP3-driven mitophagy was activated in OECM-1 CSCs than in OECM-1 cells, which also triggered a metabolic shift towards OXPHOS, and BNIP3/-L silencing by siRNA significantly attenuated OECM-1 CSCs stemness features. TCGA data analyses also revealed a higher BNIP3 expression in head and neck squamous carcinoma patients' tumour samples associated with lower patient survival. Collectively, our results revealed a BNIP3/-L-driven autophagy contributes to the OECM-1 CSCs stemness features under hypoxia, suggesting a novel therapeutic strategy involving BNIP3 and autophagy inhibition in oral CSCs.
Collapse
Affiliation(s)
- Xin Li
- Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
| | | | - Shao‐Hua Yu
- Department of Emergency MedicineChina Medical University HospitalTaichungTaiwan
| | - I‐Kuan Wang
- Division of NephrologyChina Medical University HospitalTaichungTaiwan
- Department of Internal Medicine, School of MedicineChina Medical UniversityTaichungTaiwan
| | - Tung‐Min Yu
- Division of Nephrology, Department of Internal MedicineTaichung Veterans General HospitalTaichungTaiwan
- School of MedicineChina Medical UniversityTaichungTaiwan
| | - Ya‐Wen Chuang
- Division of Nephrology, Department of Internal MedicineTaichung Veterans General HospitalTaichungTaiwan
- Department of Post‐Baccalaureate Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
| | - Kuen‐Bao Chen
- Department of AnesthesiologyChina Medical University HospitalTaichungTaiwan
| | - Feng‐Yen Lin
- Taipei Heart InstituteTaipei Medical UniversityTaipeiTaiwan
- Division of Cardiology and Cardiovascular Research CenterTaipei Medical University HospitalTaipeiTaiwan
- Department of Internal Medicine, College of Medicine, School of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Michael Yuan‐Chien Chen
- School of DentistryChina Medical UniversityTaichungTaiwan
- Department of DentistryChina Medical University HospitalTaichungTaiwan
| | - Che‐Hao Hsu
- Department of AnesthesiologyTungs' Taichung Metroharbor HospitalTaichungTaiwan
| | - Kuo‐Ting Sun
- School of DentistryChina Medical UniversityTaichungTaiwan
- Department of DentistryChina Medical University HospitalTaichungTaiwan
- Department of Pediatric DentistryChina Medical University HospitalTaichungTaiwan
| | - Chi‐Yuan Li
- Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
- Department of AnesthesiologyChina Medical University HospitalTaichungTaiwan
| |
Collapse
|
17
|
Caprara G, Pallavi R, Sanyal S, Pelicci PG. Dietary Restrictions and Cancer Prevention: State of the Art. Nutrients 2025; 17:503. [PMID: 39940361 PMCID: PMC11820753 DOI: 10.3390/nu17030503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Worldwide, almost 10 million cancer deaths occurred in 2022, a number that is expected to rise to 16.3 million by 2040. Primary prevention has long been acknowledged as a crucial approach to reducing cancer incidence. In fact, between 30 and 50 percent of all tumors are known to be preventable by eating a healthy diet, staying active, avoiding alcohol, smoking, and being overweight. Accordingly, many international organizations have created tumor prevention guidelines, which underlie the importance of following a diet that emphasizes eating plant-based foods while minimizing the consumption of red/processed meat, sugars, processed foods, and alcohol. However, further research is needed to define the relationship between the effect of specific diets or nutritional components on cancer prevention. Interestingly, reductions in food intake and dietetic restrictions can extend the lifespan of yeast, nematodes, flies, and rodents. Despite controversial results in humans, those approaches have the potential to ameliorate health via direct and indirect effects on specific signaling pathways involved in cancer onset. Here, we describe the latest knowledge on the cancer-preventive potential of dietary restrictions and the biochemical processes involved. Molecular, preclinical, and clinical studies evaluating the effects of different fasting strategies will also be reviewed.
Collapse
Affiliation(s)
- Greta Caprara
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| | - Rani Pallavi
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Shalini Sanyal
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| |
Collapse
|
18
|
Dahl-Wilkie H, Gomez J, Kelley A, Manjit K, Mansoor B, Kanumuri P, Pardo S, Molleur D, Falah R, Konakalla AR, Omiyale M, Weintraub S, Delk NA. Chronic IL-1-Exposed LNCaP Cells Evolve High Basal p62-KEAP1 Complex Accumulation and NRF2/KEAP1-Dependent and -Independent Hypersensitive Nutrient Deprivation Response. Cells 2025; 14:192. [PMID: 39936983 PMCID: PMC11816438 DOI: 10.3390/cells14030192] [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: 11/09/2024] [Revised: 01/25/2025] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
Chronic inflammation is a cancer hallmark and chronic exposure to interleukin-1 (IL-1) transforms castration-sensitive prostate cancer (PCa) cells into more fit castration-insensitive PCa cells. p62 is a scaffold protein that protects cells from nutrient deprivation via autophagy and from cytotoxic reactive oxygen via NFκB and NRF2 antioxidant signaling. Herein, we report that the LNCaP PCa cell line acquires high basal accumulation of the p62-KEAP1 complex when chronically exposed to IL-1. p62 promotes non-canonical NRF2 antioxidant signaling by binding and sequestering KEAP1 to the autophagosome for degradation. But despite high basal p62-KEAP1 accumulation, only two of several NRF2-induced genes analyzed, GCLC and HMOX1, showed high basal mRNA levels, suggesting that the high basal p62-KEAP1 accumulation does not result in overall high basal NRF2 activity. Nutrient starvation induces NRF2-dependent GCLC upregulation and HMOX1 repression, and we found that chronic IL-1-exposed LNCaP cells show hypersensitivity to serum starvation-induced GCLC and HMOX1 regulation. Thus, chronic IL-1 exposure affects cell response to nutrient stress. While HMOX1 expression remains NRF2/KEAP1-dependent in chronic IL-1-exposed LNCaP cells, GCLC expression is NRF2/KEAP1-independent. Furthermore, the high basal p62-KEAP1 complex accumulation is not required to regulate GCLC or HMOX1 expression, suggesting cells chronically exposed to IL-1 evolve a novel NRF2-independent role for the p62/KEAP1 axis.
Collapse
Affiliation(s)
- Haley Dahl-Wilkie
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Jessica Gomez
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Anastasia Kelley
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Kirti Manjit
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Basir Mansoor
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Preethi Kanumuri
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Sammy Pardo
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Dana Molleur
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Rafah Falah
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Anisha R. Konakalla
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Morolake Omiyale
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Susan Weintraub
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Nikki A. Delk
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| |
Collapse
|
19
|
Parveen S, Ikhlas S, Faruqui T, Hasan A, Khatoon A, Saeed M, Alkhathami AG, Siddiqui S, Uddin S, Mir SS. Identifying Potential Autophagy Modulators in Panch Phoron Spices (P5S): An In Silico approach. ACS OMEGA 2025; 10:871-884. [PMID: 39829452 PMCID: PMC11740127 DOI: 10.1021/acsomega.4c07924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/14/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Despite recent breakthroughs in diagnosis and treatment, cancer remains a worldwide health challenge with high mortality. Autophagy plays a major role in the progression and development. Starving cancer cells obtain nutrients through the upregulation of autophagy. Several compounds derived from natural sources, including animals, plants, and microorganisms, have been identified as potential novel anticancer drugs. Spices play an important role in human health and possess many medicinal properties. Our study aimed to identify potential autophagy modulators from panch phoron spices (P5S) through in silico approaches. Herein, we report a structure-based virtual screening of compounds isolated from P5S (i.e., cumin, fenugreek, fennel, black mustard, and black cumin) against the molecular targets of autophagy. Using various computational tools, we attempted to identify potential modulators of autophagy. Among all the screening results (such as binding energy, hydrogen bonding, drug-likeness, bioactivity, ADME properties, and toxicity), P5S, stigmasterol, and tigogenin showed the best drug-like properties and binding affinity toward the selected targets of autophagy. Furthermore, the stability of both complexes was evaluated by performing a 100 ns molecular dynamics simulation (MDS) using Schrodinger's Desmond Module. Our results provide insight into the efficacy of P5S components against cancer. Therefore, targeting autophagy using these molecules may be an effective and potential drug candidate for cancer treatment. In conclusion, stigmasterol and tigogenin may act as potential candidates for anticancer drugs by targeting autophagy.
