1
|
Liu X, Gu J, Liu S, Huang J, Li L, Wang F, He S, Mi Z, Zhang Y, Wen J, Gao Q, Yang H, Feng Y, Luo H, Zhai X, Zhang L, Zheng Y, Luo Y, Niu T. The mitochondrial protease ClpP is a promising target for multiple myeloma treatment. Biochem Pharmacol 2025; 236:116855. [PMID: 40054785 DOI: 10.1016/j.bcp.2025.116855] [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: 11/07/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Drug resistance and relapse are the major obstacles in multiple myeloma (MM) treatment, driving the search for novel therapeutics. The chemoactivation of mitochondrial caseinolytic protease P (ClpP) has shown to have anticancer effects on many tumors, but has seldom been elucidated in MM. Here we found that the CLPP expression was elevated in MM patients, and further increased in relapsed cases. After synthesizing and screening a panel of ClpP agonists, we identified a compound, 7b, as the most potent anti-MM agent in vitro. 7b activated ClpP protease activity, selectively degrading mitochondrial proteins, many of which are involved in oxidative phosphorylation (OXPHOS). As result, 7b treated MM had metabolic dysfunction, the mitochondrial membrane potential (MMP) collapse, reduced OXPHOS levels, and increased mitochondrial reactive oxygen species (ROS), leading to mitophagy-mediated MM cell death. Notably, 7b also showed efficacy against drug-resistant MM cell lines, including bortezomib- and lenalidomide-resistant cells. In vivo, 7b also exhibited remarkable anti-MM activity with tolerable side effects. In conclusion, targeting ClpP represents a promising therapeutic strategy for MM, with 7b serving as a potent anti-MM agent, especially for relapsed and refractory MM.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinlong Gu
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, China
| | - Song Liu
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, China
| | - Jingcao Huang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Linfeng Li
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Fangfang Wang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Siyao He
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Ziyue Mi
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Zhang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Wen
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China; Department of Hematology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Qianwen Gao
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China; College of Life Science, Sichuan University, Chengdu, China
| | - Haonan Yang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Feng
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Luo
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Zhai
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhuan Zheng
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China.
| | - Youfu Luo
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, China.
| | - Ting Niu
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China; State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, China; National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, China.
| |
Collapse
|
2
|
Pu K, Yang S, Sheng R, Chen J, Dai Y, Wood IC, Zhong Z, Xu S. Chuanxiong-Danggui herb pair alleviated cognitive deficits of APP/PS1 mice by promoting mitophagy. JOURNAL OF ETHNOPHARMACOLOGY 2025; 350:119988. [PMID: 40389086 DOI: 10.1016/j.jep.2025.119988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Disruption of receptor-mediated mitophagy contributes to neuronal damage in Alzheimer's disease (AD). Chuanxiong-Danggui herb pair (CDHP) is classic herbal pair applied to treating neurodegenerative diseases including AD, Amyotrophic Lateral Sclerosis, Parkinson's disease. Though studies have demonstrated the neuroprotective effects of CDHP, the underlying mechanisms by which CDHP attenuates neuronal impairment of AD remains to be elucidated. AIM OF THE STUDY The objective of this work was to investigate the anti-AD mechanism of CDHP in APP/PS1 mice. MATERIALS AND METHODS Behavioral assessments were conducted on C57BL/6J and APP/PS1 mice following CDHP treatment, alongside an evaluation of neuronal morphology in the hippocampal region. In vitro, HT-22 cells were induced by Aβ25-35 before being treated with CDHP. The mechanisms of CDHP were investigated using transmission electron microscopy, Golgi staining, immunofluorescence, and Western blot analysis. RESULTS Results from the passive avoidance test and the Morris water maze (MWM) indicated that CDHP significantly mitigated cognitive deficits of APP/PS1 mice, accompanied by a reduction of pathological damage in the CA1 and CA3 regions of hippocampus. Further testing found that a significant reduction in dendritic spines density was rescued by CDHP. Synaptophysin (SYN) and postsynaptic density protein 95 (PSD-95) were elevated in the CDHP group, while β-amyloid (Aβ) plaques deposition was significantly reduced. Simultaneously, CDHP markedly inhibited neuronal apoptosis through a decrease of the levels of Cleaved Caspase-12 and enhanced expression of Bcl-2/Bax, both in vivo and in vitro. Additionally, CDHP improved mitochondrial morphology and function in the AD model by decreasing abnormal mitochondria and increasing the expression of COXIV. Transmission electron microscopy (TEM) revealed that clear mitophagy-autophagosomes were nearly absent in APP/PS1 mice, while the expression of p62 and LC3B were elevated following CDHP treatment. Furthermore, CDHP increased the expression of the FUNDC1 and PGAM5 in APP/PS1 mice and AD-like cell models. CONCLUSION These findings suggest that CDHP mitigated cognitive dysfunction in APP/PS1 mice by enhancing mitophagy to reduce neuronal injury.
Collapse
Affiliation(s)
- Keting Pu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Simin Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Ruilin Sheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jie Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yuan Dai
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; School of Health Preservation and Rehabilitation. Chengdu University of Traditional Chinese Medicine.Chengdu, 611137, China
| | - Ian C Wood
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT, Leeds, UK.
| | - Zhanqiong Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT, Leeds, UK.
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
3
|
Wang H, Gong F, Zhao W, Huang Y, Zhou C, Wang J. Melatonin Inhibiting Neuronal Cells Ferroptosis Through Lipid Metabolic Reprogramming. Mol Neurobiol 2025:10.1007/s12035-025-05035-9. [PMID: 40366559 DOI: 10.1007/s12035-025-05035-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
This study aims to identify the underlying mechanism by which melatonin protects neurons. Firstly, the inhibitory effect of melatonin on ferroptosis was verified by treating HT22 cells with melatonin, Erastin, and Ferrostatin-1. Secondly, transcriptomic and metabolomic analyses were performed. Melatonin-related hub genes were identified by differential gene expression analysis, and lipid metabolism-related critical signaling pathways and biological processes (BPs) were determined by gene set enrichment analysis (GSEA). Finally, the expression of hub genes was verified by quantitative real-time PCR (qRT-PCR) or Western Blot (WB), and the involvement of Tribble 3 (Trib3) in the regulation of lipid metabolism and ferroptosis by melatonin was confirmed by Cell Counting Kit 8 (CCK-8) assay, ROS analysis, and WB. Assay results showed that melatonin significantly increased Gpx4 activity, decreased ROS generation, and inhibited ferroptosis in HT22 cells. The hub gene Trib3 was obtained by transcriptomic analysis, and its expression was upregulated with Erastin treatment. Lipid metabolomic analysis suggested that the regulation of lipid metabolism by melatonin was associated with glycerophospholipids. In vitro experiments showed that Trib3 was regulated by the upstream factor Atf4, and the protein levels of Trib3 and Atf4 were significantly increased after Erastin treatment. However, melatonin can reduce the protein levels of Trib3 and Atf4, increase the survival rate of HT-22 cells and the activity of GPX4, and reduce the ROS content. Melatonin inhibits neuronal ferroptosis by affecting the Atf4/Trib3 axis via the modulation of lipid metabolism.
Collapse
Affiliation(s)
- Haifeng Wang
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang Province, China.
| | - Fanyong Gong
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang Province, China
| | - Wenhui Zhao
- Psychosomatic Medical Center, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang Province, China.
| | - Yi Huang
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang Province, China
| | - Chenhui Zhou
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang Province, China
| | - Jianyong Wang
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang Province, China
| |
Collapse
|
4
|
Wu X, Huang L, Meng L, Luo S, Jablonska PA, Zhang C, Zhang A, Li P, Gong X. FOXO3a regulation of non-small cell lung cancer radiotherapy resistance through the PINK1/Parkin pathway of protective mitophagy. Transl Lung Cancer Res 2025; 14:1320-1339. [PMID: 40386735 PMCID: PMC12082242 DOI: 10.21037/tlcr-2025-181] [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: 02/23/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
Background Radiotherapy resistance has become one of the major causes of radiotherapy failure among patients with non-small cell lung cancer (NSCLC), but its underlying mechanism remains unclear. In recent years, the influence of mitochondrial autophagy on the radiotherapy resistance in treated tumor cells and its regulatory mechanism has become a hotspot in research, which is also the subject of our group research effort. The primary objective of our study is to investigate the mitophagy-associated pathway and the regulatory mechanisms underlying radiotherapy resistance in NSCLC. Methods We developed biologically stable radiotherapy-resistant NSCLC cell models A549/X and H520/X and verified the radioresistance of these cells. Subsequently, through high-throughput transcriptomic sequencing analysis and experimental verification, we found that the Forkhead box O 3a (FOXO3a) gene and the PINK1/Parkin mitochondrial autophagy pathway in NSCLC radiotherapy-resistant cell lines were consistent and upregulated more reactively than those of parent cells. The effect of gene expression status of the FOXO3a-PINK1/Parkin pathway on the survival outcomes of NSCLC was analyzed in The Cancer Genome Atlas (TCGA) database. Next, we inoculated nude mouse xenografts with small interfering RNA to interfere with the FOXO3a gene and short hairpin RNA to construct radiotherapy-resistant stable strains of NSCLC with stable knockdown of FOXO3a gene. Subsequently, the association and regulation of FOXO3a gene expression levels with radioresistance and mitochondrial autophagy PINK1/Parkin pathway at the cellular and animal levels were determined. Results The expression level of FOXO3a gene in NSCLC radioresistant cells was significantly positively correlated with the level of mitophagy and the expression level of PINK1/Parkin pathway. Higher expression levels of genes in the FOXO3a-PINK1/Parkin pathway had a negative effect on survival outcomes in NSCLC and were positively correlated with the radioresistance of cells. Conclusions FOXO3a regulates NSCLC radioresistance by modulating the mitochondrial autophagy PINK1/Parkin pathway, which may serve as a new molecular intervention target and therapeutic entry point for intervening and improving the radioresistance of patients with NSCLC in clinical practice.
Collapse
Affiliation(s)
- Xiaoting Wu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Litang Huang
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Meng
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shilan Luo
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Paola Anna Jablonska
- Radiation Oncology Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Chi Zhang
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anqi Zhang
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Li
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaomei Gong
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Yang Y, Jiang B, Shi L, Wang L, Yang Y, Li Y, Zhang Y, Zhu Z, Zhang X, Liu X. The potential of natural herbal plants in the treatment and prevention of non-small cell lung cancer: An encounter between ferroptosis and mitophagy. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119555. [PMID: 40015539 DOI: 10.1016/j.jep.2025.119555] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 02/15/2025] [Accepted: 02/22/2025] [Indexed: 03/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine constitutes a substantial cultural and scientific resource for the Chinese nation, attracting considerable scholarly interest due to its intrinsic characteristics of "multi-component, multi-target, and multi-pathway" interactions. Simultaneously, it aligns accurately with the intricate and continuously evolving progression of non-small cell lung cancer (NSCLC). Furthermore, contemporary pharmacological studies indicate that natural herbaceous plants and their bioactive compounds exhibit a diverse array of biological activities, including antioxidant, anti-inflammatory, and anti-tumor effects, among others. Additionally, these substances have been demonstrated to possess a degree of safety, particularly in terms of exhibiting comparatively lower levels of toxicity to the liver and kidneys when contrasted with conventional Western medicine. Thus, the development of herbal plants, which includes both single herbs and composite formulations, as well as their bioactive constituents, through the targeted regulation of ferroptosis and mitophagy, presents substantial potential and instills considerable hope for individuals diagnosed with NSCLC. AIM OF THE REVIEW This review aims to conduct a critical analysis of the ethnopharmacological applications of natural herbaceous plants in relation to ferroptosis and mitophagy in NSCLC. The objective is to evaluate the potential advantages of prioritizing specific phytochemical constituents found in these plants, which may serve as novel therapeutic candidates informed by ethnobotanical knowledge. Additionally, this study seeks to enhance the current pharmacological applications of natural herbaceous plants. METHODS An investigation into natural herbal remedies for NSCLC was conducted, with a particular emphasis on the ferroptosis and mitophagy pathways. This study utilized traditional medical texts and ethnomedicinal literature as primary sources. Furthermore, relevant information related to ethnobotany, phytochemistry, and pharmacology is obtained from online databases, including PubMed and the China National Knowledge Infrastructure (CNKI), among others. "Traditional Chinese medicine compound preparations", "single herb extracts", "active compounds", "NSCLC", "ferroptosis", and "mitophagy" were used as keywords when searching the databases. Consequently, pertinent articles published in recent years were collected and analyzed. RESULTS Given the complex etiology of NSCLC, treatment strategies that concentrate exclusively on ferroptosis or mitophagy often demonstrate limitations. In this regard, the utilization of herbal plants offers unique benefits in the management of NSCLC. The rationale can be summarized within the following two dimensions: Firstly, due to the molecular mechanisms of ferroptosis and mitophagy involving multiple signaling pathways (including PINK1/Parkin, HMGB1, system Xc-/GPX4/GSH, FSP1/CoQ10/NAD (P) H, and so on), sometimes drugs with a single target are difficult to involve multiple pathways. Fortunately, there is an expanding body of evidence suggesting that various herbaceous plants and their bioactive compounds can affect multiple biological targets. Moreover, these compounds seem to interact with several targets associated with ferroptosis and mitophagy in NSCLC (such as NIX, BNIP3, FUNDC1, GPX4, FSP1, P53, Nrf2, LncRNA, and so on). Secondly, Herbaceous plants and their bioactive compounds have been shown to possess a favorable safety profile, particularly with respect to reduced hepatotoxicity and nephrotoxicity in comparison to conventional Western medicine. For example, Numerous compound formulations, such as Fangji Huangqi decoction, Mufangji decoction, Qiyu Sanlong decoction, and Fuzheng Kangai decoction, have been employed in China for millennia, and their clinical efficacy appears to be quite promising. Notably, In recent years, numerous researchers have sought to isolate active constituents from clinically effective compound formulations through the application of chemical methodologies. This endeavor has been driven by the necessity to tackle challenges related to complex ingredient compositions and sophisticated processing. These active compounds have been employed in cellular and animal studies to elucidate the molecular mechanisms underlying these formulations. CONCLUSIONS The Asian region has a long-standing historical tradition of employing natural herbaceous plants for traditional medicinal purposes. Phytochemical and pharmacological studies have shown that various compound preparations derived from traditional Chinese medicine, along with individual herb extracts and their active constituents, display a range of bioactive effects. These effects encompass anti-tumor, anti-inflammatory, antibacterial, and antioxidant properties, among others. Numerous traditional compound formulations originating from China have emerged as promising candidates for the development of pharmacological agents targeting NSCLC. It is noteworthy that a variety of compound formulations aimed at the ferroptosis and mitophagy pathways, which demonstrate unique therapeutic effects on NSCLC, are presently under extensive investigation by an increasing number of researchers. Therefore, it is imperative to consider in vitro mechanistic studies, in vivo pharmacological evaluations, and assessments of clinical efficacy. Furthermore, it is essential to conduct a comprehensive assessment of plant resources, implement quality control measures, and engage in toxicological research to ensure that the data is appropriate for further examination.
