1
|
Leote RJB, Barsan MM, Sanz CG, Diculescu VC. Electrochemical bienzymatic biosensor for pyruvate kinase activity evaluation and inhibitor screening. Talanta 2025; 291:127886. [PMID: 40056645 DOI: 10.1016/j.talanta.2025.127886] [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: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/10/2025]
Abstract
This study describes the development of a pyruvate kinase (PyK)-biosensor for the evaluation of PyK activity, as a diagnostic tool for early cancer screening and detection of kinase inhibitors used in cancer treatment, with the evaluation of the inhibition mechanism. The biosensor was constructed by co-immobilizing the enzymes PyK and pyruvate oxidase (PyOx) on Au film electrodes by crosslinking with glutaraldehyde (GA) and evaluated electrochemically by cyclic voltammetry (CV) and fixed potential amperometry (CA). First, the experimental conditions were optimized in terms of applied potential, enzyme ratio PyK:PyOx and enzyme substrate concentration: phosphoenolpyruvate (PEP) and adenosine diphosphate (ADP). The biosensor sensitivity towards PEP detection was 2.11 ± 0.08 μA mM-1 cm-2, with very high reproducibility and repeatability, which made it suitable for inhibition studies of PyK inhibitor. The inhibition mechanism of shikonin was determined in relation to both PEP and ADP, with the calculation of IC50 values and binding constants (Ki). Detection of shikonin was possible at very low concentrations in the linear range of 0.1-4.0 pM. The electrochemical results were validated by UV-Vis spectrophotometry. The developed biosensor is a valuable tool for drug screening by enabling enzyme catalytic function examination with applicability to identify inhibitors, estimate their affinity, inhibition mechanism linked to their molecular mechanisms of action and evaluate selectivity, of great interest in both pharmaceutical and medical domains.
Collapse
Affiliation(s)
- Ricardo Jose Branco Leote
- National Institute of Materials Physics, Str. Atomistilor 405A, 077125, Măgurele, Romania; Faculty of Physics, University of Bucharest, Atomistilor 405, 077125, Măgurele, Romania
| | - Madalina Maria Barsan
- National Institute of Materials Physics, Str. Atomistilor 405A, 077125, Măgurele, Romania
| | - Caroline G Sanz
- National Institute of Materials Physics, Str. Atomistilor 405A, 077125, Măgurele, Romania
| | - Victor C Diculescu
- National Institute of Materials Physics, Str. Atomistilor 405A, 077125, Măgurele, Romania.
| |
Collapse
|
2
|
Román-Trufero M, Kleijn IT, Blighe K, Zhou J, Saavedra-García P, Gaffar A, Christoforou M, Bellotti A, Abrahams J, Atrih A, Lamont D, Gierlinski M, Jayaprakash P, Michel AM, Aboagye EO, Yuneva M, Masson GR, Shahrezaei V, Auner HW. An ISR-independent role of GCN2 prevents excessive ribosome biogenesis and mRNA translation. Life Sci Alliance 2025; 8:e202403014. [PMID: 40032489 DOI: 10.26508/lsa.202403014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
The integrated stress response (ISR) is a corrective physiological programme to restore cellular homeostasis that is based on the attenuation of global protein synthesis and a resource-enhancing transcriptional programme. GCN2 is the oldest of four kinases that are activated by diverse cellular stresses to trigger the ISR and acts as the primary responder to amino acid shortage and ribosome collisions. Here, using a broad multi-omics approach, we uncover an ISR-independent role of GCN2. GCN2 inhibition or depletion in the absence of discernible stress causes excessive protein synthesis and ribosome biogenesis, perturbs the cellular translatome, and results in a dynamic and broad loss of metabolic homeostasis. Cancer cells that rely on GCN2 to keep protein synthesis in check under conditions of full nutrient availability depend on GCN2 for survival and unrestricted tumour growth. Our observations describe an ISR-independent role of GCN2 in regulating the cellular proteome and translatome and suggest new avenues for cancer therapies based on unleashing excessive mRNA translation.
Collapse
Affiliation(s)
- Mónica Román-Trufero
- Division of Haematology and Central Haematology Laboratory, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Hugh and Josseline Langmuir Centre for Myeloma Research, Department of Immunology and Inflammation, Imperial College London, London, UK
- The Francis Crick Institute, London, UK
| | - Istvan T Kleijn
- Department of Mathematics, Imperial College London, London, UK
| | | | - Jinglin Zhou
- Hugh and Josseline Langmuir Centre for Myeloma Research, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Paula Saavedra-García
- Hugh and Josseline Langmuir Centre for Myeloma Research, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Abigail Gaffar
- Hugh and Josseline Langmuir Centre for Myeloma Research, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Marilena Christoforou
- Hugh and Josseline Langmuir Centre for Myeloma Research, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Axel Bellotti
- Division of Haematology and Central Haematology Laboratory, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Joel Abrahams
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Abdelmadjid Atrih
- FingerPrints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee, UK
| | - Douglas Lamont
- FingerPrints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee, UK
| | - Marek Gierlinski
- Data Analysis Group, Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | | | | | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Glenn R Masson
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee, UK
| | | | - Holger W Auner
- Division of Haematology and Central Haematology Laboratory, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Hugh and Josseline Langmuir Centre for Myeloma Research, Department of Immunology and Inflammation, Imperial College London, London, UK
- The Francis Crick Institute, London, UK
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Markov N, Sabirova S, Sharapova G, Gomzikova M, Brichkina A, Barlev NA, Egger M, Rizvanov A, Simon HU. Mitochondrial, metabolic and bioenergetic adaptations drive plasticity of colorectal cancer cells and shape their chemosensitivity. Cell Death Dis 2025; 16:253. [PMID: 40185729 PMCID: PMC11971274 DOI: 10.1038/s41419-025-07596-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
The extent of mitochondrial heterogeneity and the presence of mitochondrial archetypes in cancer remain unknown. Mitochondria play a central role in the metabolic reprogramming that occurs in cancer cells. This process adjusts the activity of metabolic pathways to support growth, proliferation, and survival of cancer cells. Using a panel of colorectal cancer (CRC) cell lines, we revealed extensive differences in their mitochondrial composition, suggesting functional specialisation of these organelles. We differentiated bioenergetic and mitochondrial phenotypes, which point to different strategies used by CRC cells to maintain their sustainability. Moreover, the efficacy of various treatments targeting metabolic pathways was dependent on the respiration and glycolysis levels of cancer cells. Furthermore, we identified metabolites associated with both bioenergetic profiles and cell responses to treatments. The levels of these molecules can be used to predict the therapeutic efficacy of anti-cancer drugs and identify metabolic vulnerabilities of CRC. Our study indicates that the efficacy of CRC therapies is closely linked to mitochondrial status and cellular bioenergetics.
Collapse
Affiliation(s)
- Nikita Markov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Sirina Sabirova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Laboratory of Intercellular Communication, Kazan Federal University, Kazan, Russia
| | - Gulnaz Sharapova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Marina Gomzikova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Laboratory of Intercellular Communication, Kazan Federal University, Kazan, Russia
| | - Anna Brichkina
- Institute of Systems Immunology, Center for Tumor Biology and Immunology, Philipps University of Marburg, Marburg, Germany
| | - Nick A Barlev
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Marcel Egger
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Albert Rizvanov
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany.
| |
Collapse
|
4
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
5
|
Yi L, Shahatiaili A, Zhang L, He H, Chen L, Zhang Z, Gao F, Shao F, Gao Y, He J. USP13: A therapeutic target for combating tumorigenesis and antitumor therapy resistance. Int J Biol Macromol 2025; 304:140608. [PMID: 39900156 DOI: 10.1016/j.ijbiomac.2025.140608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Ubiquitin-specific peptidase 13 (USP13) has emerged as a key regulator of proteins critical to the hallmarks of cancer, playing an essential role in cellular regulation. This deubiquitinating enzyme, often overexpressed in malignancies, wields its molecular scissors precisely, snipping ubiquitin tags to rescue oncoproteins from degradation. Our review highlights the dual role of USP13 in cancer biology: while it predominantly fuels tumor growth and metastasis, USP13 occasionally functions as a tumor suppressor. USP13 is as a formidable factor in cancer therapy, fortifying tumors against an arsenal of treatments. It bolsters DNA repair mechanisms, ignites prosurvival autophagy, and even reprograms cell lineages to evade targeted therapies. However, USP13 is also a promising target in the treatment of cancer. We highlight burgeoning strategies to neutralize USP13, from small molecule inhibitors to innovative protein degraders, which may disarm cancer resistance mechanisms. We also offer suggestions for future USP13 research, emphasizing the need for structural insights and more potent inhibitors. This review highlights the critical role of USP13 in cancer and underscores its potential as a therapeutic target for advancing cancer treatment.