Collapse
Affiliation(s)
- Sana Parveen
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow, Uttar Pradesh 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow, Uttar
Pradesh 226026, India
| | - Shoeb Ikhlas
- Department
of Cell Biology, Albert Einstein College
of Medicine, 1300, Moris
Park Ave, New York, New York 10461, United States
| | - Tabrez Faruqui
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow, Uttar
Pradesh 226026, India
| | - Adria Hasan
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow, Uttar Pradesh 226026, India
- Department
of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow, Uttar
Pradesh 226026, India
| | - Aisha Khatoon
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow, Uttar Pradesh 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow, Uttar
Pradesh 226026, India
| | - Mohd Saeed
- Department
of Biology College of Science, University
of Hail, Hail 2240, Saudi Arabia
| | - Ali G. Alkhathami
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62521, Saudi Arabia
| | - Samra Siddiqui
- Department
of Health Services Management, College of Public Health and Health
Informatics, University of Hail, Hail 2240, Saudi Arabia
| | - Shahab Uddin
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow, Uttar
Pradesh 226026, India
- Translational
Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Snober S. Mir
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow, Uttar Pradesh 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow, Uttar
Pradesh 226026, India
| |
Collapse
|
20
|
Li F, Yu Y, Jiang M, Zhang H. Targets for improving prostate tumor response to radiotherapy. Eur J Pharmacol 2025; 986:177149. [PMID: 39577551 DOI: 10.1016/j.ejphar.2024.177149] [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: 09/11/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Prostate cancer is a prevalent malignancy that is frequently managed with radiotherapy. However, resistance to radiotherapy remains a significant challenge in controlling this disease. Early radiotherapy is employed for locally confined prostate cancer (PCa), while recurrent disease post-surgery and metastatic castration-resistant prostate cancer (mCRPC) are treated with late-stage radiotherapy, including radium-223. Combination therapies to integrate radiotherapy and chemotherapy have demonstrated enhanced treatment efficacy. Nonetheless, both modalities can induce severe local and systemic toxicities. Consequently, selectively sensitizing prostate tumors to radiotherapy could improve therapeutic outcomes while minimizing systemic side effects. The mechanisms underlying radioresistance in prostate cancer are multifaceted, including DNA damage repair (DDR) pathways, hypoxia, angiogenesis, androgen receptor (AR) signaling, and immune evasion. The advent of 177Lu-PSMA-617, which was approved in 2022, has shown promise in targeting prostate-specific membrane antigen (PSMA) in advanced prostate cancer. Experimental and clinical studies have yielded promising results in suppressing prostate tumors by targeting these pathways. This paper reviews potential targets for sensitizing prostate tumors to radiotherapy. We discuss cellular and molecular mechanisms contributing to therapy resistance and examine findings from experimental and clinical trials on promising targets and drugs that can be used in combination with radiotherapy.
Collapse
Affiliation(s)
- Fengguang Li
- Department of Urology, Yantaishan Hospital, Shandong, 264000, China
| | - Yizhi Yu
- Department of Urology, Yantaishan Hospital, Shandong, 264000, China
| | - Maozhu Jiang
- Department of Radiotherapy, Yantaishan Hospital, Shandong, 264000, China
| | - Haiying Zhang
- Department of Urology, Yantaishan Hospital, Shandong, 264000, China.
| |
Collapse
|
21
|
Sun S, Jing X, Tong G, Chen C, Xie S, Wang C, Chen D, Zhao J, Qi Y, Zhang W, Liu C, Zhang G, Zhang J, Sun B, Wang Y, Lv Y. Loss of DDX24 inhibits lung cancer progression by stimulating IKBKG splicing-mediated autophagy. Theranostics 2025; 15:1879-1895. [PMID: 39897555 PMCID: PMC11780526 DOI: 10.7150/thno.102425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/21/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Lung cancer remains a major global health burden with limited therapeutic options. Alternative splicing, a critical post-transcriptional process, contributes to lung cancer progression through autophagy, although the underlying mechanisms remain largely unexplored. This study aims to elucidate the role of DDX24 as a splicing factor that contributes to lung cancer progression via autophagy. Methods: To establish the link between DDX24 and lung cancer progression, we performed colony formation assays, growth curve analyses, and xenograft tumor models in nude mice. Mass spectrometry and RNA sequencing were employed to investigate the involvement of DDX24 in alternative splicing, with a specific focus on the splicing of IKBKG. The mechanisms by which DDX24 regulates autophagy were further explored using co-immunoprecipitation and luciferase reporter assays. Results: The splicing factor DDX24 is significantly elevated in lung cancer tissues. Loss of DDX24 suppresses lung cancer growth by promoting autophagy. We identified DDX24 as a splicing factor that plays critical roles in the regulation of alternative splicing. Mechanistically, DDX24 regulates the alternative splicing of autophagy-related genes, including IKBKG. We demonstrate that DDX24 directly binds to IKBKG pre-mRNA, whereas DDX24 ablation stimulates the generation of the long splicing isoform of IKBKG, thereby promoting autophagy through activating of the NF-kB signaling pathway and the transcription of the BECN1 gene. Functional rescue experiments confirm that the long IKBKG isoform-mediated autophagy confers the anti-tumor effects of DDX24 depletion. In addition, IKBKG-L is positively associated with improved survival in lung cancer patients. Conclusions: This study uncovers a novel regulatory axis involving DDX24, IKBKG splicing, and autophagy in lung cancer. Our findings suggest that targeting DDX24 may represent a promising therapeutic strategy for lung cancer treatment, offering new insights into the molecular underpinnings of this disease.
Collapse
Affiliation(s)
- Siwen Sun
- Department of Oncology & Sino-US Research Center for Cancer Translational Medicine, the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, China
| | - Xiaomeng Jing
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| | - Guangquan Tong
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Chaoqun Chen
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| | - Shuaijun Xie
- Department of Pathology, the First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Chong Wang
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| | - Dan Chen
- Department of Pathology, the First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Jinyao Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Yangfan Qi
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Wenjing Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Congcong Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Ge Zhang
- Department of Immunology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Jinrui Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Bing Sun
- Department of Thoracic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Yang Wang
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| | - Yuesheng Lv
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| |
Collapse
|
22
|
Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, Cañadas I, Sethi G, Tergaonkar V, Wang L, Lin Y, Kang D, Klionsky DJ. Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat 2025; 78:101170. [PMID: 39603146 DOI: 10.1016/j.drup.2024.101170] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Macroautophagy/autophagy is a highly conserved evolutionary mechanism involving lysosomes for the degradation of cytoplasmic components including organelles. The constitutive, basal level of autophagy is fundamental for preserving cellular homeostasis; however, alterations in autophagy can cause disease pathogenesis, including cancer. The role of autophagy in cancer is particularly complicated, since this process acts both as a tumor suppressor in precancerous stages but facilitates tumor progression during carcinogenesis and later stages of cancer progression. This shift between anti-tumor and pro-tumor roles may be influenced by genetic and environmental factors modulating key pathways such as those involving autophagy-related proteins, the PI3K-AKT-MTOR axis, and AMPK, which often show dysregulation in tumors. Autophagy regulates various cellular functions, including metabolism of glucose, glutamine, and lipids, cell proliferation, metastasis, and several types of cell death (apoptosis, ferroptosis, necroptosis and immunogenic cell death). These multifaceted roles demonstrate the potential of autophagy to affect DNA damage repair, cell death pathways, proliferation and survival, which are critical in determining cancer cells' response to chemotherapy. Therefore, targeting autophagy pathways presents a promising strategy to combat chemoresistance, as one of the major reasons for the failure in cancer patient treatment. Furthermore, autophagy modulates immune evasion and the function of immune cells such as T cells and dendritic cells, influencing the tumor microenvironment and cancer's biological behavior. However, the therapeutic targeting of autophagy is complex due to its dual role in promoting survival and inducing cell death in cancer cells, highlighting the need for strategies that consider both the beneficial and detrimental effects of autophagy modulation in cancer therapy. Hence, both inducers and inhibitors of autophagy have been introduced for the treatment of cancer. This review emphasizes the intricate interplay between autophagy, tumor biology, and immune responses, offering insights into potential therapeutic approaches that deploy autophagy in the cancer suppression.
Collapse
Affiliation(s)
- Xuegang Niu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qi You
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Kaijian Hou
- School of Public Health(Long Hu people hospital), Shantou University, Shantou, 515000, Guangdong, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, IL 60532, USA
| | - Penghui Wei
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Bin Gao
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Alireza Kalbasi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Israel Cañadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A⁎STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Lingzhi Wang
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Yuanxiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Dezhi Kang
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
23
|
Yang J, Ni S, Wang A, Wang K, Deng J, Li Z, Cai Y, Chen Y, Chen G, Lin D. Myrtenol promotes skin flap survival by inhibiting apoptosis and promoting autophagy via the MEK/ERK pathway. Arch Biochem Biophys 2025; 763:110230. [PMID: 39603374 DOI: 10.1016/j.abb.2024.110230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/05/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
Skin flaps are often used for repair and reconstruction, including oral cavity and palate. However, postoperative flap necrosis limited applications. Myrtenol, a plant-derived bicyclic monoterpene, has pharmacological effects including inhibiting apoptosis and promoting autophagy. But any impact on skin flaps survival remains unclear. Thus, we established modified McFarlane flaps on 24 Sprague-Dawley rats and applied myrtenol. They were randomly divided into low-dose myrtenol (L-Myr), high-dose myrtenol (H-Myr), inhibitor and control groups. On postoperative day 7, flap survival rate was increased and Laser Doppler images showed blood circulation improvement under myrtenol treatment. Hematoxylin and eosin staining (H&E) results indicated that it increased micro vessel density (MVD) and decreased neutrophil numbers. Besides, kits detection showed that it improved anti-oxidant stress factors activities and reduced pro-oxidant stress factors contents. Moreover, immunofluorescence and Western blot results demonstrated that it upregulated the expression of pro-angiogenic factors, anti-apoptotic proteins, pro-autophagic proteins, mitogen-activated protein kinase 1/2 (MEK1/2) and extracellular signal-regulated kinases 1/2 (ERK1/2) and downregulated the expression of pro-inflammatory cytokines, pro-apoptotic proteins and anti-autophagic proteins. The specific inhibitor U0126 of MEK/ERK pathway partially reversed these effects. Overall, Myrtenol promoted angiogenesis, reduced oxidative stress, ameliorated inflammation, inhibited apoptosis and upregulated autophagy via MEK/ERK pathway to promote flap survival.