Collapse
Affiliation(s)
- Yujie Yang
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Lijuan Shi
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Lili Wang
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Yaru Yang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Yongyu Li
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Yanmei Zhang
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Zhongbo Zhu
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Xuhui Zhang
- Department of Pulmonary Diseases, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, 730030, China.
| | - Xiping Liu
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
6
|
Chen K, Chen J, Cong Y, He Q, Liu C, Chen J, Li H, Ju Y, Chen L, Song Y, Xing Y. TTK promotes mitophagy by regulating ULK1 phosphorylation and pre-mRNA splicing to inhibit mitochondrial apoptosis in bladder cancer. Cell Death Differ 2025:10.1038/s41418-025-01492-w. [PMID: 40269198 DOI: 10.1038/s41418-025-01492-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/20/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025] Open
Abstract
Bladder cancer (BC) remains a major global health challenge, with poor prognosis and limited therapeutic options in advanced stages. TTK protein kinase (TTK), a serine/threonine kinase, has been implicated in the progression of various cancers, but its role in BC has not been fully elucidated. In this study, we show that TTK is significantly upregulated in BC tissues and cell lines, correlating with poor patient prognosis. Functional assays revealed that TTK promotes proliferation and inhibits apoptosis of BC cells. Mechanistically, TTK enhances mitophagy by directly phosphorylating ULK1 at Ser477, thereby activating the ULK1/FUNDC1-mediated mitophagy pathway. TTK knockdown disrupts mitophagy, leading to impaired clearance of damaged mitochondria, excessive accumulation of mitochondrial reactive oxygen species (mtROS), and activation of mitochondrial apoptosis. Furthermore, TTK phosphorylates SRSF3 at Ser108, preventing ULK1 exon 5 skipping and maintaining ULK1 mRNA stability. These findings show that TTK plays a key role in maintaining mitophagy in BC cells. Targeting TTK could offer a promising new approach for BC treatment by disrupting mitophagy and inducing mitochondrial apoptosis.
Collapse
Affiliation(s)
- Kang Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinyu Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yukun Cong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingliu He
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chunyu Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunjie Ju
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yarong Song
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yifei Xing
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
7
|
Lv Z, Jiao J, Xue W, Shi X, Wang R, Wu J. Activation-induced cytidine deaminase in tertiary lymphoid structures: dual roles and implications in cancer prognosis. Front Oncol 2025; 15:1555491. [PMID: 40270606 PMCID: PMC12014437 DOI: 10.3389/fonc.2025.1555491] [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: 01/04/2025] [Accepted: 03/25/2025] [Indexed: 04/25/2025] Open
Abstract
Activation-induced cytidine deaminase (AID) serves as a critical molecular orchestrator in the germinal center (GC) reaction within secondary lymphoid organs (SLOs), driving the production of high-affinity antibodies through somatic hypermutation. While its pathological implications are well-documented - including ectopic expression in non-B cell populations and transcriptional dysregulation linked to hematological malignancies and solid tumorigenesis - the cellular provenance of AID in solid tumors remains an unresolved paradox. This review advances two principal hypotheses: (1) AID may derive from tertiary lymphoid structures (TLSs), ectopic immune niches mirroring SLO organization, and (2) exhibits context-dependent transcriptional duality, capable of both potentiating and suppressing gene expression based on microenvironmental cues. Through systematic analysis of AID/GC involvement across cancer subtypes, we delineate mechanistic connections between lymphoid neogenesis and tumor progression. Our examination extends to TLS architecture, revealing three critical dimensions: (i) structural organization and cellular heterogeneity, (ii) developmental trajectories, and (iii) bidirectional interactions with tumor microenvironments. Crucially, we establish functional parallels between tumor-infiltrating B cells (TIL-Bs) in SLOs versus TLSs, while elucidating the differential roles of AID in canonical GC versus TLS-associated GC formation. This synthesis ultimately proposes that AID's functional dichotomy - acting as both oncogenic collaborator and tumor suppressor - underlies the paradoxical prognostic associations observed with TLS presence across malignancies. The review thereby provides a conceptual framework reconciling AID's dual functionality with the context-dependent immunobiology of tumor-associated lymphoid structures.
Collapse
Affiliation(s)
- Zhuangwei Lv
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Junna Jiao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, China
| | - Wuyang Xue
- Department of Laboratory Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiaoyu Shi
- School of Junji College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Ruihan Wang
- School of Junji College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinhua Wu
- School of Junji College, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
8
|
Xu L, Zhang T, Zhu B, Tao H, Liu Y, Liu X, Zhang Y, Meng X. Mitochondrial quality control disorder in neurodegenerative disorders: Potential and advantages of traditional Chinese medicines. J Pharm Anal 2025; 15:101146. [PMID: 40291018 PMCID: PMC12032916 DOI: 10.1016/j.jpha.2024.101146] [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: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 04/30/2025] Open
Abstract
Neurodegenerative disorders (NDDs) are prevalent chronic conditions characterized by progressive synaptic loss and pathological protein alterations. Increasing evidence suggested that mitochondrial quality control (MQC) serves as the key cellular process responsible for clearing misfolded proteins and impaired mitochondria. Herein, we provided a comprehensive analysis of the mechanisms through which MQC mediates the onset and progression of NDDs, emphasizing mitochondrial dynamic stability, the clearance of damaged mitochondria, and the generation of new mitochondria. In addition, traditional Chinese medicines (TCMs) and their active monomers targeting MQC in NDD treatment have been demonstrated. Consequently, we compiled the TCMs that show great potential in the treatment of NDDs by targeting MQC, aiming to offer novel insights and a scientific foundation for the use of MQC stabilizers in NDD prevention and treatment.
Collapse
Affiliation(s)
- Lei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Baojie Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Honglin Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yue Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianfeng Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, Sichuan, 620032, China
| |
Collapse
|
9
|
Zhang H, Zhang J, Zhu K, Li S, Liu J, Guan B, Zhang H, Chen C, Liu Y. Identification and characterization of mitochondrial autophagy-related genes in osteosarcoma and predicting clinical prognosis. Sci Rep 2025; 15:10158. [PMID: 40128298 PMCID: PMC11933398 DOI: 10.1038/s41598-025-95173-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/19/2025] [Indexed: 03/26/2025] Open
Abstract
Osteosarcoma (OS), the most prevalent primary malignant bone tumor, is characterized by a poor prognosis and high metastatic potential. Mitochondrial autophagy has been implicated in cancer suppression. This study aimed to identify prognostic genes associated with mitochondrial autophagy in OS. Public datasets, including TARGET-OS, GSE99671, and GSE21257, were retrieved for analysis. Differentially expressed genes (DEGs1) between OS and normal samples were identified from GSE99671. Single-sample Gene Set Enrichment Analysis (ssGSEA) was applied to quantify the enrichment scores of 29 mitochondrial autophagy-related genes (MARGs) in OS samples from TARGET-OS, categorizing them into high- and low-score groups to extract DEGs2. The intersection of DEGs1 and DEGs2 yielded mitochondrial autophagy-associated differentially expressed genes (MDGs). Prognostic genes were subsequently screened through a multi-step regression analysis, and a risk score was computed. TARGET-OS samples were stratified into high- and low-risk groups based on the optimal cutoff value of the risk score. GSEA was conducted between the two risk groups. Additionally, associations between prognostic genes and the immune microenvironment were explored. A total of 31 MDGs were identified from the overlap of 3,207 DEGs1 and 622 DEGs2. Five prognostic genes-KLK2, NRXN1, HES5, OR2W3, and HS3ST4-were further selected. Kaplan-Meier survival analysis indicated significantly reduced survival in the high-risk group. GSEA revealed enrichment in ABC transporter activity and glycolysis/gluconeogenesis pathways. Immunoanalysis demonstrated significant differences in 11 immune cell populations and three immune functions between risk groups, notably myeloid-derived suppressor cells (MDSCs) and Type 1 T helper cells. HS3ST4 exhibited the strongest positive correlation with macrophages, whereas NRXN1 showed the most pronounced negative correlation with memory B cells. Expressions of HAVCR2 and PDCD1LG2 were elevated in the low-risk group. Functional analysis indicated significant differences in dysfunction patterns between risk groups. This study identified five mitochondrial autophagy-related prognostic genes and constructed a risk model, offering novel insights into OS diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Jingyu Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Kai Zhu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Shuang Li
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Jinwei Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Boya Guan
- Department of Pharmacy, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Hong Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Changbao Chen
- Department of Spinal Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Yancheng Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
| |
Collapse
|
10
|
Hu X, Lv J, Zhao Y, Li X, Qi W, Wang X. Important regulatory role of mitophagy in diabetic microvascular complications. J Transl Med 2025; 23:269. [PMID: 40038741 PMCID: PMC11877814 DOI: 10.1186/s12967-025-06307-7] [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: 12/13/2024] [Accepted: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
Microvascular complications of diabetes pose a significant threat to global health, mainly including diabetic kidney disease (DKD), diabetic retinopathy (DR), diabetic peripheral neuropathy (DPN), and diabetic cardiomyopathy (DCM), which can ultimately lead to kidney failure, blindness, disability, and heart failure. With the increasing prevalence of diabetes, the search for new therapeutic targets for diabetic microvascular complications is imminent. Mitophagy is a widespread and strictly maintained process of self-renewal and energy metabolism that plays an important role in reducing inflammatory responses, inhibiting reactive oxygen species accumulation, and maintaining cellular energy metabolism. Hyperglycemia results in impaired mitophagy, which leads to mitochondrial dysfunction and ultimately exacerbates disease progression. This article summarizes the relevant molecular mechanisms of mitophagy and reviews the current status of research on regulating mitophagy as a potential treatment for diabetic microvascular complications, attempting to give new angles on the treatment of diabetic microvascular complications.
Collapse
Affiliation(s)
- Xiangjie Hu
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jiao Lv
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yunyun Zhao
- Endocrinology Department, First Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin, 130021, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130017, China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130017, China.
| | - Xiuge Wang
- Endocrinology Department, First Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin, 130021, China.
| |
Collapse
|
11
|
Xi X, Chen S, Zhao X, Zhou Z, Zhu S, Ren X, Wang X, Wu J, Mu S, Li X, Shan E, Cui Y. TUBB4A Inhibits Glioma Development by Regulating ROS-PINK1/Parkin-Mitophagy Pathway. Mol Neurobiol 2025; 62:3125-3142. [PMID: 39230869 DOI: 10.1007/s12035-024-04459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 08/26/2024] [Indexed: 09/05/2024]
Abstract
Glioma is a refractory malignant tumor with a powerful capacity for invasiveness and a poor prognosis. This study aims to investigate the role and mechanism of tubulin beta class IVA (TUBB4A) in glioma progression. The differential expression of TUBB4A in humans was obtained from databases and analyzed. Glioma cells U251-MG and U87-MG were intervened by pcDNA3.1(+) and TUBB4A overexpression plasmid. MTT, CCK8, LDH, wound healing, transwell, and western blotting were used to explore whether TUBB4A participates in the development of glioma. Reactive oxygen species (ROS) were detected by the DCFH-DA probe. Mitochondrial membrane potential (MMP) was examined by JC-1. It was found that TUBB4A expression level correlated with tumor grade, IDH1 status, 1p/19q status, and poor survival in glioma patients. In addition, TUBB4A overexpression inhibited the proliferation, migration, and invasion of U251-MG and U87-MG, while increasing the degree of apoptosis. Notably, TUBB4A overexpression promotes ROS generation and MMP depolarization, and induces mitophagy through the PINK1/Parkin pathway. Interestingly, mitochondria-targeted ROS scavenger reversed the effect of TUBB4A overexpression on PINK1/Parkin expression and mitophagy, whereas mitophagy inhibitor did not affect ROS production. And the effect of TUBB4A overexpression on mitophagy and glioma progression was consistent with that of PINK1/Parkin agonist. In conclusion, TUBB4A is a molecular marker for predicting the prognosis of glioma patients and an effective target for inhibiting glioma progression by regulating ROS-PINK1/Parkin-mitophagy pathway.
