1
|
Ye R, Mao YM, Fei YR, Shang Y, Zhang T, Zhang ZZ, Liu YL, Li JY, Chen SL, He YB. Targeting ferroptosis for the treatment of female reproductive system disorders. J Mol Med (Berl) 2025; 103:381-402. [PMID: 40100417 DOI: 10.1007/s00109-025-02528-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/28/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
Ferroptosis, a regulated form of cell death driven by iron-dependent lipid peroxidation, has emerged as a critical factor in female reproductive health and has been implicated in disorders such as polycystic ovary syndrome, premature ovarian insufficiency, endometriosis, and ovarian cancer. This review explores the intricate molecular mechanisms underlying ferroptosis, emphasizing its reliance on iron metabolism and oxidative stress, which disrupt key processes in reproductive tissues, including granulosa cell function, folliculogenesis, and embryo implantation. Increasing evidence linking ferroptosis to these conditions offers new therapeutic opportunities, with iron chelators, lipid peroxidation inhibitors, and antioxidants showing the potential to alleviate reproductive dysfunction by modulating ferroptotic pathways. In ovarian cancer, ferroptosis inducers combined with conventional cancer therapies, such as chemotherapy, provide promising strategies to overcome drug resistance. This review synthesizes current knowledge on ferroptosis and highlights its importance as a therapeutic target in reproductive health, emphasizing the need for further research to refine and expand treatment options, evaluate their applicability in clinical settings, and explore their role in fertility preservation. By advancing our understanding of ferroptosis regulation, these therapeutic approaches could lead to novel treatments for reproductive disorders and cancers, offering new hope for improving outcomes in women's health and cancer therapy.
Collapse
Affiliation(s)
- Rui Ye
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi-Ming Mao
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Zhejiang Province, China
| | - Yi-Ran Fei
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Yue Shang
- Reproductive Center, Hainan Branch, Shanghai Children'S Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Ting Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhe-Zhong Zhang
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Yong-Lin Liu
- Reproductive Center, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jun-Yu Li
- Department of Pharmacy, Hainan Branch, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Shi-Liang Chen
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| | - Yi-Bo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| |
Collapse
|
2
|
Power K, Leandri R, Federico G, De Vico G, Leonardi L. Ferritinophagy: a possible new iron-related metabolic target in canine osteoblastic osteosarcoma. Front Vet Sci 2025; 12:1546872. [PMID: 40196812 PMCID: PMC11973301 DOI: 10.3389/fvets.2025.1546872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Canine osteosarcomas (COS) are the most common bone tumors in dogs, characterized by high metastatic rates, poor prognosis, and poor responsiveness to routine therapies, which highlights the need for new treatment targets. In this context, the metabolism of neoplastic cells represents an increasingly studied element, as cancer cells depend on particular metabolic pathways that are also elements of vulnerability. Among these, tumor cells (TCs) show higher iron requirements to sustain proliferation (so-called iron addiction), which are achieved by increasing iron uptake and/or by activating ferritinophagy, a process mediated by the Nuclear receptor Co-Activator 4 (NCOA4) leading to iron mobilization from ferritin (Ft) deposits. Previous studies have shown that COS cells overexpress Transferrin Receptor 1 (TfR1) to increase iron uptake. In this study we evaluated the immunohistochemical expression of ferritinophagy-related proteins, namely Ferritin Heavy chain (FTH1) and NCOA4, and proliferating cell nuclear antigen (PCNA) in canine normal bone and canine osteoblastic osteosarcoma (COOS) samples. Normal samples revealed negative/weak immunoreactivity for FTH1, NCOA4 and PCNA in <10% of osteocytes. In COOS samples the majority of neoplastic cells showed immunoreactivity to FTH1, NCOA4 and PCNA. Our data suggest that the activation of ferritinophagy by COOS cells responds to the need for feed their "iron addiction." These data, though preliminary, further suggest that targeting iron metabolism represents a new potential strategy worthy of further study to be transferred into clinical practice.
Collapse
Affiliation(s)
- Karen Power
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Rebecca Leandri
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Giorgia Federico
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Gionata De Vico
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Leonardo Leonardi
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
3
|
Srivastava A, Beyer M, Hladun C, Tardif R, Arshad A, Darie CC, Zoo Y, Papaefthymiou GC, Liu W, Viner R, Arosio P, Bou-Abdallah F. Characterization of the Iron-Sulfur Cluster in the NCOA4 Fragment (383-522) and Its Interaction with Ferritin. ACS Chem Biol 2025; 20:731-745. [PMID: 40014030 DOI: 10.1021/acschembio.4c00877] [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: 02/28/2025]
Abstract
Ferritin degradation pathways, particularly NCOA4-mediated ferritinophagy, are crucial for maintaining iron homeostasis. Here, we demonstrate the coexistence of two NCOA4 isoforms, one iron-sulfur cluster-free and one iron-sulfur cluster-bound, in oxygenated cell cultures. Using a combination of spectroscopic and analytical techniques, in vitro characterization of the NCOA4 fragment (383-522), denoted NCOA4-D, revealed a predominance of monomeric species with a relatively stable [2Fe-2S] cluster under normoxic conditions. The results demonstrate distinct interactions between NCOA4-D isoforms and ferritin, underscoring the influence of cellular oxygen and iron concentrations on NCOA4's regulatory functions, pathways, and ferritin's fate. Our findings suggest that different NCOA4-initiated degradation pathways may concurrently occur in cells and highlight the necessity of further exploring the role of the Fe-S cluster in NCOA4 as an iron-sensing mechanism for maintaining cellular iron homeostasis.
Collapse
Affiliation(s)
- Ayush Srivastava
- Department of Chemistry, State University of New York, Potsdam, New York 13676, United States
| | - Maximilian Beyer
- Department of Chemistry, State University of New York, Potsdam, New York 13676, United States
| | - Colby Hladun
- Department of Chemistry, State University of New York, Potsdam, New York 13676, United States
| | - Rebekah Tardif
- Department of Chemistry, State University of New York, Potsdam, New York 13676, United States
| | - Aneeta Arshad
- Department of Chemistry & Biochemistry, Biochemistry & Proteomics Laboratories, Clarkson University, Potsdam, New York 13699, United States
| | - Costel C Darie
- Department of Chemistry & Biochemistry, Biochemistry & Proteomics Laboratories, Clarkson University, Potsdam, New York 13699, United States
| | - Yeonni Zoo
- Department of Physics, Villanova University, Villanova, Pennsylvania 19085, United States
| | | | - Weijing Liu
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134, United States
| | - Rosa Viner
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134, United States
| | - Paolo Arosio
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Fadi Bou-Abdallah
- Department of Chemistry, State University of New York, Potsdam, New York 13676, United States
| |
Collapse
|
4
|
Meng C, Li S, Ma Y, Yu H, Song J, Zhi J, Zhu B, Shao L, Liu X, Yang L, Zhang M, Zhang Y, Li G. Assembling Ruthenium Complexes to Form Ruthenosome Unleashing Ferritinophagy-Mediated Tumor Suppression. ACS NANO 2025; 19:10207-10219. [PMID: 40040589 PMCID: PMC11925053 DOI: 10.1021/acsnano.4c17344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
We introduce ruthenosomes, a fusion of liposomal and reactive oxygen species (ROS)-generating properties meticulously engineered as potent ferroptosis inducers (FINs), marking a significant advancement in metallodrug design for cancer therapy. Formed through the self-assembly of oleate-conjugated ruthenium complexes, these ruthenosomes exhibit exceptional cellular uptake, selectively accumulating in mitochondria and causing substantial disruption. This targeted mitochondrial damage significantly elevates ROS levels, triggering autophagy and selectively activating ferritinophagy. Together, these processes sensitize cancer cells to ferroptosis. In vivo, ruthenosomes effectively suppress colorectal tumor growth, underscoring their therapeutic potential. Our study pioneers a design strategy that transforms ruthenium complexes into liposome-like structures capable of inducing ferroptosis independent of light activation. By leveraging ruthenosomes as multifunctional nanocarriers, this research offers a versatile and powerful platform for ROS-mediated, ferroptosis-driven cancer cell eradication.
Collapse
Affiliation(s)
- Caiting Meng
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Shuaijun Li
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- The
Second
Affiliated Hospital, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Yana Ma
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department
of Interventional Radiology, The Affiliated
Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Hongwen Yu
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jiaqi Song
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Junchao Zhi
- Active
Soft
Matter Group, Songshan Lake Materials Laboratory, Dongguan, Guangdong 523008, China
| | - Bin Zhu
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Liang Shao
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Xinling Liu
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Lulu Yang
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Mingzhen Zhang
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Ye Zhang
- Active
Soft
Matter Group, Songshan Lake Materials Laboratory, Dongguan, Guangdong 523008, China
| | - Guanying Li
- Department
of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- The
Second
Affiliated Hospital, Health Science Centre, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| |
Collapse
|
5
|
Li X, Li W, Xie X, Fang T, Yang J, Shen Y, Wang Y, Wang H, Tao L, Zhang H. ROS Regulate Rotenone-induced SH-SY5Y Dopamine Neuron Death Through Ferroptosis-mediated Autophagy and Apoptosis. Mol Neurobiol 2025:10.1007/s12035-025-04824-6. [PMID: 40097764 DOI: 10.1007/s12035-025-04824-6] [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: 09/04/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Rotenone, a plant-derived natural insecticide, is widely used to induce Parkinson's disease (PD) models. However, the mechanisms of rotenone-induced cell death remain unclear. Here, we found that rotenone (0.01, 0.1, or 1 μmol/L) suppressed SH-SY5Y dopamine neuron viability and led to PD-like pathological changes, such as reduced tyrosine hydroxylase (TH) but increased α-synuclein. Rotenone increased the levels of intracellular reactive oxygen species (ROS) and mitochondrial ROS, as well as the levels of the antioxidants nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), ultimately resulting in oxidative stress. Moreover, rotenone significantly downregulated the expression of GPX4 and xCT but upregulated the expression of COX2 and NCOA4, which are markers of ferroptosis. Furthermore, rotenone decreased phosphorylated mTOR level but increased Beclin-1, ATG5, LC3 and p62 expression, suggesting that rotenone enhances autophagy and reduces autophagy flux. Additionally, rotenone reduced Bcl-2 levels and the mitochondrial membrane potential (MMP) while promoting BAX and Caspase-3 expression, thus initiating cell apoptosis. N-acetylcysteine (NAC), a ROS scavenger, and ferrostatin-1 (Fer-1) and deferoxamine (DFO), two ferroptosis inhibitors, significantly eliminated rotenone-induced autophagy and apoptosis. Moreover, ML385, a specific inhibitor of Nrf2, suppressed rotenone-induced ferroptosis. Our results demonstrated that ROS might mediate rotenone-induced PD-like pathological changes by regulating iron death, autophagy, and apoptosis. Inhibiting ferroptosis blocked the rotenone-induced increase in autophagy and apoptosis. Thus, the ability of ROS to regulate rotenone-induced death through autophagy and apoptosis is dependent on ferroptosis. The findings require validation in multiple neuronal cell lines and in vivo.