Collapse
Affiliation(s)
- Jialong Yang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Shenchuyue Ni
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325000, China
| | - An Wang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Kaitao Wang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jiapeng Deng
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zijie Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, The First School of Clinical Medical, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yizhen Cai
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yiqi Chen
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Guodong Chen
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Dingsheng Lin
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
24
|
Hua X, Xiang D, Xu J, Zhang S, Wu S, Tian Z, Zhu J, Huang C. ISO-upregulated BECN1 specifically promotes LC3B-dependent autophagy and anticancer activity in invasive bladder cancer. Transl Oncol 2025; 51:102178. [PMID: 39489089 PMCID: PMC11565558 DOI: 10.1016/j.tranon.2024.102178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/18/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
Isorhapontigenin (ISO), an active compound isolated from the Chinese herb Gnetum Cleistostachyum, exhibited strong preventive and therapeutic effects on bladder cancer (BC) both in vitro and in vivo. Our previous studies revealed that ISO-induced autophagy is crucial for its anti-cancer activity. However, the underlying mechanism remains unclear. Here, we showed that BECN1, an important autophagic protein, was induced by ISO treatment and played crucial roles in ISO-induced late phase of LC3B-dependent, and LC3A-independent autophagy, as well as anti-cancer activity. Downregulation of BECN1 was observed in human BCs and BBN-induced mouse invasive BC tissues, whereas co-treatment with ISO completely reversed BECN1 downregulation in BBN-induced mouse invasive BCs. Consistently, ISO treatment significantly increased BECN1 expression in vitro in a dose- and time-dependent manner. Depletion of BECN1 significantly impaired LC3B-dependent autophagy following ISO treatment, as well as abolished the inhibitory effect of ISO on anchorage-independent growth of human BC cells. Mechanistic studies revealed that BECN1 induction was mediated by ISO downregulation of c-Myc, which resulted in miR-613 reduction, in turn leading to increased NCL translation and further promoting NCL binding to BECN1 mRNA, subsequently stabilizing BECN1 mRNA. In conclusion, our results demonstrate that by activating c-Myc/miR-613/NCL axis, ISO treatment results in BECN1 posttranscriptional upregulation, which specifically initiates LC3B-dependent autophagy and anti-cancer activity. Our findings further strengths our application of ISO for therapy of high-grade invasive BC (HGIBC) patients.
Collapse
Affiliation(s)
- Xiaohui Hua
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; School of Laboratory Medicine and Life Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Daimin Xiang
- Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jiheng Xu
- School of Laboratory Medicine and Life Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shouyue Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Shuai Wu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Zhongxian Tian
- School of Laboratory Medicine and Life Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Junlan Zhu
- School of Laboratory Medicine and Life Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chuanshu Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; School of Laboratory Medicine and Life Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
25
|
Jin Y, Zhao L, Zhang Y, Chen T, Shi H, Sun H, Ding S, Chen S, Cao H, Zhang G, Li Q, Gao J, Xiao M, Sheng C. BIN1 deficiency enhances ULK3-dependent autophagic flux and reduces dendritic size in mouse hippocampal neurons. Autophagy 2025; 21:223-242. [PMID: 39171951 DOI: 10.1080/15548627.2024.2393932] [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: 07/14/2023] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024] Open
Abstract
Genome-wide association studies identified variants around the BIN1 (bridging integrator 1) gene locus as prominent risk factors for late-onset Alzheimer disease. In the present study, we decreased the expression of BIN1 in mouse hippocampal neurons to investigate its neuronal function. Bin1 knockdown via RNAi reduced the dendritic arbor size in primary cultured hippocampal neurons as well as in mature Cornu Ammonis 1 excitatory neurons. The AAV-mediated Bin1 RNAi knockdown also generated a significant regional volume loss around the injection sites at the organ level, as revealed by 7-Tesla structural magnetic resonance imaging, and an impaired spatial reference memory performance in the Barnes maze test. Unexpectedly, Bin1 knockdown led to concurrent activation of both macroautophagy/autophagy and MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1). Autophagy inhibition with the lysosome inhibitor chloroquine effectively mitigated the Bin1 knockdown-induced dendritic regression. The subsequent molecular studydemonstrated that increased expression of ULK3 (unc-51 like kinase 3), which is MTOR-insensitive, supported autophagosome formation in BIN1 deficiency. Reducing ULK3 activity with SU6668, a receptor tyrosine kinase inhibitor, or decreasing neuronal ULK3 expression through AAV-mediated RNAi, significantly attenuated Bin1 knockdown-induced hippocampal volume loss and spatial memory decline. In Alzheimer disease patients, the major neuronal isoform of BIN1 is specifically reduced. Our work suggests this reduction is probably an important molecular event that increases the autophagy level, which might subsequently promote brain atrophy and cognitive impairment through reducing dendritic structures, and ULK3 is a potential interventional target for relieving these detrimental effects.Abbreviations: AV: adeno-associated virus; Aβ: amyloid-β; ACTB: actin, beta; AD: Alzheimer disease; Aduk: Another Drosophila Unc-51-like kinase; AKT1: thymoma viral proto-oncogene 1; AMPK: AMP-activated protein kinase; AP: autophagosome; BafA1: bafilomycin A1; BDNF: brain derived neurotrophic factor; BIN1: bridging integrator 1; BIN1-iso1: BIN1, isoform 1; CA1: cornu Ammonis 1; CA3: cornu Ammonis 3; CLAP: clathrin and adapter binding; CQ: chloroquine; DMEM: Dulbecco's modified Eagle medium; EGFP: enhanced green fluorescent protein; GWAS: genome-wide association study; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MRI: magnetic resonance imaging; MTOR; mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; PET: positron emission tomography; qRT-PCR: real-time quantitative reverse transcription PCR; ROS: reactive oxygen species; RPS6KB1: ribosomal protein S6 kinase B1; TFEB: transcription factor EB; ULK1: unc-51 like kinase 1; ULK3: unc-51 like kinase 3.
Collapse
Affiliation(s)
- Yuxi Jin
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lin Zhao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yanli Zhang
- Department of Psychiatry, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Tingzhen Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huili Shi
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Huaiqing Sun
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shixin Ding
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Sijia Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Haifeng Cao
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Guannan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Qian Li
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Chengyu Sheng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Yuan S, Zhao E. Recent advances of lipid droplet-targeted AIE-active materials for imaging, diagnosis and therapy. Biosens Bioelectron 2025; 267:116802. [PMID: 39332250 DOI: 10.1016/j.bios.2024.116802] [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: 07/22/2024] [Revised: 08/25/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Lipid droplets (LDs) are cellular organelles specialized in the storage and regulating the release of lipids critical for energy metabolism. As investigation on LDs deepens, the complex biological functions of LDs are revealed and their relationships with various diseases such as atherosclerosis, fatty liver, obesity, and cancer are uncovered. Fluorescence-based techniques with simple operations, visible results and high non-invasiveness are ideal tools for investigating LD-related biological processes and diseases. Materials with aggregation-induced emission (AIE) characteristics have emerged as promising candidates for investigating LDs due to their high signal-to-noise ratio (S/N), strong photostability, and large Stokes shift. This review discusses the principles and advantages of LD-targeting AIE probes for imaging LDs, diagnosis of LD-associated diseases including atherosclerotic plaques, liver diseases, acute kidney diseases and cancer, therapies with LD-targeting AIE-active photosensitizers and other relevant fields in the past five years. Through typical examples, we illustrate the status of investigating LD-related imaging, diagnosis of diseases and therapy with AIE materials. This review is expected to attract attentions from scientists with different research backgrounds and contribute to the further development of LD-targeting AIE materials.
Collapse
Affiliation(s)
- Sisi Yuan
- School of Science, Harbin Institute of Technology, Shenzhen, Guangdong, 518055, China
| | - Engui Zhao
- School of Science, Harbin Institute of Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
27
|
Raza S, Siddiqui JA, Srivastava A, Chattopadhyay N, Sinha RA, Chakravarti B. Autophagy as a Therapeutic Target in Breast Tumors: The Cancer stem cell perspective. AUTOPHAGY REPORTS 2024; 3:27694127.2024.2358648. [PMID: 39006309 PMCID: PMC7616179 DOI: 10.1080/27694127.2024.2358648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 07/16/2024]
Abstract
Breast cancer is a heterogeneous disease, with a subpopulation of tumor cells known as breast cancer stem cells (BCSCs) with self-renewal and differentiation abilities that play a critical role in tumor initiation, progression, and therapy resistance. The tumor microenvironment (TME) is a complex area where diverse cancer cells reside creating a highly interactive environment with secreted factors, and the extracellular matrix. Autophagy, a cellular self-digestion process, influences dynamic cellular processes in the tumor TME integrating diverse signals that regulate tumor development and heterogeneity. Autophagy acts as a double-edged sword in the breast TME, with both tumor-promoting and tumor-suppressing roles. Autophagy promotes breast tumorigenesis by regulating tumor cell survival, migration and invasion, metabolic reprogramming, and epithelial-mesenchymal transition (EMT). BCSCs harness autophagy to maintain stemness properties, evade immune surveillance, and resist therapeutic interventions. Conversely, excessive, or dysregulated autophagy may lead to BCSC differentiation or cell death, offering a potential avenue for therapeutic exploration. The molecular mechanisms that regulate autophagy in BCSCs including the mammalian target of rapamycin (mTOR), AMPK, and Beclin-1 signaling pathways may be potential targets for pharmacological intervention in breast cancer. This review provides a comprehensive overview of the relationship between autophagy and BCSCs, highlighting recent advancements in our understanding of their interplay. We also discuss the current state of autophagy-targeting agents and their preclinical and clinical development in BCSCs.
