1
|
Lei P, Walker T, Ayton S. Neuroferroptosis in health and diseases. Nat Rev Neurosci 2025:10.1038/s41583-025-00930-5. [PMID: 40389615 DOI: 10.1038/s41583-025-00930-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/21/2025]
Abstract
Ferroptosis is a type of cell death process defined by iron-dependent peroxidation of phospholipids leading to the destruction of cellular membranes and death of the cell. Ferroptosis occurs throughout the body, but a considerable research focus on ferroptosis in the brain - neuroferroptosis - has been driven by the rich lipid and iron content of the brain as well as its high oxygen consumption. Neurons also have an exceptionally large surface area and metabolic demand, which necessitates specific mechanisms (such as lipid antioxidants) to engage constantly to protect the plasma membrane against lipid peroxidation. Ferroptosis has been extensively linked to neurodegeneration and ischaemia and is increasingly implicated in physiological processes such as neuronal reprogramming. Astrocytes provide metabolic support to neurons, enabling them to defend against ferroptosis, yet ferroptotic signals in microglia can propagate damage to astrocytes and neurons, highlighting the complex intercellular (patho)physiology of neuroferroptosis.
Collapse
Affiliation(s)
- Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| | - Tara Walker
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Nedaeinia R, Dianat-Moghadam H, Movahednasab M, Khosroabadi Z, Keshavarz M, Amoozgar Z, Salehi R. Therapeutic and prognostic values of ferroptosis signature in glioblastoma. Int Immunopharmacol 2025; 155:114597. [PMID: 40239336 DOI: 10.1016/j.intimp.2025.114597] [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: 01/03/2025] [Revised: 03/15/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025]
Abstract
Ferroptosis is a regulated cell death process that results in decreased tumor growth and aggressiveness when targeted in various cancer cells. Studying the impact of ferroptosis in glioblastoma (GBM) will provide important knowledge about tumor biology and potential treatment strategies. The high metabolic activity resulting in ROS production, iron content and active lipid metabolism of glioblastoma cells make them particularly susceptible to ferroptosis. Single-cell RNA sequencing reveals the molecular signature of GBM and its tumor microenvironment, introducing ferroptosis-related biomarkers pathways and drug resistance mechanisms to enhance treatment outcomes for GBM patients. The relationship between ferroptosis and the immune landscape in GBM is complex and can have either positive or negative effects. These effects can be identified through single-cell RNA sequencing to develop targeted chemo-, radio- and immuno- therapies against glioma stem cells and tumor-supportive immune cells. Additionally, the implication of oncolytic virotherapy in combination with ferroptosis induction can lead to improved treatment of GBM in a clinical setting.
Collapse
Affiliation(s)
- Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Dianat-Moghadam
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maedeh Movahednasab
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Khosroabadi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
3
|
Hu S, Tang S, Liu D, Xia R, Wang X. Walnut oil as a dietary intervention for atherosclerosis: Efficacy and mechanistic pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159607. [PMID: 40064415 DOI: 10.1016/j.bbalip.2025.159607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND AIMS Walnut oil (WO) and peanut oil (PO) are common vegetable oils rich in unsaturated fatty acids, known to alleviate atherosclerosis (AS) and reduce the risk of cardiovascular diseases (CVD). WO contains a higher proportion of polyunsaturated fatty acids (PUFAs) compared to PO. This study aimed to explore the influence of WO on AS and elucidate its potential mechanisms, providing a theoretical basis for enhancing the application of WO in functional foods and pharmaceuticals. METHODS AS was established in rats using a high-fat diet and vitamin D3 injections. Rats with AS were administered WO or PO via gavage at a dose of 1.2 g/kg for 4 weeks. Serum lipid levels and arterial injury were assessed, and transcriptomic and metabolomic analyses of the rat vasculature were performed. RESULTS Both WO and PO significantly lowered serum lipid levels and the atherogenic index (AI) in rats, reducing arterial wall injury and plaque formation. WO exhibited a more pronounced effect, particularly in decreasing serum levels of TG, TC, HDLC, and LDL-C. Transcriptomic analysis indicated that fatty acid, amino acid metabolism were crucial in AS development due to a high-fat diet. Metabolomic analysis indicated significant changes in the metabolism of arginine, proline, cysteine, methionine, glycine, serine and threonine in rats treated with WO. CONCLUSION WO and PO help alleviate AS by regulating lipid metabolism and influencing pivotal metabolic pathways like TCA cycle and cysteine-methionine metabolism. The more significant impact of WO indicates its potential as a dietary supplement for preventing and treating AS.
Collapse
Affiliation(s)
- Shujuan Hu
- School of Education and Physical Education, Yangtze University, Jingzhou, Hubei 434023, China
| | - Si Tang
- Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Dang Liu
- Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Ruohan Xia
- Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Xianwang Wang
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China; Shannan Maternal and Child Health Hospital, Shannan, Xizang 856100, China.
| |
Collapse
|
4
|
Shan G, Bian Y, Ren S, Hu Z, Pan B, Zeng D, Zheng Z, Fan H, Bi G, Yao G, Zhan C. Sarcosine sensitizes lung adenocarcinoma to chemotherapy by dual activation of ferroptosis via PDK4/PDHA1 signaling and NMDAR-mediated iron export. Exp Hematol Oncol 2025; 14:60. [PMID: 40275333 PMCID: PMC12023509 DOI: 10.1186/s40164-025-00657-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Ferroptosis, a regulated cell death driven by iron-dependent lipid peroxidation, is associated with chemoresistance in lung adenocarcinoma (LUAD). This study aims to investigate the role of sarcosine in ferroptosis and its underlying mechanisms. METHODS An RSL3-induced ferroptosis model was used to screen a library of 889 human endogenous metabolites and metabolomic profiling was harnessed to identify metabolites associated with ferroptosis. Cell viability, lipid-reactive oxygen species (ROS), ferrous iron, malondialdehyde (MDA), and mitochondrial integrity were assessed to evaluate sarcosine's effects on ferroptosis. Metabolic fate was studied using 15N-labeled sarcosine. Next, we used untargeted metabolomic profiling and next-generation sequencing to dissect metabolic and transcriptomic changes upon sarcosine supplementation. The effects of sarcosine on ferroptosis and chemotherapy were further validated in patient-derived organoids (PDOs), xenograft models, and LUAD tissues. RESULTS Sarcosine emerged as a potent ferroptosis inducer in the metabolic library screening, which was further confirmed via cell viability, lipid-ROS, ferrous iron, and MDA measurements. Metabolic flux analysis showed limited conversion of sarcosine to other metabolites in LUAD cells, while untargeted metabolomic profiling and seahorse assays indicated a metabolic shift from glycolysis to oxidative phosphorylation. Sarcosine enhanced pyruvate dehydrogenase activity to generate more ROS by interacting with PDK4, reducing PDHA1 phosphorylation. As a co-activator of N-methyl-D-aspartate receptor (NMDAR), sarcosine also exerted its pro-ferroptosis effect via regulating ferrous export through the NMDAR/MXD3/SLC40A1 axis. Given the significance of ferroptosis in chemotherapy, we validated that sarcosine enhanced the sensitization of cisplatin by promoting ferroptosis in LUAD cells, PDOs, and xenograft models. CONCLUSION Sarcosine promotes ferroptosis and enhances chemosensitivity, suggesting its potential as a therapeutic agent in treating LUAD.
Collapse
Affiliation(s)
- Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Shencheng Ren
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Binyang Pan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Dejun Zeng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Zhaolin Zheng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), No. 668 Jinhu Road, Huli District, Xiamen, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China.
| | - Guangyu Yao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China.
| |
Collapse
|
5
|
Sahu U, Mullarkey MP, Murphy SA, Anderson JC, Putluri V, Kamal AHM, Park JH, Lee TJ, Ling AL, Kaipparettu BA, Sharma A, Putluri N, Wenzel PL, Willey CD, Chiocca EA, Markert JM, Kaur B. IDH status dictates oHSV mediated metabolic reprogramming affecting anti-tumor immunity. Nat Commun 2025; 16:3874. [PMID: 40274791 PMCID: PMC12022073 DOI: 10.1038/s41467-025-58911-2] [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: 07/03/2024] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Identification of isocitrate dehydrogenase (IDH) mutations has uncovered the crucial role of metabolism in gliomagenesis. Oncolytic herpes virus (oHSV) initiates direct tumor debulking by tumor lysis and activates anti-tumor immunity, however, little is known about the role of glioma metabolism in determining oHSV efficacy. Here we identify that oHSV rewires central carbon metabolism increasing glucose utilization towards oxidative phosphorylation and shuttling glutamine towards reductive carboxylation in IDH wildtype glioma. The switch in metabolism results in increased lipid synthesis and cellular ROS. PKC induces ACSL4 in oHSV treated cells leading to lipid peroxidation and ferroptosis. Ferroptosis is critical to launch an anti-tumor immune response which is important for viral efficacy. Mutant IDH (IDHR132H) gliomas are incapable of reductive carboxylation and hence ferroptosis. Pharmacological blockade of IDHR132H induces ferroptosis and anti-tumor immunity. This study provides a rationale to use an IDHR132H inhibitor to treat high grade IDH-mutant glioma patients undergoing oHSV treatment.
Collapse
Affiliation(s)
- Upasana Sahu
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA, USA.
- Georgia Cancer Center at Augusta University, Augusta, GA, USA.
| | - Matthew P Mullarkey
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sara A Murphy
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Georgia Cancer Center at Augusta University, Augusta, GA, USA
- University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Joshua C Anderson
- Department of Radiation Oncology, Marnix E. Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vasanta Putluri
- Advanced Technology Cores, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Abu Hena Mostafa Kamal
- Advanced Technology Cores, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jun Hyoung Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Tae Jin Lee
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Alexander L Ling
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Benny A Kaipparettu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Nagireddy Putluri
- Advanced Technology Cores, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Pamela L Wenzel
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christopher D Willey
- Department of Radiation Oncology, Marnix E. Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA
| | - James M Markert
- Department of Neurosurgery, Marnix E. Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Balveen Kaur
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA, USA.
- Georgia Cancer Center at Augusta University, Augusta, GA, USA.
| |
Collapse
|
6
|
Kolligundla LP, Sullivan KM, Mukhi D, Andrade-Silva M, Liu H, Guan Y, Gu X, Wu J, Doke T, Hirohama D, Guarnieri P, Hill J, Pullen SS, Kuo J, Inamoto M, Susztak K. Glutathione-specific gamma-glutamylcyclotransferase 1 ( CHAC1) increases kidney disease risk by modulating ferroptosis. Sci Transl Med 2025; 17:eadn3079. [PMID: 40267214 DOI: 10.1126/scitranslmed.adn3079] [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: 12/03/2023] [Revised: 08/20/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Genome-wide association studies (GWASs) have identified more than 1000 loci where genetic variants correlate with kidney function. However, the specific genes, cell types, and mechanisms influenced by these genetic variants remain largely uncharted. Here, we identified glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) on chromosome 15 as affected by GWAS variants by analyzing human kidney gene expression and methylation information. Both CHAC1 RNA and protein were expressed in the loop of Henle region in mouse and human kidneys, and CHAC1 expression was higher in patients carrying disease risk variants. Using CRISPR technology, we created mice with a single functional copy of the Chac1 gene (Chac1+/-) that displayed no baseline phenotypic alterations in kidney structure or function. These mice demonstrated resilience to kidney disease in multiple models, including folic acid-induced nephropathy, adenine-induced chronic kidney disease, and uninephrectomy-streptozotocin-induced diabetic nephropathy. We further showed that CHAC1 plays a critical role in degrading the cellular antioxidant glutathione. Tubule cells isolated from Chac1+/- mice showed increased glutathione, decreased lipid peroxidation, improved cell viability, and protection against ferroptosis. Expression of ferroptosis-associated genes was also lower in mice with only one copy of Chac1. Higher CHAC1 protein also correlated with ferroptosis-related protein abundance in kidney biopsies from patients with kidney disease. This study positions CHAC1 as an important mediator of kidney disease that influences glutathione concentrations and ferroptosis, suggesting potential avenues to explore for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Lakshmi P Kolligundla
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Katie M Sullivan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Pediatrics, Medical College of Wisconsin Pediatric Nephrology, Milwaukee, WI 53226, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Magaiver Andrade-Silva
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Yuting Guan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Xiangchen Gu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Junnan Wu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Daigoro Hirohama
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Paolo Guarnieri
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jon Hill
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jay Kuo
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | | | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| |
Collapse
|
7
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
8
|
Li Y, Zhang M, Zhang K, Niu H, Li H, Wu W. Ginsenosides modulate immunity via TLR4/MyD88/NF-κB pathway and gut microbiota. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156763. [PMID: 40252438 DOI: 10.1016/j.phymed.2025.156763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Ginsenosides, the primary active compounds in Panax ginseng C. A. Mey., are well known for their potent immunomodulatory effects. However, their precise mechanisms, particularly concerning the "intestinal-metabolism-immune axis", have yet to be fully elucidated. PURPOSE This study aims to investigate how ginsenosides protect immune function through the regulation of the gut-metabolism-immune axis. STUDY DESIGN A CTX-induced immunodeficient mouse model was established to assess the effects of ginsenosides on immune function, gut microbiota, and metabolic pathways. METHODS The immune organ indices (spleen and thymus), levels of immune cytokines (TNF-α, IFN-γ, IL-6, IL-1β), and immunoglobulins (IgM, IgA) were assessed. Intestinal microbial diversity was analyzed using 16S rRNA sequencing, and metabolomics was employed to identify disruptions in amino acid and lipid metabolic pathways. Spearman correlation analysis and Western blotting were conducted to explore the involvement of the TLR4/MyD88/NF-κB signaling pathway. RESULTS Ginsenosides significantly restored immune organ indices and enhanced cytokines and immunoglobulins. 16S rRNA sequencing revealed an increase in probiotic levels and a reduction in potentially harmful bacteria, thereby enhancing intestinal microbiota diversity. Metabolomics analysis showed that ginsenosides ameliorated CTX-induced metabolic disorders and stimulated the production of short-chain fatty acids (SCFAs) and bile acids. Western blot analysis confirmed the upregulation of TLR4, MyD88, and NF-κB p-p65 expression. CONCLUSION This study systematically elucidates the mechanism by which ginsenosides enhance immune function by regulating gut microbiota, restoring metabolic balance, and activating the TLR4/MyD88/NF-κB signaling pathway. These findings provide a molecular foundation for the potential use of ginsenosides in the prevention and treatment of immune-related diseases.