Collapse
Affiliation(s)
- Lina Yi
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China; Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Akezhouli Shahatiaili
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Haihua He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leifeng Chen
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fushan Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Shao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yibo Gao
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, China; Department of Gastroenterology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancers Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| | - Jie He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, China.
| |
Collapse
|
6
|
Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M, Xia Y. Metabolic reprogramming and interventions in angiogenesis. J Adv Res 2025; 70:323-338. [PMID: 38704087 PMCID: PMC11976431 DOI: 10.1016/j.jare.2024.05.001] [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: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Endothelial cell (EC) metabolism plays a crucial role in the process of angiogenesis. Intrinsic metabolic events such as glycolysis, fatty acid oxidation, and glutamine metabolism, support secure vascular migration and proliferation, energy and biomass production, as well as redox homeostasis maintenance during vessel formation. Nevertheless, perturbation of EC metabolism instigates vascular dysregulation-associated diseases, especially cancer. AIM OF REVIEW In this review, we aim to discuss the metabolic regulation of angiogenesis by EC metabolites and metabolic enzymes, as well as prospect the possible therapeutic opportunities and strategies targeting EC metabolism. KEY SCIENTIFIC CONCEPTS OF REVIEW In this work, we discuss various aspects of EC metabolism considering normal and diseased vasculature. Of relevance, we highlight that the implications of EC metabolism-targeted intervention (chiefly by metabolic enzymes or metabolites) could be harnessed in orchestrating a spectrum of pathological angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Yun Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yikun Li
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yating Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Miaomiao Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
7
|
Wang Z, He Z, Chang X, Xie L, Song Y, Wu H, Zhang H, Wang S, Zhang X, Bai Y. Mitochondrial damage-associated molecular patterns: New perspectives for mitochondria and inflammatory bowel diseases. Mucosal Immunol 2025; 18:290-298. [PMID: 39920995 DOI: 10.1016/j.mucimm.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/17/2024] [Accepted: 01/30/2025] [Indexed: 02/10/2025]
Abstract
Mitochondria are key regulators of inflammatory responses and mitochondrial dysfunction is closely linked to various inflammatory diseases. Increasing genetic and experimental evidence suggests that mitochondria play a critical role in inflammatory bowel disease (IBD). In the complex environment of the intestinal tract, intestinal epithelial cells (IECs) and their mitochondria possess unique phenotypic features, shaping each other and regulating intestinal homeostasis and inflammation through diverse mechanisms. Here, we focus on intestinal inflammation in IBD induced by mitochondrial damage-associated molecular patterns (mtDAMPs), which comprise mitochondrial components and metabolic products. The pathogenic mechanisms of mtDAMP signaling pathways mediated by two major mtDAMPs, mitochondrial DNA (mtDNA) and mitochondrial reactive oxygen species (mtROS), are discussed.
Collapse
Affiliation(s)
- Zhijie Wang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China; National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zixuan He
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Xin Chang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Lu Xie
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China
| | - Yihang Song
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Haicong Wu
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Hao Zhang
- The Sixth Student Team, College of Basic Medical Sciences, Naval Medical University, Shanghai 200433, China
| | - Shuling Wang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China.
| | - Yu Bai
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
8
|
Kjaergaard U, Lund A, Redda M, Kristensen MH, Aastrup M, Bøgh N, Sivesgaard K, Ohliger MA, Vigneron DB, Bertelsen LB, Alstrup AKO, Hansen ESS, Mortensen FV, Laustsen C. Regional quantification of metabolic liver function using hyperpolarized [1- 13C] pyruvate MRI. Sci Rep 2025; 15:10482. [PMID: 40140493 PMCID: PMC11947255 DOI: 10.1038/s41598-025-93725-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Assessment of liver function is essential before partial hepatectomy to predict the risk of post hepatectomy liver failure, a severe and life-threatening complication. Traditional methods have focused on expected future liver remnant (FLR) volume estimation. However, liver volume does not always correlate with function. We suggest that metabolism might be a better surrogate for function than volume. Therefore, we aimed to investigate the metabolic changes in a porcine model of partial portal vein ligation (PVL) using hyperpolarized magnetic resonance imaging (HP-MRI). Specifically, we sought to quantify and compare the pyruvate metabolism in the FLR and the deportalized liver (DL).Six pigs underwent PVL. HP-MRI with [1-13C] pyruvate was performed at baseline, post-surgery, and 1 week after surgery. Metabolic conversion was quantified with kinetic modelling of the rate constants of pyruvate to lactate (kPL) and pyruvate to alanine (kPA). Mean kPL was increased in FLR compared to DL at post-surgery and 1 week after surgery (P = 0.002), while kPA was unaltered (P = 0.761). These findings indicate a metabolic shift towards glycolysis in the FLR. This non-invasive metabolic imaging technique could serve as a powerful tool for evaluation of regional liver function prior to partial hepatectomy and consequently improve patient outcomes.
Collapse
Affiliation(s)
- Uffe Kjaergaard
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Surgery, Section for HPB Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark.
- Department of Surgery, Horsens Regional Hospital, Horsens, Denmark.
| | - Andrea Lund
- Department of Surgery, Section for HPB Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Mohsen Redda
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Malene Aastrup
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nikolaj Bøgh
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Kim Sivesgaard
- Department of Radiology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Michael A Ohliger
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Daniel B Vigneron
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Lotte Bonde Bertelsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Aage Kristian Olsen Alstrup
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Frank Viborg Mortensen
- Department of Surgery, Section for HPB Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
9
|
Zhou S, Zhang L, You Y, Yu K, Tie X, Gao Y, Chen Y, Yao F, Zhang R, Hao X, Fang C, Li X, Li Q, Wang X. eIF3f promotes tumour malignancy by remodelling fatty acid biosynthesis in hepatocellular carcinoma. J Hepatol 2025:S0168-8278(25)00206-5. [PMID: 40154622 DOI: 10.1016/j.jhep.2025.02.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND & AIMS Fatty acid metabolism is closely associated with hepatocellular carcinoma (HCC). Elucidating the molecules that influence fatty acid metabolism in HCC is important for developing precise therapy. However, elucidating its molecular mechanisms in tumour cells is challenging. In this study, we aimed to determine the characteristics and differences of fatty acid metabolism in HCC. METHODS We employed organoid models, single-cell RNA sequencing, and spatial transcriptomics to identify key genes involved in tumour fatty acid metabolism. Metabolomics, proteomics, metabolic flux analysis, and transmission electron microscopy were utilized to evaluate this metabolic process. Tumour malignancy was characterized using multi-species models. Changes in the immune microenvironment were analyzed by time-of-flight mass cytometry and multiplexed immunohistochemistry. Gene knockdown targeting the liver was achieved using lipid nanoparticles. RESULTS Eukaryotic translation initiation factor 3 subunit f (eIF3f) is upregulated in HCC tissues and is associated with poor prognosis. eIF3f directly interacted with and stabilised long chain acyl CoA synthetase 4 (ACSL4) through K48-linked deubiquitination, promoting fatty acid biosynthesis (FAB) and tumour malignancy. The increased fatty acid levels in the tumour microenvironment indirectly reduced CD8+ T-cell infiltration. In addition, phosphorylated eIF3f enhanced the interaction between eIF3f and ACSL4. CONCLUSIONS Targeting the eIF3f-ACSL4-FAB axis could decelerate HCC malignancy and enhance anti-programmed cell death-1 efficacy, suggesting that eIF3f is a potential target for precision therapy in HCC.