Collapse
Affiliation(s)
- Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | - Jawed Akhtar Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Anubhav Srivastava
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | - Bandana Chakravarti
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| |
Collapse
|
28
|
Meng Y, Shao H, Wu L. Exosomes derived from cancer-associated fibrolasts mediated ciplatin resistance. Cytojournal 2024; 21:74. [PMID: 39916998 PMCID: PMC11801690 DOI: 10.25259/cytojournal_149_2024] [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: 08/07/2024] [Accepted: 11/20/2024] [Indexed: 02/09/2025] Open
Abstract
Objective Nasopharyngeal carcinoma (NPC), a highly invasive form of head and neck cancer, carries a significant risk of distant metastasis. NPC is particularly prevalent in Asia and has a high incidence in southern China. Cisplatin-diamminedichloroplatinum (DDP), a chemotherapy agent, is commonly employed in NPC treatment. Despite DDP's efficacy, many patients eventually develop resistance to it over the course of their therapy, which significantly hinders treatment outcomes. Cancer-associated fibroblasts (CAFs) are key components of the tumor micro-environment and contribute to tumor progression and chemotherapy resistance. Exosomes secreted by CAFs serve as crucial mediators of intercellular communication and participate in modulating diverse biological processes. This study aimed to explore how exosomes derived from CAFs contribute to DDP resistance in NPC. Material and Methods An in vitro coculture system was used to simulate the interaction between CAFs and NPC cells, and exosomes secreted by CAFs were isolated and characterized. The expression of autophagy hallmark proteins was detected by Western blot and quantitative real-time polymerase chain reaction. Autophagy intensity was quantified using monodansylcadaverine staining, and cell proliferation was assessed by colony formation assays and methylthiazolyldiphenyl-tetrazolium assays. NPC cells were treated with autophagy inducers (rapamycin), and the expression of Ras homologue enriched in brain (Rheb), mammalian target of rapamycin complex (mTORC1), and UNC51-like kinase was detected. Immunofluorescence was used to determine the cellular localization and expression intensity of mTORC1, and the effect on DDP sensitivity was evaluated through cell proliferation rates. In addition, the exosome-mediated resistance mechanism was further validated using an in vivo xenograft tumor model. Results Coculture of CAFs with NPC cells significantly promoted the proliferation of NPC cells (P < 0.01), significantly elevated the IC50 value of DDP (P < 0.01), and elevated the resistance of NPC cells to DDP. CAF-derived exosomes elevated autophagy hallmark proteins light chain 3B-II, Beclin, and increased the autophagy intensity (P < 0.01). CAF-derived exosomes promoted autophagy by inhibiting mTORC1 (P < 0.01). In the in vitro model, exosomes promoted the growth of tumor tissues (P < 0.01), and the inhibition of exosome secretion reversed the promotion effect of autophagy (P < 0.01) and elevated the sensitivity of NPC cells to DDP. Conclusion CAF-derived exosomes promote protective autophagy in NPC cells through the Rheb/mTOR axis, and result in DDP resistance in NPC.
Collapse
Affiliation(s)
- Yiyu Meng
- Department of Otolaryngology, Lishui People’s Hospital, Lishui, China
| | - Hui Shao
- Department of Ophthalmology, Lishui People’s Hospital, Lishui, China
| | - Lijun Wu
- Department of Otolaryngology, Lishui People’s Hospital, Lishui, China
| |
Collapse
|
29
|
Lei J, Zhu Q, Guo J, Chen J, Qi L, Cui M, Jiang Z, Fan C, Wang L, Lai T, Jin Y, Si L, Liu Y, Yang Q, Bao D, Guo R. TEFM facilitates uterine corpus endometrial carcinoma progression by activating ROS-NFκB pathway. J Transl Med 2024; 22:1151. [PMID: 39731053 DOI: 10.1186/s12967-024-05833-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/31/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Mitochondrial transcription elongation factor (TEFM) is a recently discovered factor involved in mitochondrial DNA replication and transcription. Previous studies have reported that abnormal TEFM expression can disrupt the assembly of mitochondrial respiratory chain and thus mitochondrial function. However, the role of TEFM on Uterine corpus endometrial carcinoma (UCEC) progression remains unclear. The present study aims to investigate the expression of TEFM in tumor tissue of UCEC and the effect of abnormal TEFM expression on malignant phenotype of UCEC cells. METHODS The expressions of TEFM were measured in tumor tissues and cell lines of UCEC by immunohistochemistry, Western blotting, and real-time quantitative PCR assays. Besides, the effects of TEFM knockdown or overexpression on UCEC cell growth, metastasis, apoptosis, and autophagy were also determined using EdU, colony formation, flow cytometry, TUNEL, and transmission electron microscopy assays. Xenograft model was used to confirm the role of TEFM on proliferative potential of UECE cells in vivo. RESULTS Our bioinformatics analysis of CPTAC data showed that TEFM is abnormally overexpressed in UCEC and its upregulation was significantly associated with poor survival of patients with UCEC. We found that TEFM upregulation significantly promoted the growth and metastasis of UCEC cells. Mechanically, TEFM upregulation impaired the function of mitochondria, decreased their membrane potential and activated the AKT-NFκB pathway by promoting reactive oxygen species (ROS) production, leading to enhanced intracellular autophagy and thus UCEC growth and metastasis. CONCLUSION This study demonstrates that TEFM positively regulates autophagy to promote the growth and metastasis of UCEC cells, which provides a potential prognostic biomarker and therapeutic target for the treatment of UCEC.
Collapse
Affiliation(s)
- Jia Lei
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Radiotheraphy Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Qingguo Zhu
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Jianghao Guo
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Jiaxing Chen
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Lixia Qi
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Mengmeng Cui
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Zhixiong Jiang
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Chunhui Fan
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Lin Wang
- Radiotheraphy Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Tianjiao Lai
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Yuxi Jin
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Lulu Si
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Yana Liu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Qi Yang
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China.
- School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Dengke Bao
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China.
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, Henan, 475004, China.
| | - Ruixia Guo
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
30
|
Mengistu BA, Tsegaw T, Demessie Y, Getnet K, Bitew AB, Kinde MZ, Beirhun AM, Mebratu AS, Mekasha YT, Feleke MG, Fenta MD. Comprehensive review of drug resistance in mammalian cancer stem cells: implications for cancer therapy. Cancer Cell Int 2024; 24:406. [PMID: 39695669 DOI: 10.1186/s12935-024-03558-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer remains a significant global challenge, and despite the numerous strategies developed to advance cancer therapy, an effective cure for metastatic cancer remains elusive. A major hurdle in treatment success is the ability of cancer cells, particularly cancer stem cells (CSCs), to resist therapy. These CSCs possess unique abilities, including self-renewal, differentiation, and repair, which drive tumor progression and chemotherapy resistance. The resilience of CSCs is linked to certain signaling pathways. Tumors with pathway-dependent CSCs often develop genetic resistance, whereas those with pathway-independent CSCs undergo epigenetic changes that affect gene regulation. CSCs can evade cytotoxic drugs, radiation, and apoptosis by increasing drug efflux transporter activity and activating survival mechanisms. Future research should prioritize the identification of new biomarkers and signaling molecules to better understand drug resistance. The use of cutting-edge approaches, such as bioinformatics, genomics, proteomics, and nanotechnology, offers potential solutions to this challenge. Key strategies include developing targeted therapies, employing nanocarriers for precise drug delivery, and focusing on CSC-targeted pathways such as the Wnt, Notch, and Hedgehog pathways. Additionally, investigating multitarget inhibitors, immunotherapy, and nanodrug delivery systems is critical for overcoming drug resistance in cancer cells.
Collapse
Affiliation(s)
- Bemrew Admassu Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tirunesh Tsegaw
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayew Demessie
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Kalkidan Getnet
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebe Belete Bitew
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Mebrie Zemene Kinde
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Asnakew Mulaw Beirhun
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yesuneh Tefera Mekasha
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melaku Getahun Feleke
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melkie Dagnaw Fenta
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
31
|
Zhu Y, He Y, Gan R. Wnt Signaling in Hepatocellular Carcinoma: Biological Mechanisms and Therapeutic Opportunities. Cells 2024; 13:1990. [PMID: 39682738 PMCID: PMC11640042 DOI: 10.3390/cells13231990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/19/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC), characterized by significant morbidity and mortality rates, poses a substantial threat to human health. The expression of ligands and receptors within the classical and non-classical Wnt signaling pathways plays an important role in HCC. The Wnt signaling pathway is essential for regulating multiple biological processes in HCC, including proliferation, invasion, migration, tumor microenvironment modulation, epithelial-mesenchymal transition (EMT), stem cell characteristics, and autophagy. Molecular agents that specifically target the Wnt signaling pathway have demonstrated significant potential for the treatment of HCC. However, the precise mechanism by which the Wnt signaling pathway interacts with HCC remains unclear. In this paper, we review the alteration of the Wnt signaling pathway in HCC, the mechanism of Wnt pathway action in HCC, and molecular agents targeting the Wnt pathway. This paper provides a theoretical foundation for identifying molecular agents targeting the Wnt pathway in hepatocellular carcinoma.