Collapse
Affiliation(s)
- Ying Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China; Shandong Academy of Chinese Medicine, Jinan, Shandong, 250014, China
| | - Meng Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Kaiyue Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Huazhou Niu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Hui Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
| | - Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
| |
Collapse
|
9
|
Lee WC, Dixon SJ. Mechanisms of ferroptosis sensitization and resistance. Dev Cell 2025; 60:982-993. [PMID: 40199240 DOI: 10.1016/j.devcel.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/08/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis is an iron-dependent and oxidative form of non-apoptotic cell death with roles in development, homeostasis, and disease. Ferroptosis sensitivity can vary between cells, often for reasons that are not well understood. In this perspective, we describe the core ferroptosis mechanism and outline how changes in iron, redox, and lipid metabolism can alter ferroptosis sensitivity. We propose the concept of a ferroptosis sensitivity-resistance continuum to describe how different intrinsic and extrinsic factors interact to push cells toward a more ferroptosis-sensitive or ferroptosis-resistant state, with effects on development and diseases such as cancer.
Collapse
Affiliation(s)
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
10
|
Fan Y, Wang Y, Dan W, Zhang Y, Nie L, Ma Z, Zhuang Y, Liu B, Li M, Liu T, Wang Z, Ye L, Wei Y, Lei Y, Guo C, An J, Wang C, Zhang Y, Zeng J, Wei W, Gan B, Li L. PRMT5-mediated arginine methylation stabilizes GPX4 to suppress ferroptosis in cancer. Nat Cell Biol 2025; 27:641-653. [PMID: 40033101 DOI: 10.1038/s41556-025-01610-3] [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: 05/03/2024] [Accepted: 01/03/2025] [Indexed: 03/05/2025]
Abstract
The activation of ferroptosis has shown great potential for cancer therapy from an unconventional perspective, but revealing the mechanisms underlying the suppression of tumour-intrinsic ferroptosis to promote tumorigenesis remains a challenging task. Here we report that methionine is metabolized into S-adenosylmethionine, which functions as a methyl-group donor to trigger symmetric dimethylation of glutathione peroxidase 4 (GPX4) at the conserved arginine 152 (R152) residue, along with a prolonged GPX4 half-life. Inhibition of protein arginine methyltransferase 5 (PRMT5), which catalyses GPX4 methylation, decreases GPX4 protein levels by impeding GPX4 methylation and increasing ferroptosis inducer sensitivity in vitro and in vivo. This methylation prevents Cullin1-FBW7 E3 ligase binding to GPX4, thereby abrogating the ubiquitination-mediated GPX4 degradation. Notably, combining PRMT5 inhibitor treatment with ferroptotic therapies markedly suppresses tumour progression in mouse tumour models. In addition, the levels of GPX4 are negatively correlated with the levels of FBW7 and a poor prognosis in patients with human carcinoma. In summary, we found that PRMT5 functions as a target for improving cancer therapy efficacy, by acting to reduce the counteraction of ferroptosis by tumour cells by means of PRMT5-enhanced GPX4 stability.
Collapse
Affiliation(s)
- Yizeng Fan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yuzhao Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Weichao Dan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yilei Zhang
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Li Nie
- State Key Laboratory for Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Zhiqiang Ma
- Department of Medical Oncology, Senior Department of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, Beijing, China
| | - Yanxin Zhuang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Bo Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Mengxing Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Zixi Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Leihong Ye
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yi Wei
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yuzeshi Lei
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Chendong Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Jiale An
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Chi Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yulin Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China.
| |
Collapse
|
11
|
Koizume S, Miyagi Y. Adaptation mechanisms in cancer: Lipid metabolism under hypoxia and nutrient deprivation as a target for novel therapeutic strategies (Review). Mol Med Rep 2025; 31:83. [PMID: 39886950 PMCID: PMC11799873 DOI: 10.3892/mmr.2025.13448] [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: 10/30/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025] Open
Abstract
Tumor tissues generally exist in a relatively hypovascular state, and cancer cells must adapt to severe tissue conditions with a limited molecular oxygen and nutrient supply for their survival. Lipid metabolism serves a role in this adaptation. Lipids are supplied not only through the bloodstream but also through autonomous synthesis by cancer cells, and they function as sources of adenosine triphosphate and cell components. Although cancer‑associated lipid metabolism has been widely reviewed, how this metabolism responds to the tumor environment with poor molecular oxygen and nutrient supply remains to be fully discussed. The main aim of the present review was to summarize the findings on this issue and to provide insights into how cancer cells adapt to better cope with metabolic stresses within tumors. It may be suggested that diverse types of lipid metabolism have a role in enabling cancer cells to adapt to both hypoxia and nutrient‑poor conditions. Gaining a deeper understanding of these molecular mechanisms may reveal novel possibilities of exploration for cancer treatment.
Collapse
Affiliation(s)
- Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa 241-8515, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, Kanagawa 241-8515, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa 241-8515, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, Kanagawa 241-8515, Japan
| |
Collapse
|
12
|
Rowland EC, D'Antuono M, Jermakowicz AM, Ayad NG. Methionine cycle inhibition disrupts antioxidant metabolism and reduces glioblastoma cell survival. J Biol Chem 2025; 301:108349. [PMID: 40015640 PMCID: PMC11994328 DOI: 10.1016/j.jbc.2025.108349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/04/2025] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
Glioblastoma (GBM) is a highly aggressive primary malignant adult brain tumor that inevitably recurs with a fatal prognosis. This is due in part to metabolic reprogramming that allows tumors to evade treatment. Therefore, we must uncover the pathways mediating these adaptations to develop novel and effective treatments. We searched for genes that are essential in GBM cells as measured by a whole-genome pan-cancer CRISPR screen available from DepMap and identified the methionine metabolism genes MAT2A and AHCY. We conducted genetic knockdown, evaluated mitochondrial respiration, and performed targeted metabolomics to study the function of these genes in GBM. We demonstrate that MAT2A or AHCY knockdown induces oxidative stress, hinders cellular respiration, and reduces the survival of GBM cells. Furthermore, selective MAT2a or AHCY inhibition reduces GBM cell viability, impairs oxidative metabolism, and shifts the cellular metabolic profile towards oxidative stress and cell death. Mechanistically, MAT2a and AHCY regulate spare respiratory capacity, the redox buffer cystathionine, lipid and amino acid metabolism, and prevent oxidative damage in GBM cells. Our results point to the methionine metabolic pathway as a novel vulnerability point in GBM.
Collapse
Affiliation(s)
- Emma C Rowland
- Georgetown University, Lombardi Comprehensive Cancer Center, Washington, District of Columbia, USA
| | - Matthew D'Antuono
- Georgetown University, Lombardi Comprehensive Cancer Center, Washington, District of Columbia, USA
| | - Anna M Jermakowicz
- Georgetown University, Lombardi Comprehensive Cancer Center, Washington, District of Columbia, USA
| | - Nagi G Ayad
- Georgetown University, Lombardi Comprehensive Cancer Center, Washington, District of Columbia, USA.
| |
Collapse
|
13
|
Goldberg AR, Dovas A, Torres D, Pereira B, Viswanathan A, Das Sharma S, Mela A, Merricks EM, Megino-Luque C, McInvale JJ, Olabarria M, Shokooh LA, Zhao HT, Chen C, Kotidis C, Calvaresi P, Banu MA, Razavilar A, Sudhakar TD, Saxena A, Chokran C, Humala N, Mahajan A, Xu W, Metz JB, Bushong EA, Boassa D, Ellisman MH, Hillman EMC, Hargus G, Bravo-Cordero JJ, McKhann GM, Gill BJA, Rosenfeld SS, Schevon CA, Bruce JN, Sims PA, Peterka DS, Canoll P. Glioma-induced alterations in excitatory neurons are reversed by mTOR inhibition. Neuron 2025; 113:858-875.e10. [PMID: 39837324 PMCID: PMC11925689 DOI: 10.1016/j.neuron.2024.12.026] [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] [Revised: 09/27/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025]
Abstract
Gliomas are aggressive neoplasms that diffusely infiltrate the brain and cause neurological symptoms, including cognitive deficits and seizures. Increased mTOR signaling has been implicated in glioma-induced neuronal hyperexcitability, but the molecular and functional consequences have not been identified. Here, we show three types of changes in tumor-associated neurons: (1) downregulation of transcripts encoding excitatory and inhibitory postsynaptic proteins and dendritic spine development and upregulation of cytoskeletal transcripts via neuron-specific profiling of ribosome-bound mRNA, (2) marked decreases in dendritic spine density via light and electron microscopy, and (3) progressive functional alterations leading to neuronal hyperexcitability via in vivo calcium imaging. A single acute dose of AZD8055, a combined mTORC1/2 inhibitor, reversed these tumor-induced changes. These findings reveal mTOR-driven pathological plasticity in neurons at the infiltrative margin of glioma and suggest new strategies for treating glioma-associated neurological symptoms.