Collapse
Affiliation(s)
- Xinying Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Weiran Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Clinical Medicine, School of Medicine, Qinghai University, Xining, China
| | - Xinying Xie
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ting Fang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Jingwen Yang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yue Shen
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yicheng Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hongyan Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Liqing Tao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
6
|
Hao Y, Wang X, Ni Z, Ma Y, Wang J, Su W. Analysis of ferritinophagy-related genes associated with the prognosis and regulatory mechanisms in non-small cell lung cancer. Front Med (Lausanne) 2025; 12:1480169. [PMID: 40124684 PMCID: PMC11925780 DOI: 10.3389/fmed.2025.1480169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Lung cancer remains a major global health issue, with non-small cell lung cancer (NSCLC) constituting approximately 85% of cases. Ferritinophagy, a pivotal autophagic process in ferroptosis, plays an essential role in tumor initiation and progression. However, the specific contributions of ferritinophagy-related genes (FRGs) to NSCLC pathogenesis remain incompletely understood. In this study, weighted gene co-expression network analysis (WGCNA) was employed to identify key modular genes associated with FRG scores. Genes overlapping between these modules and differentially expressed genes (DEGs) were selected for further investigation. Prognostic genes were identified through univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analysis, with subsequent validation using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) on both clinical samples and the TCGA-NSCLC dataset. A nomogram incorporating clinicopathological features and risk scores was developed to predict patient outcomes. Further analyses focused on functional enrichment, drug sensitivity, and the immune microenvironment. Cross-referencing 2,142 key modular genes with 2,764 DEGs revealed 600 candidate genes. Univariate Cox regression and LASSO analysis of these candidates identified eight prognostic genes: KLK8, MFI2, B3GNT3, MYRF, CREG2, GLB1L3, AHNAK2, and NLRP10. Two distinct risk groups exhibited significant survival differences. Both the risk score and pathological N stage were found to be independent prognostic factors, forming the basis for the nomogram. Notable correlations were observed between certain immune cells, prognostic genes, and immune responses, affecting the efficacy of immunotherapy and drug sensitivity. qRT-PCR confirmed that, except for NLRP10, all prognostic genes exhibited expression patterns consistent with TCGA-NSCLC data. This study highlights the significant role of FRGs in NSCLC prognosis and regulation, offering novel insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Yuan Hao
- Clinical Trials Center, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Xin Wang
- Clinical Trials Center, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Zerong Ni
- Clinical Trials Center, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Yuhui Ma
- Department of Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Tongji Medical College Huazhong University Science of and Technology, Taiyuan, China
| | - Jing Wang
- Department of Pathology, Shanxi Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Wen Su
- Department of Immunology, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| |
Collapse
|
7
|
Zhang Y, Du W, Kong T, Hua T, Ma H, Hu Y, Pan S, Ling B, Yang M, Cheng C. Targeted temperature management alleviates post-resuscitation myocardial dysfunction by inhibiting ferroptosis. Cell Death Discov 2025; 11:71. [PMID: 39984437 PMCID: PMC11845627 DOI: 10.1038/s41420-025-02356-5] [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: 08/15/2024] [Revised: 12/19/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
Targeted temperature management (TTM) is a vital intervention for cardiac arrest survivors to mitigate post-resuscitation myocardial dysfunction (PRMD). However, the optimal temperature for TTM remains a topic of debate. This study investigates the effects of TTM at different temperatures and explores the underlying mechanisms using in vivo and in vitro models of myocardial ischemia/reperfusion (I/R) injury following cardiac arrest (CA) and cardiopulmonary resuscitation (CPR). We found that TTM at 33 °C significantly improved post-resuscitation hemodynamics and myocardial function, reducing both myocardial and mitochondrial damage in the rat model of CA/CPR. Additionally, Deferoxamin (DFO), as an iron chelating agent, also demonstrated protective effects against PRMD. Both in vitro and in vivo experiments confirmed that hypothermia at 33 °C and DFO mitigated mitochondrial damage, oxidative stress, lipid peroxidation, and iron overload, while suppressing ferritinophagy and ferroptosis. Furthermore, TTM at 33 °C and DFO facilitated the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), with Nrf2 activation leading to inhibited ferritinophagy and enhanced iron export. Our findings indicate that TTM at 33° C, as opposed to 36° C, significantly alleviates PRMD and reduced myocardial damage by inhibiting ferroptosis. Theses protective effects are associated with Nrf2 activation and modulation of iron homeostasis. Moreover, DFO not only suppressed ferroptosis through its iron chelation properties but also by activating the Nrf2 axis.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Weiwei Du
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Ting Kong
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- Department of General Practice Department, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Tianfeng Hua
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- The Second Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Honghao Ma
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Yan Hu
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- The Second Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Sinong Pan
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- The Second Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Bingrui Ling
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- The Second Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Min Yang
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
- The Second Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| | - Cheng Cheng
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
- The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
- Department of Cardiology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230088, Anhui, China.
| |
Collapse
|
8
|
Lai L, Tan M, Hu M, Yue X, Tao L, Zhai Y, Li Y. Important molecular mechanisms in ferroptosis. Mol Cell Biochem 2025; 480:639-658. [PMID: 38668809 DOI: 10.1007/s11010-024-05009-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/07/2024] [Indexed: 02/19/2025]
Abstract
Ferroptosis is a type of cell death that is caused by the oxidation of lipids and is dependent on the presence of iron. It was first characterized by Brent R. Stockwell in 2012, and since then, research in the field of ferroptosis has rapidly expanded. The process of ferroptosis-induced cell death is genetically, biochemically, and morphologically distinct from other forms of cellular death, such as apoptosis, necroptosis, and non-programmed cell death. Extensive research has been devoted to comprehending the intricate process of ferroptosis and the various factors that contribute to it. While the majority of these studies have focused on examining the effects of lipid metabolism and mitochondria on ferroptosis, recent findings have highlighted the significant involvement of signaling pathways and associated proteins, including Nrf2, P53, and YAP/TAZ, in this process. This review provides a concise summary of the crucial signaling pathways associated with ferroptosis based on relevant studies. It also elaborates on the drugs that have been employed in recent years to treat ferroptosis-related diseases by targeting the relevant signaling pathways. The established and potential therapeutic targets for ferroptosis-related diseases, such as cancer and ischemic heart disease, are systematically addressed.
Collapse
Affiliation(s)
- Lunmeng Lai
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Menglei Tan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Mingming Hu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Xiyue Yue
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Lulu Tao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Yanru Zhai
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Yunsen Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China.
| |
Collapse
|
9
|
Lin H, Liu H, Xi H, Li D, Jiang P, Wang Y, Cheng S, Jiang H, Deng H, Zhou X, Yu L. Oxygen-Independent Photodynamic Therapy-Mediated Selective Consumption of M1 Macrophage Against Ventricular Arrhythmias via Sympathetic Neuromodulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409244. [PMID: 39711260 DOI: 10.1002/smll.202409244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/12/2024] [Indexed: 12/24/2024]
Abstract
The occurrence of myocardial infarction (MI)-induced malignant ventricular arrhythmias (VAs) is closely associated with the hyperactivation of left stellate ganglion (LSG). Proinflammatory M1 macrophage is reported to aggravate sympathetic overactivation and cause VAs. Therefore, the depletion of M1 macrophage is anticipated to inhibit LSG overactivation and alleviate MI-induced VAs. Herein, oxygen-independent photodynamic therapy (Oi-PDT) combined with M1 macrophage targeting is applied to selectively deplete M1 macrophage in LSG and further treat MI-induced VAs. Oi-PDT, which overcomes the limitation of extremely dependence on oxygen content in traditional PDT, is constructed through the generation of oxidizing photogenerated holes (h+) under the irradiation of near-infrared (NIR) light on the prepared Oi-PDT agent (PPSCD). Meanwhile, PPSCD targets M1 macrophage through conjunction with SR-A receptor. The selective consumption of M1 macrophage is attributed to both apoptosis and ferroptosis induced by h+, 1O2, and O2 •- generated in Oi-PDT. In vivo tests indicated neural activity experienced a notable reduction from 104.5 ± 2.9 to 51.5 ± 6.7 after MI with Oi-PDT treatment, thereby significantly inhibited VAs. The implementation of this study provides a promising strategy for selective consumption of M1 macrophages and treatment of VAs induced by MI.
Collapse
Affiliation(s)
- Heng Lin
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Autonomic Nervous System Modulation, Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, P. R. China
| | - Hengyang Liu
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Autonomic Nervous System Modulation, Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, P. R. China
| | - Haosong Xi
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Autonomic Nervous System Modulation, Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, P. R. China
| | - Dangwei Li
- Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Engineering Center of Natural Polymers-based Medical Materials, School of Resource and Environmental Science, Wuhan University, Wuhan, 430079, P. R. China
| | - Pengcheng Jiang
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Autonomic Nervous System Modulation, Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, P. R. China
| | - Yijun Wang
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Autonomic Nervous System Modulation, Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, P. R. China
| | - Siyi Cheng
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Autonomic Nervous System Modulation, Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, P. R. China
| | - Hong Jiang
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Autonomic Nervous System Modulation, Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, P. R. China
| | - Hongbing Deng
- Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Engineering Center of Natural Polymers-based Medical Materials, School of Resource and Environmental Science, Wuhan University, Wuhan, 430079, P. R. China
| | - Xue Zhou
- Key Laboratory of Environment and Health, Ministry of Education, Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Lilei Yu
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Autonomic Nervous System Modulation, Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, P. R. China
| |
Collapse
|
10
|
Zhao P, Yin S, Qiu Y, Sun C, Yu H. Ferroptosis and pyroptosis are connected through autophagy: a new perspective of overcoming drug resistance. Mol Cancer 2025; 24:23. [PMID: 39825385 PMCID: PMC11740669 DOI: 10.1186/s12943-024-02217-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 12/25/2024] [Indexed: 01/20/2025] Open
Abstract
Drug resistance is a common challenge in clinical tumor treatment. A reduction in drug sensitivity of tumor cells is often accompanied by an increase in autophagy levels, leading to autophagy-related resistance. The effectiveness of combining chemotherapy drugs with autophagy inducers/inhibitors has been widely confirmed, but the mechanisms are still unclear. Ferroptosis and pyroptosis can be affected by various types of autophagy. Therefore, ferroptosis and pyroptosis have crosstalk via autophagy, potentially leading to a switch in cell death types under certain conditions. As two forms of inflammatory programmed cell death, ferroptosis and pyroptosis have different effects on inflammation, and the cGAS-STING signaling pathway is also involved. Therefore, it also plays an important role in the progression of some chronic inflammatory diseases. This review discusses the relationship between autophagy, ferroptosis and pyroptosis, and attempts to uncover the reasons behind the evasion of tumor cell death and the nature of drug resistance.
Collapse
Affiliation(s)
- Peng Zhao
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shuangshuang Yin
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuling Qiu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261053, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, China.
| | - Haiyang Yu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
11
|
Ying H, Kimmelman AC, Bardeesy N, Kalluri R, Maitra A, DePinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 2025; 39:36-63. [PMID: 39510840 PMCID: PMC11789498 DOI: 10.1101/gad.351863.124] [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] [Indexed: 11/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a grim prognosis for patients. Recent multidisciplinary research efforts have provided critical insights into its genetics and tumor biology, creating the foundation for rational development of targeted and immune therapies. Here, we review the PDAC genomic landscape and the role of specific oncogenic events in tumor initiation and progression, as well as their contributions to shaping its tumor biology. We further summarize and synthesize breakthroughs in single-cell and metabolic profiling technologies that have illuminated the complex cellular composition and heterotypic interactions of the PDAC tumor microenvironment, with an emphasis on metabolic cross-talk across cancer and stromal cells that sustains anabolic growth and suppresses tumor immunity. These conceptual advances have generated novel immunotherapy regimens, particularly cancer vaccines, which are now in clinical testing. We also highlight the advent of KRAS targeted therapy, a milestone advance that has transformed treatment paradigms and offers a platform for combined immunotherapy and targeted strategies. This review provides a perspective summarizing current scientific and therapeutic challenges as well as practice-changing opportunities for the PDAC field at this major inflection point.
Collapse
Affiliation(s)
- Haoqiang Ying
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Alec C Kimmelman
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, New York 10016, USA
- Department of Radiation Oncology, New York University Grossman School of Medicine, New York, New York 10016, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts 02114, USA
- The Cancer Program, Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Raghu Kalluri
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Anirban Maitra
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Sheikh Ahmed Pancreatic Cancer Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA;
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
12
|
Yang L, Zhang M, Liu M, Yu Y, Zhang Y, Yang J, Xing L, Shao Z, Wang H. Loss of FTH1 Induces Ferritinophagy-Mediated Ferroptosis in Anaemia of Myelodysplastic Syndromes. J Cell Mol Med 2025; 29:e70350. [PMID: 39804099 PMCID: PMC11726652 DOI: 10.1111/jcmm.70350] [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: 12/24/2023] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Single-cell sequencing of lineage negative (Lin-) cells from patients with myelodysplastic syndromes (MDS) revealed a reduction in ferritin heavy chain 1 (FTH1) levels, yet the significance of this decrease in FTH1 in the pathophysiology of MDS remains unclear. In this study, we evaluated the role of FTH1 in patients with MDS. The mRNA expression of FTH1 in GlycoA+ nucleated erythrocytes from MDS patients was significantly lower than that in control group. FTH1 was implicated in both ferritinophagy and ferroptosis in MDS patients, processes that are linked to the development of anaemia. To further validate our observations, we employed shRNA to knock down the FTH1 gene in K562 and SKM1 cells. This knockdown confirmed that the elevated ferroptosis levels observed after FTH1 depletion were indeed due to the induction of ferritinophagy. Hemin stimulation promoted the differentiation of K562 cells, while downregulation of FTH1 gene expression had an impact on erythroid differentiation and haemoglobin synthesis. Taken together, our results suggest that FTH1-mediated ferritinophagy may represent a novel therapeutic target for MDS.