Collapse
Affiliation(s)
| | | | - Runliang Gan
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421001, China; (Y.Z.); (Y.H.)
| |
Collapse
|
32
|
Guo Y, Tian Y, Xia P, Zhou X, Hu X, Guo Z, Ji P, Yuan X, Fu D, Yin K, Shen R, Wang D. Exploring the Function of OPTN From Multiple Dimensions. Cell Biochem Funct 2024; 42:e70029. [PMID: 39670654 DOI: 10.1002/cbf.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
Autophagy is an essential intracellular degradation system responsible for delivering cytoplasmic components to lysosomes. Within this intricate process, optineurin (OPTN), an autophagy receptor, has attracted extensive attention due to its multifaceted roles in the autophagy process. OPTN is regulated by various posttranslational modifications and actively participates in numerous signaling pathways and cellular processes. By exploring the regulatory mechanism of OPTN posttranslational modification, we can further understand the critical role of protein posttranslational modification in biological progress, such as autophagy. Additionally, OPTN is implicated in many human diseases, including rheumatoid arthritis, osteoporosis, and infectious diseases. And we delve into the inflammatory pathways regulated by OPTN and clarify how it regulates inflammatory diseases and cancer. We aim to enhance the understanding of OPTN's multifaceted functions in cellular processes and its implications in the pathogenesis of inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Yanan Guo
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Yixiao Tian
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Peng Xia
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xinyue Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaohui Hu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Zhao Guo
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Pengfei Ji
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xinyi Yuan
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Daosen Fu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Keyu Yin
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Rong Shen
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Degui Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
33
|
Peng F, He R, Liu Y, Xie Y, Xiong G, Li X, Wang M, Zhao C, Zhang H, Xu S, Qin R. MiR-200b-3p elevates 5-FU sensitivity in cholangiocarcinoma cells via autophagy inhibition by targeting KLF4. Noncoding RNA Res 2024; 9:1098-1110. [PMID: 39022678 PMCID: PMC11254509 DOI: 10.1016/j.ncrna.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 07/20/2024] Open
Abstract
Cholangiocarcinoma is one of the most lethal human cancers, and chemotherapy failure is a major cause of recurrence and poor prognosis. We previously demonstrated that miR-200 family members are downregulated in clinical samples of cholangiocarcinoma and inhibit cholangiocarcinoma tumorigenesis and metastasis. However, the role of differentially expressed miR-200b-3p in 5-fluorouracil chemosensitivity remains unclear. Here, we examined how miR-200b-3p modulates 5-fluorouracil chemosensitivity in cholangiocarcinoma. We observed that miR-200b-3p was associated with 5-fluorouracil sensitivity in cholangiocarcinoma and increased 5-fluorouracil-induced mitochondrial apoptosis in cholangiocarcinoma cells. Mechanistically, miR-200b-3p suppressed autophagy in cholangiocarcinoma cells to mediate 5-fluorouracil sensitivity. Further, we identified KLF4 as an essential target of miR-200b-3p in cholangiocarcinoma. Notably, the miR-200b-3p/KLF4/autophagy pathway augmented the chemosensitivity of cholangiocarcinoma cells to 5-fluorouracil. Our findings underscore the key role of miR-200b-3p in chemosensitivity to 5-fluorouracil and highlight the miR-200b-3p/KLF4/autophagy axis as a potential therapeutic target for cholangiocarcinoma.
Collapse
Affiliation(s)
- Feng Peng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ruizhi He
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuhui Liu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Xie
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guangbing Xiong
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xu Li
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunle Zhao
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hang Zhang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Simiao Xu
- Division of Endocrinology, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Branch of National Clinical Research Center for Metabolic Disease, Wuhan, Hubei, 430030, China
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
34
|
Asghari N, Saei AK, Cordani M, Nayeri Z, Moosavi MA. Drug repositioning identifies potential autophagy inhibitors for the LIR motif p62/SQSTM1 protein. Comput Biol Chem 2024; 113:108235. [PMID: 39369612 DOI: 10.1016/j.compbiolchem.2024.108235] [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: 07/21/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Autophagy is a critical cellular process for degrading damaged organelles and proteins under stressful conditions and has casually been shown to contribute to tumor survival and drug resistance. Sequestosome-1 (SQSTM1/p62) is an autophagy receptor that interacts with its binding partners via the LC3-interacting region (LIR). The p62 protein has been a highly researched target for its critical role in selective autophagy. In this study, we aimed to identify FDA-approved drugs that bind to the LIR motif of p62 and inhibit its LIR function, which could be useful targets for modulating autophagy. To this, the homology model of the p62 protein was predicted using biological data, and docking analysis was performed using Molegro Virtual Docker and PyRx softwares. We further assessed the toxicity profile of the drugs using the ProTox-II server and performed dynamics simulations on the effective candidate drugs identified. The results revealed that the kanamycin, velpatasvir, verteporfin, and temoporfin significantly decreased the binding of LIR to the p62 protein. Finally, we experimentally confirmed that Kanamycin can inhibit autophagy-associated acidic vesicular formation in breast cancer MCF-7 and MDA-MB 231 cells. These repositioned drugs may represent novel autophagy modulators in clinical management, warranting further investigation.
Collapse
Affiliation(s)
- Narjes Asghari
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box 14965/161, Tehran, Iran
| | - Ali Kian Saei
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box 14965/161, Tehran, Iran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, Madrid 28040, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid 28040, Spain
| | - Zahra Nayeri
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box 14965/161, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box 14965/161, Tehran, Iran.
| |
Collapse
|
35
|
Gao S, Wang X, Huang Y, You L. Calreticulin-driven autophagy enhances cell proliferation in laryngeal squamous cell carcinoma. Tissue Cell 2024; 91:102603. [PMID: 39550898 DOI: 10.1016/j.tice.2024.102603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Calreticulin (CALR) is a multifunctional calcium-binding protein. Recent studies have revealed that CALR contributes to tumor development and promotes cancer cell proliferation. However, how CALR affects the development of laryngeal squamous cell carcinoma (LSCC) remains mysterious. Thus, this study aimed to explore the effect of CALR on LSCC development and uncover its underlying mechanisms. METHODS CALR expression in LSCC cell lines and tissues was examined by qRT-PCR and western blot analysis and its functional role was detected via in vivo and in vitro assays. Cell proliferation was discriminated with CCK-8 and colony formation assays, while apoptosis was analyzed using flow cytometry. Autophagy levels were measured via LC3 immunofluorescence, and western blot assay was conducted to assess apoptosis- and autophagy-related proteins. Additionally, a mouse xenograft model was employed to determine the impact of CALR knockdown on tumor growth. RESULTS We found that CALR knockdown reduced LSCC cell viability and proliferation while enhancing apoptosis, whereas CALR overexpression showed opposite effects. In vivo experiments verified that CALR knockdown suppressed tumor growth. In addition, elevated CALR expression induced autophagy in LSCC cells, while autophagy inhibitor 3-MA (2.5 mM) reversed the anti-apoptosis effects of CALR overexpression. CONCLUSION Our study identifies CALR as an oncogene in LSCC, where it promotes tumor progression by inducing autophagy and inhibiting apoptosis. Targeting CALR or modulating autophagy may represent novel therapeutic strategies for LSCC.
Collapse
Affiliation(s)
- Shufeng Gao
- Department of ENT & HN Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China.
| | - Xintao Wang
- Department of ENT & HN Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Yun Huang
- Department of ENT & HN Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Longgui You
- Department of ENT & HN Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| |
Collapse
|
36
|
Pimentel JM, Zhou JY, Wu GS. Autophagy and cancer therapy. Cancer Lett 2024; 605:217285. [PMID: 39395780 DOI: 10.1016/j.canlet.2024.217285] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Autophagy is an intracellular degradation process that sequesters cytoplasmic components in double-membrane vesicles known as autophagosomes, which are degraded upon fusion with lysosomes. This pathway maintains the integrity of proteins and organelles while providing energy and nutrients to cells, particularly under nutrient deprivation. Deregulation of autophagy can cause genomic instability, low protein quality, and DNA damage, all of which can contribute to cancer. Autophagy can also be overactivated in cancer cells to aid in cancer cell survival and drug resistance. Emerging evidence indicates that autophagy has functions beyond cargo degradation, including roles in tumor immunity and cancer stem cell survival. Additionally, autophagy can also influence the tumor microenvironment. This feature warrants further investigation of the role of autophagy in cancer, in which autophagy manipulation can improve cancer therapies, including cancer immunotherapy. This review discusses recent findings on the regulation of autophagy and its role in cancer therapy and drug resistance.