Collapse
Affiliation(s)
- Alexander R Goldberg
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Daniela Torres
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brianna Pereira
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ashwin Viswanathan
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sohani Das Sharma
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Edward M Merricks
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cristina Megino-Luque
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10027, USA
| | - Julie J McInvale
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Markel Olabarria
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | - Hanzhi T Zhao
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Cady Chen
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Corina Kotidis
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter Calvaresi
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Matei A Banu
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aida Razavilar
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tejaswi D Sudhakar
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ankita Saxena
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cole Chokran
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nelson Humala
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aayushi Mahajan
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Weihao Xu
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Jordan B Metz
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Eric A Bushong
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniela Boassa
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Elizabeth M C Hillman
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10027, USA
| | - Guy M McKhann
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brian J A Gill
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | - Catherine A Schevon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Darcy S Peterka
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
14
|
Srivastava S, Anbiaee R, Houshyari M, Laxmi, Sridhar SB, Ashique S, Hussain S, Kumar S, Taj T, Akbarnejad Z, Taghizadeh-Hesary F. Amino acid metabolism in glioblastoma pathogenesis, immune evasion, and treatment resistance. Cancer Cell Int 2025; 25:89. [PMID: 40082966 PMCID: PMC11908050 DOI: 10.1186/s12935-025-03721-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025] Open
Abstract
Glioblastoma (GBM) ranks among the most lethal primary tumors of the central nervous system. This is partly due to its complex intracellular metabolism and interactions with the surrounding tumor microenvironment (TME). Compelling evidence represents that altered amino acids (AAs) metabolism plays a crucial role in both areas. The role of AAs and their metabolites in glioma biology is an emerging topic. Therefore, this review was conducted to summarize the current knowledge about the molecular mechanisms by which AAs participate in the GBM pathogenesis. AAs can directly influence tumor progression by affecting tumor cell metabolism or indirectly by releasing bioactive agents through particular metabolic pathways. This review begins by examining the metabolic pathways of essential AAs, such as tryptophan, tyrosine, and phenylalanine, which contribute to synthesizing critical neurotransmitters and shape tumor metabolism signatures. We explore how these pathways impact tumor growth and immune modulation, focusing on how AAs and their metabolites can promote malignant properties in GBM cells. AAs also play a pivotal role in reprogramming the TME, contributing to immune evasion and resistance to therapy. The review further discusses how tumor metabolism signatures, influenced by AA metabolism, can enhance the immunosuppressive microenvironment, providing new avenues for targeted immunotherapies. Finally, we outline potential therapeutic strategies to modulate AA metabolism and emphasize critical opportunities for future research to improve GBM management.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, 203201, India
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi, 110017, India
| | - Robab Anbiaee
- Radio Oncology Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Houshyari
- Radio Oncology Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Laxmi
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, 203201, India
| | | | - Sumel Ashique
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, 711316, West Bengal, India
| | - Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, Uttarakhand, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi, 110017, India
| | - Tahreen Taj
- Department of Pharmacology, Yenepoya Pharmacy college and research centre, Yenepoya (Deemed to be) university, Mangalore, 575018, India
| | - Zeinab Akbarnejad
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Clinical Oncology Department, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Keane L, Cryan JF, Gleeson JP. Exploiting the gut microbiome for brain tumour treatment. Trends Mol Med 2025; 31:213-223. [PMID: 39256110 DOI: 10.1016/j.molmed.2024.08.008] [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: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Increasing evidence suggests that the gut microbiome plays a key role in a host of pathological conditions, including cancer. Indeed, the bidirectional communication that occurs between the gut and the brain, known as the 'gut-brain axis,' has recently been implicated in brain tumour pathology. Here, we focus on current research that supports a gut microbiome-brain tumour link with emphasis on high-grade gliomas, the most aggressive of all brain tumours, and the impact on the glioma tumour microenvironment. We discuss the potential use of gut-brain axis signals to improve responses to current and future therapeutic approaches. We highlight that the success of novel treatment strategies may rely on patient-specific microbiome profiles, and these should be considered for personalised treatment approaches.
Collapse
Affiliation(s)
- Lily Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Jack P Gleeson
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland; CUH/UCC Cancer Centre, Cork University Hospital, Cork, Ireland.
| |
Collapse
|
16
|
Teng Y, Huang Y, Tao X, Fan Y, You J. Emerging role of ferroptosis in ultraviolet radiation-driven skin photoaging: a narrative review. Photochem Photobiol Sci 2025; 24:531-542. [PMID: 40063311 DOI: 10.1007/s43630-025-00691-1] [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: 12/06/2024] [Accepted: 02/05/2025] [Indexed: 04/05/2025]
Abstract
Photoaging is characterized by chronic inflammation in response to ultraviolet (UV) radiation. UV radiation causes skin cells to produce reactive oxygen species (ROS), which causes oxidative stress and inflammation. ROS can reversibly or irreversibly destroy different cellular compounds, including nucleic acids, proteins, free amino acids, lipids, lipoproteins, carbohydrates, and connective tissue macromolecules. Ferroptosis is a kind of programmed cell death caused by iron dependence and lipid peroxidation and has been recently discovered. Its occurrence is primarily related to iron metabolism, antioxidants, lipid peroxidation, and other processes. In addition, high levels of ROS can trigger oxidative stress, altering the redox balance within cells and thus initiating ferroptosis. Ferroptosis has been implicated in UV-driven skin photoaging. Moreover, UV radiation from sunlight can regulate numerous ferroptosis-linked genes. This review will focus on the function of ferroptosis in UV radiation-damaged skin cells. We hope to draw attention to the significance of ferroptosis regulation in the prevention and treatment of skin photoaging.
Collapse
Affiliation(s)
- Yan Teng
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Youming Huang
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Xiaohua Tao
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Yibin Fan
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Jianhua You
- Dermatology Hospital of Zhejiang Province, No. 61 Wuyuan Road, Wukang Street, Deqing, Huzhou, 313200, Zhejiang, China.
| |
Collapse
|
17
|
Hu C, Xu J, Zhang Y, Zhang R, Pan S, Chen J, Wang Y, Zhao Q, Wang Y, Zhu W, Cao M, Zhang S, Zu D, Xu Z, Jing J, Cheng X. Inhibition of glutathione peroxidase 4 suppresses gastric cancer peritoneal metastasis via regulation of RCC2 homeostasis. Redox Biol 2025; 80:103519. [PMID: 39908861 PMCID: PMC11847474 DOI: 10.1016/j.redox.2025.103519] [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: 01/12/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Gastric cancer (GC) is one of the most lethal malignancies due to high metastatic rate, making the identification of new therapeutic targets critical for developing effective anti-GC treatments. Glutathione peroxidase 4 (GPx4), a key regulator of ferroptosis and redox homeostasis, contributes to progression and influences patient survival. However, the molecular mechanism by which GPx4 drives GC progression has not been fully illuminated. In this study, we found that GPx4 was overexpressed and negatively associated with poor prognosis and distant metastasis, as confirmed by single-cell RNA sequencing (scRNA-seq) and validation with retrospective clinical samples. GPx4 knockdown suppressed GC invasion, migration and peritoneal metastasis in vitro and in vivo. Proteomic analysis revealed that GPx4 expression regulated the Homeostasis of RCC2, an oncogene link to epithelial-mesenchymal transition (EMT). Furthermore, we demonstrated that the reactive oxygen species (ROS) accumulation induced by GPx4 inhibition or knockdown activated aurora A phosphorylation, leading to RCC2 ubiquitination and degradation, thereby suppressing peritoneal metastasis in GC. We also identified that the Thr418 phosphorylation site is crucial for RCC2 ubiquitination at the K377, initiating its degradation in response to ROS. In conclusion, our results indicate that GPx4 acts as an oncogene in GC, and that suppressing GPx4 prevents GC progression and metastasis by promoting ROS-induced RCC2 ubiquitination and degradation.
Collapse
Affiliation(s)
- Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Jingli Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Ruolan Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Siwei Pan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Jiahui Chen
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Yan Wang
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Qianyu Zhao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Yuqi Wang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Weiwei Zhu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Mengxuan Cao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Shengjie Zhang
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Dan Zu
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Ji Jing
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| |
Collapse
|
18
|
Jiang P, Chipurupalli S, Yoo BH, Liu X, Rosen KV. Inactivation of necroptosis-promoting protein MLKL creates a therapeutic vulnerability in colorectal cancer cells. Cell Death Dis 2025; 16:118. [PMID: 39979285 PMCID: PMC11842741 DOI: 10.1038/s41419-025-07436-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
Mortality from colorectal cancer (CRC) is significant, and novel CRC therapies are needed. A pseudokinase MLKL typically executes necroptotic cell death, and MLKL inactivation protects cells from such death. However, we found unexpectedly that MLKL gene knockout enhanced CRC cell death caused by a protein synthesis inhibitor homoharringtonine used for chronic myeloid leukemia treatment. In an effort to explain this finding, we observed that MLKL gene knockout reduces the basal CRC cell autophagy and renders such autophagy critically dependent on the presence of VPS37A, a component of the ESCRT-I complex. We further found that the reason why homoharringtonine enhances CRC cell death caused by MLKL gene knockout is that homoharringtonine activates p38 MAP kinase and thereby prevents VPS37A from supporting autophagy in MLKL-deficient cells. We observed that the resulting inhibition of the basal autophagy in CRC cells triggers their parthanatos, a cell death type driven by poly(ADP-ribose) polymerase hyperactivation. Finally, we discovered that a pharmacological MLKL inhibitor necrosulfonamide strongly cooperates with homoharringtonine in suppressing CRC cell tumorigenicity in mice. Thus, while MLKL promotes cell death during necroptosis, MLKL supports the basal autophagy in CRC cells and thereby protects them from death. MLKL inactivation reduces such autophagy and renders the cells sensitive to autophagy inhibitors, such as homoharringtonine. Hence, MLKL inhibition creates a therapeutic vulnerability that could be utilized for CRC treatment.
Collapse
Affiliation(s)
- Peijia Jiang
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Sandhya Chipurupalli
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Byong Hoon Yoo
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Xiaoyang Liu
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Kirill V Rosen
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
19
|
Shen X, Chen Y, Tang Y, Lu P, Liu M, Mao T, Weng Y, Yu F, Liu Y, Tang Y, Wang L, Niu N, Xue J. Targeting pancreatic cancer glutamine dependency confers vulnerability to GPX4-dependent ferroptosis. Cell Rep Med 2025; 6:101928. [PMID: 39879992 PMCID: PMC11866519 DOI: 10.1016/j.xcrm.2025.101928] [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: 01/30/2024] [Revised: 07/17/2024] [Accepted: 01/02/2025] [Indexed: 01/31/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) relies heavily on glutamine (Gln) utilization to meet its metabolic and biosynthetic needs. How epigenetic regulators contribute to the metabolic flexibility and PDAC's response and adaptation to Gln scarcity in the tumor milieu remains largely unknown. Here, we elucidate that prolonged Gln restriction or treatment with the Gln antagonist, 6-diazo-5-oxo-L-norleucine (DON), leads to growth inhibition and ferroptosis program activation in PDAC. A CRISPR-Cas9 screen identifies an epigenetic regulator, Paxip1, which promotes H3K4me3 upregulation and Hmox1 transcription upon DON treatment. Additionally, ferroptosis-related repressors (e.g., Slc7a11 and Gpx4) are increased as an adaptive response, thereby predisposing PDAC cells to ferroptosis upon Gln deprivation. Moreover, DON sensitizes PDAC cells to GPX4 inhibitor-induced ferroptosis, both in vitro and in patient-derived xenografts (PDXs). Taken together, our findings reveal that targeting Gln dependency confers susceptibility to GPX4-dependent ferroptosis via epigenetic remodeling and provides a combination strategy for PDAC therapy.
Collapse
Affiliation(s)
- Xuqing Shen
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueyue Chen
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Tang
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Lu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Liu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiebo Mao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yawen Weng
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feier Yu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimei Liu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujie Tang
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, China.
| | - Liwei Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ningning Niu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jing Xue
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
20
|
Huang S, Sun J, Shen C, He G. Dietary and nutritional interventions for human diseases: their modulatory effects on ferroptosis. Food Funct 2025; 16:1186-1204. [PMID: 39866046 DOI: 10.1039/d4fo05606j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
A balanced diet is essential for maintaining human health. Increasing evidence suggests that dietary and nutritional interventions contribute to disease management and are associated with reduced healthcare costs and economic burden. Ferroptosis, a novel type of regulated cell death (RCD) driven by lipid peroxidation, has been shown to be involved in various pathological conditions, including diabetes, ischemia/reperfusion (I/R) injury, inflammation-related diseases, and cancer. Therefore, specifically targeting the uncontrolled ferroptosis process may offer new therapeutic opportunities. Of note, certain interventions, such as small-molecule compounds, natural products, herbal medicines, and non-pharmacological approaches, have been reported to prevent and treat multiple human diseases by reversing the dysregulation of ferroptosis. In this review, we present the key molecular mechanisms that regulate ferroptosis. Importantly, interventions targeting ferroptosis are summarized from the perspective of dietary patterns, food and nutrients. By understanding these advances, innovative ideas can be provided for individualized dietary interventions and treatment strategies.