Collapse
Affiliation(s)
- Liyan Yang
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| | - Mengying Zhang
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| | - Mengyuan Liu
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| | - Yating Yu
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| | - Yue Zhang
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| | - Jinyue Yang
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| | - Limin Xing
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| | - Zonghong Shao
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| | - Huaquan Wang
- Department of Hematology, General HospitalTianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjinChina
| |
Collapse
|
13
|
Liu Y, Carbonetto P, Willwerscheid J, Oakes SA, Macleod KF, Stephens M. Dissecting tumor transcriptional heterogeneity from single-cell RNA-seq data by generalized binary covariance decomposition. Nat Genet 2025; 57:263-273. [PMID: 39747597 DOI: 10.1038/s41588-024-01997-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/18/2024] [Indexed: 01/04/2025]
Abstract
Profiling tumors with single-cell RNA sequencing has the potential to identify recurrent patterns of transcription variation related to cancer progression, and to produce therapeutically relevant insights. However, strong intertumor heterogeneity can obscure more subtle patterns that are shared across tumors. Here we introduce a statistical method, generalized binary covariance decomposition (GBCD), to address this problem. We show that GBCD can decompose transcriptional heterogeneity into interpretable components-including patient-specific, dataset-specific and shared components relevant to disease subtypes-and that, in the presence of strong intertumor heterogeneity, it can produce more interpretable results than existing methods. Applied to data on pancreatic ductal adenocarcinoma, GBCD produced a refined characterization of existing tumor subtypes, and identified a gene expression program prognostic of poor survival independent of tumor stage and subtype. The gene expression program is enriched for genes involved in stress responses, and suggests a role for the integrated stress response in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Yusha Liu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Peter Carbonetto
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Jason Willwerscheid
- Department of Mathematics and Computer Science, Providence College, Providence, RI, USA
| | - Scott A Oakes
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Kay F Macleod
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Matthew Stephens
- Departments of Statistics and Human Genetics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
14
|
Yan A, Li Z, Gao Y, Hu F, Han S, Liu F, Liu Z, Chen J, Yuan C, Zhou C. Isobicyclogermacrenal ameliorates hippocampal ferroptosis involvement in neurochemical disruptions and neuroinflammation induced by sleep deprivation in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156306. [PMID: 39647468 DOI: 10.1016/j.phymed.2024.156306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Sleep deprivation (SLD) is a widespread condition that disrupts physiological functions and may increase mortality risk. Valeriana officinalis, a traditional medicinal herb known for its sedative and hypnotic properties, contains isobicyclogermacrenal (IG), a newly isolated active compound. However, research on the therapeutic potential of IG for treating SLD remains limited. METHODS In this study, IG was extracted and characterized from Valeriana officinalis, and an SLD model was established in rats using p-chlorophenylalanine (PCPA). Behavioral tests and pathological studies were conducted to assess the effects of IG on SLD, and transcriptomic and metabolomic analyses were utilized to investigate its underlying mechanisms. RESULTS IG administration significantly improved the cognitive performance of SLD rats in behavioral tests and ameliorated histological injuries in the hippocampus and cerebral cortex. IG treatment increased the levels of brain-derived neurotrophic factor (BDNF) and neurotransmitters such as serotonin (5-HT) in SLD rats. Additionally, IG directly targets TFRC, thereby improving iron metabolism in the hippocampus. Comprehensive transcriptomic and metabolomic analyses revealed that the improvements from IG stemmed from the mitigation of abnormalities in iron metabolism, cholesterol metabolism, and glutathione metabolism, leading to reduced oxidative stress, ferroptosis, and neuroinflammation in the hippocampus caused by SLD. CONCLUSIONS Collectively, these findings suggest that IG has the potential to ameliorate neurological damage and cognitive impairment caused by SLD, offering a novel strategy for protection against the adverse effects of SLD.
Collapse
Affiliation(s)
- Ao Yan
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Zhejin Li
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Yuanwei Gao
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Fanglong Hu
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Shuo Han
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Fengjie Liu
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Zhongcheng Liu
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Jinting Chen
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang 050017, China
| | - Chunmao Yuan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China.
| | - Chengyan Zhou
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China.
| |
Collapse
|
15
|
Sui X, Gao B, Zhang L, Wang Y, Ma J, Wu X, Zhou C, Liu M, Zhang L. Scutellaria barbata D.Don and Hedyotis diffusa Willd herb pair combined with cisplatin synergistically inhibits ovarian cancer progression through modulating oxidative stress via NRF2-FTH1 autophagic degradation pathway. J Ovarian Res 2024; 17:246. [PMID: 39702302 DOI: 10.1186/s13048-024-01570-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Cisplatin (DDP) is one of the most effective anticancer drugs, commonly used to treat advanced ovarian cancer (OC). However, DDP has significant limitations of platinum-based drugs, including chemical resistance and high-dose toxic side effects. Traditional Chinese medicines (TCMs) often presented in the form of formula, in which the herb pair was the basic unit. Scutellaria barbata D.Don and Hedyotis diffusa Willd (SB-HD) are famous TCMs herb pair that have been shown to help treat multiple types of cancers. However, the synergistic effects and mechanism of combination of SB-HD and DDP to enhance DDP chemosensitivity in OC are still unknown. RESULTS In vitro, we found that the optimal proportion of SB-HD to inhibit the proliferation of OC cells was 2:1, SB-HD and DDP were shown to synergistically reduce the viability of OC cells, inhibit the colony formation, promote cell cycle arrest and apoptosis, as well as inhibit cell migration and invasion. In vivo, combination treatment significantly inhibited the growth of subcutaneous tumors in BALB/c nude mice and reduced the toxic side effects of DDP. Mechanistically, SB-HD and DDP combination treatment significantly promoted oxidative stress response, decreased MMP, inhibited ATP production, decreased ROS levels and increased SOD activity, increased the expression of NRF2, HO-1, ATG5 and LC3, decreased the expression of p62 and FTH1 both in OC cells and tumor tissue of mice. Inhibitor 3-MA (Methyladenine, autophagy inhibitor) and Fer-1 (Ferrostatin-1, iron ion inhibitor) can effectively reverse the expression changes of the key target proteins, but not ZnPP (Zinc protoporphyrin, HO-1 inhibitor). Through bioinformatics analysis, it was found that the abnormal expression level of NRF2 and FTH1 mRNA has a high prognostic value, at the same time, the other four key proteins respectively or interacting with NRF2 and FTH1, also play important roles in the occurrence and development of OC. CONCLUSION Our findings uncover a synergistic effect of SB-HD and DDP against OC through modulating oxidative stress via NRF2-FTH1 autophagic degradation pathway, which may provide an important theoretical foundation for the use of SB-HD and a new strategy for enhancing DDP chemosensitivity as well as reducing toxic side effects.
Collapse
Affiliation(s)
- Xue Sui
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Bingqing Gao
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- School of Pharmacy, Anhui Xinhua University, Hefei, 230088, China
| | - Liu Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- Department of Dermatology, Dalian Lvshunkou District Hospital of Traditional Chinese Medicine, Dalian, 116041, China
| | - Yanmin Wang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Junnan Ma
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xingchen Wu
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Chenyu Zhou
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Min Liu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| | - Lin Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
16
|
Haines M, Thorup JR, Gohsman S, Ctortecka C, Newton C, Rohrer DC, Hostetter G, Mani DR, Gillette MA, Satpathy S, Carr SA. High-throughput proteomic and phosphoproteomic analysis of formalin-fixed paraffin-embedded tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.17.624038. [PMID: 39605438 PMCID: PMC11601474 DOI: 10.1101/2024.11.17.624038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Formalin-fixed, paraffin-embedded (FFPE) patient tissues are a valuable resource for proteomic studies with the potential to associate the derived molecular insights with clinical annotations and outcomes. Here we present an optimized, partially automated workflow for FFPE proteomics combining pathology-guided macro-dissection, Adaptive Focused Acoustics (AFA) sonication for lysis and decrosslinking, S-Trap digestion to peptides, and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis using Orbitrap, Astral or timsTOF HT instrumentation. The workflow enables analysis of up to 96 dissected FFPE tissue samples or 10 μm scrolls, identifying 8,000-10,000 unique proteins per sample with median CVs <20%. Key optimizations include improved tissue lysis strategies, protein quantification for normalization, and peptide cleanup prior to LC-MS/MS analysis. Application to lung adenocarcinoma (LUAD) FFPE blocks confirms the platform's effectiveness in processing complex, clinically relevant samples, achieving deep proteome coverage and quantitative robustness comparable to TMT-based methods. Using the newly released Orbitrap Astral with short, 24-minute gradients, the workflow identifies up to ~10,000 unique proteins and ~11,000 localized phosphosites in LUAD FFPE tissue. This high-throughput, scalable workflow advances biomarker discovery and proteomic research in archival tissue samples.
Collapse
Affiliation(s)
- Moe Haines
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | | | | | - D. R. Mani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Shankha Satpathy
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Current address: AstraZeneca, Waltham, MA, USA
| | | |
Collapse
|
17
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
18
|
Zhu J, Yuan A, Le Y, Chen X, Guo J, Liu J, Chen H, Wang CY, Lu D, Lu K. Yi-Qi-Jian-Pi-Xiao-Yu formula inhibits cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated ferritinophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156189. [PMID: 39515100 DOI: 10.1016/j.phymed.2024.156189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/28/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The kidneys are the primary excretory organs for platinum drugs, making them susceptible to damage from these drugs. Cisplatin-induced acute kidney injury (CIAKI) is the most common side effect observed in patients undergoing clinical cisplatin treatment. A traditional Chinese medicinal preparation, the Yi-Qi-Jian-Pi-Xiao-Yu formula (YQJPXY), which is a modified formulation of the classical Chinese medicine formula Buyang Huanwu Decoction, has long been used in the treatment of clinical kidney diseases. It is expected to be used to ameliorate cisplatin-induced acute kidney injury. However, the mechanism of this YQJPXY for the treatment of cisplatin-induced acute kidney injury remains unclear. PURPOSE The objective of this study is to examine the impact of the YQJPXY on the inhibition of ferroptosis in cisplatin-induced acute kidney injury and to elucidate the underlying mechanisms. METHODS The active components of YQJPXY were analysed using UPLC-MS/MS. A comprehensive investigation was conducted to elucidate the effects and regulatory mechanisms of YQJPXY on CIAKI and ferroptosis in mice subjected to acute cisplatin treatment and in mice receiving cisplatin treatment after STING expression was inhibited using the STING inhibitor C176. The renoprotective effect of YQJPXY on cisplatin-treated mice was evaluated by measuring tissue damage, inflammation and pro-fibrosis. In addition, we employed network pharmacology and molecular docking methodologies to analyse the principal regulatory targets of YQJPXY. Furthermore, the expression of key proteins and markers of ferroptosis and iron metabolism, as well as the levels of key indicators related to STING-associated ferritinophagy, were examined by immunoblotting, immunohistochemistry, immunoprecipitation, quantitative real-time PCR (qPCR) and specific probes. RESULTS The results demonstrated that YQJPXY reduced the levels of indicators of injury, inflammation and pro-fibrosis in CIAKI mice, with renoprotective effects. Network pharmacological analyses revealed that ferroptosis might be the main biological process regulated by YQJPXY. Furthermore, molecular docking results indicated that STING might be a potential regulatory target of YQJPXY. Furthermore, YQJPXY treatment resulted in a significant reduction in MDA and 4-HNE levels, as well as the inhibition of ferroptosis and improvement in iron metabolic processes. Concomitantly, YQJPXY exhibited a robust protective effect on ferroptosis and iron metabolism homeostasis, as evidenced by its inhibitory action on ferritinophagy. Validation experiments utilising the cisplatin inhibitor C176 demonstrated that YQJPXY inhibits cisplatin-induced ferroptosis in kidney via STING-mediated ferritinophagy. CONCLUSION These suggest that YQJPXY alleviates cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated Ferritinophagy.
Collapse
Affiliation(s)
- Ji Zhu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 330061, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Aini Yuan
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yifei Le
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaohui Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Jianan Guo
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jing Liu
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hang Chen
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Cai-Yi Wang
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Dezhao Lu
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Keda Lu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 330061, China.
| |
Collapse
|
19
|
Liu J, Wang Y, Zeng L, Yu C, Kang R, Klionsky DJ, Jiang J, Tang D. Extracellular NCOA4 is a mediator of septic death by activating the AGER-NFKB pathway. Autophagy 2024; 20:2616-2631. [PMID: 38916095 PMCID: PMC11587848 DOI: 10.1080/15548627.2024.2372215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024] Open
Abstract
Sepsis, a life-threatening condition resulting from a dysregulated response to pathogen infection, poses a significant challenge in clinical management. Here, we report a novel role for the autophagy receptor NCOA4 in the pathogenesis of sepsis. Activated macrophages and monocytes secrete NCOA4, which acts as a mediator of septic death in mice. Mechanistically, lipopolysaccharide, a major component of the outer membrane of Gram-negative bacteria, induces NCOA4 secretion through autophagy-dependent lysosomal exocytosis mediated by ATG5 and MCOLN1. Moreover, bacterial infection with E. coli or S. enterica leads to passive release of NCOA4 during GSDMD-mediated pyroptosis. Upon release, extracellular NCOA4 triggers the activation of the proinflammatory transcription factor NFKB/NF-κB by promoting the degradation of NFKBIA/IκB molecules. This process is dependent on the pattern recognition receptor AGER, rather than TLR4. In vivo studies employing endotoxemia and polymicrobial sepsis mouse models reveal that a monoclonal neutralizing antibody targeting NCOA4 or AGER delays animal death, protects against organ damage, and attenuates systemic inflammation. Furthermore, elevated plasma NCOA4 levels in septic patients, particularly in non-survivors, correlate positively with the sequential organ failure assessment score and concentrations of lactate and proinflammatory mediators, such as TNF, IL1B, IL6, and HMGB1. These findings demonstrate a previously unrecognized role of extracellular NCOA4 in inflammation, suggesting it as a potential therapeutic target for severe infectious diseases. Abbreviation: BMDMs: bone marrow-derived macrophages; BUN: blood urea nitrogen; CLP: cecal ligation and puncture; ELISA: enzyme-linked immunosorbent assay; LPS: lipopolysaccharide; NO: nitric oxide; SOFA: sequential organ failure assessment.