Collapse
Affiliation(s)
- Suiqing Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Liren Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Yue You
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Kai Yu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Xiaofeng Tie
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Yun Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Yining Chen
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Feifan Yao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Ruizhi Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Xiaopei Hao
- Department of Hepatobiliopancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Chunyao Fang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Xiangdong Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China
| | - Qing Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China.
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Key Laboratory of Hepatobiliary Tumors, National Health Commission, Jiangsu Provincial Medical Innovation Center, Jiangsu Provincial Medical Key Laboratory, Nanjing, Jiangsu Province, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
10
|
Wu B, Woo JS, Hasiakos S, Pan C, Cokus S, Benincá C, Stiles L, Sun Z, Pellegrini M, Shirihai OS, Lusis AJ, Srikanth S, Gwack Y. Mitochondrial reactive oxygen species regulate acetyl-CoA flux between cytokine production and fatty acid synthesis in effector T cells. Cell Rep 2025; 44:115430. [PMID: 40088449 DOI: 10.1016/j.celrep.2025.115430] [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: 08/08/2024] [Revised: 01/13/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Genetic and environmental factors shape an individual's susceptibility to autoimmunity. To identify genetic variations regulating effector T cell functions, we used a forward genetics screen of inbred mouse strains and uncovered genomic loci linked to cytokine expression. Among the candidate genes, we characterized a mitochondrial inner membrane protein, TMEM11, as an important determinant of Th1 responses. Loss of TMEM11 selectively impairs Th1 cell functions, reducing autoimmune symptoms in mice. Mechanistically, Tmem11-/- Th1 cells exhibit altered cristae architecture, impaired respiration, and increased mitochondrial reactive oxygen species (mtROS) production. Elevated mtROS hindered histone acetylation while promoting neutral lipid accumulation. Further experiments using genetic, biochemical, and pharmacological tools revealed that mtROS regulate acetyl-CoA flux between histone acetylation and fatty acid synthesis. Our findings highlight the role of mitochondrial cristae integrity in directing metabolic pathways that influence chromatin modifications and lipid biosynthesis in Th1 cells, providing new insights into immune cell metabolism.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jin Seok Woo
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Spyridon Hasiakos
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shawn Cokus
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cristiane Benincá
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Linsey Stiles
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zuoming Sun
- Department of Immunology & Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Cellular Integrative Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aldon J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
11
|
Wang M, Xu S, Xu J, Wei J, Wu Y. WTAP contributes to platinum resistance in high-grade serous ovarian cancer by up-regulating malic acid: insights from liquid chromatography and mass spectrometry analysis. Cancer Metab 2025; 13:14. [PMID: 40098185 PMCID: PMC11916999 DOI: 10.1186/s40170-025-00383-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
High-grade serous cancer (HGSC) is the most prevalent and aggressive subtype of ovarian cancer. In this study, we utilized liquid chromatography and mass spectrometry analysis to investigate metabolic alterations in HGSC. Among the 1353 metabolites identified, 140 were significantly differed between HGSC and normal ovarian tissue. KEGG pathway enrichment analysis revealed 23 distinct metabolic pathways, including the alanine/aspartate/glutamate metabolism, pyruvate metabolism, biosynthesis of amino acids, and citrate cycle, etc. Of the significantly differentiated metabolites, malic acid, fumarate, and phosphoenolpyruvate were found in the citrate cycle and glycolysis. In further analysis, 22 differentially expressed genes (DEGs) of glucose metabolism were found between HGSC and normal controls. Multivariate Cox analysis of the 22 DEGs showed that ME1, ALDOC, and RANBP2 were associated with overall survival in the TCGA cohort.Bioinformatic analysis indicated WTAP is strongly correlated to the expression of ME1, which is a rate-limiting enzyme that regulates the shuttle of malic acid in mitochondria and cytoplasm. After the knockdown of WTAP in A2780 and OVCAR-3 cells, the activity of the malic enzyme decreased which led to the accumulation of malic acid and citric acid, and the reduction of pyruvate and lactic acid. In A2780 and OVCAR-3 cells, the IC50 to platinum was increased after the knockdown of WTAP. After the knockdown of WTAP, the expression of ME1 was down-regulated and the m6A modification was down-regulated in ovarian cell lines. On the SRAMP website, there were two binding sites with high m6A scores at the 5 '-UTR 177 and 970 of ME1 mRNA. WTAP contributes to the platinum resistance through regulating the conversion from aerobic glycolysis to OXPHOS by upregulating the expression of ME1.
Collapse
Affiliation(s)
- Ming Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Shuiqing Xu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Jianqing Xu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Jiahui Wei
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Yumei Wu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China.
| |
Collapse
|
12
|
Jiang M, Huang J, Guo X, Fu W, Peng L, Wang Y, Liu W, Liu J, Zhou L, Xiao Y. HIF-3α/PPAR-γ Regulates Hypoxia Tolerance by Altering Glycolysis and Lipid Synthesis in Blunt Snout Bream ( Megalobrama amblycephala). Int J Mol Sci 2025; 26:2613. [PMID: 40141255 PMCID: PMC11942064 DOI: 10.3390/ijms26062613] [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: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Hypoxic stress causes cell damage and serious diseases in organisms, especially in aquatic animals. It is important to elucidate the changes in metabolic function caused by hypoxia and the mechanisms underlying these changes. This study focuses on the low oxygen tolerance feature of a new blunt snout bream strain (GBSBF1). Our data show that GBSBF1 has a different lipid and carbohydrate metabolism pattern than wild-type bream, with altering glycolysis and lipid synthesis. In GBSBF1, the expression levels of phd2 and vhl genes are significantly decreased, while the activation of HIF-3α protein is observed to have risen significantly. The results indicate that enhanced HIF-3α can positively regulate gpd1ab and gpam through PPAR-γ, which increases glucose metabolism and reduces lipolysis of GBSBF1. This research is beneficial for creating new aquaculture strains with low oxygen tolerance traits.
Collapse
Affiliation(s)
- Minggui Jiang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jing Huang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Xing Guo
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Wen Fu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Liangyue Peng
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Wenbin Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jinhui Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Yamei Xiao
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| |
Collapse
|
13
|
Cui Y. Analysis of Long Noncoding RNA in Fatty Acid Metabolism to Identify Prognostic Markers and Predict Immunotherapy Response in Low-Grade Glioma. World Neurosurg 2025; 196:123723. [PMID: 39952400 DOI: 10.1016/j.wneu.2025.123723] [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: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Low-grade gliomas (LGGs) are notorious for their difficult early-stage diagnosis, limited treatment options, and poor prognosis, making them a focal point in cancer research. Long noncoding RNAs (lncRNAs) have been identified as regulators of metabolic reprogramming in tumor cells, offering new directions for LGG treatment. METHODS This study employed data from The Cancer Genome Atlas, focusing on key fatty acid metabolism-related lncRNA. A risk scoring model was developed using univariate/multifactorial and least absolute shrinkage and selection operator Cox regression. Additionally, the study evaluated the role of these prognostic lncRNAs in LGG progression by assessing associations between LGG immune markers and tumor drug resistance. Finally, functional enrichment analysis highlighted the molecular roles of these lncRNAs. RESULTS In this study, a total of 14 prognostic lncRNAs were obtained. The risk model demonstrated excellent validity and reliability, making it a superior predictor of prognosis among patients with varying LGG risks. Among the identified lncRNAs, GHET-1 was notably associated with LGG sensitivity to current chemotherapy options and might be a crucial lncRNA affecting LGG progression. High-risk patients exhibited T-helper cell-mediated immunosuppression, potentially paving new paths for future LGG immunotherapy. CONCLUSIONS Focusing on lncRNA regulation and fatty acid metabolism reprogramming, this study established an innovative prognostic prediction model for LGGs, showing outstanding validity and reliability. The findings offer new molecular and cellular targets for the future development of LGG treatments.