Collapse
Affiliation(s)
- Xueru Xi
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Suqin Chen
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Xiaoli Zhao
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Zimu Zhou
- The Cancer Research Institute, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Shanjie Zhu
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Xurui Ren
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Xiaomei Wang
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Jing Wu
- Department of Anesthesiology, The First Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Shuai Mu
- Department of Oncology, Senior Department of Oncology, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xianwen Li
- School of Nursing, Nanjing Medical University, Nanjing, China.
| | - Enfang Shan
- School of Nursing, Nanjing Medical University, Nanjing, China.
| | - Yan Cui
- School of Nursing, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
Li R, Ma Y, He A, Pu Y, Wan X, Sun H, Wang N, Luo M, Wang G, Xia Y. Fasting enhances the efficacy of Sorafenib in breast cancer via mitophagy mediated ROS-driven p53 pathway. Free Radic Biol Med 2025; 229:350-363. [PMID: 39864757 DOI: 10.1016/j.freeradbiomed.2025.01.047] [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: 11/28/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
The multi-kinase inhibitor sorafenib has shown potential to inhibit tumor cell growth and intra-tumoral angiogenesis by targeting several kinases, including VEGFR2 and RAF. Abnormal activation of the Ras/Raf/MAPK/ERK kinase cascade and the VEGF pathway is a common feature in breast cancer. However, the efficacy of sorafenib in breast cancer treatment remains limited. Recently, fasting has emerged as a promising non-pharmacological approach to modulate cancer metabolism and enhance the effectiveness of cancer therapies. In this study, we found that fasting significantly enhances the anti-cancer effects of sorafenib monotherapy and its combination with immunotherapy in breast cancer models without causing obvious side effects. This combined treatment effectively inhibits tumor cell proliferation and intra-tumoral angiogenesis. The fasting-induced reduction in peripheral blood glucose levels strongly correlated with enhanced sensitivity to sorafenib. Mechanistically, the combined treatment induced mitophagy, characterized by mitochondrial dysfunction and activation of the PINK1-Parkin pathway. Consequently, increased mitochondrial ROS levels promoted p53 expression, amplifying cell cycle arrest and apoptosis in breast cancer cells. Furthermore, fasting reduced lactate levels within the tumor, and the consequent glucose limitation synergized with sorafenib to activate AMPK, which in turn elevated PD-L1 expression in tumor cells, potentially enhancing their sensitivity to immunotherapy. In summary, our findings demonstrate that fasting and sorafenib, as a rational combination therapy, induce mitophagy, thereby enhancing sorafenib's efficacy in treating breast cancer through the ROS-driven p53 pathway. This study underscores the potential of fasting in breast cancer therapy and provides a foundation for optimizing the clinical application of sorafenib.
Collapse
Affiliation(s)
- Ru Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China
| | - Yimei Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Anqi He
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yamin Pu
- Department of Biotherapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xuanting Wan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Hongbao Sun
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Min Luo
- Department of Biotherapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China.
| | - Yong Xia
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Stevens M, Wang Y, Bouley SJ, Mandigo TR, Sharma A, Sengupta S, Housden A, Perrimon N, Walker JA, Housden BE. Inhibition of autophagy as a novel treatment for neurofibromatosis type 1 tumors. Mol Oncol 2025; 19:825-851. [PMID: 39129390 PMCID: PMC11887668 DOI: 10.1002/1878-0261.13704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/29/2024] [Accepted: 07/19/2024] [Indexed: 08/13/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is a genetic disorder caused by mutation of the NF1 gene that is associated with various symptoms, including the formation of benign tumors, called neurofibromas, within nerves. Drug treatments are currently limited. The mitogen-activated protein kinase kinase (MEK) inhibitor selumetinib is used for a subset of plexiform neurofibromas (PNs) but is not always effective and can cause side effects. Therefore, there is a clear need to discover new drugs to target NF1-deficient tumor cells. Using a Drosophila cell model of NF1, we performed synthetic lethal screens to identify novel drug targets. We identified 54 gene candidates, which were validated with variable dose analysis as a secondary screen. Pathways associated with five candidates could be targeted using existing drugs. Among these, chloroquine (CQ) and bafilomycin A1, known to target the autophagy pathway, showed the greatest potential for selectively killing NF1-deficient Drosophila cells. When further investigating autophagy-related genes, we found that 14 out of 30 genes tested had a synthetic lethal interaction with NF1. These 14 genes are involved in multiple aspects of the autophagy pathway and can be targeted with additional drugs that mediate the autophagy pathway, although CQ was the most effective. The lethal effect of autophagy inhibitors was conserved in a panel of human NF1-deficient Schwann cell lines, highlighting their translational potential. The effect of CQ was also conserved in a Drosophila NF1 in vivo model and in a xenografted NF1-deficient tumor cell line grown in mice, with CQ treatment resulting in a more significant reduction in tumor growth than selumetinib treatment. Furthermore, combined treatment with CQ and selumetinib resulted in a further reduction in NF1-deficient cell viability. In conclusion, NF1-deficient cells are vulnerable to disruption of the autophagy pathway. This pathway represents a promising target for the treatment of NF1-associated tumors, and we identified CQ as a candidate drug for the treatment of NF1 tumors.
Collapse
Affiliation(s)
- Megan Stevens
- Living Systems InstituteUniversity of ExeterUK
- Department of Clinical and Biomedical ScienceUniversity of ExeterUK
| | - Yuanli Wang
- Living Systems InstituteUniversity of ExeterUK
- The First People's Hospital of QinzhouChina
| | | | - Torrey R. Mandigo
- Center for Genomic MedicineMassachusetts General HospitalBostonMAUSA
| | - Aditi Sharma
- Center for Genomic MedicineMassachusetts General HospitalBostonMAUSA
| | - Sonali Sengupta
- Living Systems InstituteUniversity of ExeterUK
- Department of Clinical and Biomedical ScienceUniversity of ExeterUK
| | - Amy Housden
- Living Systems InstituteUniversity of ExeterUK
| | - Norbert Perrimon
- Department of Genetics, Blavatnik InstituteHarvard Medical SchoolBostonMAUSA
- Howard Hughes Medical InstituteNew YorkNYUSA
| | - James A. Walker
- Center for Genomic MedicineMassachusetts General HospitalBostonMAUSA
- Cancer ProgramBroad Institute of MIT and HarvardCambridgeMAUSA
- Department of Neurology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Benjamin E. Housden
- Living Systems InstituteUniversity of ExeterUK
- Department of Clinical and Biomedical ScienceUniversity of ExeterUK
| |
Collapse
|
14
|
Wang J, Xue J, Ma B, Zhu Y, Li J, Tian C. MEK5-ERK5 pathway mediates mitophagy by regulating Nur77 to promote tumorigenesis of osteosarcoma cells. Eur J Med Res 2025; 30:117. [PMID: 39972514 PMCID: PMC11837295 DOI: 10.1186/s40001-025-02312-0] [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: 12/10/2024] [Accepted: 01/19/2025] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVES To investigate the influence of MEK5/ERK5 pathway on mitophagy in osteosarcoma (OS), as well as the involved molecular mechanisms. METHODS The overlapped genes of mitophagy-related genes from MSigDB database and DEGs between metastatic and primary OS groups from GSE32981 were identified. GSVA of mitophagy-related pathways between the metastatic and primary groups were analyzed. The relationships between Nur77 and mitophagy-related pathways, prognosis, immune infiltrating cells, immune response gene sets were investigated. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blotting were utilized to assess the expression levels of MEK5, ERK5, Nur77, PINK1, and Parkin. Cellular behaviors and mitochondrial potential were evaluated via CCK-8, Transwell assay and JC-1 staining. RESULTS Total 4 overlapped genes were obtained as mitophagy-related DEGs, including GABARAPL1, HIF1A, PINK1, and RB1CC1. The activity scores of 3 mitophagy-related pathways exhibited significant differences between metastatic and primary groups. Importantly, Nur77 was significantly negatively correlated with a mitophagy-related pathway (GOBP MITOPHAGY: R = - 0.48, P = 0.02). The Nur77 expression in metastatic group was remarkedly higher than that in the primary group (P < 0.001). Patients with high Nur77 expression had poor prognosis, with AUC values all above 0.615 in predicting 1-, 3-, and 5-year survival. In addition, Nur77 was closely related to numerous immune cells, including activated dendritic cells, activated mast cells and M0 macrophages, and immune response gene sets chemokines and cytokines (all P < 0.05). In addition, MEK5/ERK5 pathway is activated in OS, and Nur77 is overexpressed in OS, and MEK5/ERK pathway promotes Nur77 expression, tumorigenesis and mitochondrial function in U2OS cells. Cytosporone B implement significantly increased the tumorigenesis of U2OS cells in sh-MEK5 group, and inhibited the weaken in mitochondrial membrane potential caused by MEK5 downregulation, and reversed the protein levels of mitophagy markers PINK1 and Parkin in sh-MEK5 group. CONCLUSIONS MEK5-ERK5 pathway mediates mitophagy by regulating Nur77 to promote tumorigenesis of OS cells. These findings offered promising therapeutic targets for OS.
Collapse
Affiliation(s)
- Jianshu Wang
- Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Jinxu Xue
- Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Baijing Ma
- Department of Medical Molecular Biology Research Center, Gansu Provincial Academic Institute for Medical Research, Xiaoxihu East Street, Lanzhou, 730050, China
| | - Yanqi Zhu
- Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Jing Li
- Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Caiping Tian
- Department of Medical Molecular Biology Research Center, Gansu Provincial Academic Institute for Medical Research, Xiaoxihu East Street, Lanzhou, 730050, China.
| |
Collapse
|
15
|
Liang Z, Zhao S, Liu Y, Cheng C. The promise of mitochondria in the treatment of glioblastoma: a brief review. Discov Oncol 2025; 16:142. [PMID: 39924629 PMCID: PMC11807951 DOI: 10.1007/s12672-025-01891-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
Glioblastoma (GBM) is a prevalent and refractory type of brain tumor. Over the past two decades, there have been minimal advancements in GBM therapy. The current standard treatment involves surgical excision followed by radiation and chemotherapy. Compared to other tumors, GBM is more challenging to treat due to the presence of glioma stem-like cells (GSCs) and the blood-brain barrier, resulting in an extremely low survival rate. Mitochondria play a critical role in tumor respiration, metabolism, and multiple signaling pathways involved in tumor formation, progression, and cell apoptosis. Consequently, mitochondria represent promising targets for developing novel anticancer agents, including those targeting oxidative phosphorylation, reactive oxygen species (ROS), mitochondrial transfer, and mitophagy. This review outlines the mitochondrial-related therapeutic targets in GBM, highlighting the potential of mitochondria as a target for GBM treatment.
Collapse
Affiliation(s)
- Zhuo Liang
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Songyun Zhao
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yuankun Liu
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Chao Cheng
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
| |
Collapse
|
16
|
Klaßmüller T, Lengauer F, Blenninger J, Geisslinger F, Bartel K, Bracher F. Zephycandidine A and Synthetic Analogues-Synthesis and Evaluation of Biological Activity. Molecules 2025; 30:752. [PMID: 39942855 PMCID: PMC11820727 DOI: 10.3390/molecules30030752] [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: 01/11/2025] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
A convenient total synthesis of the imidazo[1,2-f]phenanthridine-type Amaryllidaceae alkaloid zephycandidine A (3) was developed, which further allowed us to perform modifications of substituents on benzenoid ring A and imidazole ring D. The biological activities of all synthesized compounds were evaluated, and it was reported that activities against cancer cells of the parent alkaloid were poorly reproducible, while the closely related analogue THK-121 (11) showed a strong inhibitory effect on proliferation. Additionally, our novel analogue significantly induced cell death via the intrinsic apoptosis pathway, evident by the loss of mitochondrial membrane potential, increased mitochondrial oxidative stress, and disrupted mitochondrial structure in the same cells. At the same time, healthy cells were less affected by the treatment with THK-121 (11), indicating a potential therapeutic margin.
Collapse
|
17
|
Vieira J, Barros M, López-Fernández H, Glez-Peña D, Nogueira-Rodríguez A, Vieira CP. Predicting Which Mitophagy Proteins Are Dysregulated in Spinocerebellar Ataxia Type 3 (SCA3) Using the Auto-p2docking Pipeline. Int J Mol Sci 2025; 26:1325. [PMID: 39941093 PMCID: PMC11818632 DOI: 10.3390/ijms26031325] [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: 11/29/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Dysfunctional mitochondria are present in many neurodegenerative diseases, such as spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD). SCA3/MJD, the most frequent neurodegenerative ataxia worldwide, is caused by the abnormal expansion of the polyglutamine tract (polyQ) at ataxin-3. This protein is known to deubiquitinate key proteins such as Parkin, which is required for mitophagy. Ataxin-3 also interacts with Beclin1 (essential for initiating autophagosome formation adjacent to mitochondria), as well as with the mitochondrial cristae protein TBK1. To identify other proteins of the mitophagy pathway (according to the KEGG database) that can interact with ataxin-3, here we developed a pipeline for in silico analyses of protein-protein interactions (PPIs), called auto-p2docking. Containerized in Docker, auto-p2docking ensures reproducibility and reduces the number of errors through its simplified configuration. Its architecture consists of 22 modules, here used to develop 12 protocols but that can be specified according to user needs. In this work, we identify 45 mitophagy proteins as putative ataxin-3 interactors (53% are novel), using ataxin-3 interacting regions for validation. Furthermore, we predict that ataxin-3 interactors from both Parkin-independent and -dependent mechanisms are affected by the polyQ expansion.
Collapse
Affiliation(s)
- Jorge Vieira
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (J.V.); (M.B.); (A.N.-R.)
- Instituto de Biologia Molecular e Celular (IBMC), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Mariana Barros
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (J.V.); (M.B.); (A.N.-R.)
- Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Hugo López-Fernández
- Department of Computer Science, CINBIO, ESEI—Escuela Superior de Ingeniería Informática, Universidade de Vigo, 32004 Ourense, Spain; (H.L.-F.); (D.G.-P.)
- SING Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain
| | - Daniel Glez-Peña
- Department of Computer Science, CINBIO, ESEI—Escuela Superior de Ingeniería Informática, Universidade de Vigo, 32004 Ourense, Spain; (H.L.-F.); (D.G.-P.)