Collapse
Affiliation(s)
- Shiqiong Huang
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha 410000, China.
| | - Ji Sun
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha 410000, China.
| | - Chaozan Shen
- Department of Clinical Pharmacy, The Second People's Hospital of Huaihua, Huaihua 418000, China.
| | - Gefei He
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha 410000, China.
| |
Collapse
|
21
|
Zheng X, Diao M, Tong S, Yang S, Lin J, Zhuo S, Wang T, Dai J, Chen S, Wang K. Global research landscape and hotspots for ferroptosis in glioma: A comprehensive bibliometric and visual analysis. Heliyon 2025; 11:e42242. [PMID: 39931474 PMCID: PMC11808509 DOI: 10.1016/j.heliyon.2025.e42242] [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: 05/20/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025] Open
Abstract
Studying ferroptosis is crucial for understanding the mechanisms underlying the onset and progression of glioblastoma, identifying therapeutic targets, and improving prognosis assessment and diagnostic methods. While recent research has explored the link between ferroptosis and glioblastoma, there is a lack of comprehensive bibliometric analyses specifically addressing this relationship and its connection to glioblastoma. To address this gap, we conducted a thorough analysis of 225 relevant articles on glioma and ferroptosis obtained from the Web of Science database covering the period from 2012 to 2023, employing rigorous exclusion criteria. Visual and statistical analyses were performed using CiteSpace, VOSviewer, R Studio Plotting, and Scimago Graphica Beta. Our findings revealed a significant exponential growth in the number of studies during the last decade. China, the United States, and Germany made the most substantial contributions to research in this field, collectively accounting for 76.2 % of the total research output. Notably, Central South University, Shandong University, and Zhejiang University emerged as leaders in both literature production and research collaboration. Frontiers in Oncology stood out as the most prolific journal, encompassing a wide array of topics from molecular mechanisms to potential therapeutic strategies. Visual keyword analysis highlighted "tumor biology" "cell death mechanisms" and "gene expression and metabolic processes" as central themes in the research network. This study offers a comprehensive visual perspective on the global publication landscape of ferroptosis in glioma, providing valuable insights for researchers seeking to understand the current state of the field and identify potential directions for future studies.
Collapse
Affiliation(s)
- Xinyue Zheng
- School of Basic Medicine and Life Sciences, Key Laboratory of Tropical Translational Medicine of Ministry of Education, International Center for Aging and Cancer, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Mengyao Diao
- School of Basic Medicine and Life Sciences, Key Laboratory of Tropical Translational Medicine of Ministry of Education, International Center for Aging and Cancer, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Shan Tong
- Center of Geriatrics, the Hainan Affiliated Hospital (Hainan General Hospital), Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Shuo Yang
- School of Basic Medicine and Life Sciences, Key Laboratory of Tropical Translational Medicine of Ministry of Education, International Center for Aging and Cancer, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Jing Lin
- School of Basic Medicine and Life Sciences, Key Laboratory of Tropical Translational Medicine of Ministry of Education, International Center for Aging and Cancer, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Shenghua Zhuo
- International Center for Aging and Cancer, Department of Neurosurgery, the First Affiliated Hospital, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Ting Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jian Dai
- School of Basic Medicine and Life Sciences, Key Laboratory of Tropical Translational Medicine of Ministry of Education, International Center for Aging and Cancer, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Shenbo Chen
- International Center for Aging and Cancer, Department of Neurosurgery, the First Affiliated Hospital, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Kai Wang
- International Center for Aging and Cancer, Department of Hematology, Department of Spine Surgery (Hainan Province Spinal Surgery Clinical Medical Center), the First Affiliated Hospital, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| |
Collapse
|
22
|
Jiang Y, Tao Q, Qiao X, Yang Y, Peng C, Han M, Dong K, Zhang W, Xu M, Wang D, Zhu W, Li X. Targeting amino acid metabolism to inhibit gastric cancer progression and promote anti-tumor immunity: a review. Front Immunol 2025; 16:1508730. [PMID: 40018041 PMCID: PMC11864927 DOI: 10.3389/fimmu.2025.1508730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
The incidence of gastric cancer remains high and poses a serious threat to human health. Recent comprehensive investigations into amino acid metabolism and immune system components within the tumor microenvironment have elucidated the functional interactions between tumor cells, immune cells, and amino acid metabolism. This study reviews the characteristics of amino acid metabolism in gastric cancer, with a particular focus on the metabolism of methionine, cysteine, glutamic acid, serine, taurine, and other amino acids. It discusses the relationship between these metabolic processes, tumor development, and the body's anti-tumor immunity, and analyzes the importance of targeting amino acid metabolism in gastric cancer for chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yuchun Jiang
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Qing Tao
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuehan Qiao
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yufei Yang
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chen Peng
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Miao Han
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kebin Dong
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wei Zhang
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Min Xu
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Deqiang Wang
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Wen Zhu
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
23
|
Liu C, Liu Z, Dong Z, Liu S, Kan H, Zhang S. Multifaceted interplays between the essential players and lipid peroxidation in ferroptosis. J Genet Genomics 2025:S1673-8527(25)00024-4. [PMID: 39862922 DOI: 10.1016/j.jgg.2025.01.009] [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: 09/07/2024] [Revised: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Ferroptosis, a type of programmed cell death, represents a distinct paradigm in cell biology. It is characterized by the iron-dependent accumulation of reactive oxygen species, which induce lipid peroxidation (LPO), and is orchestrated by the interplay between iron, lipid peroxides, and glutathione. In this review, we emphasize the frequently overlooked role of iron in LPO beyond the classical iron-driven Fenton reaction in several crucial processes that regulate cellular iron homeostasis, including iron intake and export as well as ferritinophagy, and the emerging roles of endoplasmic reticulum-resident flavoprotein oxidoreductases, especially P450 oxidoreductases, in modulating LPO. We summarize how various types of fatty acids (FAs), including saturated, monounsaturated, and polyunsaturated FAs, differentially influence ferroptosis when incorporated into phospholipids. Furthermore, we highlight the therapeutic potential of targeting LPO to mitigate ferroptosis and discuss the regulatory mechanisms of endogenous lipophilic radical-trapping antioxidants that confer resistance to ferroptosis, shedding light on therapeutic avenues for ferroptosis-associated diseases.
Collapse
Affiliation(s)
- Conghe Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zhihao Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zheng Dong
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Sijin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
24
|
Ma M, Zhang Y, Pu K, Tang W. Nanomaterial-enabled metabolic reprogramming strategies for boosting antitumor immunity. Chem Soc Rev 2025; 54:653-714. [PMID: 39620588 DOI: 10.1039/d4cs00679h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Immunotherapy has become a crucial strategy in cancer treatment, but its effectiveness is often constrained. Most cancer immunotherapies focus on stimulating T-cell-mediated immunity by driving the cancer-immunity cycle, which includes tumor antigen release, antigen presentation, T cell activation, infiltration, and tumor cell killing. However, metabolism reprogramming in the tumor microenvironment (TME) supports the viability of cancer cells and inhibits the function of immune cells within this cycle, presenting clinical challenges. The distinct metabolic needs of tumor cells and immune cells require precise and selective metabolic interventions to maximize therapeutic outcomes while minimizing adverse effects. Recent advances in nanotherapeutics offer a promising approach to target tumor metabolism reprogramming and enhance the cancer-immunity cycle through tailored metabolic modulation. In this review, we explore cutting-edge nanomaterial strategies for modulating tumor metabolism to improve therapeutic outcomes. We review the design principles of nanoplatforms for immunometabolic modulation, key metabolic pathways and their regulation, recent advances in targeting these pathways for the cancer-immunity cycle enhancement, and future prospects for next-generation metabolic nanomodulators in cancer immunotherapy. We expect that emerging immunometabolic modulatory nanotechnology will establish a new frontier in cancer immunotherapy in the near future.
Collapse
Affiliation(s)
- Muye Ma
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Dr 2, Singapore, 117545, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Dr, Singapore, 117597, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Wei Tang
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
- Department of Pharmacy and Pharmaceutic Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| |
Collapse
|
25
|
Cao Y, Wang Y, Li Y, Liu S, Wang L, Zhou L, Zhu T. Angelic acid triggers ferroptosis in colorectal cancer cells via targeting and impairing NRF2 protein stability. J Nat Med 2025; 79:82-94. [PMID: 39433724 DOI: 10.1007/s11418-024-01849-4] [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: 08/17/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024]
Abstract
Ferroptosis is a unique programmed cell death driven by iron-dependent phospholipid peroxidation. Tumor cells that escape from the conventional therapies appear more sensitive to ferroptosis. Therefore, it is extremely urgent to find safe and efficient active ingredients that induce ferroptosis in tumor cells. Herein, we identified that angelic acid, as a potent anti-tumor active ingredient in Angelica sinensis, profoundly sensitizes CRC cells to ferroptosis via a natural compound library screen. We revealed that angelic acid treatment is sufficient to predispose CRC cells to ferroptosis phenotype, evidenced by malondialdehyde (MDA) accumulation, lipid peroxidation, and upregulation of ferroptosis-associated markers CHAC1 and PTGS2, which is abolished by ferroptosis inhibitor Fer-1. Moreover, the results of network pharmacology showed that NRF2 is critical for angelic acid-mediated CRC cell ferroptosis. Mechanistically, angelic acid exerted its ferroptosis induction via directly binding and facilitating the degradation of NRF2. In addition, the syngeneic mouse models revealed that angelic acid boosts CRC cell sensitivity to ferroptosis inducer sulfasalazine with essentially no toxicity in vivo. Collectively, our findings highlighted a previously unrecognized anti-tumor mechanism of angelic acid and represented an appealing therapeutic strategy for CRC treatment.
Collapse
Affiliation(s)
- Yongyi Cao
- Department of Oncology, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390 Huaihe Road, Hefei, 230071, Anhui, China
| | - Yu Wang
- Department of Oncology, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390 Huaihe Road, Hefei, 230071, Anhui, China
| | - Yueyang Li
- Department of Oncology, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390 Huaihe Road, Hefei, 230071, Anhui, China
| | - Sihan Liu
- Department of Oncology, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390 Huaihe Road, Hefei, 230071, Anhui, China
| | - Lizhe Wang
- Department of Oncology, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390 Huaihe Road, Hefei, 230071, Anhui, China
| | - Li Zhou
- Department of Oncology, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390 Huaihe Road, Hefei, 230071, Anhui, China
| | - Ting Zhu
- Department of Oncology, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390 Huaihe Road, Hefei, 230071, Anhui, China.
| |
Collapse
|
26
|
Zhang H, Hou W, He Y, Liu Y, Ju Y, Shi X, Zhang Y, Qiao L, He J, Jiang J. Enhanced protection for interfacial lipid ozonolysis by sulfur-containing amino acids. J Colloid Interface Sci 2025; 677:244-249. [PMID: 39094485 DOI: 10.1016/j.jcis.2024.07.217] [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: 05/19/2024] [Revised: 07/10/2024] [Accepted: 07/27/2024] [Indexed: 08/04/2024]
Abstract
Sulfur-containing amino acids have been proposed as drugs for lipid oxidation associated with diseases for a long time, but the molecular-level mechanism on the effectiveness of sulfur-containing amino acids against lipid oxidation remains elusive. In this work, with the interfacial sensitivity mass spectrometry method, oxidation of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphatidylglycerol (POPG), a widely used model lipid, was significantly inhibited on hung droplet surface in presence of sulfur-containing amino acids, such as cysteine (Cys) and methionine (Met). Both the Cys and Met showed a self-sacrificing protection. The amino acids with -S-R tails (R referring to methyl or t-butyl group) showed more effective against POPG oxidation than those with -SH tails, and this process was not related to the conformations of amino acids. The low effectiveness of Cys during the interfacial chemistry was proved to arise from the formation of disulfide bond. This study extends the current understanding of chemistry of sulfur-containing amino acids and provides insights to aid the sulfur-containing amino acids against cell oxidation.
Collapse
Affiliation(s)
- Hong Zhang
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China; State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150090, China
| | - Wenhao Hou
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China; Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Yuwei He
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China
| | - Yaqi Liu
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China
| | - Yun Ju
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China; State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150090, China
| | - Xiaohui Shi
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China
| | - Yuexin Zhang
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China
| | - Lina Qiao
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Jing He
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China; State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150090, China.
| | - Jie Jiang
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai, Shandong 264209, China; State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150090, China
| |
Collapse
|
27
|
Zhu Q, Fu D, Zhu Z, Wu J, Li C, Chen J, Chen C, Li Y, Guo Y, Li X. Potential involvement of cuproptosis induced by m6A-modified autophagy gene ATG10 in KICH : Cuproptosis induced by m6A-modified ATG10 in KICH. BMC Cancer 2024; 24:1591. [PMID: 39736548 PMCID: PMC11686867 DOI: 10.1186/s12885-024-13300-8] [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: 07/02/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
Kidney Chromophobe (KICH) is the third most prevalent renal malignancy, with research challenges due to a dearth of cell lines and clinical samples. There is no specific treatment regimen tailored exclusively for KICH. This study employed gene expression analysis, immunohistochemistry (IHC), Spearman's correlation, immune cell infiltration assessment, and molecular network construction to investigate the autophagy gene ATG10 in KICH. ATG10 was uniquely downregulated in KICH, predominantly regulated by RNA m6A methylation. This downregulation correlated with patient survival, suggesting a potential tumor-regulatory role. ATG10's involvement in the protein lipidation pathway, essential for cuproptosis, was identified. B cells and CD8 + T cells were key immune cells in KICH tumorigenesis associated with ATG10. Examination of molecular networks identified several key molecules and mechanisms, including ceRNA, interplaying proteins, and transcription factors. Additionally, drug targeting analysis pointed to specific amino acids and metabolites as potential therapeutic agents. This study elucidates the significance of ATG10 in KICH, implicating m6A methylation and cuproptosis as novel targets for therapeutic intervention. The identification of B cells and CD8 + T cells as key immune components, along with specific amino acids, suggests that a combination of targeted immune therapies and dietary interventions could provide a multifaceted approach to KICH treatment. Given the limited understanding of KICH pathogenesis, our analysis has unveiled new theoretical insights and potential clinical significances for KICH, expected to broaden the research horizon in this field.