Collapse
Affiliation(s)
- Julio M Pimentel
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA; Institutional Research Academic Career Development Award Program, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jun Ying Zhou
- Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, 48201, USA; Department of Oncology, Wayne State University, Detroit, MI, 48201, USA
| | - Gen Sheng Wu
- Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, 48201, USA; Department of Oncology, Wayne State University, Detroit, MI, 48201, USA; Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
37
|
Wang X, Fang L, Xiao L, Zhong G, Han M, Wang B, Ren J, Zang Y. Research on the effect of LAMP1 in the development and progression of ccRCC and its potential mechanism with LC3C-mediated autophagy. Front Immunol 2024; 15:1494005. [PMID: 39669571 PMCID: PMC11634794 DOI: 10.3389/fimmu.2024.1494005] [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: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Background Lysomembrane-associated protein 1 (LAMP1), known to exhibit differential expression in various tumor types and play a crucial role in the development of tumors. Clear cell Renal Cell Carcinoma (ccRCC) is still the most common pathological type of renal carcinoma with poor prognosis. However, the expression of LAMP1 and its underlying molecular mechanism with ccRCC remain elusive. Methods Firstly, the expression of LAMP1 in ccRCC and its clinical significance were analyzed using various databases. Next, Weston Blot was performed to detect the expression of LAMP1 protein in cancer tissues and adjacent tissues from 60 pairs of clinical ccRCC patients. The correlation between LAMP1 expression and different clinical indicators as well as the relationship with patient prognosis was analyzed. Furthermore, molecular cell biology experiments were conducted to validate the effects of LAMP1 gene expression on cell proliferation, invasion and migration. Additionally, we investigated the impact of VHL, a key gene in renal cancer, and LC3C, an autophagy-related gene, on LAMP1 expression through molecular biology experiments to elucidate the potential underlying mechanism. Results Bioinformatics analysis revealed significant underexpression of LAMP1 in ccRCC (P<0.001), which correlated with poorer prognosis. In multivariate survival analysis, LAMP1 emerged as an independent prognostic marker for overall survival(OS)(P<0.05). Analysis of cancer and paracancer tissue samples from ccRCC patients demonstrated significantly lower levels of LAMP1 in tumors compared to paracancerous tissues (P<0.001), confirming its prognostic impact. Cell functionality experiment revealed that elevated LAMP1 inhibited cell proliferation, migration, and invasion. LAMP1 expression remained unchanged during autophagy modulation but decreased with LC3C knockdown and vice versa. Notably, VHL(+) cells expressed less LAMP1 than VHL(-) cells. Conclusions These findings indicate that low expression levels of LAMP1 is associated with poor prognosis in ccRCC. Therefore, LAMP1 emerges as a novel biomarker associated with the diagnosis and prognosis of renal cancer. Furthermore, we have also described the potential mechanism of action of LAMP1 in renal cancer. LAMP1 is a promising target for the treatment of ccRCC.
Collapse
Affiliation(s)
- Xiongbao Wang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Liang Fang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lixiang Xiao
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Guangxin Zhong
- Department of Urology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Minghao Han
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Bingshen Wang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Juchao Ren
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuanwei Zang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
38
|
Hosseinkhani S, Amandadi M, Ghanavatian P, Zarein F, Ataei F, Nikkhah M, Vandenabeele P. Harnessing luciferase chemistry in regulated cell death modalities and autophagy: overview and perspectives. Chem Soc Rev 2024; 53:11557-11589. [PMID: 39417351 DOI: 10.1039/d3cs00743j] [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: 10/19/2024]
Abstract
Regulated cell death is a fate of cells in (patho)physiological conditions during which extrinsic or intrinsic signals or redox equilibrium pathways following infection, cellular stress or injury are coupled to cell death modalities like apoptosis, necroptosis, pyroptosis or ferroptosis. An immediate survival response to cellular stress is often induction of autophagy, a process that deals with removal of aggregated proteins and damaged organelles by a lysosomal recycling process. These cellular processes and their regulation are crucial in several human diseases. Exploiting high-throughput assays which discriminate distinct cell death modalities and autophagy are critical to identify potential therapeutic agents that modulate these cellular responses. In the past few years, luciferase-based assays have been widely developed for assessing regulated cell death and autophagy pathways due to their simplicity, sensitivity, known chemistry, different spectral properties and high-throughput potential. Here, we review basic principles of bioluminescent reactions from a mechanistic perspective, along with their implication in vitro and in vivo for probing cell death and autophagy pathways. These include applying luciferase-, luciferin-, and ATP-based biosensors for investigating regulated cell death modalities. We discuss multiplex bioluminescence platforms which simultaneously distinguish between the various cell death phenomena and cellular stress recovery processes such as autophagy. We also highlight the recent technological achievements of bioluminescent tools for the prediction of drug effectiveness in pathways associated with regulated cell death.
Collapse
Affiliation(s)
- Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mojdeh Amandadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Parisa Ghanavatian
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Fateme Zarein
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farangis Ataei
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Maryam Nikkhah
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Peter Vandenabeele
- Cell Death and Inflammation Unit, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
39
|
Ju S, Singh MK, Han S, Ranbhise J, Ha J, Choe W, Yoon KS, Yeo SG, Kim SS, Kang I. Oxidative Stress and Cancer Therapy: Controlling Cancer Cells Using Reactive Oxygen Species. Int J Mol Sci 2024; 25:12387. [PMID: 39596452 PMCID: PMC11595237 DOI: 10.3390/ijms252212387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer is a multifaceted disease influenced by various mechanisms, including the generation of reactive oxygen species (ROS), which have a paradoxical role in both promoting cancer progression and serving as targets for therapeutic interventions. At low concentrations, ROS serve as signaling agents that enhance cancer cell proliferation, migration, and resistance to drugs. However, at elevated levels, ROS induce oxidative stress, causing damage to biomolecules and leading to cell death. Cancer cells have developed mechanisms to manage ROS levels, including activating pathways such as NRF2, NF-κB, and PI3K/Akt. This review explores the relationship between ROS and cancer, focusing on cell death mechanisms like apoptosis, ferroptosis, and autophagy, highlighting the potential therapeutic strategies that exploit ROS to target cancer cells.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jyotsna Ranbhise
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02453, Republic of Korea;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
40
|
程 永, 沈 亦, 王 学, 李 丹, 樊 春, 古丽巴哈·买买提, 严 媚. [Mechanism by which mycobacterial antigen 85B inhibits autophagy and promotes apoptosis in Hodgkin lymphoma cells]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:1218-1224. [PMID: 39587752 PMCID: PMC11601109 DOI: 10.7499/j.issn.1008-8830.2404153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/05/2024] [Indexed: 11/27/2024]
Abstract
OBJECTIVES To investigate the mechanism by which mycobacterial antigen 85B (Ag85B) inhibits autophagy and promotes apoptosis in Hodgkin lymphoma (HL) cells. METHODS The clinical data and pathological tissue slides were retrospectively collected from 80 HL children and 30 children with reactive lymphadenopathy (control group) treated at the First Affiliated Hospital of Xinjiang Medical University. Immunohistochemical analysis was performed to assess the expression of microtubule-associated protein 1 light chain 3 (LC3), sequestosome 1 (P62/SQSTM1), and Beclin-1 in the pathological tissues of HL and control groups. Human Hodgkin lymphoma cells (HDLM-2) were divided into the HDLM-2 group and the HDLM-2+Ag85B groups (with Ag85B concentrations of 0.5, 1, 2, and 4 μg/mL). The CCK8 method was used to measure HDLM-2 cell proliferation; qRT-PCR was employed to detect the expression of LC3, P62, Beclin-1, Akt, and mTOR mRNA in cells. An apoptosis kit was used to detect cell apoptosis. RESULTS The positive expression of LC3 and Beclin-1 in the HL group were higher than those in the control group (P<0.05), while the positive expression of P62 was lower than that in the control group (P<0.05). In stages III-IV compared to stages I-II, the positive expression of LC3 and Beclin-1 increased, while the positive expression of P62 decreased (P<0.05). Cell experiment results showed that the HDLM-2+Ag85B group had suppressed cell proliferation compared to the HDLM-2 group, with decreased mRNA expression of LC3 and Beclin-1, and increased mRNA expression of P62, PI3K, Akt, and mTOR, leading to increased cell apoptosis. Notably, when Ag85B was at a concentration of 2 μg/mL, it had the strongest effect on HDLM-2 cells after 24 hours (P<0.05). CONCLUSIONS Autophagy is enhanced in children with HL and increases with disease stage. Ag85B can inhibit the proliferation and autophagy of HL tumor cells and promote apoptosis, possibly related to the activation of the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
| | - 亦平 沈
- 哈佛大学医学院波士顿儿童医院,美国波士顿02115
| | | | | | | | | | | |
Collapse
|
41
|
Pizzimenti C, Fiorentino V, Ruggeri C, Franchina M, Ercoli A, Tuccari G, Ieni A. Autophagy Involvement in Non-Neoplastic and Neoplastic Endometrial Pathology: The State of the Art with a Focus on Carcinoma. Int J Mol Sci 2024; 25:12118. [PMID: 39596186 PMCID: PMC11594225 DOI: 10.3390/ijms252212118] [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: 09/27/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Autophagy is a cellular process crucial for maintaining homeostasis by degrading damaged proteins and organelles. It is stimulated in response to stress, recycling nutrients and generating energy for cell survival. In normal endometrium, it suppresses tumorigenesis by preventing toxic accumulation and maintaining cellular homeostasis. It is involved in the cyclic remodelling of the endometrium during the menstrual cycle and contributes to decidualisation for successful pregnancy. Such a process is regulated by various signalling pathways, including PI3K/AKT/mTOR, AMPK/mTOR, and p53. Dysregulation of autophagy has been associated with benign conditions like endometriosis and endometrial hyperplasia but also with malignant neoplasms such as endometrial carcinoma. In fact, it has emerged as a crucial player in endometrial carcinoma biology, exhibiting a dual role in both tumour suppression and tumour promotion, providing nutrients during metabolic stress and allowing cancer cell survival. It also regulates cancer stem cells, metastasis and therapy resistance. Targeting autophagy is therefore a promising therapeutic strategy in endometrial carcinoma and potential for overcoming resistance to standard treatments. The aim of this review is to delve into the intricate details of autophagy's role in endometrial pathology, exploring its mechanisms, signalling pathways and potential therapeutic implications.