- SING Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain
| | - Alba Nogueira-Rodríguez
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (J.V.); (M.B.); (A.N.-R.)
- Department of Computer Science, CINBIO, ESEI—Escuela Superior de Ingeniería Informática, Universidade de Vigo, 32004 Ourense, Spain; (H.L.-F.); (D.G.-P.)
- SING Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain
| | - Cristina P. Vieira
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (J.V.); (M.B.); (A.N.-R.)
- Instituto de Biologia Molecular e Celular (IBMC), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| |
Collapse
|
18
|
Huang L, Xue R, Zhu M, Xu S, Luo Y, Qin C, Yang C, Jia L, Tang K, Tan Q. Triptolide's impact on ACER1 signaling: Inducing autophagy for triple-negative breast cancer suppression. Pathol Res Pract 2025; 266:155823. [PMID: 39879681 DOI: 10.1016/j.prp.2025.155823] [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: 10/19/2024] [Revised: 12/24/2024] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Given the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (Her-2) in triple-negative breast cancer (TNBC) cells, the efficacy of targeted therapies is limited. In this study, we uncovered that triptolide (TP) effectively suppresses the migration and invasiveness of MDA-MB-231 cells by activating autophagic pathways. Western blotting analysis revealed that TP significantly reduced the expression levels of p62 protein, while simultaneously markedly increasing the expression levels of LC3B-II, BNIP3, BNIP3L, ATG5, and ULK1 proteins, strongly suggesting an enhancement of autophagic activity in the cells. Based on PCR array screening, we identified the ACER1 gene as exhibiting notable expression alterations post-TP treatment. Overexpression of ACER1 gene enhanced the TP-induced apoptosis in MDA-MB-231 cells and augmented the regulation of autophagy-related proteins p62 and LC3B-II, leading to an increase in autophagosome numbers and a marked reduction in cellular migration and invasiveness. Conversely, ACER1 gene knockdown reversed these effects. In vivo experiments demonstrated that TP effectively inhibits the growth of MDA-MB-231 xenograft tumors, concurrently upregulating ACER1 and LC3B-II expression in tumor tissues, while p62 protein levels were notably decreased. Hematoxylin and eosin (H&E) staining results indicated no evident toxicity in liver and kidney tissues of BALB/c mice at a TP dose of 0.4 mg/kg. This study, for the first time, elucidates a novel mechanism by which TP inhibits TNBC through an autophagic process mediated by ACER1.
Collapse
Affiliation(s)
- Lingyue Huang
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Rui Xue
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Mingfei Zhu
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Siyuan Xu
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Yuqin Luo
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Chuling Qin
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Chang Yang
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Lulu Jia
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Ke Tang
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Qinyou Tan
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, the Affiliated Hospital of Guilin Medical University, Guilin 541001, China; China-USA Lipids in Health and Disease Research Center, Guilin Medical University,Guilin 541001, China; Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, China.
| |
Collapse
|
19
|
Li S, Chen Y, Feng S, Liu Z, Gan L, Wang Q. Autophagy-targeted Pt(IV) agents: a new horizon in antitumor drug development. Dalton Trans 2025; 54:1770-1778. [PMID: 39804164 DOI: 10.1039/d4dt03162h] [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: 01/29/2025]
Abstract
Pt(IV) complexes as prodrugs of Pt(II) drugs exhibit numerous advantages such as enhanced stability, reduced toxicity, increased oral bioavailability, and efficacy in overcoming the drug resistance of Pt(II) compounds, which underscore their significant potential in the advancement of novel Pt anticancer agents. Furthermore, protective autophagy is pivotal in sustaining tumor cell homeostasis and modulating the tumor microenvironment (TME), thereby representing a critical target for the development of antitumor drugs. Specific inhibition or activation of autophagy during chemotherapy would break the internal homeostasis in the TME and increase antitumor activities. Consequently, developing novel Pt(IV) antitumor agents with autophagy-targeting capabilities by incorporating autophagy-regulating moieties into the Pt(IV) framework has emerged as a hot topic in the discovery of novel Pt drugs. Herein, the research progress in novel Pt(IV) antitumor drugs with autophagy-targeted properties is systematically reviewed based on the literature. The future challenges and perspectives of this fascinating class of conjugates are also discussed, aiming to provide new insights and approaches for the future design and investigation of novel Pt antitumor drugs.
Collapse
Affiliation(s)
- Suying Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Yan Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, P.R. China
| | - Shuaiqi Feng
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Linling Gan
- Chongqing Engineering Research Center of Pharmaceutical Sciences, School of Pharmacy, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, P. R. China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| |
Collapse
|
20
|
Zhang X, Chen T, Zhang F, Shi H, Li X, Wang Z, Wang D, Hou C. METTL1 coordinates cutaneous squamous cell carcinoma progression via the m7G modification of the ATF4 mRNA. Cell Death Discov 2025; 11:27. [PMID: 39870616 PMCID: PMC11772585 DOI: 10.1038/s41420-025-02304-3] [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: 07/05/2024] [Revised: 12/11/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Methyltransferase-like 1 (METTL1)-mediated m7G modification is a common occurrence in various RNA species, including mRNAs, tRNAs, rRNAs, and miRNAs. Recent evidence suggests that this modification is linked to the development of several cancers, making it a promising target for cancer therapy. However, the specific role of m7G modification in cutaneous squamous cell carcinoma (cSCC) is not well understood. In this study, we observed conspicuously elevated levels of METTL1 in cSCC tumors and cell lines. Inhibiting METTL1 led to reduced survival, migration, invasion, and xenograft tumor growth in cSCC cells. Mechanistically, through a combination of RNA sequencing, m7G methylated immunoprecipitation (MeRIP)-qPCR, and mRNA stability assays, we discovered that METTL1 is responsible for the m7G modification of ATF4 mRNA, leading to increased expression of ATF4. Importantly, we demonstrated that this modification is dependent on the methyltransferase activity of METTL1. Additionally, we observed a positive association between ATF4 expression and METTL1 levels in cSCC tumors. Intriguingly, restoring ATF4 expression in cSCC cells not only promoted glycolysis but also reversed the anti-tumor effects of METTL1 knockdown. In conclusion, our results underscore the critical role of METTL1 and m7G modification in cSCC tumorigenesis, suggesting a promising target for future cSCC therapies.
Collapse
Affiliation(s)
- Xinru Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Tong Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Fanrong Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Huanhuan Shi
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xiang Li
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zhijuan Wang
- The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Dong Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Chao Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
21
|
Sun Y, Zhu G, Zhao R, Li Y, Li H, Liu Y, Jin N, Li X, Li Y, Liu T. Deapioplatycodin D inhibits glioblastoma cell proliferation by inducing BNIP3L-mediated incomplete mitophagy. Cancer Cell Int 2025; 25:11. [PMID: 39800710 PMCID: PMC11726947 DOI: 10.1186/s12935-025-03636-x] [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: 07/06/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Deapioplatycodin D (DPD) is a triterpenoid saponin natural compound isolated from the Chinese herb Platycodon grandiflorum that has antiviral and antitumor properties. This study aimed to investigate the effects of DPD on glioblastoma (GBM) cells and to determine its intrinsic mechanism of action. Using a CCK8 assay, it was found that DPD significantly inhibited the growth of GBM cells. DPD-treated GBM cells contained swollen and degenerated mitochondria with empty vesicular bilayer membrane-like autophagic vesicle structures in the periphery of the mitochondria under transmission electron microscopy. DPD activated autophagy in GBM cells and induced a blockage of autophagic flux in the late stage. Transcriptomics identified differences in mitophagy-related genes, and analysis of the levels of the corresponding proteins indicated that mitophagy in GBM cells was induced mainly through BNIP3L. Increased expression of BNIP3L disrupts the Bcl-2-Beclin-1 complex, thereby releasing Beclin-1 and activating autophagy. Autophagy was inhibited after silencing of BNIP3L and overexpression of Bcl-2 in GBM cells, and the growth inhibitory effect of DPD was significantly reduced. This result demonstrated that DPD induces mitophagy in GBM cells through BNIP3L. Finally, activation of incomplete mitophagy in GBM cells by DPD through BNIP3L in vivo was demonstrated by establishing a mouse subcutaneous xenograft tumor model. In this study, in vitro and in vivo experiments established that DPD inhibited GBM cell growth by inducing BNIP3L-mediated incomplete mitophagy, which provides an experimental basis for studying new treatments of GBM.
Collapse
Affiliation(s)
- Yu Sun
- Department of Blood Transfusion, China-Japan, Union Hospital of Jilin University, Changchun, 130033, P.R. China
- Department of Neurology, Jilin Central Hospital, Jilin, 132011, P.R. China
| | - Guangze Zhu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, P.R. China
| | - Renshuang Zhao
- Medical College, Yanbian University, Yanji, 133002, P.R. China
| | - Yaru Li
- Medical College, Yanbian University, Yanji, 133002, P.R. China
| | - Hongyang Li
- Department of Blood Transfusion, China-Japan, Union Hospital of Jilin University, Changchun, 130033, P.R. China
| | - Yunyun Liu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, P.R. China
| | - Ningyi Jin
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, P.R. China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, P.R. China
| | - Xiao Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, P.R. China.
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, P.R. China.
| | - Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, P.R. China.
| | - Tiemei Liu
- Department of Blood Transfusion, China-Japan, Union Hospital of Jilin University, Changchun, 130033, P.R. China.
- Jingyue Economic & Technological Development Zone, No. 1035, Boshuo road, Changchun, Jilin, P. R. China.
| |
Collapse
|
22
|
Liu X, Tuerxun H, Zhao Y, Li Y, Wen S, Li X, Zhao Y. Crosstalk between ferroptosis and autophagy: broaden horizons of cancer therapy. J Transl Med 2025; 23:18. [PMID: 39762980 PMCID: PMC11702107 DOI: 10.1186/s12967-024-06059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Ferroptosis and autophagy are two main forms of regulated cell death (RCD). Ferroptosis is a newly identified RCD driven by iron accumulation and lipid peroxidation. Autophagy is a self-degradation system through membrane rearrangement. Autophagy regulates the metabolic balance between synthesis, degradation and reutilization of cellular substances to maintain intracellular homeostasis. Numerous studies have demonstrated that both ferroptosis and autophagy play important roles in cancer pathogenesis and cancer therapy. We also found that there are intricate connections between ferroptosis and autophagy. In this article, we tried to clarify how different kinds of autophagy participate in the process of ferroptosis and sort out the common regulatory pathways between ferroptosis and autophagy in cancer. By exploring the complex crosstalk between ferroptosis and autophagy, we hope to broaden horizons of cancer therapy.
Collapse
Affiliation(s)
- Xingyu Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Halahati Tuerxun
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yixin Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yawen Li
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shuhui Wen
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xi Li
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yuguang Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
23
|
Sun J, Ding J, Yue H, Xu B, Sodhi A, Xue K, Ren H, Qian J. Hypoxia-induced BNIP3 facilitates the progression and metastasis of uveal melanoma by driving metabolic reprogramming. Autophagy 2025; 21:191-209. [PMID: 39265983 DOI: 10.1080/15548627.2024.2395142] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 09/14/2024] Open
Abstract
Uveal melanoma (UM) is an aggressive intraocular malignancy derived from melanocytes in the uvea tract of the eye. Up to 50% of patients with UM develop distant metastases which is usually fatal within one year; preventing metastases is therefore essential. Metabolic reprogramming plays a critical role in UM progression and metastasis. However, the metabolic phenotype of UM cells in the hypoxic tumor is not well understood. Here, we report that hypoxia-induced BNIP3 reprograms tumor cell metabolism, promoting their survival and metastasis. In response to hypoxia, BNIP3-mediated mitophagy alleviates mitochondrial dysfunction and enhances mitochondrial oxidative phosphorylation (OXPHOS) while simultaneously reducing mitochondrial reactive oxygen species (mtROS) production. This, in turn, impairs HIF1A/HIF-1α protein stability and inhibits glycolysis. Inhibition of mitophagy significantly suppresses BNIP3-induced UM progression and metastasis in vitro and in vivo. Collectively, these observations demonstrate a novel mechanism whereby BNIP3 promotes UM metabolic reprogramming and malignant progression by mediating hypoxia-induced mitophagy and suggest that BNIP3 could be an important therapeutic target to prevent metastasis in patients with UM.Abbreviations: AOD: average optical density; BNIP3: BCL2/adenovirus E1B interacting protein 3; CQ: chloroquine; CoCl2: cobalt chloride; GEPIA: Gene Expression Profiling Interactive Analysis; HIF1A: hypoxia inducible factor 1, alpha subunit; IHC: immunohistochemistry; mtROS: mitochondrial reactive oxygen species; NAC: N-acetylcysteine; OCR: oxygen consumption rate; OXPHOS: oxidative phosphorylation; ROS: reactive oxygen species; TCGA: The Cancer Genome Atlas; UM: uveal melanoma.