Collapse
Affiliation(s)
- Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yichun Wang
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Critical Care Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Chongqing, China
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Chongqing, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
20
|
Zhang M, Cui Y, Jia R, Cai Z, Shen Y, Yu W, Pang Z, Ding S, Gao X, Huang Y, Song M, Lou Y, Ye F, Ao H, Zhu A. Hesperidin alleviated dendritic spines through inhibiting ferritinophagy via HERC2-NCOA4 ubiquitination in CUMS mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156132. [PMID: 39522250 DOI: 10.1016/j.phymed.2024.156132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 09/01/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Depression has been linked to ferritinophagy-induced synaptic damage, which affects the emotional circuitry and can ultimately lead to depressive symptoms. It has been suggested that Hesperidin might improve depression disorders. However, the relationship between the therapeutic effects of the sympathetic nervous system in alleviating depression-like behaviors and ferritinophagy is still unknown. PURPOSE The objective of this study is to investigate the possible impact of Hesperidin in alleviating dendritic spines through the inhibition of ferritinophagy via HERC2-NCOA4 ubiquitination in mice exposed to chronic unpredictable mild stress (CUMS). METHODS C57BL/6 and NCOA4+/+ mice were exposed to CUMS for 42 days. During the last 3 weeks of the CUMS procedure, the mice were administered Hesperidin (50, 100, 200 g/kg/d) or fluoxetine (10 mg/kg/d) once daily. Following the behavioral tests, Golgi staining, tissue iron concentration test, and perls staining were conducted to assess the therapeutic effect of Hesperidin. Additionally, ultrahigh-performance liquid chromatography-high-resolution tandem mass spectrometry (UPLC-Q-TOF/MS) and ultrahigh-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) was utilized to analyze the metabolic components of Hesperidin in both blood and brain tissue. To investigate mechanism of Hesperidin, the cells were subjected to different concentrations of Hesperidin (25, 50, 100 µM), its metabolites (Eriodictyol (10, 20, 50 µM), Homoeriodictyol (0.1, 0.5, 1 µM)) and si-HERC2. Furthermore, HERC2-NCOA4 ubiquitination, and ferritinophagy-related proteins was explored through techniques such as western blot, immunofluorescence, co-immunoprecipitation, and molecular docking. RESULTS Hesperidin has demonstrated the potential to alleviate symptoms of depression by regulating dendritic spines through the inhibition of NCOA4-ferritinophagy, while NCOA4 overexpression could reverse these results. Importantly, the content of Hesperidin metabolites (Homoeriodictyol and Eriodictyol) was relatively high in brain tissue. The Hesperidin and its metabolites, Eriodictyol and Homoeriodictyol, were able to regulate GluR2 and SYN protein expression. Additionally, they inhibited ferritinophagy involving NCOA4, P62, LC3, and FTH. but this phenomenon was reversed by si-HERC2 following Hesperidin and its metabolite administration. Furthermore, the binding of HERC2 and NCOA4 protein was found to be inhibited by Hesperidin and its metabolites. CONCLUSIONS Hesperidin alleviated dendritic spines through inhibiting ferritinophagy via HERC2-NCOA4 ubiquitination in CUMS mice.
Collapse
Affiliation(s)
- Mingjia Zhang
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; Southern Medicine University, Sch Tradit Chinese Med, Guangzhou, China
| | - Yixuan Cui
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, China
| | - Ruiting Jia
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ziling Cai
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yineng Shen
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wumin Yu
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zixin Pang
- Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuning Ding
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xing Gao
- Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yike Huang
- Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Maolin Song
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, China
| | - Yahui Lou
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fa Ye
- Sleep Medicine Center, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, China.
| | - Haiqing Ao
- School of Public Health and Management, Guangzhou University of Chinese Medicin, China.
| | - Aisong Zhu
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Engineering Research Center for "Preventive Treatment" Smart Health of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
21
|
Meng K, Song J, Qi F, Li J, Fang Z, Song L. MT1G promotes iron autophagy and inhibits the function of gastric cancer cell lines by intervening in GPX4/SQSTM1. Sci Rep 2024; 14:28539. [PMID: 39558129 PMCID: PMC11574125 DOI: 10.1038/s41598-024-80160-4] [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: 08/15/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024] Open
Abstract
Gastric cancer (GC) is the fifth most common cancer and the third most common cause of cancer death globally, with high invasiveness, high recurrence rate, and poor prognosis. Multiple studies have shown that Metallothionein-1G (MT1G) is closely associated with oxidative stress, ferroptosis, and autophagy. However, the role and potential mechanisms of MT1G in GC have not been fully elucidated. This study aims to explore the biological functions and regulatory mechanisms of MT1G in GC. Perform bioinformatics analysis using the TCGA database to investigate the expression of MT1G in GC. RT-qPCR and Western blot were used to detect the expression of MT1G, ferroptosis related proteins, autophagy related proteins and ARNTL clock autophagy related proteins in Hgc27, MKN45 and AGS cell lines. Exploring the biological functions of MT1G overexpressing GC cell lines through wound healing and transwell experiments. Use specific fluorescence probes to examine mitochondrial membrane potential and Fe2+ fluorescence intensity. Using immunoprecipitation analysis (CO-IP) to elucidate the association between GC cell lines GPX4, SQSTM and ARNTL. Use flow cytometry to detect ROS expression. Observation of autophagy related morphological changes in cells using transmission electron microscopy. Compared with gastric mucosal cell lines, the expression of MT1G is decreased in three gastric cancer cell lines (Hgc27, MKN45 and AGS). Overexpression of MT1G inhibits the proliferation, migration, and invasion functions of GC cells, reduces SOD and GSH content, increases MDA content, cause the mitochondrial membrane potential to weaken and promote the transformation of JC-1 aggregates to JC-1 monomer, increases Fe2+, affects ROS, and reduces GPX4 and SLC7A11 protein expression, promoting ferroptosis. Overexpression of MT1G promotes the transformation of LC3B I to LC3B II, reduces SQSTM1 protein expression, and leads to the appearance of more autophagosomes and autolysosomes at low magnification. At high magnification, mitochondrial autophagy, endoplasmic reticulum autophagy, lipid droplet autophagy, and wrinkled mitochondrial cristae are observed, promoting autophagy. Overexpression of MT1G inhibits GPX4, thereby affecting SQSTM1 as a vector to promote ARNTL autophagy and EGLN2, promoting ARNTL clock autophagy through the GPX4/SQSTM1 axis. Our research findings elucidate that overexpression of MT1G promotes iron autophagy centered around ARNTL in GC cells via the GPX4/SQSTM1 axis, thereby inhibiting GC cell function and providing a new molecular mechanism and therapeutic target for the development of GC.
Collapse
Affiliation(s)
- Kaiqiang Meng
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Jian Song
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China.
| | - Fan Qi
- College of Integrated Traditional Chinese and Western Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Jiamin Li
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Zhichao Fang
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Liang Song
- Basic Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| |
Collapse
|
22
|
Liu Z, Liu C, Fan C, Li R, Zhang S, Liu J, Li B, Zhang S, Guo L, Wang X, Qi Z, Shen Y. E3 ubiquitin ligase DTX2 fosters ferroptosis resistance via suppressing NCOA4-mediated ferritinophagy in non-small cell lung cancer. Drug Resist Updat 2024; 77:101154. [PMID: 39366066 DOI: 10.1016/j.drup.2024.101154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/10/2024] [Accepted: 09/21/2024] [Indexed: 10/06/2024]
Abstract
Non-small cell lung cancer (NSCLC) remains the foremost contributor to cancer-related fatalities globally, with limited effective therapeutic modalities. Recent research has shed light on the role of ferroptosis in various types of cancers, offering a potential avenue for improving cancer therapy. Herein, we identified E3 ubiquitin ligase deltex 2 (DTX2) as a potential therapeutic target candidate implicated in promoting NSCLC cell growth by inhibiting ferroptosis. Our investigation revealed a significant upregulation of DTX2 in NSCLC cells and tissues, which was correlated with poor prognosis. Downregulation of DTX2 suppressed NSCLC cell growth both in vitro and in vivo, while its overexpression accelerated cell proliferation. Moreover, knockdown of DTX2 promoted ferroptosis in NSCLC cells, which was mitigated by DTX2 overexpression. Mechanistically, we uncovered that DTX2 binds to nuclear receptor coactivator 4 (NCOA4), facilitating its ubiquitination and degradation via the K48 chain, which subsequently dampens NCOA4-driven ferritinophagy and ferroptosis in NSCLC cells. Notably, DTX2 knockdown promotes cisplatin-induced ferroptosis and overcomes drug resistance of NSCLC cells. These findings underscore the critical role of DTX2 in regulating ferroptosis and NCOA4-mediated ferritinophagy, suggesting its potential as a novel therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Zhuang Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin Medical University, Tianjin 300203, China
| | - Chang Liu
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Caihong Fan
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Runze Li
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Shiqi Zhang
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Jia Liu
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Bo Li
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Shengzheng Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Lihong Guo
- Department of Gastroenterology, Shengli Oilfield Central Hospital 257000, China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin Medical University, Tianjin 300203, China.
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300000, China; Department of Gastroenterology, Shengli Oilfield Central Hospital 257000, China; The First Department of Critical Care Medicine, The First Affiliated Hospital, Shihezi University 832000, China.
| | - Yanna Shen
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China.
| |
Collapse
|
23
|
Li X, Tian M, Yu L, Qian J, Yang J, Wang X, Lu C, Xiao C, Liu Y. The role of ferroptosis resistance in lymph-associated tumour metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189200. [PMID: 39426689 DOI: 10.1016/j.bbcan.2024.189200] [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: 05/30/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Tumour metastasis is a crucial factor in determining clinically challenging tumours. In this respect, the lymphatic system may act as potential entry portals for tumour metastasis, whilst, clinical detection of tumour-infiltrated lymph nodes also indicates poorer prognosis and higher metastatic risk. Whether tumour cells gain ferroptosis resistance in lymph that make them exhibit a stronger propensity for lymphatic dissemination compared to hematogenous spread might be a breakthrough for elucidating lymph-associated tumour metastasis. This review discusses how the lymphatic system endows tumour cells with ferroptosis resistance character, which makes them more propensity for lymph node pre-metastasis and distant metastasis through lymphatic circulation. Comprehensively considering the distinct structure and property of lymph and the unique metabolic characteristics of tumours, all of the lymphatic vessels, intestinal lymph and lymph nodes collectively manipulate an intricate interaction with the hematogenous system and afford substances exchange with tumour cells and extracellular vesicles, upon which make a ferroptosis resistant microenvironment for subsequent metastasis in distant organs and lymph nodes.
Collapse
Affiliation(s)
- Xiaoyu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Meng Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Liuchunyang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - JinXiu Qian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jue Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiangpeng Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
24
|
Osman EEA, Neamati N. Ironing Out the Mechanism of gp130 Signaling. Pharmacol Rev 2024; 76:1399-1443. [PMID: 39414364 DOI: 10.1124/pharmrev.124.001245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 10/18/2024] Open
Abstract
gp130 functions as a shared signal-transducing subunit not only for interleukin (IL)-6 but also for eight other human cytokine receptor complexes. The IL-6 signaling pathway mediated through gp130 encompasses classical, trans, or cluster signaling, intricately regulated by a diverse array of modulators affecting IL-6, its receptor, and gp130. Currently, only a limited number of small molecule antagonists and agonists for gp130 are known. This review aims to comprehensively examine the current knowledge of these modulators and provide insights into their pharmacological properties, particularly in the context of cancer and other diseases. Notably, the prominent gp130 modulators SC144, bazedoxifene, and raloxifene are discussed in detail, with a specific focus on the discovery of SC144's iron-chelating properties. This adds a new dimension to the understanding of its pharmacological effects and therapeutic potential in conditions where iron homeostasis is significant. Our bioinformatic analysis of gp130 and genes related to iron homeostasis reveals insightful correlations, implicating the role of iron in the gp130 signaling pathway. Overall, this review contributes to the evolving understanding of gp130 modulation and its potential therapeutic applications in various disease contexts. SIGNIFICANCE STATEMENT: This perspective provides a timely and comprehensive analysis of advancements in gp130 signaling research, emphasizing the therapeutic implications of the currently available modulators. Bioinformatic analysis demonstrates potential interplay between gp130 and genes that regulate iron homeostasis, suggesting new therapeutic avenues. By combining original research findings with a broader discussion of gp130's therapeutic potential, this perspective significantly contributes to the field.
Collapse
Affiliation(s)
- Essam Eldin A Osman
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan (E.E.A.O., N.N.) and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt (E.E.A.O.)