Collapse
Affiliation(s)
- Yang Cui
- Department of Neurosurgery, Hebei Yanda Hospital, Langfang, He Bei, China.
| |
Collapse
|
14
|
Zhang J, Xu S, Fang H, Wu D, Ouyang C, Shi Y, Hu Z, Zhang M, Zhong Y, Zhao J, Gan Y, Zhang S, Liu X, Yin J, Li Y, Tang M, Wang Y, Li L, Chan WC, Horne D, Feng M, Huang W, Gu Y. CAMKIIδ Reinforces Lipid Metabolism and Promotes the Development of B Cell Lymphoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409513. [PMID: 39840457 PMCID: PMC11905072 DOI: 10.1002/advs.202409513] [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: 08/12/2024] [Revised: 12/14/2024] [Indexed: 01/23/2025]
Abstract
The most prevalent types of lymphomas are B cell lymphomas (BCL). Newer therapies for BCL have improved the prognosis for many patients. However, approximately 30% with aggressive BCL either remain refractory or ultimately relapse. These patients urgently need other options. This study shows how calcium/calmodulin-dependent protein kinase II delta (CAMKIIδ) is pivotal for BCL development. In BCL cells, ablation of CAMKIIδ inhibits both lipolysis from lipid droplets and oxidative phosphorylation (OXPHOS). With lipolysis blocked, BCL progression is markedly suppressed in two distinct BCL mouse models: MYC-driven EµMyc mice and Myc/Bcl2 double-expressed mice. When CAMKIIδ is present, it destabilizes transcription factor Forkhead Box O3A (FOXO3A) by phosphorylating it at Ser7 and Ser12. This then permits transcription of downstream gene IRF4 - a master transcription factor of lipid metabolism. The CAMKIIδ/FOXO3A axis bolsters lipid metabolism, mitochondrial respiration, and tumor fitness in BCL under metabolic stress. This study also evaluates Tetrandrine (TET), a small molecule compound, as a potent CAMKIIδ inhibitor. TET attenuates metabolic fitness and elicits therapeutic responses both in vitro and in vivo. Collectively, this study highlights how CAMKIIδ is critical in BCL progression. The results also pave the way for innovative therapeutic strategies for treating aggressive BCL.
Collapse
Affiliation(s)
- Jiawei Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Senlin Xu
- Molecular and Cellular Biology of Cancer Program & Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Center for Genetic Medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Hui Fang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Dehao Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Department of Digestive, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Ching Ouyang
- Integrative Genomic Core, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Yunfei Shi
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Zhenkang Hu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Mingfeng Zhang
- Molecular and Cellular Biology of Cancer Program & Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Yaoyao Zhong
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Junwei Zhao
- Center for Genetic Medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yichao Gan
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Shize Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoqian Liu
- Molecular and Cellular Biology of Cancer Program & Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Department of Hematology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, 264000, China
| | - Jie Yin
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yuan Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Mengyue Tang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yingda Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ling Li
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Wing C Chan
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - David Horne
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Mingye Feng
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Department of Immuno-oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Wendong Huang
- Molecular and Cellular Biology of Cancer Program & Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Ying Gu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Center for Genetic Medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Zhejiang Provincial Key Lab of Genetic and Developmental Disorder, Hangzhou, 310058, China
| |
Collapse
|
15
|
Han Z, Yan Z, Ma Z, Wang Y, Beus M, Lu J, Weidenhammer LB, Lakhani K, Lee J, Civils JD, Furdui CM, Liu L, Wu J, Kang Y, Bieberich E, Boise LH, Nikiforov MA. Targeting ABCD1-ACOX1-MET/IGF1R axis suppresses multiple myeloma. Leukemia 2025; 39:720-733. [PMID: 39885295 DOI: 10.1038/s41375-025-02522-9] [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] [Received: 07/12/2024] [Revised: 01/03/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Multiple myeloma (MM) remains an incurable hematological malignancy that necessitates the identification of novel therapeutic strategies. Here, we report that intracellular levels of very long chain fatty acids (VLCFAs) control the cytotoxicity of MM chemotherapeutic agents. Inhibition of VLCFA biosynthesis reduced cell death in MM cells caused by the proteasome inhibitor, bortezomib. Conversely, inhibition of VLCFA degradation via suppression of peroxisomal acyl-CoA oxidase 1 (ACOX1) increased the cytotoxicity of bortezomib, its next-generation analog, carfilzomib, and the immunomodulatory agent lenalidomide. Furthermore, treatment with an orally available ACOX1 inhibitor cooperated with bortezomib in suppressing the growth of bortezomib-resistant MM xenografts in mice. Increased VLCFA levels caused by genetic or pharmacological inhibition of VLCFA degradation reduced the activity of two major kinases involved in MM pathogenesis, MET proto-oncogene (MET) and insulin-like growth factor 1 receptor (IGF1R). Mechanistically, inhibition of ACOX1 promoted the accumulation of VLCFA-containing cerebrosides, altered MET and IGF1R interaction with a cerebroside analog, and selectively inhibited the association of these kinases with the plasma membrane signaling platforms, importantly, without disrupting the platforms' integrity. Our study revealed a specific metabolic vulnerability of MM cells and identified a targetable axis linking VLCFA metabolism to the regulation of MET and IGF1R activity.
Collapse
Affiliation(s)
- Zhannan Han
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Zhibo Yan
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Zhehan Ma
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Yihui Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Maja Beus
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Junqi Lu
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Loren B Weidenhammer
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kiran Lakhani
- Department of Hematology and Medical Oncology Emory School of Medicine and the Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Jingyun Lee
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - John D Civils
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Liang Liu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Jian Wu
- Division of Hematologic Malignancies & Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yubin Kang
- Division of Hematologic Malignancies & Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky College of Medicine, 741 S. Limestone BBSRB Room 269, Lexington, KY, 40536, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology Emory School of Medicine and the Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Mikhail A Nikiforov
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
16
|
Bathe OF. Tumor metabolism as a factor affecting diversity in cancer cachexia. Am J Physiol Cell Physiol 2025; 328:C908-C920. [PMID: 39870605 DOI: 10.1152/ajpcell.00677.2024] [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: 09/16/2024] [Revised: 09/21/2024] [Accepted: 01/20/2025] [Indexed: 01/29/2025]
Abstract
Cancer cachexia is a multifaceted metabolic syndrome characterized by muscle wasting, fat redistribution, and metabolic dysregulation, commonly associated with advanced cancer but sometimes also evident in early-stage disease. More subtle body composition changes have also been reported in association with cancer, including sarcopenia, myosteatosis, and increased fat radiodensity. Emerging evidence reveals that body composition changes including sarcopenia, myosteatosis, and increased fat radiodensity, arise from distinct biological mechanisms and significantly impact survival outcomes. Importantly, these features often occur independently, with their combined presence exacerbating poor prognoses. Tumor plays a pivotal role in driving these host changes, either by acting as a metabolic parasite or by releasing mediators that disrupt normal tissue function. This review explores the diversity of tumor metabolism. It highlights the potential for tumor-specific metabolic phenotypes to influence systemic effects, including fat redistribution and sarcopenia. Addressing this tumor-host metabolic interplay requires personalized approaches that disrupt tumor metabolism while preserving host health. Promising strategies include targeted pharmacological interventions and anticachexia agents like growth differentiation factor 15 (GDF-15) inhibitors. Nutritional modifications such as ketogenic diets and omega-3 fatty acid supplementation also merit further investigation. In addition to preserving muscle, these therapies will need to be evaluated for their capability to improve survival and quality of life. This review underscores the need for further research into tumor-driven metabolic effects on the host and the development of integrative treatment strategies to address the interconnected challenges of cancer progression and cachexia.