Collapse
Affiliation(s)
- Jie Sun
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Shenzhen Eye Hospital, Jinan University, Shenzhen, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jie Ding
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Han Yue
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Binbin Xu
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Akrit Sodhi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kang Xue
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Hui Ren
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jiang Qian
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
24
|
Lei J, Zhai J, Qi J, Sun C. Identification of sepsis-associated encephalopathy biomarkers through machine learning and bioinformatics approaches. Sci Rep 2024; 14:31717. [PMID: 39738412 PMCID: PMC11685593 DOI: 10.1038/s41598-024-82885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025] Open
Abstract
Sepsis-associated encephalopathy (SAE) is common in septic patients, characterized by acute and long-term cognitive impairment, and is associated with higher mortality. This study aimed to identify SAE-related biomarkers and evaluate their diagnostic potential. We analyzed three SAE-related sequencing datasets, using two as training sets and one as a validation set. Weighted Gene Co-expression Network Analysis and four machine learning methods-Elastic Net regression, LASSO, random forest, and XGBoost-were employed, dentifying 18 biomarkers with significant expression changes. External validation and in vitro experiments confirmed the differential expression of these biomarkers. These findings provide insights into SAE pathogenesis and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Jingchao Lei
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Jia Zhai
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Jing Qi
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Chuanzheng Sun
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| |
Collapse
|
25
|
Guo J, Wang J, Fan S, Gao M, Liu G, Xia Y. The Novel Elemene Derivative, OMe-Ph-Elemene, Attenuates Oxidative Phosphorylation and Facilitates Apoptosis by Inducing Intracellular Reactive Oxygen Species. Antioxidants (Basel) 2024; 13:1499. [PMID: 39765827 PMCID: PMC11672920 DOI: 10.3390/antiox13121499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/01/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The incidence and mortality rates of colorectal cancer have been steadily increasing, making it one of the most prevalent cancers globally. Although current chemotherapy drugs have shown some efficacy in treating this disease, their associated side effects necessitate the development of more effective treatments and medications. The clinical application of elemene is widely utilized in tumor treatment; however, its efficacy is hindered by the requirement for high dosage and suboptimal anticancer effects. Thus, we have made modifications and enhancements to elemene, resulting in the development of a novel compound named (E)-8-(3,4,5-OMe-Ph)-β-Elemene (abbreviated as OMe-Ph-Elemene) that demonstrates significantly enhanced efficacy in suppressing colorectal cancer. We conducted an in vivo study and demonstrated the potential of OMe-Ph-Elemene in suppressing the growth of colorectal cancer xenograft tumors in zebrafish. The in vitro experiments revealed that OMe-Ph-Elemene effectively inhibited the proliferation and migration of colorectal cancer SW480 and HT-29 cells by inducing reactive oxygen species (ROS)-caused apoptosis and inhibiting mitochondrial oxidative phosphorylation. The mechanism was elucidated through high-throughput proteomic analysis and molecular biological analysis, revealing that OMe-Ph-Elemene induced cellular oxidative stress by downregulating CISD3 and promoted cell apoptosis by downregulating TRIAP1 and upregulating HMOX1. Furthermore, OMe-Ph-Elemene suppressed colorectal cancer cells' mitochondrial oxidative phosphorylation by downregulating NDUFA7. In summary, the utilization of the elemene parent nucleus structure has led to the derivation of a novel tumor suppressor compound characterized by high efficacy and low toxicity, thereby providing a significant reference for the development of innovative drugs for colorectal cancer treatment.
Collapse
Affiliation(s)
- Jianhua Guo
- Cheeloo College of Medicine, Shandong University, Jinan 250012, China;
- Key Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, College of Medical Engineering, Jining Medical University, Jining 272067, China; (J.W.); (S.F.); (M.G.)
| | - Jiayi Wang
- Key Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, College of Medical Engineering, Jining Medical University, Jining 272067, China; (J.W.); (S.F.); (M.G.)
| | - Shuhao Fan
- Key Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, College of Medical Engineering, Jining Medical University, Jining 272067, China; (J.W.); (S.F.); (M.G.)
| | - Mucong Gao
- Key Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, College of Medical Engineering, Jining Medical University, Jining 272067, China; (J.W.); (S.F.); (M.G.)
| | - Guodu Liu
- Inner Mongolia Key Laboratory of Fine Organic Synthesis, College of Chemistry and Chemical Engineering, Inner Mongolia University (South Campus), Hohhot 010030, China
| | - Yong Xia
- Cheeloo College of Medicine, Shandong University, Jinan 250012, China;
- Key Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, College of Medical Engineering, Jining Medical University, Jining 272067, China; (J.W.); (S.F.); (M.G.)
| |
Collapse
|
26
|
Chiarelli R, Caradonna F, Naselli F. Autophagy and nutrigenomics: a winning team against chronic disease and tumors. Front Nutr 2024; 11:1409142. [PMID: 39703336 PMCID: PMC11655209 DOI: 10.3389/fnut.2024.1409142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Autophagy, a vital cell process, has garnered attention for its role in various diseases and potential therapeutic interventions. Dysregulation of autophagy contributes to conditions such as metabolic diseases, neurodegenerative disorders, and cancer. In diseases such as diabetes, autophagy plays a crucial role in islet β-cell maintenance and glucose homeostasis, offering potential targets for therapeutic intervention. Nutrigenomics, which explores how dietary components interact with the genome, has emerged as a promising avenue for disease management. It sheds light on how diet influences gene expression and cellular processes, offering personalized approaches to disease prevention and management. Studies have showed the impact of specific dietary components, such as polyphenols and omega-3 fatty acids, on autophagy processes, suggesting their potential therapeutic benefits in neurodegenerative conditions and metabolic disorders. In cancer, autophagy's dual role in either suppressing tumorigenesis or promoting cancer cell survival underscores the importance of understanding its modulation through dietary interventions. Combined with conventional chemotherapy drugs, dietary compounds show synergistic effects in cancer treatment. Furthermore, phytochemicals such as indicaxanthin have been found to epigenetically regulate genes involved in autophagy, offering novel insights into personalized cancer therapies. This comprehensive review has the aim to study the autophagy in a combined view with nutrigenomics effects of some dietary molecules in maintaining cellular homeostasis and responding to pathological stimuli. Overall, the intersection of autophagy and nutrigenomics effect of bioactive compounds holds promise for developing targeted interventions for various diseases, emphasizing the significance of dietary interventions in disease prevention and management.
Collapse
Affiliation(s)
- Roberto Chiarelli
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Fabio Caradonna
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Flores Naselli
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| |
Collapse
|
27
|
Chen F, Xue Y, Zhang W, Zhou H, Zhou Z, Chen T, YinWang E, Li H, Ye Z, Gao J, Wang S. The role of mitochondria in tumor metastasis and advances in mitochondria-targeted cancer therapy. Cancer Metastasis Rev 2024; 43:1419-1443. [PMID: 39307891 PMCID: PMC11554835 DOI: 10.1007/s10555-024-10211-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/08/2024] [Indexed: 11/05/2024]
Abstract
Mitochondria are central actors in diverse physiological phenomena ranging from energy metabolism to stress signaling and immune modulation. Accumulating scientific evidence points to the critical involvement of specific mitochondrial-associated events, including mitochondrial quality control, intercellular mitochondrial transfer, and mitochondrial genetics, in potentiating the metastatic cascade of neoplastic cells. Furthermore, numerous recent studies have consistently emphasized the highly significant role mitochondria play in coordinating the regulation of tumor-infiltrating immune cells and immunotherapeutic interventions. This review provides a comprehensive and rigorous scholarly investigation of this subject matter, exploring the intricate mechanisms by which mitochondria contribute to tumor metastasis and examining the progress of mitochondria-targeted cancer therapies.
Collapse
Affiliation(s)
- Fanglu Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yucheng Xue
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wenkan Zhang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hao Zhou
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhiyi Zhou
- The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Eloy YinWang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
28
|
Wang Y, Liu Y, Su X, Niu L, Li N, Xu C, Sun Z, Guo H, Shen S, Yu M. Non-pathogenic Trojan horse Nissle1917 triggers mitophagy through PINK1/Parkin pathway to discourage colon cancer. Mater Today Bio 2024; 29:101273. [PMID: 39415764 PMCID: PMC11480251 DOI: 10.1016/j.mtbio.2024.101273] [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: 04/18/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Bacteria-mediated antitumor therapy has gained widespread attention for its innate tumor-targeting capability and excellent immune activation properties. Nevertheless, the clinical approval of bacterial therapies remains elusive primarily due to the formidable challenge of balancing safety with enhancing in vivo efficacy. In this study, leveraging the probiotic Escherichia coli Nissle1917 (EcN) emerges as a promising approach for colon cancer therapy, offering a high level of safety attributed to its lack of virulence factors and its tumor-targeting potential owing to its obligate anaerobic nature. Specifically, we delineate the erythrocyte (RBC) membrane-camouflaged EcN, termed as Trojan horse EcN@RBC, which triggers apoptosis in tumor cells by mitigating mitochondrial membrane potential (MMP) and subsequently activating the PINK1/Parkin pathway associated with mitophagy. Concurrently, the decline in MMP induced by mitophagy disrupts the mitochondrial permeability transition pore (MPTP), leading to the release of Cytochrome C and subsequent apoptosis induction. Moreover, synergistic effects were observed through the combination of the autophagy activator rapamycin, bolstering the antitumor efficacy in vivo. These findings offer novel insights into probiotic-mediated antitumor mechanisms and underscore the therapeutic potential of EcN@RBC for colon cancer patients.
Collapse
Affiliation(s)
- Yang Wang
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Yao Liu
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xiaomin Su
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Lili Niu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Nannan Li
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Ce Xu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Zanya Sun
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Huishu Guo
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, Shanghai, 201399, China
| |
Collapse
|
29
|
Huang Y, Li W, Sun H, Guo X, Zhou Y, Liu J, Liu F, Fan Y. Mitochondrial transfer in the progression and treatment of cardiac disease. Life Sci 2024; 358:123119. [PMID: 39395616 DOI: 10.1016/j.lfs.2024.123119] [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: 08/13/2024] [Revised: 09/27/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
Mitochondria are the primary site for energy production and play a crucial role in supporting normal physiological functions of the human body. In cardiomyocytes (CMs), mitochondria can occupy up to 30 % of the cell volume, providing sufficient energy for CMs contraction and relaxation. However, some pathological conditions such as ischemia, hypoxia, infection, and the side effect of drugs, can cause mitochondrial dysfunction in CMs, leading to various myocardial injury-related diseases including myocardial infarction (MI), myocardial hypertrophy, and heart failure. Self-control of mitochondria quality and conversion of metabolism pathway in energy production can serve as the self-rescue measure to avoid autologous mitochondrial damage. Particularly, mitochondrial transfer from the neighboring or extraneous cells enables to mitigate mitochondrial dysfunction and restore their biological functions in CMs. Here, we described the homeostatic control strategies and related mechanisms of mitochondria in injured CMs, including autologous mitochondrial quality control, mitochondrial energy conversion, and especially the exogenetic mitochondrial donation. Additionally, this review emphasizes on the therapeutic effects and potential application of utilizing mitochondrial transfer in reducing myocardial injury. We hope that this review can provide theoretical clues for the developing of advanced therapeutics to treat cardiac diseases.
Collapse
Affiliation(s)
- Yaqing Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Wanling Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Hongyu Sun
- The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Xin Guo
- The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| | - Yonghong Fan
- The General Hospital of Western Theater Command, Chengdu 610083, China.
| |
Collapse
|
30
|
Lu D, Li Y, Niu X, Sun J, Zhan W, Shi Y, Yu K, Huang S, Liu X, Xie L, Ma X, Liu B. STAT2/SLC27A3/PINK1-Mediated Mitophagy Remodeling Lipid Metabolism Contributes to Pazopanib Resistance in Clear Cell Renal Cell Carcinoma. RESEARCH (WASHINGTON, D.C.) 2024; 7:0539. [PMID: 39600540 PMCID: PMC11588985 DOI: 10.34133/research.0539] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024]
Abstract
Background: Clear cell renal cell carcinoma (ccRCC) is a prevalent malignant tumor of the urinary system. While tyrosine kinase inhibitors (TKIs) are currently the first-line treatments for advanced/metastatic ccRCC, patients often develop resistance after TKI therapy. Lipid metabolic reprogramming, a hallmark of tumor progression, contributes to acquired drug resistance in various malignant tumors. Mitophagy, a process that maintains mitochondrial homeostasis, aids tumor cells in adapting to microenvironmental changes and consequently developing drug resistance. Solute carrier family 27 member 3 (SLC27A3), highly expressed in lipid-rich tumors like ccRCC, has been associated with poor prognosis. However, the impact of SLC27A3 and the transcription factor complex containing STAT2 on lipid metabolic reprogramming, mitophagy in ccRCC, and their role in TKI resistance remain unexplored. Methods: 786-O to pazopanib resistance was induced by gradient increase of concentration, and the genes related to lipid metabolism were screened by RNA sequencing. Bioinformatics was used to analyze the differential expression of SLC27A3 and its effect on patient prognosis, and to predict the activated pathway in pazopanib-resistant cells. Lipid droplets (LDs) were detected by Red Oil O and BODIPY probe. Micro-targeted lipidomic of acyl-coenzyme A (CoA) and lipid metabolomics were performed to screen potential metabolites of SLC27A3. The differential expression of SLC27A3 was detected in clinical samples. The differential expression of SLC27A3 and its effect on drug resistance of ccRCC tumor were detected in vitro and in vivo. Mitophagy was detected by electron microscopy, Mtphagy probe, and Western blot. The mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) levels were detected by JC-1 and DCF probes. The binding site of the transcription factor complex to the SLC27A3 promoter was detected by dual-luciferase reporter gene assay. Results: SLC27A3, highly expressed in lipid-rich tumors such as ccRCC and glioblastoma, predicts poor prognosis. SLC27A3 expression level also increased in pazopanib-resistant 786-O cells (786-O-PR) with more LD accumulation compared to parental cells. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis from RNA sequencing showed that PINK1/Parkin-mediated mitophagy pathway was enriched in 786-O-PR. Knockdown of SLC27A3 markedly suppressed LD accumulation and mitophagy, and overcame pazopanib resistance in vitro and in vivo. Moreover, SLC27A3 functions as an acyl-CoA ligase catalyzing the formation of acyl-CoA, which refers to fatty acid oxidation accompanied by ROS production and synthesis of lipid. Overproduced acyl-CoA oxidation in mitochondria resulted in MMP decrease and amounts of ROS production, subsequently triggering PINK1/Parkin-mediated mitophagy. Moreover, mitophagy inhibition led to more ROS accumulation and cell death, indicating that mitophagy can keep ROS at an appropriate level by negative feedback. Mitophagy, simultaneously, prevented fatty acid oxidation in mitochondria by consuming CPT1A, forcing synthesis of triglycerides and cholesterol esters stored in LDs by transforming acyl-CoA, to support ccRCC progression. Besides, we found that STAT2 expression was positively correlated to SLC27A3. Transcriptional factor complex containing STAT2 could bind to the promoter of SLC27A3 mRNA to promote SLC27A3 transcription proved by dual-luciferase reporter assay, which also regulated LD metabolism and activated mitophagy during pazopanib resistance. Conclusion: SLC27A3 is up-regulated in pazopanib-resistant ccRCC and predicts poor prognosis. High expression of SLC27A3 produces excessive metabolites of various long-chain fatty acyl-CoA (12:0-, 16:0-, 17:0-, 20:3-CoA) to enter mitochondria for β-oxidation and produce amounts of ROS activating mitophagy. Subsequent mitophagy/ROS negative feedback controls ROS homeostasis and consumes CPT1A protein within mitochondria to suppress fatty acid β-oxidation, forcing acyl-CoA storage in LDs, mediating pazopanib resistance in ccRCC. Furthermore, STAT2 was identified as a core component of a potential upstream transcriptional factor complex for SLC27A3. Our findings shed new light on the underlying mechanism of SLC27A3 in ccRCC TKI resistance, which may provide a novel therapeutic target for the management of ccRCC.