Collapse
Affiliation(s)
- Cristina Pizzimenti
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| | - Vincenzo Fiorentino
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| | - Chiara Ruggeri
- Section of Gynecology and Obstetrics, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.R.); (A.E.)
| | - Mariausilia Franchina
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| | - Alfredo Ercoli
- Section of Gynecology and Obstetrics, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.R.); (A.E.)
| | - Giovanni Tuccari
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| | - Antonio Ieni
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| |
Collapse
|
42
|
Huang Y, Luo G, Peng K, Song Y, Wang Y, Zhang H, Li J, Qiu X, Pu M, Liu X, Peng C, Neculai D, Sun Q, Zhou T, Huang P, Liu W. Lactylation stabilizes TFEB to elevate autophagy and lysosomal activity. J Cell Biol 2024; 223:e202308099. [PMID: 39196068 PMCID: PMC11354204 DOI: 10.1083/jcb.202308099] [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: 08/21/2023] [Revised: 02/04/2024] [Accepted: 04/03/2024] [Indexed: 08/29/2024] Open
Abstract
The transcription factor TFEB is a major regulator of lysosomal biogenesis and autophagy. There is growing evidence that posttranslational modifications play a crucial role in regulating TFEB activity. Here, we show that lactate molecules can covalently modify TFEB, leading to its lactylation and stabilization. Mechanically, lactylation at K91 prevents TFEB from interacting with E3 ubiquitin ligase WWP2, thereby inhibiting TFEB ubiquitination and proteasome degradation, resulting in increased TFEB activity and autophagy flux. Using a specific antibody against lactylated K91, enhanced TFEB lactylation was observed in clinical human pancreatic cancer samples. Our results suggest that lactylation is a novel mode of TFEB regulation and that lactylation of TFEB may be associated with high levels of autophagy in rapidly proliferating cells, such as cancer cells.
Collapse
Affiliation(s)
- Yewei Huang
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Gan Luo
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Kesong Peng
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yue Song
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yusha Wang
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Hongtao Zhang
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jin Li
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Xiangmin Qiu
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Maomao Pu
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Xinchang Liu
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Chao Peng
- National Center for Protein Science Shanghai, Institute of Biochemistry and Cell Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dante Neculai
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qiming Sun
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Tianhua Zhou
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Pintong Huang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
43
|
Ferraresi A, Ghezzi I, Salwa A, Esposito A, Dhanasekaran DN, Isidoro C. NKX3-2 Induces Ovarian Cancer Cell Migration by HDAC6-Mediated Repositioning of Lysosomes and Inhibition of Autophagy. Cells 2024; 13:1816. [PMID: 39513923 PMCID: PMC11544992 DOI: 10.3390/cells13211816] [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: 08/28/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Several soluble factors secreted by the stromal cells and cancer cells within the tumor microenvironment facilitate the progression and invasiveness of ovarian cancer. In ovarian cancer cells, lysophosphatidic acid (LPA) modulates the transcriptome profile and promotes cell invasiveness by the downregulation of autophagy. Here, we further elucidate this mechanism by focusing on the molecular and cellular events regulating autophagy. Transcriptomic and Western blotting analyses revealed NKX3-2, a transcriptional factor, to be among the genes hyperexpressed in LPA-stimulated ovarian cancer cells. Bioinformatic analyses revealed that in ovarian cancer patients, the expression of NKX3-2 positively correlates with genes involved in cell motility and migration, while it negatively correlates with macromolecular catabolic pathways. In various ovarian cancer cell lines, NKX3-2 silencing abrogated LPA-induced cell migration. Mechanistically, this effect is linked to the restoration of the HDAC6-mediated relocation of the lysosomes in the para-golgian area, and this results in an increase in autolysosome formation and the overall upregulation of autophagy. Silencing the expression of ATG7 or BECN1, two autophagy genes, rescued the migratory phenotype of the NKX3-2-silenced ovarian cancer cells. Taken together, these data reveal the mechanism by which the LPA-NKX3-2 axis promotes the invasiveness of ovarian cancer cells and supports the possibility of targeting NKX3-2 to reduce the migratory capacity of cancer cells in response to a permissive microenvironment.
Collapse
Affiliation(s)
- Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (I.G.); (A.S.); (A.E.)
| | - Ian Ghezzi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (I.G.); (A.S.); (A.E.)
| | - Amreen Salwa
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (I.G.); (A.S.); (A.E.)
| | - Andrea Esposito
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (I.G.); (A.S.); (A.E.)
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (I.G.); (A.S.); (A.E.)
| |
Collapse
|
44
|
Yamakawa T, Tanaka A, Miron C, Nakamura K, Kajiyama H, Toyokuni S, Mizuno M, Hori M, Tanaka H. Effects of autophagy on the selective death of human breast cancer cells exposed to plasma-activated Ringer's lactate solution. Free Radic Res 2024; 58:758-769. [PMID: 39625787 DOI: 10.1080/10715762.2024.2433965] [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: 07/07/2024] [Revised: 10/23/2024] [Accepted: 11/17/2024] [Indexed: 12/28/2024]
Abstract
Plasma-activated Ringer's lactate (PAL) solution prepared by irradiating an intravenous solution with a non-equilibrium atmospheric pressure plasma is a potential new cancer therapy having no side effects. However, the induction of autophagy to avoid cell death has been confirmed to occur following exposure to PAL solution. It is thought that the antitumor effect of PAL solution could be weakened by this process, which is meant to maintain homeostasis in cells and assists tumorigenesis. Thus, it would be helpful to devise PAL-based cancer therapies that inhibit autophagy. Unfortunately, it is not yet clear which substances in PAL solution promote autophagy. The present work examined the mechanism by which PAL solution induces autophagy when treating MCF-7 human breast cancer cells. Autophagy was found to be temporarily induced upon exposure to PAL solution, suggesting that this effect contributes to cell proliferation. Although autophagy is associated with reactive oxygen and nitrogen species and/or acidic environments, in this study, significant autophagy was observed using a PAL solution diluted 1/256x without these stressors. Acetate, glyoxylate and 2,3-dimethyltartrate in the PAL solution were determined to promote autophagy. Interestingly, 2,3-dimethyltartrate was found to either induce cell death or autophagy depending on the concentration.
Collapse
Affiliation(s)
- Taishi Yamakawa
- Department of Electronics, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Ayako Tanaka
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Camelia Miron
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Kae Nakamura
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinya Toyokuni
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaaki Mizuno
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Masaru Hori
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Hiromasa Tanaka
- Department of Electronics, Graduate School of Engineering, Nagoya University, Nagoya, Japan
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
45
|
Gholami F, Seyedalipour B, Heidari-Kalvani N, Nabi-Afjadi M, Yaghoubzad-Maleki M, Fathi Z, Alipourfard I, Barjesteh F, Bahreini E. Catharanthine, an anticancer vinca alkaloid: an in silico and in vitro analysis of the autophagic system as the major mechanism of cell death in liver HepG2 cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8879-8892. [PMID: 38856913 DOI: 10.1007/s00210-024-03191-8] [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: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 06/11/2024]
Abstract
Catharanthine, a component of the anticancer drug vinblastine along with vindoline, disrupts the cell cycle by interfering with mitotic spindle formation. Apart from their antioxidant properties, vinca alkaloids like catharanthine inhibit phosphodiesterase activity and elevate intracellular cAMP levels. The aim of this study was to investigate how catharantine affects apoptosis and autophagy. This study conducted experiments on HepG2 liver carcinoma cells with varying doses of catharanthine to evaluate cell death rates and viability and determine the IC50 concentration via MTT assays. The apoptotic and autophagic effects of catharanthine were assessed using flow cytometry with annexin V and PI staining, while the expression of autophagy-related genes was analyzed through quantitative PCR. Additionally, molecular docking and molecular dynamics simulations were employed to further investigate catharanthine's impact on autophagy mechanisms. The study showed that catharanthine reduced oxidative stress and triggered apoptosis in HepG2 cells in a dose-dependent manner. Catharanthine also upregulated the expression of autophagy-related genes like LC3, Beclin1, and ULK1. Notably, catharanthine increased sirtuin-1 levels, a known autophagy inducer, while decreasing Akt expression compared to untreated cells. Molecular docking results indicated rapamycin had a stronger binding affinity with FRB (-10.7 KJ/mol-1) than catharanthine (-7.3 KJ/mol-1). Additionally, molecular dynamics simulations revealed that catharanthine interacted effectively with the FRB domain of mTOR, displaying stability and a strong binding affinity, although not as potent as rapamycin. In summary, besides its cytotoxic and pro-apoptotic effects, catharanthine activates autophagy signaling pathways and induces autophagic necrosis by inhibiting mTOR.
Collapse
Affiliation(s)
- Farnoosh Gholami
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Bagher Seyedalipour
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Nafiseh Heidari-Kalvani
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Yaghoubzad-Maleki
- Division of Biochemistry, Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zeinab Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Fereshte Barjesteh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Wang T, Hu Q, Li B, Fan G, Jing D, Xu J, Hu Y, Dang Q, Ji S, Zhou C, Zhuo Q, Xu X, Qin Y, Yu X, Li Z. Transcription factor EB reprograms branched-chain amino acid metabolism and promotes pancreatic cancer progression via transcriptional regulation of BCAT1. Cell Prolif 2024; 57:e13694. [PMID: 38938061 PMCID: PMC11533072 DOI: 10.1111/cpr.13694] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024] Open
Abstract
Pancreatic cancer cells have a much higher metabolic demand than that of normal cells. However, the abundant interstitium and lack of blood supply determine the lack of nutrients in the tumour microenvironment. Although pancreatic cancer has been reported to supply extra metabolic demand for proliferation through autophagy and other means, the specific regulatory mechanisms have not yet been elucidated. In this study, we focused on transcription factor EB (TFEB), a key factor in the regulation of autophagy, to explore its effect on the phenotype and role in the unique amino acid utilisation pattern of pancreatic cancer cells (PCCs). The results showed that TFEB, which is generally highly expressed in pancreatic cancer, promoted the proliferation and metastasis of PCCs. TFEB knockdown inhibited the proliferation and metastasis of PCCs by blocking the catabolism of branched-chain amino acids (BCAAs). Concerning the mechanism, we found that TFEB regulates the catabolism of BCAAs by regulating BCAT1, a key enzyme in BCAA metabolism. BCAA deprivation alone did not effectively inhibit PCC proliferation. However, BCAA deprivation combined with eltrombopag, a drug targeting TFEB, can play a two-pronged role in exogenous supply deprivation and endogenous utilisation blockade to inhibit the proliferation of pancreatic cancer to the greatest extent, providing a new therapeutic direction, such as targeted metabolic reprogramming of pancreatic cancer.