Collapse
Affiliation(s)
- Oliver F Bathe
- Department of Surgery and Oncology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
17
|
Yang X, Ma B, Liu Y, Zhou J, Guo J, Peng Y, Bai Y, Wu J, Hu D. SSBP1 positively regulates RRM2, affecting epithelial mesenchymal transition and cell cycle arrest in human lung adenocarcinoma cells. Cell Signal 2025; 127:111552. [PMID: 39643024 DOI: 10.1016/j.cellsig.2024.111552] [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: 10/15/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Progression of lung adenocarcinoma (LUAD) is frequently associated with alterations in epithelial-mesenchymal transition (EMT) and cell cycle. Our study analyzed the Cancer Genome Atlas (TCGA) database and identified a positive correlation between high expression of SSBP1 in LUAD tumor tissues and poor prognosis (p < 0.05), with an AUC of 0.853, suggesting that SSBP1 could serve as a prognostic biomarker. In vitro experiments, including siRNA-mediated SSBP1 knockdown and subsequent cell cloning and Transwell assays, revealed significant inhibition of proliferation, migration, and cell cycle progression in LUAD cells (p < 0.05). In vivo mouse model experiments further confirmed that SSBP1 knockdown inhibits tumor burden (p < 0.05). Mechanistic investigations, integrating pathway enrichment analysis with molecular biology techniques, identified RRM2 as a downstream target of SSBP1, and RRM2 knockdown similarly suppressed LUAD cell proliferation, migration, and cell cycle progression (p < 0.05). These findings indicate that SSBP1 promotes EMT and cell cycle progression in LUAD cells by positively regulating RRM2, thereby accelerating disease progression. Collectively, our study not only confirms the potential role of SSBP1 in LUAD but also provides a theoretical foundation for therapeutic strategies targeting the SSBP1/RRM2 axis, potentially offering new therapeutic targets for LUAD patients.
Collapse
Affiliation(s)
- Xingyu Yang
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Bingfeng Ma
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Yafeng Liu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jiawei Zhou
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jianqiang Guo
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Yuanyuan Peng
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Ying Bai
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, School of Medicine, Anhui University of Science and Technology, Huainan City, China.
| | - Jing Wu
- Joint Research Center for Occupational Medicine and Health of IHM, School of Medicine, Anhui University of Science and Technology, Huainan City, China.
| | - Dong Hu
- The First Affiliated Hospital of Anhui University of Science and Technology (Huainan First People's Hospital), School of Medicine, Huainan City, China; Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei City, China.
| |
Collapse
|
18
|
Dhar S, Sarkar T, Bose S, Pati S, Chakraborty D, Roy D, Panda AK, Guin A, Mukherjee S, Jana K, Sarkar DK, Sa G. FOXP3 Transcriptionally Activates Fatty Acid Scavenger Receptor CD36 in Tumour-Induced Treg Cells. Immunology 2025; 174:296-309. [PMID: 39736083 DOI: 10.1111/imm.13887] [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: 01/20/2024] [Revised: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
The host immune system is adapted in a variety of ways by tumour microenvironment and growing tumour interacts to promote immune escape. One of these adaptations is manipulating the metabolic processes of cells in the tumour microenvironment. The growing tumour aggressively utilise glucose, its primary energy source available in tumour site, and produce lactate by Warburg effect. In such a hostile environment, tumour-infiltrating immune cells are unable to survive metabolically. Tumour-infiltrating CD4+ Treg cells, on the other hand, adapted to an alternative energy-generating system, switching from the highly-competitive glucose to the fatty-acid metabolic pathway, by down-regulating glucose-metabolising genes and up-regulating fatty-acid metabolising genes. Tregs with high-levels of the fatty acid scavenger receptor CD36, a key component of the fatty-acid metabolic pathway, aided this metabolic shift. Treg cell formation was hampered when the fatty-acid metabolic pathway was disrupted, showing that it is necessary for Treg cell development. FOXP3, the Treg lineage-specific transcription factor, regulates fatty-acid metabolism by inducing CD36 transcription. A high-fat diet enhanced Treg development while suppressing anti-tumour immunity, whereas a low-fat diet suppressed Treg development. The altered metabolism of tumour-infiltrating Treg cells enables their rapid generation and survival in the hostile tumour microenvironment, aiding cancer progression. Fascinatingly, mice fed with a low-fat diet showed a positive prognosis with chemotherapy than mice fed with a high-fat diet. Thus, a maximum efficacy of chemotherapy might be achieved by altering diet composition during chemotherapy, providing a promising indication for future cancer treatment.
Collapse
Affiliation(s)
- Subhanki Dhar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Tania Sarkar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sayantan Bose
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Subhadip Pati
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Dia Roy
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Abir K Panda
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Aharna Guin
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sumon Mukherjee
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
19
|
Huang X, Li Q, Xu M, Sun S, Gong Y, Luan R, Wang M, Shao Y, Li X. The interplay between metabolic reprogramming, mitochondrial impairment, and steroid response in proliferative vitreoretinopathy. Free Radic Biol Med 2025; 229:485-498. [PMID: 39826818 DOI: 10.1016/j.freeradbiomed.2025.01.033] [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: 09/26/2024] [Revised: 12/23/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Proliferative vitreoretinopathy (PVR) is a major cause of rhegmatogenous retinal detachment repair failure. Despite many attempts to find therapeutics for PVR, no pharmacotherapy has been proven effective. Steroids, as the epitome, show uncertain clinical effectiveness, which lacks an explanation and hints at unappreciated mechanisms of PVR. In this study, we investigated the involvement of metabolic reprogramming, mitochondrial impairment, and their association with steroid effectiveness in PVR using dexamethasone (Dex) as an example. Proteomics of vitreous samples from PVR patients demonstrated an upregulation in the glycolysis pathway. Transcriptomics of PVR tissues (dataset GSE179603) revealed downregulations in oxidative phosphorylation (OXPHOS), mitochondrial respiration, and mitochondrial quality control-related pathways. Transcriptomics of TGFβ and TNFα (TNT)-induced retinal pigment epithelial (RPE) cell model (GSE176513) confirmed the changes in glycolysis, OXPHOS, and mitochondria and also revealed downregulation of Dex response pathway with increased duration of TNT exposure. Transcriptomics of mouse RPE/choroid following Dex intravitreal injections (GSE49872) showed that glycolysis decreased at 1-week postinjection but increased at 1-month postinjection; OXPHOS increased but gradually decreased with treatment duration. The dispase-induced mouse PVR model revealed that a simultaneous Dex injection could alleviate PVR severity rather than an injection 5 days after the PVR induction. The TGFβ2-induced RPE cell model demonstrated the enhancement of EMT, oxidative stress, and mitochondrial impairment, which could be alleviated by Dex: Cellular ROS were accumulated; the mRNA expressions of antioxidases (GPX, SOD1 and TXN2) were decreased; mitochondrial morphology and dynamics were impaired, exhibiting decreases in mitochondrial heterogeneity, mitochondrial length and MFN2 expression; Mitochondrial membrane potential showed an elevation; and mitophagy was decreased, related to reduced Parkin recruitment. These results demonstrate the essential roles of metabolic reprogramming and mitochondrial dysfunction in PVR pathology, which is associated with the therapeutic effect of steroids. Steroid intervention might benefit the treatment of PVR in the early rather than late stages.