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan (E.E.A.O., N.N.) and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt (E.E.A.O.)
| |
Collapse
|
25
|
Tan J, Liao S, Yuan B, Liu X, Yu W, Zhan H, Jiang Y, Liu Y. Targeting SMYD2 promotes ferroptosis and impacts the progression of pancreatic cancer through the c-Myc/NCOA4 axis-mediated ferritinophagy. Biochim Biophys Acta Gen Subj 2024; 1868:130683. [PMID: 39089637 DOI: 10.1016/j.bbagen.2024.130683] [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: 02/27/2024] [Revised: 07/10/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is characterized by a poor prognosis and limited treatment options. Ferroptosis plays an important role in cancer, SET and MYND domain-containing protein 2 (SMYD2) is widely expressed in various cancers. However, the role of SMYD2 in regulating ferroptosis in PC remains unexplored. This study aimed to investigate the role of SMYD2 in mediating ferroptosis and its mechanistic implications in PC progression. METHODS The levels of SMYD2, c-Myc, and NCOA4 were assessed in PC tissues, and peritumoral tissues. SMYD2 expression was further analyzed in human PC cell lines. In BxPC3 cells, the expression of c-Myc, NCOA4, autophagy-related proteins, and mitochondrial morphology, was evaluated following transfection with si-SMYD2 and treatment with autophagy inhibitors and ferroptosis inhibitors. Ferroptosis levels were quantified using flow cytometry and ELISA assays. RNA immunoprecipitation was conducted to elucidate the interaction between c-Myc and NCOA4 mRNA. A xenograft mouse model was constructed to validate the impact of SMYD2 knockdown on PC growth. RESULTS SMYD2 and c-Myc were found to be highly expressed in PC tissues, while NCOA4 showed reduced expression. Among the PC cell lines studied, BxPC3 cells exhibited the highest SMYD2 expression. SMYD2 knockdown led to decreased c-Myc levels, increased NCOA4 expression, reduced autophagy-related protein expression, mitochondrial shrinkage, and heightened ferroptosis levels. Additionally, an interaction between c-Myc and NCOA4 was identified. In vivo, SMYD2 knockdown inhibited tumor growth. CONCLUSIONS Targeting SMYD2 inhibits PC progression by promoting ferritinophagy-dependent ferroptosis through the c-Myc/NCOA4 axis. These findings provide insights into potential diagnostic and therapeutic strategies for PC.
Collapse
Affiliation(s)
- Juan Tan
- Department of Pathology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Shan Liao
- Department of Pathology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Bowen Yuan
- Department of Pathology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Xinrong Liu
- Department of Pathology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Wentao Yu
- Department of Pathology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Han Zhan
- 921 Hospital of the Chinese People's Liberation Army Joint Logistic Support Force, Changsha, China
| | - Yan Jiang
- Department of Pathology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Yang Liu
- Department of Pathology, Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
26
|
Zhu Q, Zhai J, Chen Z, Guo Z, Wang N, Zhang C, Deng H, Wang S, Yang G. Ferritinophagy: Molecular mechanisms and role in disease. Pathol Res Pract 2024; 262:155553. [PMID: 39180800 DOI: 10.1016/j.prp.2024.155553] [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: 02/07/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Ferritinophagy is a regulatory pathway of iron homeostasis. It is a process in which nuclear receptor coactivator 4 (NCOA4) carries ferritin to autophagolysosomes for degradation. After ferritin is degraded by autophagy, iron ions are released, which promotes the labile iron pool (LIP) to drive the Fenton reaction to cause lipid peroxidation. Furthermore, ferroptosis promoted by the accumulation of lipid reactive oxygen species (ROS) induced by ferritinophagy can cause a variety of systemic diseases. In clinical studies, targeting the genes regulating ferritinophagy can prevent and treat such diseases. This article describes the key regulatory factors of ferritinophagy and the mechanism of ferritinophagy involved in ferroptosis. It also reviews the damage of ferritinophagy to the body, providing a theoretical basis for further finding clinical treatment methods.
Collapse
Affiliation(s)
- Qi Zhu
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Jianan Zhai
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Zhengguo Chen
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Zhifang Guo
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Ningning Wang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Cong Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Haoyuan Deng
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Shaopeng Wang
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guang Yang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China.
| |
Collapse
|
27
|
Bou-Abdallah F, Boumaiza M, Srivastava AK. Effects of ferritin iron loading, subunit composition, and the NCOA4-iron sulfur cluster on ferritin-NCOA4 interactions: An isothermal titration calorimetry study. Int J Biol Macromol 2024; 278:135044. [PMID: 39182888 DOI: 10.1016/j.ijbiomac.2024.135044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Ferritin is a 24-mer protein nanocage that stores iron and regulates intracellular iron homeostasis. The nuclear receptor coactivator-4 (NCOA4) binds specifically to ferritin H subunits and facilitates the autophagic trafficking of ferritin to the lysosome for degradation and iron release. Using isothermal titration calorimetry (ITC), we studied the thermodynamics of the interactions between ferritin and the soluble fragment of NCOA4 (residues 383-522), focusing on the effects of the recently identified FeS cluster bound to NCOA4, ferritin subunit composition, and ferritin-iron loading. Our findings show that in the presence of the FeS cluster, the binding is driven by a more favorable enthalpy change and a decrease in entropy change, indicating a key role for the FeS cluster in the structural organization and stability of the complex. The ferritin iron core further enhances this association, increasing binding enthalpy and stabilizing the NCOA4-ferritin complex. The ferritin subunit composition primarily affects binding stoichiometry of the reaction based on the number of H subunits in the ferritin H/L oligomer. Our results demonstrate that both the FeS cluster and the ferritin iron core significantly affect the binding thermodynamics of the NCOA4-ferritin interactions, suggesting regulatory roles for the FeS cluster and ferritin iron content in ferritinophagy.
Collapse
Affiliation(s)
- Fadi Bou-Abdallah
- Department of Chemistry, State University of New York, Potsdam, NY 13676, USA.
| | - Mohamed Boumaiza
- Department of Chemistry, State University of New York, Potsdam, NY 13676, USA
| | - Ayush K Srivastava
- Department of Chemistry, State University of New York, Potsdam, NY 13676, USA
| |
Collapse
|
28
|
Mao Z, Hu Y, Zhao Y, Zhang X, Guo L, Wang X, Zhang J, Miao M. The Mutual Regulatory Role of Ferroptosis and Immunotherapy in Anti-tumor Therapy. Apoptosis 2024; 29:1291-1308. [PMID: 38853203 PMCID: PMC11416416 DOI: 10.1007/s10495-024-01988-9] [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] [Accepted: 05/25/2024] [Indexed: 06/11/2024]
Abstract
Ferroptosis is a form of cell death that is triggered by the presence of ferrous ions and is characterized by lipid peroxidation induced by these ions. The mechanism exhibits distinct morphological characteristics compared to apoptosis, autophagy, and necrosis. A notable aspect of ferroptosis is its ability to inhibit uncontrolled tumor replication and immortalization, especially in malignant, drug-resistant, and metastatic tumors. Additionally, immunotherapy, a novel therapeutic approach for tumors, has been found to have a reciprocal regulatory relationship with ferroptosis in the context of anti-tumor therapy. A comprehensive analysis of ferroptosis and immunotherapy in tumor therapy is presented in this paper, highlighting the potential for mutual adjuvant effects. Specifically, we discuss the mechanisms underlying ferroptosis and immunotherapy, emphasizing their ability to improve the tumor immune microenvironment and enhance immunotherapeutic effects. Furthermore, we investigate how immunotherapeutic factors may increase the sensitivity of tumor cells to ferroptosis. We aim to provide a prospective view of the promising value of combined ferroptosis and immunotherapy in anticancer therapy by elucidating the mutual regulatory network between each.
Collapse
Affiliation(s)
- Zhiguo Mao
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Yilong Hu
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Yinan Zhao
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Xiaolei Zhang
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Lin Guo
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Xiaoran Wang
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Jinying Zhang
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Mingsan Miao
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China.
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China.
| |
Collapse
|
29
|
Peiyin F, Yuxian W, Jiali Z, Jian X. Research progress of ferroptosis in female infertility. J Ovarian Res 2024; 17:183. [PMID: 39267109 PMCID: PMC11391650 DOI: 10.1186/s13048-024-01508-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/30/2024] [Indexed: 09/14/2024] Open
Abstract
Ferroptosis is a novel type of programmed cell death dependent on iron and characterized by the accumulation of lipid peroxides in cells and is closely related to various diseases. Female infertility is a global health concern, which is associated with a variety of factors. The etiology remains unknown in many women with infertility. With further investigation into the pathogenesis of infertility, a growing number of studies have demonstrated the close connections between infertility and ferroptosis. Through a literature review, it is found that ferroptosis is closely involved in endometriosis- and polycystic ovarian syndrome (PCOS)-associated infertility and tubal factor infertility. Iron overload increases the resistance to ferroptosis, and ferroptosis in some cells accelerates endometrial lesion growth. Moreover, iron overload may be hazardous to oocytes. This review may shed some light on the diagnosis and treatment of female infertility.
Collapse
Affiliation(s)
- Fan Peiyin
- Reproductive Medicine Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Wang Yuxian
- Reproductive Medicine Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Zhang Jiali
- Reproductive Medicine Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Xu Jian
- Reproductive Medicine Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China.
| |
Collapse
|
30
|
Hansen FJ, Mittelstädt A, Clausen FN, Knoedler S, Knoedler L, Klöckner S, Kuchenreuther I, Mazurie J, Arnold LS, Anthuber A, Jacobsen A, Merkel S, Weisel N, Klösch B, Karabiber A, Tacyildiz I, Czubayko F, Reitberger H, Gendy AE, Brunner M, Krautz C, Wolff K, Mihai S, Neufert C, Siebler J, Grützmann R, Weber GF, David P. CD71 expressing circulating neutrophils serve as a novel prognostic biomarker for metastatic spread and reduced outcome in pancreatic ductal adenocarcinoma patients. Sci Rep 2024; 14:21164. [PMID: 39256468 PMCID: PMC11387421 DOI: 10.1038/s41598-024-70916-3] [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: 04/25/2024] [Accepted: 08/22/2024] [Indexed: 09/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, presenting a persisting global health burden. Neutrophils have a double-edged role in tumor progression exhibiting both pro-tumor and anti-tumor functions. CD71, also known as transferrin receptor 1, performs a critical role in cellular iron uptake and is highly expressed on proliferating cells, and especially on activated immune cells. CD71 is known to be elevated in various types of solid cancers and is associated with poor prognosis, however, the expression of CD71 on neutrophils in PDAC and its potential clinical impact is still unknown. Therefore, we analyzed CD71 on circulating neutrophils in PDAC and clinical control patients and found a significant increased expression in PDAC patients. High expression of CD71 on neutrophils in PDAC patients was associated with reduced outcome compared to low expression. CD71 on neutrophils correlated positively with the levels of proinflammatory cytokines IL-6, IFN-γ, and growth factor ligands CD40-L, and BAFF in plasma of PDAC patients. Finally, we have demonstrated that high expression of CD71 on neutrophils was also associated with an increased expression of CD39 and CD25 on circulating T-cells. Based on our findings, we hypothesize that CD71 on neutrophils is associated with tumor progression in PDAC. Further studies are required to investigate the distinct functionality of CD71 expressing neutrophils and their potential clinical application.
Collapse
Affiliation(s)
- Frederik J Hansen
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Anke Mittelstädt
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Finn-Niklas Clausen
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Samuel Knoedler
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Ingolstädter Landtsraße 1, 85764, Neuherberg, Germany
| | - Leonard Knoedler
- Division of Genetic Immunotherapy (LIT), University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Sebastian Klöckner
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Isabelle Kuchenreuther
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Johanne Mazurie
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Lisa-Sophie Arnold
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Anna Anthuber
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Anne Jacobsen
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Susanne Merkel
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Nadine Weisel
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Bettina Klösch
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Alara Karabiber
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Irem Tacyildiz
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Franziska Czubayko
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Helena Reitberger
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Amr El Gendy
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Maximilian Brunner
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Christian Krautz
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Kerstin Wolff
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- First Department of Medicine, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sidonia Mihai
- Zentrallabor im Universitätsklinikum, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstr. 12, Erlangen, Germany
| | - Clemens Neufert
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- First Department of Medicine, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Siebler
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- First Department of Medicine, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Grützmann
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Georg F Weber
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Paul David
- Department of General and Visceral Surgery, Friedrich-Alexander-University, Krankenhausstraße 12, 91054, Erlangen, Germany
| |
Collapse
|
31
|
Leandri R, Power K, Buonocore S, De Vico G. Preliminary Evidence of the Possible Roles of the Ferritinophagy-Iron Uptake Axis in Canine Testicular Cancer. Animals (Basel) 2024; 14:2619. [PMID: 39272404 PMCID: PMC11394645 DOI: 10.3390/ani14172619] [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: 07/18/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Iron is a key element in spermatogenesis; its metabolic pathway in the testis is strictly regulated. Alterations in iron metabolism are linked to various diseases, including cancer, and changes in iron metabolism-related proteins have been observed in multiple human, mouse and canine tumors. There is limited knowledge about iron metabolism in canine non-neoplastic and neoplastic testes. This study aimed to explore the immunohistochemical expression of molecules involved in iron uptake and storage [Transferrin Receptor 1 (TfR1), ferritin (FTH1), nuclear receptor coactivator 4 (NCOA4)] and PCNA in canine non-neoplastic and neoplastic testicular samples. Non-neoplastic testes showed moderate TfR1 expression in developing germ cells and Sertoli cells, high NCOA4 cytoplasmic immunostaining in the Sertoli cells and occasional cytoplasmic immunopositivity for FTH1 in the spermatogonia and Sertoli cells. In contrast, Leydig cell tumors (LCTs) and Diffuse Type Seminoma (DSEM) exhibited increased expression of TfR1, along with higher PCNA expression, suggesting a higher iron need for proliferation. Intratubular Type Seminoma (ITSEM) showed a higher FTH1 expression, indicating greater iron storage, while the increased NCOA4 expression in the LCTs and DSEM suggested ferritinophagy to release iron for proliferation. Sertoli cell tumors (SCTs) showed only NCOA4 expression. These preliminary findings highlight potential molecular targets for developing new anti-neoplastic treatments in canine testicular tumors.