Collapse
Affiliation(s)
- Qingyun Zhu
- Department of Intervention, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - Daiquan Fu
- Department of Intervention, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - Zhaohui Zhu
- Department of Hematology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - Jian Wu
- Department of Pathology, Punan Hospital, Pudong New Area, Shanghai, 200125, China
| | - Chenxi Li
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jiajun Chen
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Chuan Chen
- Department of Intervention, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - Yanxiang Li
- Department of Intervention, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - Yuehui Guo
- Department of Intervention, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China.
| | - Xiangqi Li
- Department of Endocrinology and Metabolism, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China.
| |
Collapse
|
28
|
Scales TQ, Smith B, Blanchard LM, Wixom N, Tuttle ET, Altman BJ, Peppone LJ, Munger J, Campbell TM, Campbell EK, Harris IS. A whole food, plant-based diet reduces amino acid levels in patients with metastatic breast cancer. Cancer Metab 2024; 12:38. [PMID: 39702320 DOI: 10.1186/s40170-024-00368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Amino acids are critical to tumor survival. Tumors can acquire amino acids from the surrounding microenvironment, including the serum. Limiting dietary amino acids is suggested to influence their serum levels. Further, a plant-based diet is reported to contain fewer amino acids than an animal-based diet. The extent to which a plant-based diet lowers the serum levels of amino acids in patients with cancer is unclear. METHODS Patients with metastatic breast cancer (n = 17) were enrolled in a clinical trial with an ad libitum whole food, plant-based diet for 8 weeks without calorie or portion restriction. Dietary changes by participants were monitored using a three-day food record. Serum was collected from participants at baseline and 8 weeks. Food records and serum were analyzed for metabolic changes. RESULTS We found that a whole food, plant-based diet resulted in a lower intake of calories, fat, and amino acids and higher levels of fiber. Additionally, body weight, serum insulin, and IGF were reduced in participants. The diet contained lower levels of essential and non-essential amino acids, except for arginine (glutamine and asparagine were not measured). Importantly, the lowered dietary intake of amino acids translated to reduced serum levels of amino acids in participants (5/9 essential amino acids; 4/11 non-essential amino acids). CONCLUSIONS These findings provide a tractable approach to limiting amino acid levels in persons with cancer. This data lays a foundation for studying the relationship between amino acids in patients and tumor progression. Further, a whole-food, plant-based diet has the potential to synergize with cancer therapies that exploit metabolic vulnerabilities. TRIAL REGISTRATION The clinical trial was registered with ClinicalTrials.gov identifier NCT03045289 on 2017-02-07.
Collapse
Affiliation(s)
- TashJaé Q Scales
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Bradley Smith
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Lisa M Blanchard
- Department of Family Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Nellie Wixom
- Clinical Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Emily T Tuttle
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brian J Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Luke J Peppone
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Surgery, Cancer Control, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Joshua Munger
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Thomas M Campbell
- Department of Family Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Erin K Campbell
- Department of Public Health Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Isaac S Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
29
|
Zhang Y, Tang L, Yang G, Xin H, Huang Y, Li K, Liu J, Pang J, Cao D. Coumarin-aurone based fluorescence probes for cysteine sensitive in-situ identification in living cells. Colloids Surf B Biointerfaces 2024; 244:114173. [PMID: 39191111 DOI: 10.1016/j.colsurfb.2024.114173] [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: 07/10/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Abstract
The quantification of cysteine (Cys) levels in the organisms holds paramount significance in biological research and disease diagnosis, which can give the correlation between abnormal Cys levels and diseases. In this study, two fluorescent probes, designated as DEA-OH and DEA-AC, featuring a coumarin-aurone backbone specifically engineered for Cys detection, were meticulously designed and synthesized. The diethylamino coumarin-aurone probe DEA-OH and the acrylate-substituted probe DEA-AC demonstrated remarkable sensitivity in detecting cysteine by means of copper displacement (DEA-OH) and acrylate hydrolysis mechanisms (DEA-AC) with fluorescence detection limits of 7.25 μM and 1.65 μM, respectively. Moreover, the fluorescence peak wavelength of the two probes displayed a linear relationship with solvent polarity in the ET (30) range of 30-65 kcal•mol-1, indicating the potential for monitoring changes in environmental polarity within this ET (30) range. The outstanding attributes exhibited by DEA-AC including superior photostability, remarkable selectivity, and swift response (kinetic rate constant: 0.00747 s-1), coupled with the exceptional anti-interference ability, have significantly broadened its scope of applications, for example detecting alterations in Cys within biological systems.
Collapse
Affiliation(s)
- Yan Zhang
- School of Materials Science and Engineering, University of Jinan, Jinan, China
| | - Luyao Tang
- School of Materials Science and Engineering, University of Jinan, Jinan, China
| | - Guiyi Yang
- School of Materials Science and Engineering, University of Jinan, Jinan, China
| | - Haotian Xin
- School of Materials Science and Engineering, University of Jinan, Jinan, China
| | - Yan Huang
- School of Materials Science and Engineering, University of Jinan, Jinan, China
| | - Keyi Li
- School of Materials Science and Engineering, University of Jinan, Jinan, China
| | - Jiandong Liu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China.
| | - Jiaojiao Pang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China.
| | - Duxia Cao
- School of Materials Science and Engineering, University of Jinan, Jinan, China.
| |
Collapse
|
30
|
Wu H, Cao P, Wang H, Wang W, Yu H, You C, Shen T, Yang S, Hu Z, Zhou T, Wang J, Wang Q, Qian X, Zhang J, Wang X, Cao Y, Ning L, Lin F. Postoperative Injection of a Triptolide-Preloaded Hydrogel Prevents the Recurrence of Glioblastoma by Dual-Pathway Activation of Ferroptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2406036. [PMID: 39375977 DOI: 10.1002/smll.202406036] [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: 07/18/2024] [Revised: 09/14/2024] [Indexed: 10/09/2024]
Abstract
Glioblastoma (GBM) recurrence leads to high mortality, which remains a major concern in clinical therapy. Herein, an injectable triptolide (TP)-preloaded hydrogel (TP@DNH) accompanied by a postoperative injection strategy is developed to prevent the recurrence of GBM. With a potential inhibitor of the NRF2/SLC7A11/GPX4 axis, it is demonstrated that TP can deactivate glutathione peroxidase 4 (GPX4) from the source of glutathione (GSH) biosynthesis, thereby activating ferroptosis in GBM cells by blocking the neutralization of intracellular lipid peroxide (LPO). Based on acid-sensitive Fe3+/tannic acid (TA) metal-phenolic networks (MPNs), the TP@DNH hydrogel can induce the effective generation of reactive oxygen species (ROS) through Fe3+/TA-mediated Fenton reaction and achieve controllable release of TP in resected GBM cavity. Due to ROS generation and GPX4 deactivation, postoperative injection of TP@DNH can achieve high-level ferroptosis through dual-pathway LPO accumulation, remarkably suppressing the growth of recurrent GBM and prolonging the overall survival in orthotopic GBM relapse mouse model. This work provides an alternative paradigm for regulating ferroptosis in the postoperative treatment of GBM.
Collapse
Affiliation(s)
- Hongshuai Wu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Key Laboratory for Multidisciplinary Intersection of Radiotherapy and Immunology for Gastrointestinal Tumor, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, Wuxi, Jiangsu, 214499, P. R. China
| | - Pingping Cao
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Haiyang Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Digestive Endoscopy, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Wenhong Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Hanyang Yu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Chaoqun You
- Jiangsu Key Lab for the Chemistry and Utilization of Agro-Forest Biomass, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Tianqi Shen
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Suisui Yang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ziyi Hu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Tingting Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Jing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Junxia Zhang
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Neurosurgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Xiuxing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yuandong Cao
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Radiation Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Like Ning
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Fan Lin
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Gastroenterology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, 471023, P. R. China
| |
Collapse
|
31
|
Rowland EC, D’Antuono M, Jermakowicz A, Ayad NG. MAT2a and AHCY inhibition disrupts antioxidant metabolism and reduces glioblastoma cell survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.23.624981. [PMID: 39605416 PMCID: PMC11601785 DOI: 10.1101/2024.11.23.624981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive primary malignant adult brain tumor that inevitably recurs with a fatal prognosis. This is due in part to metabolic reprogramming that allows tumors to evade treatment. We therefore must uncover the pathways mediating these adaptations to develop novel and effective treatments. We searched for genes that are essential in GBM cells as measured by a whole-genome pan-cancer CRISPR screen available from DepMap and identified the methionine metabolism genes MAT2A and AHCY. We conducted genetic knockdown, evaluated mitochondrial respiration, and performed targeted metabolomics to study the function of these genes in GBM. We demonstrate that MAT2A or AHCY knockdown induces oxidative stress, hinders cellular respiration, and reduces the survival of GBM cells. Furthermore, selective MAT2a or AHCY inhibition reduces GBM cell viability, impairs oxidative metabolism, and changes the metabolic profile of these cells towards oxidative stress and cell death. Mechanistically, MAT2a or AHCY regulates spare respiratory capacity, the redox buffer cystathionine, lipid and amino acid metabolism, and prevents DNA damage in GBM cells. Our results point to the methionine metabolic pathway as a novel vulnerability point in GBM.
Collapse
Affiliation(s)
- Emma C. Rowland
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Matthew D’Antuono
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Anna Jermakowicz
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Nagi G. Ayad
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| |
Collapse
|
32
|
Li J, Liu R, Hu H, Huang Y, Shi Y, Li H, Chen H, Cai M, Wang N, Yan T, Wang K, Liu H. Methionine deprivation inhibits glioma proliferation and EMT via the TP53TG1/miR-96-5p/STK17B ceRNA pathway. NPJ Precis Oncol 2024; 8:270. [PMID: 39572759 PMCID: PMC11582638 DOI: 10.1038/s41698-024-00763-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024] Open
Abstract
Recent research highlights the significant impact of methionine metabolism on glioma progression. An increasing amount of compelling evidence bridges long non-coding RNAs to abnormal metabolism in gliomas. However, the specific role of long non-coding RNAs in methionine metabolism regulating glioma progression remains unclear. This study reveals that methionine deprivation inhibits the proliferation, migration, and invasion capabilities of gliomas. Interestingly, the expression of TP53TG1, a long non-coding RNA, is also suppressed. TP53TG1 is highly expressed in gliomas and associated with poor patient outcomes. Subsequently, our data proves that inhibition of TP53TG1 suppresses glioma cell proliferation and the epithelial-mesenchymal transition process both in vitro and in vivo. Ultimately, we found that the underlying mechanism involves a competing endogenous RNA regulating network, in which TP53TG1 modulates the target protein STK17B by competitively binding to miR-96-5p, thus regulating glioma progression. These findings suggest that targeting methionine deprivation could be a promising approach for the clinical treatment of glioma.
Collapse
Affiliation(s)
- Jiafeng Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Ruijie Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Hong Hu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yishuai Huang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Ying Shi
- Departments of Magnetic Resonance, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Honglei Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Hao Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Meng Cai
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Ning Wang
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Tao Yan
- Central Laboratory, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Kaikai Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Province, Hangzhou, PR China.
| | - Huailei Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China.
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
33
|
Liu X, Du J, Sun J, Wang H, An J, Li Y, Hu Y, Cheng S, Feng H, Tian H, Mei X, Qiu Y, Wu C. Borneol-Functionalized Macrophage Membrane-Encapsulated Mesoporous Selenium Nanoparticles Loaded with Resveratrol for the Treatment of Spinal Cord Injury. ACS APPLIED MATERIALS & INTERFACES 2024; 16:63170-63185. [PMID: 39511843 DOI: 10.1021/acsami.4c12454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Spinal cord injury (SCI) is a serious neurological disease that can result in paralysis. After SCI occurs, the blood-spinal cord barrier (BSCB) is disrupted, and permeability is transiently elevated. However, the permeability of the BSCB returns to normal over time, which prevents many drugs from being used in subsequent treatments. In this study, we designed a borneol-functionalized macrophage membrane encapsulating mesoporous selenium nanoparticles loaded with resveratrol (MSe-Res-BMMs) for SCI treatment. In vivo animal experiments and in vitro cell experiments demonstrated that MSe-Res-BMMs were able to protect neurons from ferroptosis by reducing ROS levels and increasing glutathione peroxidase-4 (GPx-4) activity. In addition, this treatment also reduced ROS-induced inflammation and apoptosis by decreasing the expression of inflammatory factor IL-1β and apoptotic factor Cleaved Caspase-3 at the site of injury. Therefore, MSe-Res-BMMs are expected to provide new therapeutic options for SCI treatment.