Collapse
Affiliation(s)
- Xinyuan Huang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China
| | - Qingbo Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China
| | - Manhong Xu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China
| | - Shuo Sun
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China
| | - Yi Gong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China
| | - Rong Luan
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China
| | - Manqiao Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China
| | - Yan Shao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, PR China, No.251 Fu Kang Road, Nankai District, Tianjin, 300384, PR China.
| |
Collapse
|
20
|
Choi H, Gupta M, Sengupta A, Furth EE, Hensley C, Weljie AM, Lee H, Lu YT, Pantel A, Mankoff D, Zhou R. Disruption of redox balance in glutaminolytic triple negative breast cancer by inhibition of glutaminase and glutamate export. Neoplasia 2025; 61:101136. [PMID: 39938153 PMCID: PMC11869985 DOI: 10.1016/j.neo.2025.101136] [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: 10/28/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
Resistance to chemotherapy is an important challenge in the clinical management of triple-negative breast cancer (TNBC). Utilization of the amino acid glutamine as a key nutrient is a metabolic signature of TNBC featuring high glutaminase (GLS) activity and a large pool of cellular glutamate, which mediates intracellular enrichment of cystine via xCT (SLC7A11) antiporter activity. To overcome chemo-resistant TNBC, we identified a strategy of dual metabolic inhibition of GLS and xCT to sensitize resistant TNBC cells to chemotherapy. We successfully tested this strategy in a human TNBC line and its chemoresistant variant in vitro and their xenograft models in vivo. Key findings of our study include: 1. Dual metabolic inhibition induced pronounced reductions of cellular glutathione accompanying significant increases of cellular superoxide level in both parent and resistant TNBC cells. While GLS and xCT inhibition did not directly kill cells via apoptosis, they potentiated doxorubicin (DOX) and cisplatin (CIS) to induce remarkably higher levels of apoptosis than DOX or CIS alone. 2. Although the resistant TNBC cells exhibited higher capacity to mitigate oxidative stress than the parent cells, their resistance was overcome by dual metabolic inhibition combined with DOX or CIS. 3. In vivo efficacy and safety of the triple combination (GLS and xCT inhibition plus DOX or CIS) were demonstrated in both chemo sensitive and resistant TNBC tumors in mice. In conclusion, GLS and xCT inhibition resulted in unmitigated oxidative stress due to depletion of glutathione, representing a promising strategy to overcome chemoresistance in glutamine-dependent TNBC.
Collapse
Affiliation(s)
- Hoon Choi
- Department of Radiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mamta Gupta
- Department of Radiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arjun Sengupta
- Department of Systems Pharmacology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma E Furth
- Department of Pathology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Hensley
- Department of Radiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hsiaoju Lee
- Department of Radiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yu-Ting Lu
- Department of Radiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Austin Pantel
- Department of Radiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Mankoff
- Department of Radiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Zhou
- Department of Radiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Sarami I, Hekman KE, Gupta AK, Snider JM, Ivancic D, Zec M, Kandpal M, Ben-Sahra I, Menon R, Otto EA, Chilton FH, Wertheim JA. Parallel multiOMIC analysis reveals glutamine deprivation enhances directed differentiation of renal organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640060. [PMID: 40060393 PMCID: PMC11888470 DOI: 10.1101/2025.02.27.640060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Metabolic pathways play a critical role in driving differentiation but remain poorly understood in the development of kidney organoids. In this study, parallel metabolite and transcriptome profiling of differentiating human pluripotent stem cells (hPSCs) to multicellular renal organoids revealed key metabolic drivers of the differentiation process. In the early stage, transitioning from hPSCs to nephron progenitor cells (NPCs), both the glutamine and the alanine-aspartate-glutamate pathways changed significantly, as detected by enrichment and pathway impact analyses. Intriguingly, hPSCs maintained their ability to generate NPCs, even when deprived of both glutamine and glutamate. Surprisingly, single cell RNA-Seq analysis detected enhanced maturation and enrichment for podocytes under glutamine-deprived conditions. Together, these findings illustrate a novel role of glutamine metabolism in regulating podocyte development.
Collapse
Affiliation(s)
- Iman Sarami
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Hematopathology and Molecular Diagnostics Laboratory at the University of Texas MD, Anderson Cancer Center, Houston, TX
| | - Katherine E. Hekman
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Jesse Brown VA Medical Center, Chicago, IL
- Atlanta VA Healthcare System, Decatur, GA
- Department of Surgery, Emory University School of Medicine, Atlanta, GA
| | - Ashwani Kumar Gupta
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ
- Southern Arizona VA Healthcare System, Tucson, AZ
| | - Justin M. Snider
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ
| | - David Ivancic
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Jesse Brown VA Medical Center, Chicago, IL
| | - Manja Zec
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Manoj Kandpal
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Center for Clinical and Translational Science, Rockefeller University Hospital, New York, NY
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| | - Edgar A. Otto
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Floyd H. Chilton
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ
| | - Jason A. Wertheim
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ
- Southern Arizona VA Healthcare System, Tucson, AZ
| |
Collapse
|
22
|
Luo Y, Zhang N, Ye J, Wang Z, Zhou X, Liu J, Cai J, Li C, Chen L. Unveiling lactylation modification: A new hope for cancer treatment. Biomed Pharmacother 2025; 184:117934. [PMID: 39986235 DOI: 10.1016/j.biopha.2025.117934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025] Open
Abstract
This review article delves into the multifaceted role of lactylation modification in antitumor therapy, revealing the complex interplay between lactylation modification and the tumor microenvironment (TME), metabolic reprogramming, gene expression, and immunotherapy. As an emerging epigenetic modification, lactylation has a significant impact on the metabolic pathways of tumor cells, immune evasion, gene expression regulation, and sensitivity to chemotherapy drugs. Studies have shown that lactylation modification significantly alters the development and therapeutic response of tumors by affecting metabolites in the TME, immune cell functions, and signaling pathways. In the field of immunotherapy, the regulatory role of lactylation modification provides a new perspective and potential targets for tumor treatment, including modulating the sensitivity of tumors to immunotherapy by affecting the expression of immune checkpoint molecules and the infiltration of immune cells. Moreover, research progress on lactylation modification in various types of tumors indicates that it may serve as a biomarker to predict patients' responses to chemotherapy and immunotherapy. Overall, research on lactylation modification provides a theoretical foundation for the development of new tumor treatment strategies and holds promise for improving patient prognosis and quality of life. Future research will further explore the application potential of lactylation modification in tumor therapy and how to improve treatment efficacy by targeting lactylation modification.
Collapse
Affiliation(s)
- Yuxiang Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Ning Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Zuao Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Xinchi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jipeng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Chen Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi 330006, China; Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Jiangxi 330006, China; Institute of Minimally Invasive Orthopedics, Nanchang University, Jiangxi 330006, China.
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Precision Oncology Medicine Center,The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China.
| |
Collapse
|
23
|
Park S, Hall MN. Metabolic reprogramming in hepatocellular carcinoma: mechanisms and therapeutic implications. Exp Mol Med 2025; 57:515-523. [PMID: 40025169 PMCID: PMC11958682 DOI: 10.1038/s12276-025-01415-2] [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: 09/17/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 03/04/2025] Open
Abstract
Hepatocellular carcinoma features extensive metabolic reprogramming. This includes alterations in major biochemical pathways such as glycolysis, the pentose phosphate pathway, amino acid metabolism and fatty acid metabolism. Moreover, there is a complex interplay among these altered pathways, particularly involving acetyl-CoA (coenzyme-A) metabolism and redox homeostasis, which in turn influences reprogramming of other metabolic pathways. Understanding these metabolic changes and their interactions with cellular signaling pathways offers potential strategies for the targeted treatment of hepatocellular carcinoma and improved patient outcomes. This review explores the specific metabolic alterations observed in hepatocellular carcinoma and highlights their roles in the progression of the disease.