Collapse
Affiliation(s)
- Rebecca Leandri
- Department of Biology, University of Naples 'Federico II', Via Vicinale Cupa Cinthia 21, 80216 Napoli, Italy
| | - Karen Power
- Department of Biology, University of Naples 'Federico II', Via Vicinale Cupa Cinthia 21, 80216 Napoli, Italy
| | - Sara Buonocore
- Department of Biology, University of Naples 'Federico II', Via Vicinale Cupa Cinthia 21, 80216 Napoli, Italy
| | - Gionata De Vico
- Department of Biology, University of Naples 'Federico II', Via Vicinale Cupa Cinthia 21, 80216 Napoli, Italy
| |
Collapse
|
32
|
Zhang Y, Zheng BY, Zhang QF, Zhao YN, Yu QM, Liu X, Ding SY, Qian SS, Wu H, Wu QY, Zhang YH, Zheng L, Zhang XH, Zhang HF, Hao YM, Lu JC, Wang L, Wen JK, Zheng B. Nanoparticles targeting OPN loaded with BY1 inhibits vascular restenosis by inducing FTH1-dependent ferroptosis in vascular smooth muscle cells. Biomaterials 2024; 309:122613. [PMID: 38759485 DOI: 10.1016/j.biomaterials.2024.122613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Vascular restenosis following angioplasty continues to pose a significant challenge. The heterocyclic trioxirane compound [1, 3, 5-tris((oxiran-2-yl)methyl)-1, 3, 5-triazinane-2, 4, 6-trione (TGIC)], known for its anticancer activity, was utilized as the parent ring to conjugate with a non-steroidal anti-inflammatory drug, resulting in the creation of the spliced conjugated compound BY1. We found that BY1 induced ferroptosis in VSMCs as well as in neointima hyperplasia. Furthermore, ferroptosis inducers amplified BY1-induced cell death, while inhibitors mitigated it, indicating the contribution of ferroptosis to BY1-induced cell death. Additionally, we established that ferritin heavy chain1 (FTH1) played a pivotal role in BY1-induced ferroptosis, as evidenced by the fact that FTH1 overexpression abrogated BY1-induced ferroptosis, while FTH1 knockdown exacerbated it. Further study found that BY1 induced ferroptosis by enhancing the NCOA4-FTH1 interaction and increasing the amount of intracellular ferrous. We compared the effectiveness of various administration routes for BY1, including BY1-coated balloons, hydrogel-based BY1 delivery, and nanoparticles targeting OPN loaded with BY1 (TOP@MPDA@BY1) for targeting proliferated VSMCs, for prevention and treatment of the restenosis. Our results indicated that TOP@MPDA@BY1 was the most effective among the three administration routes, positioning BY1 as a highly promising candidate for the development of drug-eluting stents or treatments for restenosis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Bo-Yang Zheng
- Department of tumor biotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Qian-Fan Zhang
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ya-Nan Zhao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qi-Ming Yu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xin Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Si-Ying Ding
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuang-Shuang Qian
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Han Wu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qian-Yu Wu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yu-Han Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Lei Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xin-Hua Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China; Institution of Chinese Integrative Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Hao-Feng Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Hebei Province Key Laboratory of Innovative Drug Research and Evaluation, Shijiazhuang, 050017, China
| | - Yi-Ming Hao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jing-Chao Lu
- Department of Cardiovascular Medicine, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lei Wang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Hebei Province Key Laboratory of Innovative Drug Research and Evaluation, Shijiazhuang, 050017, China.
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
33
|
Wu H, Fu M, Wu M, Cao Z, Zhang Q, Liu Z. Emerging mechanisms and promising approaches in pancreatic cancer metabolism. Cell Death Dis 2024; 15:553. [PMID: 39090116 PMCID: PMC11294586 DOI: 10.1038/s41419-024-06930-0] [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: 04/18/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Pancreatic cancer is an aggressive cancer with a poor prognosis. Metabolic abnormalities are one of the hallmarks of pancreatic cancer, and pancreatic cancer cells can adapt to biosynthesis, energy intake, and redox needs through metabolic reprogramming to tolerate nutrient deficiency and hypoxic microenvironments. Pancreatic cancer cells can use glucose, amino acids, and lipids as energy to maintain malignant growth. Moreover, they also metabolically interact with cells in the tumour microenvironment to change cell fate, promote tumour progression, and even affect immune responses. Importantly, metabolic changes at the body level deserve more attention. Basic research and clinical trials based on targeted metabolic therapy or in combination with other treatments are in full swing. A more comprehensive and in-depth understanding of the metabolic regulation of pancreatic cancer cells will not only enrich the understanding of the mechanisms of disease progression but also provide inspiration for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hao Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengdi Fu
- Department of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhen Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiyao Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
34
|
Zuo Z, Luo M, Liu Z, Liu T, Wang X, Huang X, Li S, Wu H, Pan Q, Chen T, Yang L, Liu HF. Selenium nanoparticles alleviate renal ischemia/reperfusion injury by inhibiting ferritinophagy via the XBP1/NCOA4 pathway. Cell Commun Signal 2024; 22:376. [PMID: 39061070 PMCID: PMC11282718 DOI: 10.1186/s12964-024-01751-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Acute kidney injury (AKI) is closely related to lysosomal dysfunction and ferroptosis in renal tubular epithelial cells (TECs), for which effective treatments are urgently needed. Although selenium nanoparticles (SeNPs) have emerged as promising candidates for AKI therapy, their underlying mechanisms have not been fully elucidated. Here, we investigated the effect of SeNPs on hypoxia/reoxygenation (H/R)-induced ferroptosis and lysosomal dysfunction in TECs in vitro and evaluated their efficacy in a murine model of ischemia/reperfusion (I/R)-AKI. We observed that H/R-induced ferroptosis was accompanied by lysosomal Fe2+ accumulation and dysfunction in TECs, which was ameliorated by SeNPs administration. Furthermore, SeNPs protected C57BL/6 mice against I/R-induced inflammation and ferroptosis. Mechanistically, we found that lysosomal Fe2+ accumulation and ferroptosis were associated with the excessive activation of NCOA4-mediated ferritinophagy, a process mitigated by SeNPs through the upregulation of X-box binding protein 1 (XBP1). Downregulation of XBP1 promoted ferritinophagy and partially counteracted the protective effects of SeNPs on ferroptosis inhibition in TECs. Overall, our findings revealed a novel role for SeNPs in modulating ferritinophagy, thereby improving lysosomal function and attenuating ferroptosis of TECs in I/R-AKI. These results provide evidence for the potential application of SeNPs as therapeutic agents for the prevention and treatment of AKI.
Collapse
Affiliation(s)
- Zhenying Zuo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Mianna Luo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhongyu Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Ting Liu
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Xi Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Huang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shangmei Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hongluan Wu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Lawei Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
35
|
Larrue C, Mouche S, Angelino P, Sajot M, Birsen R, Kosmider O, Mckee T, Vergez F, Recher C, Mas VMD, Gu Q, Xu J, Tsantoulis P, Sarry JE, Tamburini J. Targeting ferritinophagy impairs quiescent cancer stem cells in acute myeloid leukemia in vitro and in vivo models. Sci Transl Med 2024; 16:eadk1731. [PMID: 39047119 DOI: 10.1126/scitranslmed.adk1731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
Acute myeloid leukemia (AML) remains a challenging hematological malignancy with poor prognosis and limited treatment options. Leukemic stem cells (LSCs) contribute to therapeutic failure, relapse, and adverse outcome. This study investigates the role of quiescence and related molecular mechanisms in AML pathogenesis and LSC functions to identify potential therapeutic targets. Transcriptomic analysis revealed that the LSC-enriched quiescent cell population has a distinct gene signature with prognostic relevance in patients with AML. Mechanistically, quiescent blasts exhibit increased autophagic activity, which contributes to their sustained viability. Proteomic profiling uncovered differential requirements for iron metabolism between quiescent and cycling cells, revealing a unique dependence of quiescent cells on ferritinophagy, a selective form of autophagy mediated by nuclear receptor coactivator 4 (NCOA4), which regulates iron bioavailability. We evaluated the therapeutic potential of inhibiting NCOA4-mediated ferritinophagy using genetic knockdown and chemical inhibition approaches. In vitro assays showed that suppression of NCOA4 was toxic to leukemic blasts, particularly the CD34+CD38- LSC-enriched population, without affecting normal CD34+ hematopoietic progenitors. In vivo studies using murine patient-derived xenograft (PDX) models of AML confirmed that NCOA4 inhibition reduced tumor burden and impaired LSC viability and self-renewal, indicating a specific vulnerability of these cells to ferritinophagy disruption. Our findings underscore the role of NCOA4-mediated ferritinophagy in maintaining LSC quiescence and function and suggest that targeting this pathway may be an effective therapeutic strategy for AML. This study highlights the potential of NCOA4 inhibition to improve AML outcomes and paves the way for future research and clinical development.
Collapse
Affiliation(s)
- Clement Larrue
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Sarah Mouche
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
| | - Paolo Angelino
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Translational Data Science, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Maxime Sajot
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Rudy Birsen
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, 75014 Paris, France
| | - Olivier Kosmider
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, 75014 Paris, France
| | - Thomas Mckee
- Division of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - François Vergez
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Christian Recher
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Véronique Mansat-De Mas
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Petros Tsantoulis
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Jerome Tamburini
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Oncology Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| |
Collapse
|
36
|
Pezacki AT, Gonciarz RL, Okamura T, Matier CD, Torrente L, Cheng K, Miller SG, Ralle M, Ward NP, DeNicola GM, Renslo AR, Chang CJ. A tandem activity-based sensing and labeling strategy reveals antioxidant response element regulation of labile iron pools. Proc Natl Acad Sci U S A 2024; 121:e2401579121. [PMID: 38968123 PMCID: PMC11252945 DOI: 10.1073/pnas.2401579121] [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: 01/23/2024] [Accepted: 05/29/2024] [Indexed: 07/07/2024] Open
Abstract
Iron is an essential element for life owing to its ability to participate in a diverse array of oxidation-reduction reactions. However, misregulation of iron-dependent redox cycling can also produce oxidative stress, contributing to cell growth, proliferation, and death pathways underlying aging, cancer, neurodegeneration, and metabolic diseases. Fluorescent probes that selectively monitor loosely bound Fe(II) ions, termed the labile iron pool, are potentially powerful tools for studies of this metal nutrient; however, the dynamic spatiotemporal nature and potent fluorescence quenching capacity of these bioavailable metal stores pose challenges for their detection. Here, we report a tandem activity-based sensing and labeling strategy that enables imaging of labile iron pools in live cells through enhancement in cellular retention. Iron green-1 fluoromethyl (IG1-FM) reacts selectively with Fe(II) using an endoperoxide trigger to release a quinone methide dye for subsequent attachment to proximal biological nucleophiles, providing a permanent fluorescent stain at sites of elevated labile iron. IG1-FM imaging reveals that degradation of the major iron storage protein ferritin through ferritinophagy expands the labile iron pool, while activation of nuclear factor-erythroid 2-related factor 2 (NRF2) antioxidant response elements (AREs) depletes it. We further show that lung cancer cells with heightened NRF2 activation, and thus lower basal labile iron, have reduced viability when treated with an iron chelator. By connecting labile iron pools and NRF2-ARE activity to a druggable metal-dependent vulnerability in cancer, this work provides a starting point for broader investigations into the roles of transition metal and antioxidant signaling pathways in health and disease.