Collapse
Affiliation(s)
- Xiaobang Liu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Jiaqun Du
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Junpeng Sun
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Han Wang
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Jinyu An
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Yingqiao Li
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Yu Hu
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Shuai Cheng
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Huicong Feng
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - He Tian
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Xifan Mei
- Liaoning Vocational College of Medicine, Shenyang, Liaoning 110101, China
| | - Yang Qiu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Chao Wu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| |
Collapse
|
34
|
Lv D, Dixit D, Cruz AF, Kim LJY, Duan L, Xu X, Wu Q, Zhong C, Lu C, Gersey ZC, Gimple RC, Xie Q, Yang K, Liu X, Fang X, Wu X, Kidwell RL, Wang X, Bao S, He HH, Locasale JW, Agnihotri S, Rich JN. Metabolic regulation of the glioblastoma stem cell epitranscriptome by malate dehydrogenase 2. Cell Metab 2024; 36:2419-2436.e8. [PMID: 39454581 PMCID: PMC11726586 DOI: 10.1016/j.cmet.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/17/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
Tumors reprogram their metabolism to generate complex neoplastic ecosystems. Here, we demonstrate that glioblastoma (GBM) stem cells (GSCs) display elevated activity of the malate-aspartate shuttle (MAS) and expression of malate dehydrogenase 2 (MDH2). Genetic and pharmacologic targeting of MDH2 attenuated GSC proliferation, self-renewal, and in vivo tumor growth, partially rescued by aspartate. Targeting MDH2 induced accumulation of alpha-ketoglutarate (αKG), a critical co-factor for dioxygenases, including the N6-methyladenosine (m6A) RNA demethylase AlkB homolog 5, RNA demethylase (ALKBH5). Forced expression of MDH2 increased m6A levels and inhibited ALKBH5 activity, both rescued by αKG supplementation. Reciprocally, targeting MDH2 reduced global m6A levels with platelet-derived growth factor receptor-β (PDGFRβ) as a regulated transcript. Pharmacological inhibition of MDH2 in GSCs augmented efficacy of dasatinib, an orally bioavailable multi-kinase inhibitor, including PDGFRβ. Collectively, stem-like tumor cells reprogram their metabolism to induce changes in their epitranscriptomes and reveal possible therapeutic paradigms.
Collapse
Affiliation(s)
- Deguan Lv
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Deobrat Dixit
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Andrea F Cruz
- Brain Tumor Biology and Therapy Lab, Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Leo J Y Kim
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Likun Duan
- Department of Pharmacology and Cancer Biology, Duke Cancer Institute, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xin Xu
- Princess Margaret Cancer Centre/University Health Network, Toronto, ON, Canada
| | - Qiulian Wu
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Cuiqing Zhong
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Chenfei Lu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Zachary C Gersey
- Brain Tumor Biology and Therapy Lab, Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Qi Xie
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Westlake University, Hangzhou 310024, Zhejiang, China
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Xiaojing Liu
- Department of Molecular and Structural Chemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - Xiaoguang Fang
- Department of Cancer Biology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Xujia Wu
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Reilly L Kidwell
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Xiuxing Wang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Shideng Bao
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Housheng H He
- Princess Margaret Cancer Centre/University Health Network, Toronto, ON, Canada
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke Cancer Institute, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sameer Agnihotri
- Brain Tumor Biology and Therapy Lab, Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| | - Jeremy N Rich
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Division of Regenerative Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
35
|
Chang J, Pan Y, Jiang F, Xu W, Wang Y, Wang L, Hu B. Mechanism of CXCL8 regulation of methionine metabolism to promote angiogenesis in gliomas. Discov Oncol 2024; 15:614. [PMID: 39488622 PMCID: PMC11531453 DOI: 10.1007/s12672-024-01467-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/16/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Gliomas are the most common malignant brain tumors characterized by angiogenesis and invasive growth. A detailed understanding of its molecular characteristics could provide potential therapeutic targets. In the present study, we sought to explore the key gene CXCL8 in methionine metabolism in gliomas and its potential role in angiogenesis. METHODS U251 glioma cells were divided into control and methionine-restriction tolerant (constructed with 1/4 of the standard level of methionine in the culture medium) groups for transcriptome and metabolome analysis. To confirm the functions and mechanism of CXCL8 in glioma, heat map, volcano map, Go enrichment, gene set enrichment analysis (GSEA), protein-protein interaction network analysis, RT-PCR, western blotting assays, chicken embryo chorioallantoic membrane (CAM) test, chicken embryo yolk sac membrane (YSM) test and transplantation tumor nude mice model were performed. The TCGA database, CGGA database and clinical tissue samples were used to analyze CXCL8's significance on prognosis for patients with glioma. RESULTS CXCL8 expression was significantly up-regulated in methionine-restricted tolerance cells, it also activated vascular system development and triggered angiogenesis. CXCL8 expression is negatively correlated with survival prognosis in gliomas. CONCLUSIONS Glioma cells promote angiogenesis in methionine-restricted environments through the activation of CXCL8, compensating for nutrient deprivation, and possibly contributing to the failure of antiangiogenic therapy.
Collapse
Affiliation(s)
- Jie Chang
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
- Precision Diagnosis and Treatment Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
- Key Laboratory of Nutrition and Metabolism Research for Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yi Pan
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
- Precision Diagnosis and Treatment Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
- Key Laboratory of Nutrition and Metabolism Research for Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Fengfeng Jiang
- Neurological Surgery Department, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Wenxia Xu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
- Precision Diagnosis and Treatment Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
- Key Laboratory of Nutrition and Metabolism Research for Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yue Wang
- Dian Diagnostics Group Co. Ltd, Hangzhou, China
| | - Lude Wang
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China.
- Precision Diagnosis and Treatment Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China.
- Key Laboratory of Nutrition and Metabolism Research for Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China.
| | - Bin Hu
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China.
| |
Collapse
|
36
|
Kalyanaraman B, Cheng G, Hardy M. The role of short-chain fatty acids in cancer prevention and cancer treatment. Arch Biochem Biophys 2024; 761:110172. [PMID: 39369836 PMCID: PMC11784870 DOI: 10.1016/j.abb.2024.110172] [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] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Short-chain fatty acids (SCFAs) are microbial metabolites in the gut that may play a role in cancer prevention and treatment. They affect the metabolism of both normal and cancer cells, regulating various cellular energetic processes. SCFAs also inhibit histone deacetylases, which are targets for cancer therapy. The three main SCFAs are acetate, propionate, and butyrate, which are transported into cells through specific transporters. SCFAs may enhance the efficacy of chemotherapeutic agents and modulate immune cell metabolism, potentially reprogramming the tumor microenvironment. Although SCFAs and SCFA-generating microbes enhance therapeutic efficacy of several forms of cancer therapy, published data also support the opposing viewpoint that SCFAs mitigate the efficacy of some cancer therapies. Therefore, the relationship between SCFAs and cancer is more complex, and this review discusses some of these aspects. Clearly, further research is needed to understand the role of SCFAs, their mechanisms, and applications in cancer prevention and treatment.
Collapse
Affiliation(s)
- Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, United States.
| | - Gang Cheng
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, United States
| | - Micael Hardy
- Aix-Marseille Univ, CNRS, ICR, UMR 7273, Marseille, 13013, France
| |
Collapse
|
37
|
Qiu Y, Xie E, Xu H, Cheng H, Li G. One-carbon metabolism shapes T cell immunity in cancer. Trends Endocrinol Metab 2024; 35:967-980. [PMID: 38925992 DOI: 10.1016/j.tem.2024.05.010] [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: 04/25/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
One-carbon metabolism (1CM), comprising folate metabolism and methionine metabolism, serves as an important mechanism for cellular energy provision and the production of vital signaling molecules, including single-carbon moieties. Its regulation is instrumental in sustaining the proliferation of cancer cells and facilitating metastasis; in addition, recent research has shed light on its impact on the efficacy of T cell-mediated immunotherapy. In this review, we consolidate current insights into how 1CM affects T cell activation, differentiation, and functionality. Furthermore, we delve into the strategies for modulating 1CM in both T cells and tumor cells to enhance the efficacy of adoptively transferred T cells, overcome metabolic challenges in the tumor microenvironment (TME), and maximize the benefits of T cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Yajing Qiu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Ermei Xie
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Haipeng Xu
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian, 350011, China
| | - Hongcheng Cheng
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| | - Guideng Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
38
|
Huang D, Mela A, Bhanu NV, Garcia BA, Canoll P, Casaccia P. PDGF-BB overexpression in p53 null oligodendrocyte progenitors increases H3K27me3 and induces transcriptional changes which favor proliferation. Neoplasia 2024; 57:101042. [PMID: 39216363 PMCID: PMC11402553 DOI: 10.1016/j.neo.2024.101042] [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: 03/28/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Proneural gliomas are brain tumors characterized by enrichment of oligodendrocyte progenitor cell (OPC) transcripts and genetic alterations. In this study we sought to identify transcriptional and epigenetic differences between OPCs with Trp53 deletion and PDGF-BB overexpression (BB-p53n) and those carrying only p53 deletion (p53n). In culture, the BB-p53n OPCs display growth characteristics more similar to glioma cells than p53n OPCs. When injected in mouse brains, BB-p53n OPC form tumors, while the p53n OPCs do not. Unbiased histone proteomics and transcriptomic analysis on these OPC populations identified higher levels of the histone H3K27me3 mark and lower levels of the histone H4K20me3. The transcriptome of the BB-p53n OPCs was characterized by higher levels of transcripts related to proliferation and cell adhesion compared to p53n OPCs. Pharmacological inhibition of the enzyme responsible for histone H3K27 trimethylation (EZH2i) in BB-p53n OPCs, reduced cell cycle transcripts and increased the expression of differentiation markers, but was not sufficient to restore their growth characteristics. This suggests that PDGF-BB overexpression in p53n OPCs favors the early stages of transformation, by promoting proliferation and halting differentiation in a H3K27me3-dependent pathway, and favoring growth characteristics in a H3K27me3 independent manner.
Collapse
Affiliation(s)
- Dennis Huang
- Program in Molecular, Cellular and Developmental Biology at The Graduate Center of The City University of New, York 365 5th Ave, New York, NY 10016, United States; Belfer Research Institute, City University of New York & Weill Cornell Medical College, 413 E 69th St, New York, NY 10021, United States; Neuroscience Initiative, Advance Science Research Center, Graduate Center of The City University of New York, 85 St Nicholas Terrace, New York, NY 10031, United States; Department of Biological Sciences, Hunter College, City University of New York, 695 Park Ave, New York, NY 10065, United States
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 W 168th St, New York, NY 10032, United States
| | - Natarajan V Bhanu
- Department Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110, United States
| | - Benjamin A Garcia
- Department Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110, United States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 W 168th St, New York, NY 10032, United States
| | - Patrizia Casaccia
- Program in Molecular, Cellular and Developmental Biology at The Graduate Center of The City University of New, York 365 5th Ave, New York, NY 10016, United States; Neuroscience Initiative, Advance Science Research Center, Graduate Center of The City University of New York, 85 St Nicholas Terrace, New York, NY 10031, United States.
| |
Collapse
|
39
|
Du K, Grocott L, Anichini G, O’Neill K, Syed N. Amino Acid Deprivation in Glioblastoma: The Role in Survival and the Tumour Microenvironment-A Narrative Review. Biomedicines 2024; 12:2481. [PMID: 39595047 PMCID: PMC11592029 DOI: 10.3390/biomedicines12112481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Glioblastoma is the most common and aggressive primary brain tumour, characterised by its invasive nature and complex metabolic profile. Emerging research highlights the role of amino acids (AAs) in glioblastoma metabolism, influencing tumour growth and the surrounding microenvironment. METHODS This narrative review synthesises recent pre-clinical studies focusing on the metabolic functions of AAs in glioblastoma. Key areas include the effects of AA deprivation on tumour growth, adaptive mechanisms, and the tumour microenvironment. RESULTS The effects related to arginine, glutamine, methionine, and cysteine deprivation have been more extensively reported. Arginine deprivation in arginine-auxotrophic glioblastomas induces apoptosis and affects cell adhesion, while glutamine deprivation disrupts metabolic pathways and enhances autophagy. Methionine and cysteine deprivation impact lipid metabolism and ferroptosis. Tumour adaptive mechanisms present challenges, and potential compensatory responses have been identified. The response of the microenvironment to AA deprivation, including immune modulation, is critical to determining therapeutic outcomes. CONCLUSIONS Targeting AA metabolism offers a promising approach for glioblastoma treatment, with potential targeted drugs showing clinical promise. However, the complexity of tumour adaptive mechanisms and their impact on the microenvironment necessitates further research to optimise combination therapies and improve therapeutic efficacy.