Collapse
Affiliation(s)
- Sujin Park
- Center for Genome Engineering, Institute for Basic Science, Daejeon, Republic of Korea.
| | | |
Collapse
|
24
|
Du W, Ouyang Y, Feng X, Yu C, Zhang H, Chen S, Liu Z, Wang B, Li X, Liu Z, Deng W. IL-10RA promotes lung cancer cell proliferation by increasing fatty acid oxidation via STAT3 signaling pathway. Pulm Pharmacol Ther 2025; 88:102344. [PMID: 39892560 DOI: 10.1016/j.pupt.2025.102344] [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: 09/20/2024] [Revised: 12/31/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
Metabolic reprogramming in tumor cells plays a crucial role in promoting cell proliferation and metastasis, and is currently recognized as a significant marker of tumor progression. Interleukin-10 receptor subunit alpha (IL-10RA), a member of the type II cytokine receptor family, is predominantly expressed on macrophages and T cells and plays a crucial role in regulating immune cell metabolism and immune response. However, its role in the energy metabolic pathways of tumor cells remains unclear. In this study, we found increased expression of IL-10RA in human non-small cell lung cancer (NSCLC), and a correlation between increased IL-10RA expression and tumor stage, tumor size, and short overall survival of patients with NSCLC. IL-10RA overexpression significantly promoted the proliferation of NSCLC cell lines and enhanced glycolysis and fatty acid oxidation (FAO), thereby boosting energy production. Correspondingly, the downregulation of IL-10RA inhibited proliferation, glycolysis, and FAO in NSCLC cell lines. Bioinformatic analyses indicated that IL-10RA upregulates the signal transducer and activator of transcription 3 (STAT3) signaling pathway. STAT3 inhibitor effectively blocked the increase in FAO levels and cell proliferation induced by IL-10RA overexpression. These findings suggest that IL-10RA accelerates NSCLC cell proliferation by increasing FAO levels via the STAT3 pathway, highlighting IL-10RA as a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Wei Du
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Yuqing Ouyang
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaofan Feng
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Chunyan Yu
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Haoke Zhang
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Siqi Chen
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Zixuan Liu
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Bo Wang
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Xueying Li
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Zihe Liu
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Weimin Deng
- Tianjin Institute of Immunology, Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
25
|
Fu Y, Sun J, Yang C, Li W, Wang Y. Diversified nanocarrier design to optimize glucose oxidase-mediated anti-tumor therapy: Strategy and progress. Int J Biol Macromol 2025; 306:141581. [PMID: 40023419 DOI: 10.1016/j.ijbiomac.2025.141581] [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: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Given the inherent complexity and heterogeneity of tumors, current therapeutic approaches often fall short in meeting prognostic requirements. Starvation therapy (ST) utilizing glucose oxidase (GOx) has emerged as a promising strategy, specifically targeting tumor glucose consumption to disrupt nutrient supply. However, the therapeutic potential of GOx is significantly hampered by its inherent limitations as a protein, particularly its poor stability and short in vivo half-life. In recent years, the development of nanocarriors has provided an effective platform for intravenous and local tumor delivery of GOx. This review systematically examines three key strategies in GOx delivery: stimulus-response, biofilm modification, and local delivery. The progress in various carrier systems for GOx-mediated tumor therapy is comprehensively summarized, providing valuable insights for nanocarrier design. Furthermore, the existing challenges and future directions to advance the development of GOx-based tumor therapies are critically analyzed.
Collapse
Affiliation(s)
- Yuhan Fu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jialin Sun
- Department of medicine, Heilongjiang Minzu College, Harbin, Heilongjiang Province, China
| | - Chunyu Yang
- Department of Pathology, Harbin 242 Hospital, Harbin, Heilongjiang Province, China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| |
Collapse
|
26
|
Pouyan A, Ghorbanlo M, Eslami M, Jahanshahi M, Ziaei E, Salami A, Mokhtari K, Shahpasand K, Farahani N, Meybodi TE, Entezari M, Taheriazam A, Hushmandi K, Hashemi M. Glioblastoma multiforme: insights into pathogenesis, key signaling pathways, and therapeutic strategies. Mol Cancer 2025; 24:58. [PMID: 40011944 DOI: 10.1186/s12943-025-02267-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor in adults, characterized by a poor prognosis and significant resistance to existing treatments. Despite progress in therapeutic strategies, the median overall survival remains approximately 15 months. A hallmark of GBM is its intricate molecular profile, driven by disruptions in multiple signaling pathways, including PI3K/AKT/mTOR, Wnt, NF-κB, and TGF-β, critical to tumor growth, invasion, and treatment resistance. This review examines the epidemiology, molecular mechanisms, and therapeutic prospects of targeting these pathways in GBM, highlighting recent insights into pathway interactions and discovering new therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Ashkan Pouyan
- Department of Neurosurgery, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Masoud Ghorbanlo
- Department of Anesthesiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Eslami
- Department of Neurosurgery, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Jahanshahi
- Department of Neurosurgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Ziaei
- Department of Neurosurgery, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Salami
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Koorosh Shahpasand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Tohid Emami Meybodi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Functional Neurosurgery Research Center, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Epidemiology, University of Tehran, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
27
|
Zhang D, Zhang X, Chang S, Zhao Y, Zhang L. E2F1 activates USP19 to affect the stability of c-Myc to facilitate the progression of hepatocellular carcinoma. Mutat Res 2025; 830:111902. [PMID: 40020513 DOI: 10.1016/j.mrfmmm.2025.111902] [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: 12/03/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common malignant tumor worldwide with a high mortality rate. Herein, this study aims to explore the molecular mechanisms of E2F transcription factor 1 (E2F1), ubiquitin specific peptidase 19 (USP19) and c-Myc in regulating HCC progression. METHODS RT-qPCR and western blotting were utilized to assess mRNA and protein expression, respectively. The behavior of cells was examined through Methylthiazolyldiphenyl-tetrazolium bromide (MTT), flow cytometry, transwell, and cell sphere formation assays. Glycolysis-related indicators were detected by kits. The interaction between USP19 and c-Myc was measured by co-immunoprecipitation (Co-IP). Dual-luciferase reporter assay and Chromatin Immunoprecipitation (ChIP) assays were used to assess the binding of E2F1 and USP19 promoter. A mouse xenograft model was established for the purpose of analysis in vivo. RESULTS High level of c-Myc was observed in HCC tissues and cells. Silencing c-Myc results in the suppression of cell migration, invasion, proliferation, and glycolysis or promotion of apoptosis. USP19 directly bound to c-Myc, and maintained its stability by removing ubiquitination on c-Myc. Overexpression of c-Myc in HCC cells rescued the anti-tumor effect of USP19 deletion. E2F1 promoted USP19 transcription, and increased USP19 expression counteracts the effects of E2F1 depletion on cell behaviors. In vivo, USP19 knockdown controlled HCC growth by modulating c-Myc. CONCLUSION E2F1 activated USP19 transcription, thereby stabilizing c-Myc via deubiquitination and accelerating HCC progression.
Collapse
Affiliation(s)
- Di Zhang
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710000, China
| | - Xinwu Zhang
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710000, China
| | - Shuai Chang
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710000, China
| | - Yao Zhao
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710000, China
| | - Li Zhang
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710000, China.
| |
Collapse
|
28
|
Du P, Li Y, Han A, Wang M, Liu J, Piao Y, Chen L. β-lapachone suppresses carcinogenesis of cervical cancer via interaction with AKT1. Front Pharmacol 2025; 16:1509568. [PMID: 40051559 PMCID: PMC11882534 DOI: 10.3389/fphar.2025.1509568] [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: 10/11/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Introduction Cervical cancer is one of the most prevalent malignant tumors affecting women worldwide, and affected patients often face a poor prognosis due to its high drug resistance and recurrence rates. β-lapachone, a quinone compound originally extracted from natural plants, is an antitumor agent that specifically targets NQO1. Methods CC cells were treated with varying concentrations of β-lapachone to examine its effects on glucose metabolism, proliferation, metastasis, angiogenesis, and EMT in vitro. The targets and action pathways of β-lapachone were identified using network pharmacology and molecular docking, with KEGG pathway enrichment analysis. Its effects and toxicity were verified in vivo using a nude mouse xenograft model. Results β-lapachone significantly inhibited the proliferation and metastasis of cervical cancer cells by regulating glucose metabolism, reducing tumor angiogenesis, and suppressing epithelial-mesenchymal transition (EMT) in cells with high NQO1 expression. Furthermore, we identified the inactivation of the PI3K/AKT/mTOR pathway as the key mechanism underlying these effects. AKT1 was identified as a potential target of β-lapachone in modulating glucose metabolism and EMT in cervical cancer cells. Conclusion These findings suggest that β-lapachone inhibits the malignant progression of cervical cancer by targeting AKT1 to regulate glucose metabolism in NQO1-overexpressing cells, providing a theoretical basis for developing novel therapeutic strategies for cervical cancer.