Collapse
Affiliation(s)
- Aidan T. Pezacki
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Ryan L. Gonciarz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Toshitaka Okamura
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Carson D. Matier
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Laura Torrente
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Ke Cheng
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Sophia G. Miller
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR97239
| | - Martina Ralle
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR97239
| | - Nathan P. Ward
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Gina M. DeNicola
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA94158
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA94720
| |
Collapse
|
37
|
Le Y, Liu Q, Yang Y, Wu J. The emerging role of nuclear receptor coactivator 4 in health and disease: a novel bridge between iron metabolism and immunity. Cell Death Discov 2024; 10:312. [PMID: 38961066 PMCID: PMC11222541 DOI: 10.1038/s41420-024-02075-3] [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: 03/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
Nuclear receptor coactivator 4 (NCOA4) has recently been recognized as a selective cargo receptor of ferritinophagy participating in ferroptosis. However, NCOA4 is also a coactivator that modulates the transcriptional activity of many vital nuclear receptors. Recent novel studies have documented the role of NCOA4 in healthy and pathogenic conditions via its modulation of iron- and non-iron-dependent metabolic pathways. NCOA4 exhibits non-ferritinophagic and iron-independent features such as promoting tumorigenesis and erythropoiesis, immunomodulation, regulating autophagy, and participating in DNA replication and mitosis. Full-length human-NCOA4 is composed of 614 amino acids, of which the N-terminal (1-237) contains nuclear-receptor-binding domains, while the C-terminal (238-614) principally contains a ferritin-binding domain. The exploration of the protein structure of NCOA4 suggests that NCOA4 possesses additional significant and complex functions based on its structural domains. Intriguingly, another three isoforms of NCOA4 that are produced by alternative splicing have been identified, which may also display disparate activities in physiological and pathological processes. Thus, NCOA4 has become an important bridge that encompasses interactions between immunity and metabolism. In this review, we outline the latest advances in the important regulating mechanisms underlying NCOA4 actions in health and disease conditions, providing insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Yue Le
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Qinjie Liu
- Department of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Jie Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
- Research Center of Surgery, BenQ Medical Center, the Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210021, China.
| |
Collapse
|
38
|
Guo JY, White E. Role of Tumor Cell Intrinsic and Host Autophagy in Cancer. Cold Spring Harb Perspect Med 2024; 14:a041539. [PMID: 38253423 PMCID: PMC11216174 DOI: 10.1101/cshperspect.a041539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Macroautophagy (autophagy hereafter) is an intracellular nutrient scavenging pathway induced by starvation and other stressors whereby cellular components such as organelles are captured in double-membrane vesicles (autophagosomes), whereupon their contents are degraded through fusion with lysosomes. Two main purposes of autophagy are to recycle the intracellular breakdown products to sustain metabolism and survival during starvation and to eliminate damaged or excess cellular components to suppress inflammation and maintain homeostasis. In contrast to most normal cells and tissues in the fed state, tumor cells up-regulate autophagy to promote their growth, survival, and malignancy. This tumor-cell-autonomous autophagy supports elevated metabolic demand and suppresses tumoricidal activation of the innate and adaptive immune responses. Tumor-cell-nonautonomous (e.g., host) autophagy also supports tumor growth by maintaining essential tumor nutrients in the circulation and tumor microenvironment and by suppressing an antitumor immune response. In the setting of cancer therapy, autophagy is a resistance mechanism to chemotherapy, targeted therapy, and immunotherapy. Thus, tumor and host autophagy are protumorigenic and autophagy inhibition is being examined as a novel therapeutic approach to treat cancer.
Collapse
Affiliation(s)
- Jessie Yanxiang Guo
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
- Department of Chemical Biology, Rutgers Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey 08544, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey 08544, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08903, USA
| |
Collapse
|
39
|
De Santis MC, Bockorny B, Hirsch E, Cappello P, Martini M. Exploiting pancreatic cancer metabolism: challenges and opportunities. Trends Mol Med 2024; 30:592-604. [PMID: 38604929 DOI: 10.1016/j.molmed.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/13/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive form of pancreatic cancer, known for its challenging diagnosis and limited treatment options. The focus on metabolic reprogramming as a key factor in tumor initiation, progression, and therapy resistance has gained prominence. In this review we focus on the impact of metabolic changes on the interplay among stromal, immune, and tumor cells, as glutamine and branched-chain amino acids (BCAAs) emerge as pivotal players in modulating immune cell functions and tumor growth. We also discuss ongoing clinical trials that explore metabolic modulation for PDAC, targeting mitochondrial metabolism, asparagine and glutamine addiction, and autophagy inhibition. Overcoming challenges in understanding nutrient effects on immune-stromal-tumor interactions holds promise for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Maria Chiara De Santis
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy.
| | - Bruno Bockorny
- BIDMC Department of Medicine, Harvard Medical School, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Miriam Martini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy.
| |
Collapse
|
40
|
Zhi HT, Lu Z, Chen L, Wu JQ, Li L, Hu J, Chen WH. Anticancer efficacy triggered by synergistically modulating the homeostasis of anions and iron: Design, synthesis and biological evaluation of dual-functional squaramide-hydroxamic acid conjugates. Bioorg Chem 2024; 147:107421. [PMID: 38714118 DOI: 10.1016/j.bioorg.2024.107421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 05/09/2024]
Abstract
Targeting the homeostasis of anions and iron has emerged as a promising therapeutic approach for the treatment of cancers. However, single-targeted agents often fall short of achieving optimal treatment efficacy. Herein we designed and synthesized a series of novel dual-functional squaramide-hydroxamic acid conjugates that are capable of synergistically modulating the homeostasis of anions and iron. Among them, compound 16 exhibited the most potent antiproliferative activity against a panel of selected cancer cell lines, and strong in vivo anti-tumor efficacy. This compound effectively elevated lysosomal pH through anion transport, and reduced the levels of intracellular iron. Compound 16 could disturb autophagy in A549 cells and trigger robust apoptosis. This compound caused cell cycle arrest at the G1/S phase, altered the mitochondrial function and elevated ROS levels. The present findings clearly demonstrated that synergistic modulation of anion and iron homeostasis has high potentials in the development of promising chemotherapeutic agents with dual action against cancers.
Collapse
Affiliation(s)
- Hai-Tao Zhi
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Zhonghui Lu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Li Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Jia-Qiang Wu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Lanqing Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Jinhui Hu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China.
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China.
| |
Collapse
|
41
|
Liu Y, Carbonetto P, Willwerscheid J, Oakes SA, Macleod KF, Stephens M. Dissecting tumor transcriptional heterogeneity from single-cell RNA-seq data by generalized binary covariance decomposition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.15.553436. [PMID: 37645713 PMCID: PMC10462040 DOI: 10.1101/2023.08.15.553436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Profiling tumors with single-cell RNA sequencing (scRNA-seq) has the potential to identify recurrent patterns of transcription variation related to cancer progression, and produce new therapeutically relevant insights. However, the presence of strong inter-tumor heterogeneity often obscures more subtle patterns that are shared across tumors, some of which may characterize clinically relevant disease subtypes. Here we introduce a new statistical method, generalized binary covariance decomposition (GBCD), to address this problem. We show that GBCD can help decompose transcriptional heterogeneity into interpretable components - including patient-specific, dataset-specific and shared components relevant to disease subtypes - and that, in the presence of strong inter-tumor heterogeneity, it can produce more interpretable results than existing methods. Applied to data from three studies on pancreatic cancer adenocarcinoma (PDAC), GBCD produces a refined characterization of existing tumor subtypes (e.g., classical vs. basal), and identifies a new gene expression program (GEP) that is prognostic of poor survival independent of established prognostic factors such as tumor stage and subtype. The new GEP is enriched for genes involved in a variety of stress responses, and suggests a potentially important role for the integrated stress response in PDAC development and prognosis.
Collapse
|
42
|
Arcos M, Liu Z, Villareal LB, Velez PK, Desai SP, Noureddine A, Zheng H, Martin DR, Brinker J, Zhang D, Xue X. Myeloid NCOA4 sequesters KEAP1 to reduce ferroptosis for protection against salmonellosis in mice. RESEARCH SQUARE 2024:rs.3.rs-4278310. [PMID: 38798412 PMCID: PMC11118698 DOI: 10.21203/rs.3.rs-4278310/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Salmonellosis, caused by Salmonella enterica serovar Typhimurium, is a significant global threat. Host immunity limits bacterial replication by inducing hepcidin, which degrades ferroportin, reducing iron transfer. However, this boosts macrophage iron storage, aiding intracellular pathogens like Salmonella. Mice lacking ferritin heavy chain (FTH1) in myeloid cells suffer worsened Salmonella infection. Nuclear receptor co-activator 4 (NCOA4) regulates iron release via FTH1 degradation during low iron, but its role in salmonellosis is unclear. Here, we reveal that myeloid NCOA4 deficiency augments spleen iron levels and increases cellular iron accumulation, oxidative stress, and ferroptosis in bone marrow-derived macrophages. This deficiency also increases susceptibility to Salmonella-induced colitis in mice. Mechanistically, NCOA4 suppresses oxidative stress by directly binding to the E3 ubiquitin ligase Kelch-like ECH-associated protein 1 (KEAP1) and stabilizing the antioxidant transcription factor nuclear factor-erythroid 2-related factor 2 (NRF2). Activation of NRF2 protects myeloid NCOA4 knockout mice from Salmonella-induced colitis. Antioxidant Tempol and myeloid cell-targeted curcumin offer protection against colitis in myeloid NCOA4-deficient mice. A low iron diet and ferroptosis inhibition also mitigate the heightened colitis in these mice. Overexpression of myeloid cell-specific NCOA4 confers protection against Salmonella-induced colitis via upregulating NRF2 signaling. Serum iron was reduced in myeloid NCOA4-overexpressing mice, but not in NCOA4-deficient mice. Targeted serum metabolomics analysis revealed that many lipids were decreased in myeloid NCOA4-deficient mice, while several of them were increased in myeloid NCOA4-overexpressing mice. Together, this study not only advances our understanding of NCOA4/KEAP1/NRF2/ferroptosis axis but also paves the way for novel myeloid cell-targeted therapies to combat salmonellosis.
Collapse
Affiliation(s)
- Mariella Arcos
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131
| | - Zhaoli Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131
| | - Luke B Villareal
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131
| | - Paloma Kai Velez
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131
| | - Sharina P Desai
- Department of Molecular Genetics Microbiology, University of New Mexico, Albuquerque, NM 87131
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131
| | - Huayu Zheng
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - David R Martin
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
| | - Jeffrey Brinker
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131
| | - Donna Zhang
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL 33458
| | - Xiang Xue
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
43
|
Wang H, Hu Q, Chen Y, Huang X, Feng Y, Shi Y, Li R, Yin X, Song X, Liang Y, Zhang T, Xu L, Dong G, Jiang F. Ferritinophagy mediates adaptive resistance to EGFR tyrosine kinase inhibitors in non-small cell lung cancer. Nat Commun 2024; 15:4195. [PMID: 38760351 PMCID: PMC11101634 DOI: 10.1038/s41467-024-48433-8] [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: 06/19/2023] [Accepted: 04/26/2024] [Indexed: 05/19/2024] Open
Abstract
Osimertinib (Osi) is a widely used epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). However, the emergence of resistance is inevitable, partly due to the gradual evolution of adaptive resistant cells during initial treatment. Here, we find that Osi treatment rapidly triggers adaptive resistance in tumor cells. Metabolomics analysis reveals a significant enhancement of oxidative phosphorylation (OXPHOS) in Osi adaptive-resistant cells. Mechanically, Osi treatment induces an elevation of NCOA4, a key protein of ferritinophagy, which maintains the synthesis of iron-sulfur cluster (ISC) proteins of electron transport chain and OXPHOS. Additionally, active ISC protein synthesis in adaptive-resistant cells significantly increases the sensitivity to copper ions. Combining Osi with elesclomol, a copper ion ionophore, significantly increases the efficacy of Osi, with no additional toxicity. Altogether, this study reveals the mechanisms of NCOA4-mediated ferritinophagy in Osi adaptive resistance and introduces a promising new therapy of combining copper ionophores to improve its initial efficacy.
Collapse
Affiliation(s)
- Hui Wang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Qianfan Hu
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Yuzhong Chen
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- Department of Oncology, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
| | - Xing Huang
- Department of Pathology, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
| | - Yipeng Feng
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Yuanjian Shi
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Rutao Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuewen Yin
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
| | - Xuming Song
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Yingkuan Liang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
| | - Te Zhang
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Lin Xu
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Jiangning District, Nanjing, China
| | - Gaochao Dong
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China.
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China.
| | - Feng Jiang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China.
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China.
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
44
|
Hoelzgen F, Nguyen TTP, Klukin E, Boumaiza M, Srivastava AK, Kim EY, Zalk R, Shahar A, Cohen-Schwartz S, Meyron-Holtz EG, Bou-Abdallah F, Mancias JD, Frank GA. Structural basis for the intracellular regulation of ferritin degradation. Nat Commun 2024; 15:3802. [PMID: 38714719 PMCID: PMC11076521 DOI: 10.1038/s41467-024-48151-1] [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: 11/14/2023] [Accepted: 04/19/2024] [Indexed: 05/10/2024] Open
Abstract
The interaction between nuclear receptor coactivator 4 (NCOA4) and the iron storage protein ferritin is a crucial component of cellular iron homeostasis. The binding of NCOA4 to the FTH1 subunits of ferritin initiates ferritinophagy-a ferritin-specific autophagic pathway leading to the release of the iron stored inside ferritin. The dysregulation of NCOA4 is associated with several diseases, including neurodegenerative disorders and cancer, highlighting the NCOA4-ferritin interface as a prime target for drug development. Here, we present the cryo-EM structure of the NCOA4-FTH1 interface, resolving 16 amino acids of NCOA4 that are crucial for the interaction. The characterization of mutants, designed to modulate the NCOA4-FTH1 interaction, is used to validate the significance of the different features of the binding site. Our results explain the role of the large solvent-exposed hydrophobic patch found on the surface of FTH1 and pave the way for the rational development of ferritinophagy modulators.