Collapse
Affiliation(s)
- Keven Du
- Imperial College School of Medicine, Imperial College London, London SW7 2AZ, UK; (K.D.); (L.G.)
| | - Leila Grocott
- Imperial College School of Medicine, Imperial College London, London SW7 2AZ, UK; (K.D.); (L.G.)
| | - Giulio Anichini
- Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK; (K.O.); (N.S.)
| | - Kevin O’Neill
- Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK; (K.O.); (N.S.)
| | - Nelofer Syed
- Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK; (K.O.); (N.S.)
| |
Collapse
|
40
|
Zou Y, Chen J, Luo X, Qu Y, Zhou M, Xia R, Wang W, Zheng X. Porphyrin-engineered nanoscale metal-organic frameworks: enhancing photodynamic therapy and ferroptosis in oncology. Front Pharmacol 2024; 15:1481168. [PMID: 39512824 PMCID: PMC11541831 DOI: 10.3389/fphar.2024.1481168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Photodynamic therapy and ferroptosis induction have risen as vanguard oncological interventions, distinguished by their precision and ability to target vulnerabilities in cancer cells. Photodynamic therapy's non-invasive profile and selective cytotoxicity complement ferroptosis' unique mode of action, which exploits iron-dependent lipid peroxidation, offering a pathway to overcome chemoresistance with lower systemic impact. The synergism between photodynamic therapy and ferroptosis is underscored by the depletion of glutathione and glutathione peroxidase four inhibitions by photodynamic therapy-induced reactive oxygen species, amplifying lipid peroxidation and enhancing ferroptotic cell death. This synergy presents an opportunity to refine cancer treatment by modulating redox homeostasis. Porphyrin-based nanoscale metal-organic frameworks have unique hybrid structures and exceptional properties. These frameworks can serve as a platform for integrating photodynamic therapy and ferroptosis through carefully designed structures and functions. These nanostructures can be engineered to deliver multiple therapeutic modalities simultaneously, marking a pivotal advance in multimodal cancer therapy. This review synthesizes recent progress in porphyrin-modified nanoscale metal-organic frameworks for combined photodynamic therapy and ferroptosis, delineating the mechanisms that underlie their synergistic effects in a multimodal context. It underscores the potential of porphyrin-based nanoscale metal-organic frameworks as advanced nanocarriers in oncology, propelling the field toward more efficacious and tailored cancer treatments.
Collapse
Affiliation(s)
- Yutao Zou
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu, China
| | - Jiayi Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xuanxuan Luo
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yijie Qu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Rui Xia
- School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
41
|
Scales TQ, Smith B, Blanchard LM, Wixom N, Tuttle ET, Altman BJ, Peppone LJ, Munger J, Campbell TM, Campbell EK, Harris IS. A whole food, plant-based diet reduces amino acid levels in patients with metastatic breast cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.09.24315165. [PMID: 39417128 PMCID: PMC11483017 DOI: 10.1101/2024.10.09.24315165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Background Amino acids are critical to tumor survival. Tumors can acquire amino acids from the surrounding microenvironment, including the serum. Limiting dietary amino acids is suggested to influence their serum levels. Further, a plant-based diet is reported to contain fewer amino acids than an animal-based diet. The extent to which a plant-based diet lowers the serum levels of amino acids in patients with cancer is unclear. Methods Patients with metastatic breast cancer (n=17) were enrolled in a clinical trial with an ad libitum whole food, plant-based diet for 8 weeks without calorie or portion restriction. Dietary changes by participants were monitored using a three-day food record. Serum was collected from participants at baseline and 8 weeks. Food records and serum were analyzed for metabolic changes. Results We found that a whole food, plant-based diet resulted in a lower intake of calories, fat, and amino acids and higher levels of fiber. Additionally, body weight, serum insulin, and IGF were reduced in participants. The diet contained lower levels of essential and non-essential amino acids, except for arginine (glutamine and asparagine were not measured). Importantly, the lowered dietary intake of amino acids translated to reduced serum levels of amino acids in participants (5/9 essential amino acids; 4/11 non-essential amino acids). Conclusions These findings provide a tractable approach to limiting amino acid levels in persons with cancer. This data lays a foundation for studying the relationship between amino acids in patients and tumor progression. Further, a whole-food, plant-based diet has the potential to synergize with cancer therapies that exploit metabolic vulnerabilities. Trial Registration The clinical trial was registered with ClinicalTrials.gov identifier NCT03045289 on 2017-02-07.
Collapse
Affiliation(s)
- TashJaé Q. Scales
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Bradley Smith
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Lisa M. Blanchard
- Department of Family Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, USA 14642
| | - Nellie Wixom
- Clinical Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Emily T. Tuttle
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Brian J. Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Luke J. Peppone
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Surgery, Cancer Control, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Joshua Munger
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Thomas M. Campbell
- Department of Family Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, USA 14642
| | - Erin K. Campbell
- Department of Public Health Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, USA,14642
| | - Isaac S. Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| |
Collapse
|
42
|
Okano Y, Yamauchi T, Fukuzaki R, Tsuruta A, Yoshida Y, Tsurudome Y, Ushijima K, Matsunaga N, Koyanagi S, Ohdo S. Oncogenic accumulation of cysteine promotes cancer cell proliferation by regulating the translation of D-type cyclins. J Biol Chem 2024; 300:107890. [PMID: 39413876 DOI: 10.1016/j.jbc.2024.107890] [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: 05/04/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
Malignant cells exhibit a high demand for amino acids to sustain their abnormal proliferation. Particularly, the intracellular accumulation of cysteine is often observed in cancer cells. Previous studies have shown that deprivation of intracellular cysteine in cancer cells results in the accumulation of lipid peroxides in the plasma membrane and induction of ferroptotic cell death, indicating that cysteine plays a critical role in the suppression of ferroptosis. Herein, we found that the oncogenic accumulation of cysteine also contributes to cancer cell proliferation by promoting the cell cycle progression, which is independent of its suppressive effect on ferroptosis. The growth ability of four types of cancer cells, including murine hepatocarcinoma cells, but not of primary hepatocytes, were dependent on the exogenous supply of cysteine. Deprivation of intracellular cysteine in cancer cells induced cell cycle arrest at the G0/G1 phase, accompanied by a decrease in the expression of cyclin D1 and D2 proteins. The cysteine deprivation-induced decrease in D-type cyclin expression was associated with the upregulation of eukaryotic translation initiation factor 4E binding protein 1, which represses the translation of cyclin D1 and D2 proteins by binding to eukaryotic translation initiation factor 4E. Similar results were observed in hepatocarcinoma cells treated with erastin, an inhibitor of cystine/glutamate antiporter, xCT. These findings reveal an unappreciated role of cysteine in regulating the growth of malignant cancer cells and deepen our understanding of the cytotoxic effect of xCT inhibitor to prevent cancer cell proliferation.
Collapse
Affiliation(s)
- Yumi Okano
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoaki Yamauchi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Runa Fukuzaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akito Tsuruta
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuya Yoshida
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuya Tsurudome
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyoonoda, Yamaguchi, Japan
| | - Kentaro Ushijima
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyoonoda, Yamaguchi, Japan
| | - Naoya Matsunaga
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoru Koyanagi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | - Shigehiro Ohdo
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
43
|
Hecht F, Zocchi M, Tuttle ET, Ward NP, Smith B, Kang YP, Cazarin J, Soares ZG, Ozgurses ME, Zhao H, Sheehan C, Alimohammadi F, Munger LD, Trivedi D, Asantewaa G, Blick-Nitko SK, Zoeller JJ, Chen Y, Vasiliou V, Turner BM, Muir A, Coloff JL, Munger J, DeNicola GM, Harris IS. Catabolism of extracellular glutathione supplies amino acids to support tumor growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617667. [PMID: 39416022 PMCID: PMC11482906 DOI: 10.1101/2024.10.10.617667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Restricting amino acids from tumors is an emerging therapeutic strategy with significant promise. While typically considered an intracellular antioxidant with tumor-promoting capabilities, glutathione (GSH) is a tripeptide of cysteine, glutamate, and glycine that can be catabolized, yielding amino acids. The extent to which GSH-derived amino acids are essential to cancers is unclear. Here, we find that GSH catabolism promotes tumor growth. We show that depletion of intracellular GSH does not perturb tumor growth, and extracellular GSH is highly abundant in the tumor microenvironment, highlighting the potential importance of GSH outside of tumors. We find supplementation with GSH can rescue cancer cell survival and growth in cystine-deficient conditions, and this rescue is dependent on the catabolic activity of γ-glutamyltransferases (GGTs). Finally, pharmacologic targeting of GGTs' activity prevents the breakdown of circulating GSH, lowers tumor cysteine levels, and slows tumor growth. Our findings indicate a non-canonical role for GSH in supporting tumors by acting as a reservoir of amino acids. Depriving tumors of extracellular GSH or inhibiting its breakdown is potentially a therapeutically tractable approach for patients with cancer. Further, these findings change our view of GSH and how amino acids, including cysteine, are supplied to cells.
Collapse
Affiliation(s)
- Fabio Hecht
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
- These authors contributed equally
| | - Marco Zocchi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
- These authors contributed equally
| | - Emily T. Tuttle
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Nathan P. Ward
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Bradley Smith
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Yun Pyo Kang
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Zamira G. Soares
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Mete Emir Ozgurses
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Huiping Zhao
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Colin Sheehan
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Fatemeh Alimohammadi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Lila D. Munger
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Dhvani Trivedi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Gloria Asantewaa
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Sara K. Blick-Nitko
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Jason J. Zoeller
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA, 02115
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA, 06510
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA, 06510
| | - Bradley M. Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Alexander Muir
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Jonathan L. Coloff
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Joshua Munger
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Gina M. DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Isaac S. Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| |
Collapse
|
44
|
Cao PHA, Dominic A, Lujan FE, Senthilkumar S, Bhattacharya PK, Frigo DE, Subramani E. Unlocking ferroptosis in prostate cancer - the road to novel therapies and imaging markers. Nat Rev Urol 2024; 21:615-637. [PMID: 38627553 PMCID: PMC12067944 DOI: 10.1038/s41585-024-00869-9] [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] [Accepted: 03/04/2024] [Indexed: 04/19/2024]
Abstract
Ferroptosis is a distinct form of regulated cell death that is predominantly driven by the build-up of intracellular iron and lipid peroxides. Ferroptosis suppression is widely accepted to contribute to the pathogenesis of several tumours including prostate cancer. Results from some studies reported that prostate cancer cells can be highly susceptible to ferroptosis inducers, providing potential for an interesting new avenue of therapeutic intervention for advanced prostate cancer. In this Perspective, we describe novel molecular underpinnings and metabolic drivers of ferroptosis, analyse the functions and mechanisms of ferroptosis in tumours, and highlight prostate cancer-specific susceptibilities to ferroptosis by connecting ferroptosis pathways to the distinctive metabolic reprogramming of prostate cancer cells. Leveraging these novel mechanistic insights could provide innovative therapeutic opportunities in which ferroptosis induction augments the efficacy of currently available prostate cancer treatment regimens, pending the elimination of major bottlenecks for the clinical translation of these treatment combinations, such as the development of clinical-grade inhibitors of the anti-ferroptotic enzymes as well as non-invasive biomarkers of ferroptosis. These biomarkers could be exploited for diagnostic imaging and treatment decision-making.