Collapse
Affiliation(s)
- Dingheng Lu
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Yuxiao Li
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Xinyang Niu
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Jiazhu Sun
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Weitao Zhan
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Yuchen Shi
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Kai Yu
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Suyuelin Huang
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Xiaoyan Liu
- Department of Pathology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Liping Xie
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
| | - Xueyou Ma
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
- Cancer Center,
Zhejiang University, Hangzhou, 310003 Zhejiang, China
| | - Ben Liu
- Department of Urology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, 310003 Zhejiang, China
- Cancer Center,
Zhejiang University, Hangzhou, 310003 Zhejiang, China
| |
Collapse
|
31
|
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
|
32
|
Xu Z, Zhao G, Zhang L, Qiao C, Wang H, Wei H, Liu R, Liu P, Zhang Y, Zhu W, You W. Tong-Xie-Yao-Fang induces mitophagy in colonic epithelial cells to inhibit colitis-associated colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118541. [PMID: 38992403 DOI: 10.1016/j.jep.2024.118541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 06/10/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Based on the core pathogenesis of hepatosplenic disorder and qi transformation disorder in ulcerative colitis, Tong-Xie-Yao-Fang (TXYF) is a classical traditional Chinese medicine commonly used to treat ulcerative colitis. Our study revealed that it has the potential to prevent colitis-associated colorectal cancer, which embodies the academic concept in traditional Chinese medicine of treating the disease before it develops. AIM OF THE STUDY This study was aimed at evaluating the therapeutic role of TXYF in treating colitis-associated colorectal cancer and exploring its possible underlying mechanisms. MATERIALS AND METHODS A colitis-associated colorectal cancer model was established in mice using azoxymethane and dextran sulfate sodium salt to examine the therapeutic effect of TXYF. The mouse body weights were observed. Hematoxylin-eosin staining was used to evaluate mouse colon histopathology. Colon cancer cells and colon epithelial cells were used to explore the potential molecular mechanisms. The proliferation and apoptosis of cells were detected by CCK8 and cell colony assays, flow cytometry and western blotting. The epithelial-mesenchymal transition (EMT) and mitophagy markers were examined by immunohistochemistry, western blotting, quantitative real-time PCR and immunofluorescence staining. RESULTS TXYF inhibited the tumorigenesis of mice with colitis-associated colorectal cancer and the growth of inflammatory colon cells. TXYF induced mitophagy in colon cancer cells through the PTEN-induced putative kinase 1 (PINK1)/Parkin pathway to reverse EMT, which was consistent with the results in mice with colitis-associated colorectal cancer. CONCLUSIONS The results of the present study demonstrated that TXYF effectively inhibited the progression of colitis-associated colorectal cancer through the PINK1/Parkin pathway, which provides new evidence for prevention strategies for this disease.
Collapse
Affiliation(s)
- Zitong Xu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Gang Zhao
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Lize Zhang
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Cuixia Qiao
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Hao Wang
- Department of President's Office, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Hongyun Wei
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Ruiqing Liu
- Department of Gastroenterological Surgery, The Affiliated Hospital of Qingdao University, Shandong, 266000, China.
| | - Penglin Liu
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Yuejuan Zhang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Shandong, 266000, China.
| | - Wei Zhu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Shandong, 266000, China.
| | - Wenli You
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| |
Collapse
|
33
|
Santos de Macedo BG, Albuquerque de Melo M, Pereira-Martins DA, Machado-Neto JA, Traina F. An updated outlook on autophagy mechanism and how it supports acute myeloid leukemia maintenance. Biochim Biophys Acta Rev Cancer 2024; 1879:189214. [PMID: 39515545 DOI: 10.1016/j.bbcan.2024.189214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
The gradual acquisition of genetic and epigenetic disturbances bestows malignant traits upon hematopoietic stem cells, subverting them into a founder and reservoir cell for de novo acute myeloid leukemia (AML) known as leukemic stem cells (LSC). Beyond its molecular heterogeneity, AML is also characterized by rewiring biological processes to support its onset and maintenance. LSC were observed to inherently and actively trigger mitochondrial turnover through selective autophagic removal such that impairing the process led to cell differentiation at the expense of its stemness. This review provides a current take on autophagy regulation mechanisms according to the current molecular characterization of the process; describes autophagy as a drug resistance mechanism, and a pivotal mechanism whereby LSC harmonize their strong reliance on mitochondrial respiration to obtain energy, and their necessity for lower internal oxidative stress to avoid exhaustion. Therefore, targeting autophagy presents a promising strategy to promote long-term remissions in AML.
Collapse
Affiliation(s)
- Brunno Gilberto Santos de Macedo
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Manuela Albuquerque de Melo
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - João Agostinho Machado-Neto
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Pharmacology, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Fabiola Traina
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
34
|
Ganguly U, Carroll T, Nehrke K, Johnson GVW. Mitochondrial Quality Control in Alzheimer's Disease: Insights from Caenorhabditis elegans Models. Antioxidants (Basel) 2024; 13:1343. [PMID: 39594485 PMCID: PMC11590956 DOI: 10.3390/antiox13111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder that is classically defined by the extracellular deposition of senile plaques rich in amyloid-beta (Aβ) protein and the intracellular accumulation of neurofibrillary tangles (NFTs) that are rich in aberrantly modified tau protein. In addition to aggregative and proteostatic abnormalities, neurons affected by AD also frequently possess dysfunctional mitochondria and disrupted mitochondrial maintenance, such as the inability to eliminate damaged mitochondria via mitophagy. Decades have been spent interrogating the etiopathogenesis of AD, and contributions from model organism research have aided in developing a more fundamental understanding of molecular dysfunction caused by Aβ and toxic tau aggregates. The soil nematode C. elegans is a genetic model organism that has been widely used for interrogating neurodegenerative mechanisms including AD. In this review, we discuss the advantages and limitations of the many C. elegans AD models, with a special focus and discussion on how mitochondrial quality control pathways (namely mitophagy) may contribute to AD development. We also summarize evidence on how targeting mitophagy has been therapeutically beneficial in AD. Lastly, we delineate possible mechanisms that can work alone or in concert to ultimately lead to mitophagy impairment in neurons and may contribute to AD etiopathology.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Trae Carroll
- Department of Pathology, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| |
Collapse
|
35
|
Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y, Chang X. Emerging insights into the pathogenesis and therapeutic strategies for vascular endothelial injury-associated diseases: focus on mitochondrial dysfunction. Angiogenesis 2024; 27:623-639. [PMID: 39060773 PMCID: PMC11564294 DOI: 10.1007/s10456-024-09938-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
As a vital component of blood vessels, endothelial cells play a key role in maintaining overall physiological function by residing between circulating blood and semi-solid tissue. Various stress stimuli can induce endothelial injury, leading to the onset of corresponding diseases in the body. In recent years, the importance of mitochondria in vascular endothelial injury has become increasingly apparent. Mitochondria, as the primary site of cellular aerobic respiration and the organelle for "energy information transfer," can detect endothelial cell damage by integrating and receiving various external stress signals. The generation of reactive oxygen species (ROS) and mitochondrial dysfunction often determine the evolution of endothelial cell injury towards necrosis or apoptosis. Therefore, mitochondria are closely associated with endothelial cell function, helping to determine the progression of clinical diseases. This article comprehensively reviews the interconnection and pathogenesis of mitochondrial-induced vascular endothelial cell injury in cardiovascular diseases, renal diseases, pulmonary-related diseases, cerebrovascular diseases, and microvascular diseases associated with diabetes. Corresponding therapeutic approaches are also provided. Additionally, strategies for using clinical drugs to treat vascular endothelial injury-based diseases are discussed, aiming to offer new insights and treatment options for the clinical diagnosis of related vascular injuries.
Collapse
Affiliation(s)
- Boxian Pang
- Beijing University of Chinese Medicine, Beijing, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | | | - Tieliang Pang
- Beijing Anding hospital, Capital Medical University, Beijing, China
| | - Xinyao Sun
- Beijing University of Chinese Medicine, Beijing, China
| | - Xin Liu
- Bioscience Department, University of Nottingham, Nottingham, UK
| | - Yifeng Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiagge, Xicheng District, Beijing, China.
| |
Collapse
|
36
|
Li S, Feng S, Chen Y, Sun B, Zhang N, Zhao Y, Han J, Liu Z, He YQ, Wang Q. Ciclopirox platinum(IV) conjugates suppress tumors by promoting mitophagy and provoking immune responses. J Inorg Biochem 2024; 260:112696. [PMID: 39142055 DOI: 10.1016/j.jinorgbio.2024.112696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/11/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Mitophagy is an important target for antitumor drugs development. A series of ciclopirox (CPX) platinum(IV) hybrids targeting PTEN induced putative kinase 1 (PINK1)/Parkin mediated mitophagy were designed and prepared as antitumor agents. The dual CPX platinum(IV) complex with cisplatin core was screened out as a candidate, which displayed promising antitumor activities both in vitro and in vivo. Mechanistically, it caused serious DNA damage in tumor cells. Then, remarkable mitochondrial damage was induced accompanied by the mitochondrial membrane depolarization and reactive oxygen species generation, which further promoted apoptosis through the Bcl-2/Bax/Caspase3 pathway. Furthermore, mitophagy was ignited via the PINK1/Parkin/P62/LC3 axis, and exhibited positive influence on promoting the apoptosis of tumor cells. The antitumor immunity was boosted by the block of immune check point programmed cell death ligand-1 (PD-L1), which further increased the density of T cells in tumors. Subsequently, the metastasis of tumor cells was inhibited by inhibiting angiogenesis in tumors.
Collapse
Affiliation(s)
- Suying Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Shuaiqi Feng
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Yan Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China; Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Bin Sun
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Yanna Zhao
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Jun Han
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China; Liaocheng High-Tech Biotechnology Co., Ltd, Liaocheng 252059, PR China
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Yan-Qin He
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China.