Collapse
Affiliation(s)
- Ting Wang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Borui Li
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Guixiong Fan
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Desheng Jing
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Junfeng Xu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Yuheng Hu
- Department of Hepatobiliary and Pancreatic SurgeryTenth People's Hospital of Tongji UniversityShanghaiChina
| | - Qin Dang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Shunrong Ji
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Chenjie Zhou
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Qifeng Zhuo
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Xiaowu Xu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Yi Qin
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Zheng Li
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| |
Collapse
|
47
|
Wang F, Liao Q, Qin Z, Li J, Wei Q, Li M, Deng H, Xiong W, Tan M, Zhou M. Autophagy: a critical mechanism of N 6-methyladenosine modification involved in tumor progression and therapy resistance. Cell Death Dis 2024; 15:783. [PMID: 39468015 PMCID: PMC11519594 DOI: 10.1038/s41419-024-07148-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024]
Abstract
N6-Methyladenosine (m6A) is an evolutionarily highly conserved epigenetic modification that affects eukaryotic RNAs, especially mRNAs, and m6A modification is commonly linked to tumor proliferation, progression, and therapeutic resistance by participating in RNA metabolism. Autophagy is an intracellular degradation and recycling biological process by which cells remove damaged organelles, protein aggregates, and other intracellular wastes, and release nutrients to maintain cell survival when energy is scarce. Recent studies have shown that m6A modification plays a critical role in the regulation of autophagy, affecting the initiation of autophagy, the formation and assembly of autophagosomes, and lysosomal function by regulating critical regulatory molecules involved in the process of autophagy. Moreover, autophagy can also affect the expression of the three types of regulators related to m6A, which in turn affects the levels of their target genes via m6A modification. Thus, m6A modification and autophagy form a sophisticated regulatory network through mutual regulation, which plays an important role in tumor progression and therapeutic resistance. In this manuscript, we reviewed the effects of m6A modification on autophagy as well as the effects of autophagy on m6A modification and the roles of the m6A-autophagy axis in tumor progression and therapy resistance. Additionally, we summarized the value and application prospects of key molecules in the m6A-autophagy axis in tumor diagnosis and therapy.
Collapse
Affiliation(s)
- Feiyang Wang
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Qiudi Liao
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Zihao Qin
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jingyi Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Qingqing Wei
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Mengna Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Hongyu Deng
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Ming Tan
- Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, China.
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China.
| |
Collapse
|
48
|
Qin W, Huang J, Zhang M, Xu M, He J, Liu Q. Nanotechnology-Based Drug Delivery Systems for Treating Acute Kidney Injury. ACS Biomater Sci Eng 2024; 10:6078-6096. [PMID: 39226188 PMCID: PMC11480945 DOI: 10.1021/acsbiomaterials.4c01385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Acute kidney injury (AKI) is a disease that is characterized by a rapid decline in renal function and has a relatively high incidence in hospitalized patients. Sepsis, renal hypoperfusion, and nephrotoxic drug exposure are the main causes of AKI. The major therapy measures currently include supportive treatment, symptomatic treatment, and kidney transplantation. These methods are supportive treatments, and their results are not satisfactory. Fortunately, many new treatments that markedly improve the AKI therapy efficiency are emerging. These include antioxidant therapy, ferroptosis therapy, anti-inflammatory therapy, autophagy therapy, and antiapoptotic therapy. In addition, the development of nanotechnology has further promoted therapeutic effects on AKI. In this review, we highlight recent advances in the development of nanocarriers for AKI drug delivery. Emphasis has been placed on the latest developments in nanocarrier modification and design. We also summarize the applications of different nanocarriers in AKI treatment. Finally, the advantages and challenges of nanocarrier applications in AKI are summarized, and several nanomedicines that have been approved for clinical trials to treat diverse kidney diseases are listed.
Collapse
Affiliation(s)
- Wanbing Qin
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Jiaqi Huang
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Manting Zhang
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Mingwei Xu
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Junbing He
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Qinghua Liu
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
- Department
of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 Guangdong, China
- NHC Key
Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong
Provincial Key Laboratory of Nephrology, Guangzhou, 510080 Guangdong, China
| |
Collapse
|
49
|
Wang C, Wang L. Resistance mechanisms and potential therapeutic strategies in relapsed or refractory natural killer/T cell lymphoma. Chin Med J (Engl) 2024; 137:2308-2324. [PMID: 39175124 PMCID: PMC11441923 DOI: 10.1097/cm9.0000000000003152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Indexed: 08/24/2024] Open
Abstract
ABSTRACT Natural killer/T cell lymphoma (NKTCL) is a malignant tumor originating from NK or T cells, characterized by its highly aggressive and heterogeneous nature. NKTCL is predominantly associated with Epstein-Barr virus infection, disproportionately affecting Asian and Latin American populations. Owing to the application of asparaginase and immunotherapy, clinical outcomes have improved significantly. However, for patients in whom first-line treatment fails, the prognosis is exceedingly poor. Overexpression of multidrug resistance genes, abnormal signaling pathways, epigenetic modifications and active Epstein-Barr virus infection may be responsible for resistance. This review summarized the mechanisms of resistance for NKTCL and proposed potential therapeutic approaches.
Collapse
Affiliation(s)
- Chengji Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
50
|
Eity TA, Bhuia MS, Chowdhury R, Ahmmed S, Salehin Sheikh, Akter R, Islam MT. Therapeutic Efficacy of Quercetin and Its Nanoformulation Both the Mono- or Combination Therapies in the Management of Cancer: An Update with Molecular Mechanisms. J Trop Med 2024; 2024:5594462. [PMID: 39380577 PMCID: PMC11461079 DOI: 10.1155/2024/5594462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024] Open
Abstract
Quercetin, a major representative of the flavonol subclass found abundantly in almost all edible vegetables and fruits, showed remarkable therapeutic properties and was beneficial in numerous degenerative diseases by preventing lipid peroxidation. Quercetin is beneficial in different diseases, such as atherosclerosis and chronic inflammation. This study aims to find out the anticancer activities of quercetin and to determine different mechanisms and pathways which are responsible for the anticancer effect. It also revealed the biopharmaceutical, toxicological characteristics, and clinical utilization of quercetin to evaluate its suitability for further investigations as a reliable anticancer drug. All of the relevant data concerning this compound with cancer was collected using different scientific search engines, including PubMed, Springer Link, Wiley Online, Web of Science, SciFinder, ScienceDirect, and Google Scholar. This review demonstrated that quercetin showed strong anticancer properties, including apoptosis, inhibition of cell proliferation, autophagy, cell cycle arrest, inhibition of angiogenesis, and inhibition of invasion and migration against various types of cancer. Findings also revealed that quercetin could significantly moderate and regulate different pathways, including PI3K/AKT-mTORC1 pathway, JAK/STAT signaling system, MAPK signaling pathway, MMP signaling pathway, NF-κB pathway, and p-Camk2/p-DRP1 pathway. However, this study found that quercetin showed poor oral bioavailability due to reduced absorption; this limitation is overcome by applying nanotechnology (nanoformulation of quercetin). Moreover, different investigations revealed that quercetin expressed no toxic effect in the investigated subjects. Based on the view of these findings, it is demonstrated that quercetin might be considered a reliable chemotherapeutic drug candidate in the treatment of different cancers. However, more clinical studies are suggested to establish the proper therapeutic efficacy, safety, and human dose.
Collapse
Affiliation(s)
- Tanzila Akter Eity
- Department of Biotechnology and Genetic EngineeringBangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Gopalganj 8100, Bangladesh
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research Center Ltd., Gopalganj, Gopalganj 8100, Bangladesh
| | - Md. Shimul Bhuia
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research Center Ltd., Gopalganj, Gopalganj 8100, Bangladesh
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Gopalganj 8100, Bangladesh
| | - Raihan Chowdhury
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research Center Ltd., Gopalganj, Gopalganj 8100, Bangladesh
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Gopalganj 8100, Bangladesh
| | - Shakil Ahmmed
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research Center Ltd., Gopalganj, Gopalganj 8100, Bangladesh
- Department of Biochemistry and Molecular BiologyBangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Salehin Sheikh
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research Center Ltd., Gopalganj, Gopalganj 8100, Bangladesh
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Gopalganj 8100, Bangladesh
| | - Rima Akter
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research Center Ltd., Gopalganj, Gopalganj 8100, Bangladesh
- Biotechnology and Genetic Engineering DisciplineKhulna University, Khulna 9208, Bangladesh
| | - Muhammad Torequl Islam
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research Center Ltd., Gopalganj, Gopalganj 8100, Bangladesh
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Gopalganj 8100, Bangladesh
- Pharmacy DisciplineKhulna University, Khulna 9208, Bangladesh
| |
Collapse
|