Collapse
Affiliation(s)
- Pham Hong Anh Cao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Abishai Dominic
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fabiola Ester Lujan
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Sanjanaa Senthilkumar
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signalling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| | - Elavarasan Subramani
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
45
|
Banu MA, Dovas A, Argenziano MG, Zhao W, Sperring CP, Cuervo Grajal H, Liu Z, Higgins DM, Amini M, Pereira B, Ye LF, Mahajan A, Humala N, Furnari JL, Upadhyayula PS, Zandkarimi F, Nguyen TT, Teasley D, Wu PB, Hai L, Karan C, Dowdy T, Razavilar A, Siegelin MD, Kitajewski J, Larion M, Bruce JN, Stockwell BR, Sims PA, Canoll P. A cell state-specific metabolic vulnerability to GPX4-dependent ferroptosis in glioblastoma. EMBO J 2024; 43:4492-4521. [PMID: 39192032 PMCID: PMC11480389 DOI: 10.1038/s44318-024-00176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/12/2024] [Accepted: 07/01/2024] [Indexed: 08/29/2024] Open
Abstract
Glioma cells hijack developmental programs to control cell state. Here, we uncover a glioma cell state-specific metabolic liability that can be therapeutically targeted. To model cell conditions at brain tumor inception, we generated genetically engineered murine gliomas, with deletion of p53 alone (p53) or with constitutively active Notch signaling (N1IC), a pathway critical in controlling astrocyte differentiation during brain development. N1IC tumors harbored quiescent astrocyte-like transformed cell populations while p53 tumors were predominantly comprised of proliferating progenitor-like cell states. Further, N1IC transformed cells exhibited increased mitochondrial lipid peroxidation, high ROS production and depletion of reduced glutathione. This altered mitochondrial phenotype rendered the astrocyte-like, quiescent populations more sensitive to pharmacologic or genetic inhibition of the lipid hydroperoxidase GPX4 and induction of ferroptosis. Treatment of patient-derived early-passage cell lines and glioma slice cultures generated from surgical samples with a GPX4 inhibitor induced selective depletion of quiescent astrocyte-like glioma cell populations with similar metabolic profiles. Collectively, these findings reveal a specific therapeutic vulnerability to ferroptosis linked to mitochondrial redox imbalance in a subpopulation of quiescent astrocyte-like glioma cells resistant to standard forms of treatment.
Collapse
Affiliation(s)
- Matei A Banu
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Wenting Zhao
- Department of System Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Colin P Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Zhouzerui Liu
- Department of System Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dominique Mo Higgins
- Department of Neurological Surgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Misha Amini
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Brianna Pereira
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ling F Ye
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Aayushi Mahajan
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Nelson Humala
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia L Furnari
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Pavan S Upadhyayula
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Fereshteh Zandkarimi
- Department of Biological Sciences, Department of Chemistry and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Trang Tt Nguyen
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Damian Teasley
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter B Wu
- Department of Neurological Surgery, UCLA Geffen School of Medicine, Los Angeles, CA, USA
| | - Li Hai
- Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA
| | - Charles Karan
- Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA
| | | | - Aida Razavilar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jan Kitajewski
- University of Illinois Cancer Center, Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, USA
| | | | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Department of Chemistry and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Peter A Sims
- Department of System Biology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Peter Canoll
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
46
|
Huang H, Lin Y, Xin J, Sun N, Zhao Z, Wang H, Duan L, Zhou Y, Liu X, Fang J, Jing B, Pan K, Zeng Y, Zeng D, Li H, Ma H, Bai Y, Wei L, Ni X. Fluoride exposure-induced gut microbiota alteration mediates colonic ferroptosis through N 6-methyladenosine (m 6A) mediated silencing of SLC7A11. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116816. [PMID: 39096685 DOI: 10.1016/j.ecoenv.2024.116816] [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: 02/08/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 08/05/2024]
Abstract
Fluoride exposure is widespread worldwide and poses a significant threat to organisms, particularly to their gastrointestinal tracts. However, due to limited knowledge of the mechanism of fluoride induced intestinal injury, it has been challenging to develop an effective treatment. To address this issue, we used a series of molecular biology in vitro and in vivo experiments. NaF triggered m6A mediated ferroptosis to cause intestinal damage. Mechanistically, NaF exposure increased the m6A level of SLC7A11 mRNA, promoted YTHDF2 binding to m6A-modified SLC7A11 mRNA, drove the degradation of SLC7A11 mRNA, and led to a decrease in its protein expression, which eventually triggers ferroptosis. Moreover, NaF aggravated ferroptosis of the colon after antibiotics destroyed the composition of gut microbiota. 16 S rRNA sequencing and SPEC-OCCU plots, Zi-Pi relationships, and Spearman correlation coefficients verified that Lactobacillus murinus (ASV54, ASV58, and ASV82) plays a key role in the response to NaF-induced ferroptosis. Collectively, NaF-induced gut microbiota alteration mediates severe intestinal cell injury by inducing m6A modification-mediated ferroptosis. Our results highlight a key mechanism of the gut in response to NaF exposure and suggest a valuable theoretical basis for its prevention and treatment.
Collapse
Affiliation(s)
- Haonan Huang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yu Lin
- Department of Gastroenterology, Southern Medical University Hospital of Integrative Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Jinge Xin
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhifang Zhao
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, Guizhou, China
| | - Hesong Wang
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lixiao Duan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yanxi Zhou
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xingmei Liu
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jing Fang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hao Li
- Plateau Brain Science Research Center, Tibet University, Lhasa, Tibet 850000, China
| | - Hailin Ma
- Plateau Brain Science Research Center, Tibet University, Lhasa, Tibet 850000, China
| | - Yang Bai
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Limin Wei
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
47
|
Yao H, Jiang W, Liao X, Wang D, Zhu H. Regulatory mechanisms of amino acids in ferroptosis. Life Sci 2024; 351:122803. [PMID: 38857653 DOI: 10.1016/j.lfs.2024.122803] [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: 03/12/2024] [Revised: 05/19/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Ferroptosis, an iron-dependent non-apoptotic regulated cell death process, is associated with the pathogenesis of various diseases. Amino acids, which are indispensable substrates of vital activities, significantly regulate ferroptosis. Amino acid metabolism is involved in maintaining iron and lipid homeostasis and redox balance. The regulatory effects of amino acids on ferroptosis are complex. An amino acid may exert contrasting effects on ferroptosis depending on the context. This review systematically and comprehensively summarized the distinct roles of amino acids in regulating ferroptosis and highlighted the emerging opportunities to develop clinical therapeutic strategies targeting amino acid-mediated ferroptosis.
Collapse
Affiliation(s)
- Heying Yao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Wei Jiang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Dongqing Wang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
48
|
Wang H, Chang H, Weng H, Zhai Y, Zeng H, Li S, Han Z. Study of Plasma Biochemistry and Plasma Metabolomics Differences in Montbéliard and Holstein Backcross and Holstein Heifers. Animals (Basel) 2024; 14:2294. [PMID: 39199828 PMCID: PMC11350815 DOI: 10.3390/ani14162294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Holstein cattle are the main breed of dairy cattle in China. However, given the high degree of purebred selection of Holstein cattle, Chinese dairy cattle are increasingly being characterized by poor disease resistance, poor quality, and declining fertility. In this study, using Montbéliard × Holstein cattle as females and Montbéliard bulls as males for backcross breeding, we sought to provide a reference for improving the quality and performance of Holstein cattle and enhancing the efficiency of dairy farming. On the basis of similar physiological status and age, we selected 24 Montbéliard and Holstein backcross heifers and 11 Holstein heifers fed the same formula for comparative analyses. Plasma samples collected for plasma biochemical index analyses revealed that the content of ALB and BUN in the Montbéliard and Holstein backcross heifers was 20.83% (31.62 g/L to 26.17 g/L) and 42.36% (6.89 mmol/L to 4.84 mmol/L) higher than in the Holsteins (p < 0.01). The ALB/GLB (0.90 to 0.60, p < 0.05) was significantly higher in Montbéliard and Holstein backcross heifers than in Holstein heifers. Similarly, the activity of CAT in the backcross heifers was 61.28% (4.29 U/mL to 2.66 U/mL) higher than that in the Holstein heifers (p < 0.05). Although the activity of GSH-Px in the backcross heifers also showed an increasing trend, the difference did not reach the level of statistical significance (p = 0.052). Compared with Holstein heifers, the concentrations of IgA, IgG, and IL-4 were elevated by 32.52% (24.90 μg/mL to 18.79 μg/mL, p < 0.01), 13.46% (234.32 μg/mL to 206.53 μg/mL, p < 0.01), and 14.59% (306.27 pg/mL to 267.28 pg/mL, p < 0.05), and the contents of IL-6 and TNF-α were decreased by 15.92% (215.71 pg/mL to 256.55 pg/mL, p < 0.01) and 32.17% (7.17 ng/mL to 10.57 ng/mL, p < 0.01) in the plasma of Montbéliard and Holstein backcross heifers. Among the experimental heifers, five animals from each of the two groups were selected for plasma metabolomic analysis based on untargeted liquid chromatography-mass spectrometry. A comparison of the differential metabolites between the two heifer breeds revealed an up-regulation of d-glucuronic acid, s-glutathionyl-l-cysteine, and oleic acid levels in the backcross cattle compared with those in the Holstein heifers. We speculate that changes in the levels of these metabolites may be associated with an enhancement of the anti-inflammatory, antioxidant, and immune systems in these backcross heifers. Collectively, our findings in this study indicate that compared with 12-month-old purebred Holstein heifers, Montbéliard and Holstein backcross heifers of the same age are characterized by higher antioxidant capacity and immunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhaoyu Han
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (H.W.); (H.C.); (H.W.); (Y.Z.); (H.Z.); (S.L.)
| |
Collapse
|
49
|
Polak R, Zhang ET, Kuo CJ. Cancer organoids 2.0: modelling the complexity of the tumour immune microenvironment. Nat Rev Cancer 2024; 24:523-539. [PMID: 38977835 DOI: 10.1038/s41568-024-00706-6] [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: 05/09/2024] [Indexed: 07/10/2024]
Abstract
The development of neoplasia involves a complex and continuous interplay between malignantly transformed cells and the tumour microenvironment (TME). Cancer immunotherapies targeting the immune TME have been increasingly validated in clinical trials but response rates vary substantially between tumour histologies and are often transient, idiosyncratic and confounded by resistance. Faithful experimental models of the patient-specific tumour immune microenvironment, capable of recapitulating tumour biology and immunotherapy effects, would greatly improve patient selection, target identification and definition of resistance mechanisms for immuno-oncology therapeutics. In this Review, we discuss currently available and rapidly evolving 3D tumour organoid models that capture important immune features of the TME. We highlight diverse opportunities for organoid-based investigations of tumour immunity, drug development and precision medicine.
Collapse
Affiliation(s)
- Roel Polak
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa T Zhang
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
50
|
Yang L, Shao Y, Gao T, Bajinka O, Yuan X. Current advances in cancer energy metabolism under dietary restriction: a mini review. Med Oncol 2024; 41:209. [PMID: 39060824 DOI: 10.1007/s12032-024-02452-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024]
Abstract
The manipulation of the energy or source of food for cancer cells has attracted significant attention in oncology research. Metabolic reprogramming of the immune system allows for a deeper understanding of cancer cell mechanisms, thereby impeding their progression. A more targeted approach is the restriction of cancer cells through dietary restriction (CR), which deprives cancer cells of the preferred energy sources within the tumor microenvironment, thereby enhancing immune cell efficacy. Although there is a plethora of CR strategies that can be employed to impede cancer progression, there is currently no comprehensive review that delineates the specific dietary restrictions that target the diverse metabolic pathways of cancer cells. This mini-review introduces amino acids as anti-cancer agents and discusses the role of dietary interventions in cancer prevention and treatment. It highlights the potential of a ketogenic diet as a therapeutic approach for cancer, elucidating its distinct mechanisms of action in tumor progression. Additionally, the potential of plant-based diets as anti-cancer agents and the role of polyphenols and vitamins in anti-cancer therapy were also discussed, along with some prospective interventions for CR as anti-tumor progression.
Collapse
Affiliation(s)
- Liuxin Yang
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, No. 24 Heping Street, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Yudian Shao
- Second Clinical Medical College, Heilongjiang University of Chinese Medicine, No. 24 Heping Street, Harbin, Heilongjiang, 150040, People's Republic of China
| | - Tingting Gao
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, No. 33 Xidazhi Street, Harbin, 150006, Heilongjiang, People's Republic of China
| | - Ousman Bajinka
- School of Medicine and Allied Health Sciences, University of The Gambia, Banjul, The Gambia
| | - Xingxing Yuan
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, No. 24 Heping Street, Harbin, 150040, Heilongjiang Province, People's Republic of China.
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, No. 33 Xidazhi Street, Harbin, 150006, Heilongjiang, People's Republic of China.
| |
Collapse
|