Collapse
Affiliation(s)
- Fabian Hoelzgen
- The Kreitman School of Advanced Graduate Studies, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Life Sciences, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Thuy T P Nguyen
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Elina Klukin
- Department of Life Sciences, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Mohamed Boumaiza
- Department of Chemistry, State University of New York at Potsdam (SUNY Potsdam), Potsdam, NY, USA
| | - Ayush K Srivastava
- Department of Chemistry, State University of New York at Potsdam (SUNY Potsdam), Potsdam, NY, USA
| | - Elizabeth Y Kim
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ran Zalk
- Ilse Katz Institute for Nanoscale Science & Technology, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anat Shahar
- Ilse Katz Institute for Nanoscale Science & Technology, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sagit Cohen-Schwartz
- The National Institute for Biotechnology in the Negev - NIBN, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Fadi Bou-Abdallah
- Department of Chemistry, State University of New York at Potsdam (SUNY Potsdam), Potsdam, NY, USA
| | - Joseph D Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Gabriel A Frank
- Department of Life Sciences, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Ilse Katz Institute for Nanoscale Science & Technology, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- The National Institute for Biotechnology in the Negev - NIBN, Marcus Family Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
45
|
Perurena N, Situ L, Cichowski K. Combinatorial strategies to target RAS-driven cancers. Nat Rev Cancer 2024; 24:316-337. [PMID: 38627557 DOI: 10.1038/s41568-024-00679-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2024] [Indexed: 05/01/2024]
Abstract
Although RAS was formerly considered undruggable, various agents that inhibit RAS or specific RAS oncoproteins have now been developed. Indeed, the importance of directly targeting RAS has recently been illustrated by the clinical success of mutant-selective KRAS inhibitors. Nevertheless, responses to these agents are typically incomplete and restricted to a subset of patients, highlighting the need to develop more effective treatments, which will likely require a combinatorial approach. Vertical strategies that target multiple nodes within the RAS pathway to achieve deeper suppression are being investigated and have precedence in other contexts. However, alternative strategies that co-target RAS and other therapeutic vulnerabilities have been identified, which may mitigate the requirement for profound pathway suppression. Regardless, the efficacy of any given approach will likely be dictated by genetic, epigenetic and tumour-specific variables. Here we discuss various combinatorial strategies to treat KRAS-driven cancers, highlighting mechanistic concepts that may extend to tumours harbouring other RAS mutations. Although many promising combinations have been identified, clinical responses will ultimately depend on whether a therapeutic window can be achieved and our ability to prospectively select responsive patients. Therefore, we must continue to develop and understand biologically diverse strategies to maximize our likelihood of success.
Collapse
Affiliation(s)
- Naiara Perurena
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Lisa Situ
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Karen Cichowski
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Ludwig Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Terzi EM, Possemato R. Iron, Copper, and Selenium: Cancer's Thing for Redox Bling. Cold Spring Harb Perspect Med 2024; 14:a041545. [PMID: 37932129 PMCID: PMC10982729 DOI: 10.1101/cshperspect.a041545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Cells require micronutrients for numerous basic functions. Among these, iron, copper, and selenium are particularly critical for redox metabolism, and their importance is heightened during oncogene-driven perturbations in cancer. In this review, which particularly focuses on iron, we describe how these micronutrients are carefully chaperoned about the body and made available to tissues, a process that is designed to limit the toxicity of free iron and copper or by-products of selenium metabolism. We delineate perturbations in iron metabolism and iron-dependent proteins that are observed in cancer, and describe the current approaches being used to target iron metabolism and iron-dependent processes.
Collapse
Affiliation(s)
- Erdem M Terzi
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York, New York 10016, USA
| | - Richard Possemato
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York, New York 10016, USA
| |
Collapse
|
47
|
Jiang J, Ruan Y, Liu X, Ma J, Chen H. Ferritinophagy Is Critical for Deoxynivalenol-Induced Liver Injury in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6660-6671. [PMID: 38501926 DOI: 10.1021/acs.jafc.4c00556] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Background: Deoxynivalenol (DON) contamination, pervasive throughout all stages of food production and processing, presents a significant threat to human health. The degradation of ferritin mediated by nuclear receptor coactivator 4 (NCOA4), termed ferritinophagy, plays a crucial role in maintaining iron homeostasis and regulating ferroptosis. Aim: This study aims to elucidate the role of ferritinophagy and ferroptosis in DON-induced liver injury. Methods: Male mice and AML12 cells were subjected to varying doses of DON, serving as in vivo and in vitro models, respectively. Protein expression was assessed by using immunofluorescence and western blot techniques. Co-immunoprecipitation was employed to investigate the protein-protein interactions. Results: Our findings demonstrate that DON triggers hepatocyte ferroptosis in a ferritinophagy-dependent manner. Specifically, DON impedes the activation of the mammalian target of rapamycin complex 1 (mTORC1) by inhibiting RAC1's binding to mTOR, thereby ultimately inducing autophagy. Concurrently, DON amplifies NCOA4's affinity for ferritin by facilitating NCOA4 phosphorylation through the ataxia-telangiectasia mutated kinase (ATM), thus promoting the autophagy-dependent degradation of ferritin. Both autophagy inhibition and NCOA4 expression suppression ameliorate DON-induced ferroptosis. Conclusion: Our study concludes that DON facilitates NCOA4-mediated ferritinophagy via the ATM-NCOA4 pathway, subsequently inducing ferroptosis in the liver.
Collapse
Affiliation(s)
- Junze Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yongbao Ruan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xiaohui Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, P. R. China
| | - Hao Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, P. R. China
| |
Collapse
|
48
|
Cheng C, Hu J, Mannan R, Bhattacharyya R, Rossiter NJ, Magnuson B, Wisniewski JP, Zheng Y, Xiao L, Li C, Awad D, He T, Bao Y, Zhang Y, Cao X, Wang Z, Mehra R, Morlacchi P, Sahai V, di Magliano MP, Shah YM, Ding K, Qiao Y, Lyssiotis CA, Chinnaiyan AM. Targeting PIKfyve-driven lipid homeostasis as a metabolic vulnerability in pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585580. [PMID: 38562800 PMCID: PMC10983929 DOI: 10.1101/2024.03.18.585580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) subsists in a nutrient-deregulated microenvironment, making it particularly susceptible to treatments that interfere with cancer metabolism12. For example, PDAC utilizes and is dependent on high levels of autophagy and other lysosomal processes3-5. Although targeting these pathways has shown potential in preclinical studies, progress has been hampered by the challenge of identifying and characterizing favorable targets for drug development6. Here, we characterize PIKfyve, a lipid kinase integral to lysosomal functioning7, as a novel and targetable vulnerability in PDAC. In human patient and murine PDAC samples, we discovered that PIKFYVE is overexpressed in PDAC cells compared to adjacent normal cells. Employing a genetically engineered mouse model, we established the essential role of PIKfyve in PDAC progression. Further, through comprehensive metabolic analyses, we found that PIKfyve inhibition obligated PDAC to upregulate de novo lipid synthesis, a relationship previously undescribed. PIKfyve inhibition triggered a distinct lipogenic gene expression and metabolic program, creating a dependency on de novo lipid metabolism pathways, by upregulating genes such as FASN and ACACA. In PDAC, the KRAS-MAPK signaling pathway is a primary driver of de novo lipid synthesis, specifically enhancing FASN and ACACA levels. Accordingly, the simultaneous targeting of PIKfyve and KRAS-MAPK resulted in the elimination of tumor burden in a syngeneic orthotopic model and tumor regression in a xenograft model of PDAC. Taken together, these studies suggest that disrupting lipid metabolism through PIKfyve inhibition induces synthetic lethality in conjunction with KRAS-MAPK-directed therapies for PDAC.
Collapse
Affiliation(s)
- Caleb Cheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Jing Hu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PRC
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rupam Bhattacharyya
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas J Rossiter
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Brian Magnuson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jasmine P Wisniewski
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Lanbo Xiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Chungen Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PRC
| | - Dominik Awad
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PRC
| | - Yi Bao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PRC
| | - Rohit Mehra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Vaibhav Sahai
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Marina Pasca di Magliano
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Yatrik M Shah
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, USA
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PRC
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
49
|
Settembre C, Perera RM. Lysosomes as coordinators of cellular catabolism, metabolic signalling and organ physiology. Nat Rev Mol Cell Biol 2024; 25:223-245. [PMID: 38001393 DOI: 10.1038/s41580-023-00676-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/26/2023]
Abstract
Every cell must satisfy basic requirements for nutrient sensing, utilization and recycling through macromolecular breakdown to coordinate programmes for growth, repair and stress adaptation. The lysosome orchestrates these key functions through the synchronised interplay between hydrolytic enzymes, nutrient transporters and signalling factors, which together enable metabolic coordination with other organelles and regulation of specific gene expression programmes. In this Review, we discuss recent findings on lysosome-dependent signalling pathways, focusing on how the lysosome senses nutrient availability through its physical and functional association with mechanistic target of rapamycin complex 1 (mTORC1) and how, in response, the microphthalmia/transcription factor E (MiT/TFE) transcription factors exert feedback regulation on lysosome biogenesis. We also highlight the emerging interactions of lysosomes with other organelles, which contribute to cellular homeostasis. Lastly, we discuss how lysosome dysfunction contributes to diverse disease pathologies and how inherited mutations that compromise lysosomal hydrolysis, transport or signalling components lead to multi-organ disorders with severe metabolic and neurological impact. A deeper comprehension of lysosomal composition and function, at both the cellular and organismal level, may uncover fundamental insights into human physiology and disease.
Collapse
Affiliation(s)
- Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.
| | - Rushika M Perera
- Department of Anatomy, University of California at San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California at San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
50
|
Wang Y, Ding H, Zheng Y, Wei X, Yang X, Wei H, Tian Y, Sun X, Wei W, Ma J, Tian D, Zheng F. Alleviated NCOA4-mediated ferritinophagy protected RA FLSs from ferroptosis in lipopolysaccharide-induced inflammation under hypoxia. Inflamm Res 2024; 73:363-379. [PMID: 38189810 DOI: 10.1007/s00011-023-01842-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/09/2024] Open
Abstract
OBJECTIVE Ferroptosis is a reactive oxygen species (ROS)- and iron-dependent form of non-apoptotic cell death process. Previous studies have demonstrated that ferroptosis participates in the development of inflammatory arthritis. However, the role of ferroptosis in rheumatoid arthritis (RA) inflammatory hypoxic joints remains unclear. This study sought to explore the underlying mechanism of ferroptosis on lipopolysaccharide (LPS)-induced RA fibroblast-like synoviocytes (FLSs). METHODS FLSs, isolated from patients with RA, were treated with LPS and ferroptosis inducer (erastin and RSL-3), and ferroptosis inhibitor (Fer-1 and DFO), respectively. The cell viability was measured by CCK-8. The cell death was detected by flow cytometer. The proteins level were tested by Western blot. The cytosolic ROS and lipid peroxidation were determined using DCFH-DA and C11-BODIPY581/591 fluorescence probes, respectively. The small interfering RNA (siRNA) was used to knock down related proteins. The levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), iron, inflammatory cytokines (IL6 and IL8), and LDH were analyzed by commercial kits. RESULTS Ferroptosis was activated by LPS in RA FLS with increased cellular damage, ROS and lipid peroxidation, intracellular Fe and IL8, which can be further amplified by ferroptosis inducer (erastin and RSL-3) and inhibited by ferroptosis inhibitor (Fer-1 and DFO). Mechanistically, LPS triggered ferroptosis via NCOA4-mediated ferritinophagy in RA FLSs, and knockdown of NCOA4 strikingly prevent the process of ferroptosis. Intriguingly, LPS-induced RA FLSs became insensitive to ferroptosis and NCOA4-mediated ferritinophagy under hypoxia compared with normoxia. Knockdown of HIF-1α reverted ferroptosis and ferritinophagy evoking by LPS-induced RA FLSs inflammation under hypoxia. In addition, low dose of auranofin (AUR) induced re-sensitization of ferroptosis and ferritinophagy through inhibiting the expression of HIF-1α under hypoxia. CONCLUSIONS NCOA4-mediated ferritinophagy was a key driver of ferroptosis in inflammatory RA FLSs. The suppression of NCOA4-mediated ferritinophagy protected RA FLSs from ferroptosis in LPS-induced inflammation under hypoxia. Targeting HIF-1α/NCOA4 and ferroptosis could be an effective and valuable therapeutic strategy for synovium hyperplasia in the patients with RA.
Collapse
Affiliation(s)
- Yang Wang
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
- Department of Clinical Laboratory, Tianjin Hospital, Tianjin University, Tianjin, China
- Department of Clinical Laboratory, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Hongmei Ding
- Department of Clinical Laboratory, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Yuqun Zheng
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xinyue Wei
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
- Department of Clinical Laboratory, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Xiaoting Yang
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Huan Wei
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yanshuang Tian
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xuguo Sun
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Wei Wei
- Department of Rheumatology, General Hospital, Tianjin Medical University, Tianjin, China.
| | - Jun Ma
- Department of Health Statistics, College of Public Health, Tianjin Medical University, Tianjin, China.
| | - Derun Tian
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China.
| | - Fang Zheng
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China.
| |
Collapse
|