| |
Collapse
|
37
|
Wang L, Zhang X, Huang X, Sha X, Li X, Zheng J, Li S, Wei Z, Wu F. Homoplantaginin alleviates high glucose-induced vascular endothelial senescence by inhibiting mtDNA-cGAS-STING pathway via blunting DRP1-mitochondrial fission-VDAC1 axis. FASEB J 2024; 38:e70127. [PMID: 39436199 DOI: 10.1096/fj.202401299rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
Vascular endothelial senescence is a major risk factor for diabetic vascular complications. Abnormal mitochondrial fission by dynamically related protein 1 (DRP1) accelerates vascular endothelial cell senescence. Homoplantaginin (Hom) is a flavonoid in Salvia plebeia R. Br. with protecting mitochondrial and repairing vascular properties. However, the relevant mechanism of Hom against diabetic vascular endothelial cell senescence remains unclear. Here, we used db/db mice and high glucose (HG)-treated human umbilical vein endothelial cells (HUVECs) to assess the anti-vascular endothelial cell senescence of Hom. We found that Hom inhibited senescence-associated β-galactosidase activity, decreased the levels of senescence markers, and senescence-associated secretory phenotype factors. Additionally, Hom inhibited the expression of cGAS-STING pathway and downstream inflammatory factors. STING inhibitor H-151 delayed endothelial senescence, whereas STING overexpression attenuated the anti-endothelial senescence effect of Hom. Furthermore, we observed that Hom reduced mitochondrial fragmentation and inhibited abnormal mitochondrial fission using transmission electron microscopy. Importantly, Hom has a stronger effect on mitochondrial fission protein than mitochondrial fusion protein, especially downregulated the expression of DRP1. DRP1 inhibitor Mdivi-1 suppressed cGAS-STING pathway and vascular endothelial senescence, yet DRP1 agonist FCCP attenuated the effect of Hom. Surprisingly, Hom blunted abnormal mitochondrial fission mediated by DRP1 mitochondrial localization, suppressed interaction of DRP1 with VDAC1 and prevented VDAC1 oligomerization, which was necessary for mtDNA escape and subsequent cGAS-STING pathway activation. These results revealed a previously unrecognized mechanism that Hom alleviated vascular endothelial senescence by inhibited mtDNA-cGAS-STING signaling pathway via blunting DRP1-mitochondrial fission-VDAC1 axis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xueying Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xi Huang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaotong Sha
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xulu Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jianmei Zheng
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shitong Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feihua Wu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
38
|
Chen C, Xiang A, Lin X, Guo J, Liu J, Hu S, Rui T, Ye Q. Mitophagy: insights into its signaling molecules, biological functions, and therapeutic potential in breast cancer. Cell Death Discov 2024; 10:457. [PMID: 39472438 PMCID: PMC11522701 DOI: 10.1038/s41420-024-02226-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
Mitophagy, a form of selective autophagy that removes damaged or dysfunctional mitochondria, plays a crucial role in maintaining mitochondrial and cellular homeostasis. Recent findings suggest that defective mitophagy is closely associated with various diseases, including breast cancer. Moreover, a better understanding of the multifaceted roles of mitophagy in breast cancer progression is crucial for the treatment of this disease. Here, we will summarize the molecular mechanisms of mitophagy process. In addition, we highlight the expression patterns and roles of mitophagy-related signaling molecules in breast cancer progression and the potential implications of mitophagy for the development of breast cancer, aiming to provide better therapeutic strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Cong Chen
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Aizhai Xiang
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Xia Lin
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jian Liu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Shufang Hu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Tao Rui
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Qianwei Ye
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
39
|
Sang W, Yan X, Wang L, Sun H, Jian Y, Wang F, Tang B, Li Y. CALCOCO2 prevents AngII-induced atrial remodeling by regulating the interaction between mitophagy and mitochondrial stress. Int Immunopharmacol 2024; 140:112841. [PMID: 39094358 DOI: 10.1016/j.intimp.2024.112841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/28/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND The biological functions of mitochondrial complexes are closely related to the development of atrial fibrillation (AF). Calcium binding and coiled-coil domain 2 (CALCOCO2) is a novel and specific receptor for mitophagy; however, its function in AF remains unknown. Therefore, this study aimed to investigate the role and molecular mechanisms of CALCOCO2 in AF, especially its regulatory mechanism in mitophagy and mitochondrial stress. METHODS Mice and HL-1 cells were treated with AngII to establish in vitro and in vivo AF models. Additionally, we examined the effect of CALCOCO2 or DAP3 Binding Cell Death Enhancer 1 (DELE1) overexpression on mitophagy and mitochondrial stress in AF models. To investigate the role of mitophagy in the regulatory effects of CALCOCO2 in AF, HL-1 cells were treated with chloroquine, a mitophagy inhibitor. Moreover, mitochondrial parameters were examined using specific fluorescent probes, transmission electron microscopy, western blotting, immunohistochemistry, and confocal microscopy. RESULTS AngII severely impaired the normal morphology and function of mitochondria; inhibited mitophagy; promoted atrial mitochondrial stress, fibrosis, and oxidative stress; and accelerated the progression of atrial remodeling in atrial myocytes. However, CALCOCO2 overexpression reversed/ameliorated these AF-induced changes. Additionally, CALCOCO2 overexpression restored mitochondrial homeostasis in atrial muscle by activating mitophagy and ameliorating mitochondrial stress. Mechanistically, DELE1 overexpression increased mitochondrial reactive oxygen species level and the expression of mitochondrial stress proteins (HRI, eIF2α, and ATF4) even in CALCOCO2-expressing in vitro AF models.. CONCLUSIONS CALCOCO2 may serve as a potential target for AF therapy to prevent or reverse the progression of atrial remodeling by regulating mitophagy and DELE1-mediated mitochondrial stress.
Collapse
Affiliation(s)
- Wanyue Sang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiaoji Yan
- Department of Emergency, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lu Wang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Huaxin Sun
- Department of Cardiology, The Third People's Hospital of Chengdu, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yi Jian
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Feifei Wang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xinjiang Emergency Center, People's Hospital of Xinjiang Uygur Autonomous Region, China
| | - Baopeng Tang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| | - Yaodong Li
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| |
Collapse
|
40
|
Wang L, Mei N, Li J, Chen H, He J, Wang R. Exploring the role of mitophagy-related genes in breast cancer: subtype classification and prognosis prediction. Int J Med Sci 2024; 21:2664-2682. [PMID: 39512680 PMCID: PMC11539391 DOI: 10.7150/ijms.100785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/04/2024] [Indexed: 11/15/2024] Open
Abstract
Background: Breast cancer (BC) is the most common cancer among women globally and poses the leading health threat to women worldwide, with persistently high incidence rates. Mitophagy is a selective autophagy process that specifically targets mitochondria within the cell, maintaining cellular energy balance and metabolic health by identifying and degrading damaged mitochondria. Although there is an understanding of the relationship between mitophagy and cancer, the specific mechanisms remain unclear due to the complexity and diversity of mitophagy, suggesting that it could be an effective and more targeted therapeutic approach for BC. Methods: In this study, we meticulously examined the BC expression and clinical pathology data from The Cancer Genome Atlas (TCGA) to assess the expression profiles, copy number variations (CNV), and to investigate the correlation, function, and prognostic impact of 34 mitophagy-related genes (MRGs). Differentially expressed genes (DEGs) were identified based on group classification. Lasso and Cox regression were used to determine prognostic genes for constructing a nomogram. Single-cell analysis mapped the distribution of these genes in BC cells. Additionally, the association between gene-derived risk scores and factors such as immune infiltration, tumor mutational burden (TMB), cancer stem cell (CSC) index, and drug responses was studied. In vitro experiments were conducted to confirm the analyses. Results: We included 34 MRGs and subsequently generated a risk score for 7 genes, including RPLP2, PCDHGA2, PRKAA2, CLIC6, FLT3, CHI3L1, and IYD. It was found that the low-risk group had better overall survival (OS) in BC, higher immune scores, but lower tumor mutational burden (TMB) and cancer stem cell (CSC) index, as well as lower IC50 values for commonly used drugs. To enhance clinical applicability, age and staging were incorporated into the risk score, and a more comprehensive nomogram was constructed to predict OS. This nomogram was validated and showed good predictive performance, with area under the curve (AUC) values for 1-year, 3-year, and 5-year OS of 0.895, 0.765, and 0.728, respectively. Conclusion: Our findings underscore the profound impact of prognostic genes on the immune response and prognostic outcomes in BC, indicating that they can provide new avenues for personalized BC treatment and potentially improve clinical outcomes.
Collapse
Affiliation(s)
- Lizhao Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Nan Mei
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jianpeng Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Heyan Chen
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jianjun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ru Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
41
|
Lin J, Chen X, Du Y, Li J, Guo T, Luo S. Mitophagy in Cell Death Regulation: Insights into Mechanisms and Disease Implications. Biomolecules 2024; 14:1270. [PMID: 39456203 PMCID: PMC11506020 DOI: 10.3390/biom14101270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/15/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Mitophagy, a selective form of autophagy, plays a crucial role in maintaining optimal mitochondrial populations, normal function, and intracellular homeostasis by monitoring and removing damaged or excess mitochondria. Furthermore, mitophagy promotes mitochondrial degradation via the lysosomal pathway, and not only eliminates damaged mitochondria but also regulates programmed cell death-associated genes, thus preventing cell death. The interaction between mitophagy and various forms of cell death has recently gained increasing attention in relation to the pathogenesis of clinical diseases, such as cancers and osteoarthritis, neurodegenerative, cardiovascular, and renal diseases. However, despite the abundant literature on this subject, there is a lack of understanding regarding the interaction between mitophagy and cell death. In this review, we discuss the main pathways of mitophagy, those related to cell death mechanisms (including apoptosis, ferroptosis, and pyroptosis), and the relationship between mitophagy and cell death uncovered in recent years. Our study offers potential directions for therapeutic intervention and disease diagnosis, and contributes to understanding the molecular mechanism of mitophagy.
Collapse
Affiliation(s)
| | | | | | | | | | - Sai Luo
- The 1st Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin 150000, China; (J.L.); (X.C.); (Y.D.); (J.L.); (T.G.)
| |
Collapse
|
42
|
ZHANG G, LIU G, GUO M, LUO F, YAN Z, GE W. Effect of phosphatase and tensin homolog-induced putative kinase 1/ E3 ubiquitin ligase Parkin mediated mitochondrial autophagy on chronic kidney disease myocardial injury and the intervention mechanism of Shenshuai recipe. J TRADIT CHIN MED 2024; 44:934-943. [PMID: 39380224 PMCID: PMC11462534 DOI: 10.19852/j.cnki.jtcm.20231231.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/10/2023] [Indexed: 10/10/2024]
Abstract
OBJECTIVE To study whether Shenshuai recipe (, SSR) can play a protective role on chronic kidney disease myocardial injury model through phosphatase and tensin homolog-induced putative kinase 1 (PINK1)/E3 ubiquitin ligase Parkin (Parkin) mitochondrial autophagy pathway. METHODS Forty-eight nephrectomized rats were randomly divided into six groups: sham-operated group, model group, Benazepril group, low, medium and high-dose groups of SSR. The rats were given the cor-responding intervention for six weeks, then were sacrificed. Serum was examined by enzyme linked immunosorbent assay (ELISA). Cardiac ultrasound was used to detect cardiac function in 5/6 nephrectomized rats. Myocardial tissue was examined by light and electron microscopy; PINK1, Parkin, microtubule-associated protein1 light chain 3 II (LC3B), sequestosome 1 (P62), BECN1 (Beclin-1) and dynamin-related protein 1 (Drp-1) were measured by real time polymerase chain reaction (RT-PCR), Western blot (WB) and immunohistochemistry (IHC). RESULTS The expression levels of blood urea nitrogen (BUN) and creatinine (SCr) in the model group were significantly higher than those in the sham-operated group, indicating that modeling was successful. SSR can protect myocardium by reducing the relative expression of creatine kinase myocardial isoenzyme and hypersensitivity cardiac troponin I (P<0.05). SSR can improve cardiac function in rats after ultrasound testing. SSR can improve the pathological manifestations of myocardial tissue after Masson staining. SSR can increase the number of autophagosomes and autophagiclysosomes in 5/6 nephrectomized rats (P<0.05). Determined by RT-PCR, WB and IHC, SSR can increase the relative expression of PINK1, Parkin, and LC3B (P<0.05), and decrease the relative expression of P62, Beclin-1 and Drp-1 (P<0.05). CONCLUSIONS The PINK1/Parkin mitochondrial autophagy pathway in myocardial tissues in 5/6 nephrectomy CKD myocardial injury rats was inhibited. SSR can activate PINK1/Parkin mitochondrial autophagy to enhance mitochondrial autophagy, and play a protective role in myocardial tissues.
Collapse
Affiliation(s)
- Gedi ZHANG
- 1 School of Graduate, Jiangxi University of Chinese Medicine, Nanchang 330000, China
| | - Gengxin LIU
- 1 School of Graduate, Jiangxi University of Chinese Medicine, Nanchang 330000, China
| | - Min GUO
- 1 School of Graduate, Jiangxi University of Chinese Medicine, Nanchang 330000, China
| | - Fuli LUO
- 2 Department of Nephrology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330000, China
| | - Ziyou YAN
- 2 Department of Nephrology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330000, China
| | - Wei GE
- 3 Department of Anorectal, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330000, China
| |
Collapse
|
43
|
Kobayashi H, Imanaka S, Yoshimoto C, Matsubara S, Shigetomi H. Rethinking the pathogenesis of endometriosis: Complex interactions of genomic, epigenetic, and environmental factors. J Obstet Gynaecol Res 2024; 50:1771-1784. [PMID: 39293995 DOI: 10.1111/jog.16089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/01/2024] [Indexed: 09/20/2024]
Abstract
AIM Endometriosis is a complex, multifactorial disease. Recent advances in molecular biology underscore that somatic mutations within the epithelial component of the normal endometrium, alongside aberrant epigenetic alterations within endometrial stromal cells, may serve as stimulators for the proliferation of endometriotic tissue within the peritoneal cavity. Nevertheless, pivotal inquiries persist: the deterministic factors driving endometriosis development in certain women while sparing others, notwithstanding comparable experiences of retrograde menstruation. Within this review, we endeavor to synopsize the current understanding of diverse pathophysiologic mechanisms underlying the initiation and progression of endometriosis and delineate avenues for future research. METHODS A literature search without time restriction was conducted utilizing PubMed and Google Scholar. RESULTS Given that aberrant clonal expansion stemming from cancer-associated mutations is common in normal endometrial tissue, only endometrial cells harboring mutations imparting proliferative advantages may be selected for survival outside the uterus. Endometriotic cells capable of engendering metabolic plasticity and modulating mitochondrial dynamics, thereby orchestrating responses to hypoxia, oxidative stress, inflammation, hormonal stimuli, and immune surveillance, and adeptly acclimating to their harsh surroundings, stand a chance at viability. CONCLUSION The genesis of endometriosis appears to reflect the evolutionary principles of mutation, selection, clonal expansion, and adaptation to the environment.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, Nara, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Medicine, Kei Oushin Clinic, Nishinomiya, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, Nara, Japan
| |
Collapse
|
44
|
Brogyanyi T, Kejík Z, Veselá K, Dytrych P, Hoskovec D, Masařik M, Babula P, Kaplánek R, Přibyl T, Zelenka J, Ruml T, Vokurka M, Martásek P, Jakubek M. Iron chelators as mitophagy agents: Potential and limitations. Biomed Pharmacother 2024; 179:117407. [PMID: 39265234 DOI: 10.1016/j.biopha.2024.117407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Mitochondrial autophagy (mitophagy) is very important process for the maintenance of cellular homeostasis, functionality and survival. Its dysregulation is associated with high risk and progression numerous serious diseases (e.g., oncological, neurodegenerative and cardiovascular ones). Therefore, targeting mitophagy mechanisms is very hot topic in the biological and medicinal research. The interrelationships between the regulation of mitophagy and iron homeostasis are now becoming apparent. In short, mitochondria are central point for the regulation of iron homeostasis, but change in intracellular cheatable iron level can induce/repress mitophagy. In this review, relationships between iron homeostasis and mitophagy are thoroughly discussed and described. Also, therapeutic applicability of mitophagy chelators in the context of individual diseases is comprehensively and critically evaluated.
Collapse
Affiliation(s)
- Tereza Brogyanyi
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - Michal Masařik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Babula
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Tomáš Přibyl
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic.
| |
Collapse
|
45
|
Rajendran P, Renu K, Ali EM, Genena MAM, Veeraraghavan V, Sekar R, Sekar AK, Tejavat S, Barik P, Abdallah BM. Promising and challenging phytochemicals targeting LC3 mediated autophagy signaling in cancer therapy. Immun Inflamm Dis 2024; 12:e70041. [PMID: 39436197 PMCID: PMC11494898 DOI: 10.1002/iid3.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Phytochemicals possess a wide range of anti-tumor properties, including the modulation of autophagy and regulation of programmed cell death. Autophagy is a critical process in cellular homeostasis and its dysregulation is associated with several pathological conditions, such as cancer, neurodegenerative diseases, and diabetes. In cancer, autophagy plays a dual role by either promoting tumor growth or suppressing it, depending on the cellular context. During autophagy, autophagosomes engulf cytoplasmic components such as proteins and organelles. LC3-II (microtubule-associated protein 1 light chain 3-II) is an established marker of autophagosome formation, making it central to autophagy monitoring in mammals. OBJECTIVE To explore the regulatory role of phytochemicals in LC3-mediated autophagy and their potential therapeutic impact on cancer. The review emphasizes the involvement of autophagy in tumor promotion and suppression, particularly focusing on autophagy-related signaling pathways like oxidative stress through the NRF2 pathway, and its implications for genomic stability in cancer development. METHODS The review focuses on a comprehensive analysis of bioactive compounds including Curcumin, Celastrol, Resveratrol, Kaempferol, Naringenin, Carvacrol, Farnesol, and Piperine. Literature on these compounds was examined to assess their influence on autophagy, LC3 expression, and tumor-related signaling pathways. A systematic literature search was conducted across databases including PubMed, Scopus, and Web of Science from inception to 2023. Studies were selected from prominent databases, focusing on their roles in cancer diagnosis and therapeutic interventions, particularly in relation to LC3-mediated mechanisms. RESULTS Phytochemicals have been shown to modulate autophagy through the regulation of LC3-II levels and autophagic flux in cancer cells. The interaction between autophagy and other cellular pathways such as oxidative stress, inflammation, and epigenetic modulation highlights the complex role of autophagy in tumor biology. For instance, Curcumin and Resveratrol have been reported to either induce or inhibit autophagy depending on cancer type, influencing tumor progression and therapeutic responses. CONCLUSION Targeting autophagy through LC3 modulation presents a promising strategy for cancer therapy. The dual role of autophagy in tumor suppression and promotion, however, necessitates careful consideration of the context in which autophagy is induced or inhibited. Future research should aim to delineate these context-specific roles and explore how phytochemicals can be optimized for therapeutic efficacy. Novel therapeutic strategies should focus on the use of bioactive compounds to fine-tune autophagy, thereby maximizing tumor suppression and inducing programmed cell death in cancer cells.
Collapse
Affiliation(s)
- Peramaiyan Rajendran
- Department of Biological Sciences, College of ScienceKing Faisal UniversityAl‐AhsaSaudi Arabia
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiTamil NaduIndia
| | - Kaviyarasi Renu
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiTamil NaduIndia
| | - Enas M. Ali
- Department of Biological Sciences, College of ScienceKing Faisal UniversityAl‐AhsaSaudi Arabia
- Department of Botany and Microbiology, Faculty of ScienceCairo UniversityCairoEgypt
| | - Marwa Azmy M. Genena
- Department of Biological Sciences, College of ScienceKing Faisal UniversityAl‐AhsaSaudi Arabia
- Agricultural Zoology Department, Faculty of AgricultureMansoura UniversityMansouraEgypt
| | - Vishnupriya Veeraraghavan
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiTamil NaduIndia
| | - Ramya Sekar
- Department of Oral & Maxillofacial Pathology and Oral MicrobiologyMeenakshi Ammal Dental College & Hospital, MAHERChennaiTamil NaduIndia
| | | | - Sujatha Tejavat
- Department of Biomedical Sciences, College of MedicineKing Faisal UniversityAl‐AhsaSaudi Arabia
| | | | - Basem M. Abdallah
- Department of Biological Sciences, College of ScienceKing Faisal UniversityAl‐AhsaSaudi Arabia
| |
Collapse
|
46
|
Wu B, Qi B, Duan L, Chen J. Lidamycin induces mitophagy in pancreatic cancer cells by regulating the expression of Mfn2. Sci Rep 2024; 14:20713. [PMID: 39237684 PMCID: PMC11377765 DOI: 10.1038/s41598-024-71377-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
Lidamycin (LDM) has been confirmed to have a strong anti-pancreatic cancer effect and can affect the mitochondrial function of pancreatic cancer cells. Mitofusin-2 (Mfn2) is located in the outer membrane of mitochondria, and Mfn2 is currently believed to play a role in cancer inhibition in pancreatic cancer. In order to explore whether the anti-pancreatic cancer effect of LDM is related to Mfn2-mediated mitophagy, Bioinformatics and in vitro cell experiments are used for experimental research. The experimental results demonstrated that Mfn2 is correlated with mitochondrial autophagy in pancreatic cancer. Lidamycin can increase the expression of Mfn2 in pancreatic cancer and affect the process of EMT, affect the level of reactive oxygen species and mitochondrial membrane potential, and increase the expression of mitochondrial autophagy marker proteins BNIP3L and Beclin1. These results demonstrate that Mfn2 affects mitophagy in pancreatic cancer cells by regulating the expression of Mfn2.
Collapse
Affiliation(s)
- Boya Wu
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China
| | - Bing Qi
- College of Life Sciences, North China University of Science and Technology, 21 Bohai Road, Caofeidian Xincheng, Tangshan, Hebei, China
| | - Liumeng Duan
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China
| | - Jing Chen
- College of Life Sciences, North China University of Science and Technology, 21 Bohai Road, Caofeidian Xincheng, Tangshan, Hebei, China.
| |
Collapse
|
47
|
Liu F, Wang G, Zhao L, Chen G, Dong L, Li Q, Zhu D. Toosendanin Induces Lung Squamous Cell Carcinoma Cell Apoptosis and Inhibits Tumor Progression via the BNIP3/AMPK Signaling Pathway. Adv Biol (Weinh) 2024; 8:e2300610. [PMID: 38773915 DOI: 10.1002/adbi.202300610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/28/2024] [Indexed: 05/24/2024]
Abstract
Lung squamous cell carcinoma (LUSC) is the second most common type of non-small cell lung cancer. Toosendanin can target critical cancer cell survival and proliferation. However, the function of toosendanin in LUSC is limited. Cancer cell proliferative capacity is detected using cell morphology, colony formation, and flow cytometry. The invasiveness of the cells is detected by a Transwell assay, western blotting, and RT-qPCR. Nude mice are injected with H226 (1×106) and received an intraperitoneal injection of toosendanin every 2 days for 21 days. RNA sequence transcriptome analysis is performed on toosendanin-treated cells to identify target genes and signaling pathways. With increasing concentrations of toosendanin, the rate of cell proliferation decreases and apoptotic cells increases. The number of migrated cells significantly reduces and epithelial-mesenchymal transition is reversed. Injection of toosendanin in nude mice leads to a reduction in tumor volume, weight, and the number of metastatic tumors. Furthermore, KEGG shows that genes related to the AMPK pathway are highly enriched. BNIP3 is the most differentially expressed gene, and its expression along with phosphorylated-AMPK significantly increases in toosendanin-treated cells. Toosendanin exerts anticancer effects, induces apoptosis in LUSC cells, and inhibits tumor progression via the BNIP3/AMPK signaling pathway.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- AMP-Activated Protein Kinases/metabolism
- Apoptosis/drug effects
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/genetics
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Disease Progression
- Drugs, Chinese Herbal/pharmacology
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Mice, Inbred BALB C
- Mice, Nude
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/genetics
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Fabing Liu
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Cardiothoracic Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, China
| | - Guangxue Wang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Liming Zhao
- Department of Emergency, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai, 200123, China
| | - Guohan Chen
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Lin Dong
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qinchuan Li
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Dongyi Zhu
- Department of Respiratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai, 200123, China
| |
Collapse
|
48
|
Cui Y, Wang Q, Shi Y, Dai Y, Liu Y. mtROS-mediated mitophagy is involved in aflatoxin-B 1 induced liver injury in ducks. Comp Biochem Physiol C Toxicol Pharmacol 2024; 283:109942. [PMID: 38810896 DOI: 10.1016/j.cbpc.2024.109942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Aflatoxin B1 (AFB1) is highly toxic to the liver and can cause excessive production of mitochondrial reactive oxygen species (mtROS) in hepatocytes, leading to oxidative stress, inflammation, fibrosis, cirrhosis, and even liver cancer. The overproduction of mtROS can induce mitophagy, but whether mtROS and mitophagy are involved in the liver injury induced by AFB1 in ducks remains unclear. In this study, we first demonstrated that overproduction of mtROS and mitophagy occurred during liver injury induced by AFB1 exposure in ducks. Then, by inhibiting mtROS and mitophagy, we found that the damage caused by AFB1 in ducks was significantly alleviated, and the overproduction of mtROS induced by AFB1 exposure could mediate the occurrence of mitophagy. These results suggested that mtROS-mediated mitophagy is involved in AFB1-induced duck liver injury, and they may be the prevention and treatment targets of AFB1 hepatotoxicity.
Collapse
Affiliation(s)
- Yilong Cui
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Qi Wang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yun Shi
- Tongliao City Animal Quarantine Technical Service Center, Tongliao 028000, China
| | - Yang Dai
- Tongliao Agriculture and Animal Husbandry Bureau, Tongliao 028000, China
| | - Yanfen Liu
- Institute of Animal Husbandry and Veterinary Medicine, Liaoning Agricultural Vocational and Technical College, Yingkou 115009, China.
| |
Collapse
|
49
|
Kovale L, Singh MK, Kim J, Ha J. Role of Autophagy and AMPK in Cancer Stem Cells: Therapeutic Opportunities and Obstacles in Cancer. Int J Mol Sci 2024; 25:8647. [PMID: 39201332 PMCID: PMC11354724 DOI: 10.3390/ijms25168647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Cancer stem cells represent a resilient subset within the tumor microenvironment capable of differentiation, regeneration, and resistance to chemotherapeutic agents, often using dormancy as a shield. Their unique properties, including drug resistance and metastatic potential, pose challenges for effective targeting. These cells exploit certain metabolic processes for their maintenance and survival. One of these processes is autophagy, which generally helps in energy homeostasis but when hijacked by CSCs can help maintain their stemness. Thus, it is often referred as an Achilles heel in CSCs, as certain cancers tend to depend on autophagy for survival. Autophagy, while crucial for maintaining stemness in cancer stem cells (CSCs), can also serve as a vulnerability in certain contexts, making it a complex target for therapy. Regulators of autophagy like AMPK (5' adenosine monophosphate-activated protein kinase) also play a crucial role in maintaining CSCs stemness by helping CSCs in metabolic reprogramming in harsh environments. The purpose of this review is to elucidate the interplay between autophagy and AMPK in CSCs, highlighting the challenges in targeting autophagy and discussing therapeutic strategies to overcome these limitations. This review focuses on previous research on autophagy and its regulators in cancer biology, particularly in CSCs, addresses the remaining unanswered questions, and potential targets for therapy are also brought to attention.
Collapse
Affiliation(s)
- Lochana Kovale
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Joungmok Kim
- Department of Oral Biochemistry and Molecular Biology, College of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| |
Collapse
|
50
|
Deepak K, Roy PK, Das CK, Mukherjee B, Mandal M. Mitophagy at the crossroads of cancer development: Exploring the role of mitophagy in tumor progression and therapy resistance. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119752. [PMID: 38776987 DOI: 10.1016/j.bbamcr.2024.119752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/27/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Preserving a functional mitochondrial network is crucial for cellular well-being, considering the pivotal role of mitochondria in ensuring cellular survival, especially under stressful conditions. Mitophagy, the selective removal of damaged mitochondria through autophagy, plays a pivotal role in preserving cellular homeostasis by preventing the production of harmful reactive oxygen species from dysfunctional mitochondria. While the involvement of mitophagy in neurodegenerative diseases has been thoroughly investigated, it is becoming increasingly evident that mitophagy plays a significant role in cancer biology. Perturbations in mitophagy pathways lead to suboptimal mitochondrial quality control, catalyzing various aspects of carcinogenesis, including establishing metabolic plasticity, stemness, metabolic reconfiguration of cancer-associated fibroblasts, and immunomodulation. While mitophagy performs a delicate balancing act at the intersection of cell survival and cell death, mounting evidence indicates that, particularly in the context of stress responses induced by cancer therapy, it predominantly promotes cell survival. Here, we showcase an overview of the current understanding of the role of mitophagy in cancer biology and its potential as a target for cancer therapy. Gaining a more comprehensive insight into the interaction between cancer therapy and mitophagy has the potential to reveal novel targets and pathways, paving the way for enhanced treatment strategies for therapy-resistant tumors in the near future.
Collapse
Affiliation(s)
- K Deepak
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India.
| | - Pritam Kumar Roy
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India.
| | - Chandan Kanta Das
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, BRBII/III, Philadelphia, PA, 19104, USA
| | - Budhaditya Mukherjee
- Infectious Disease and Immunology Lab, School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India.
| | - Mahitosh Mandal
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India.
| |
Collapse
|