Collapse
Affiliation(s)
- Pan Du
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Yue Li
- Changchun Center for Disease Control and Prevention, Changchun, China
| | - Anna Han
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Mengying Wang
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Jiajing Liu
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Yingshi Piao
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
- Cancer Research Center, Yanbian University, Yanji, China
| | - Liyan Chen
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
- Cancer Research Center, Yanbian University, Yanji, China
| |
Collapse
|
29
|
Wu H, Zhang J, Wang Q, Li Z, Li L, Xie Y. Metformin combined with CB-839 specifically inhibits KRAS-mutant ovarian cancer. Sci Rep 2025; 15:6072. [PMID: 39972191 PMCID: PMC11840008 DOI: 10.1038/s41598-025-90963-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/17/2025] [Indexed: 02/21/2025] Open
Abstract
KRAS mutations can cause metabolic reprogramming in ovarian cancer, leading to an increased metastatic capacity. This study investigated the metabolic reprogramming changes induced by KRAS mutations in ovarian cancer and the mechanism of action of metformin combined with a glutaminase 1 inhibitor (CB-839). KRAS-mutant ovarian cancer accounted for 14% of ovarian cancers. The expression of glucose metabolism-related (PFKFB3, HK2, GLUT1, and PDK2) and glutamine metabolism-related enzymes (GLS1 and ASCT2) was elevated in KRAS-mutant ovarian cancer cells compared with that in wild-type cells. KRAS-mutant cells had a higher aerobic oxidative capacity than did wild-type cells. Metformin inhibited proliferation, the expression of glucose metabolism-related enzymes, and the aerobic oxidative capacity of KRAS-mutant cells compared with those of control cells. Furthermore, it enhanced the expression of glutamine metabolism-related enzymes in KRAS-mutant cells. Metformin combined with CB-839 inhibited the proliferation and aerobic oxidation of KRAS-mutant cells to a greater extent than that observed in wild-type cells. Additionally, the inhibitory effects of metformin and CB-839 in the KRAS-mutant ovarian cancer NOD-SCID mouse model were significantly stronger than those in the drug-alone group. KRAS mutations lead to enhanced glucose and glutamine metabolism in ovarian cancer cells, which was inhibited by metformin combined with CB-839.
Collapse
Affiliation(s)
- Han Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jialin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qiujie Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zijiao Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Linlin Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ya Xie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
30
|
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 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
|
31
|
Park SH, Seo JH, Kim MY, Yun HJ, Kang BK, Kim JH, Heo SV, Lee YH, Park HR, Choi MS, Lee JH. Enhanced Antitumor Activity of Korean Black Soybean Cultivar 'Soman' by Targeting STAT-Mediated Aerobic Glycolysis. Antioxidants (Basel) 2025; 14:228. [PMID: 40002413 PMCID: PMC11852074 DOI: 10.3390/antiox14020228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Black soybeans have numerous health benefits owing to their high polyphenolic content, antioxidant activity, and antitumor effects. We previously reported that the Korean black soybean cultivar 'Soman' possesses higher anthocyanin and isoflavone contents and superior antioxidant potential than other Korean black soybean cultivars and landraces (Seoritae) do. Here, we investigated and compared the antitumor effects of Soman and Seoritae and aimed to elucidate the possible mechanisms of action. Soman inhibited cancer cell proliferation and was more potent than Seoritae. Mechanistically, Soman inhibited the phosphorylation of the signal transducer and activator of transcription (STAT1, 3, and 5) in a reactive oxygen species (ROS)-independent manner, subsequently decreasing glycolytic enzyme expression and the activities of pyruvate kinase and lactate dehydrogenase. Thus, Soman suppressed glucose uptake, lactate production, and ATP production in cancer cells. Additionally, it inhibited tumor growth in a B16F10 murine melanoma syngeneic model, accompanied by reduced STAT1 phosphorylation and decreased proliferation in Soman-treated mice, more potently than observed in Seoritae-treated mice. These findings showed that Soman exerted superior antitumor activities by suppressing STAT-mediated aerobic glycolysis and proliferation. Overall, our findings demonstrate the potent, tumor-suppressive role of Soman in human cancer and uncover a novel molecular mechanism for its therapeutic effects in cancer treatment.
Collapse
Affiliation(s)
- Su Hwan Park
- Department of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; (S.H.P.); (H.J.Y.)
| | - Jeong Hyun Seo
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Miryang 50424, Republic of Korea; (M.Y.K.); (B.K.K.); (J.H.K.); (S.V.H.); (Y.H.L.); (H.R.P.); (M.S.C.)
| | - Min Young Kim
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Miryang 50424, Republic of Korea; (M.Y.K.); (B.K.K.); (J.H.K.); (S.V.H.); (Y.H.L.); (H.R.P.); (M.S.C.)
| | - Hye Jin Yun
- Department of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; (S.H.P.); (H.J.Y.)
| | - Beom Kyu Kang
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Miryang 50424, Republic of Korea; (M.Y.K.); (B.K.K.); (J.H.K.); (S.V.H.); (Y.H.L.); (H.R.P.); (M.S.C.)
| | - Jun Hoi Kim
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Miryang 50424, Republic of Korea; (M.Y.K.); (B.K.K.); (J.H.K.); (S.V.H.); (Y.H.L.); (H.R.P.); (M.S.C.)
| | - Su Vin Heo
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Miryang 50424, Republic of Korea; (M.Y.K.); (B.K.K.); (J.H.K.); (S.V.H.); (Y.H.L.); (H.R.P.); (M.S.C.)
| | - Yeong Hoon Lee
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Miryang 50424, Republic of Korea; (M.Y.K.); (B.K.K.); (J.H.K.); (S.V.H.); (Y.H.L.); (H.R.P.); (M.S.C.)
| | - Hye Rang Park
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Miryang 50424, Republic of Korea; (M.Y.K.); (B.K.K.); (J.H.K.); (S.V.H.); (Y.H.L.); (H.R.P.); (M.S.C.)
| | - Man Soo Choi
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Miryang 50424, Republic of Korea; (M.Y.K.); (B.K.K.); (J.H.K.); (S.V.H.); (Y.H.L.); (H.R.P.); (M.S.C.)
| | - Jong-Ho Lee
- Department of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; (S.H.P.); (H.J.Y.)
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea
| |
Collapse
|
32
|
Baber MA, Gough MD, Yeomans L, Giesler K, Muzzarelli K, Chen CJ, Assar Z, Toogood PL. Identification of a selective pyruvate dehydrogenase kinase 1 (PDHK1) chemical probe by virtual screening. Eur J Med Chem 2025; 284:117210. [PMID: 39742699 DOI: 10.1016/j.ejmech.2024.117210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
PDHK1 is a non-canonical Ser/Thr kinase that negatively regulates the pyruvate dehydrogenase complex (PDC), restricting entry of acetyl-CoA into the tricarboxylic acid (TCA) cycle and downregulating oxidative phosphorylation. In many glycolytic tumors, PDHK1 is overexpressed to suppress activity of the PDC and cause a shift in metabolism toward an increased reliance on glycolysis (the Warburg effect). Genetic studies have shown that knockdown or knockout of PDHK1 reverts this phenotype and inhibits tumor growth in vitro and in vivo, but chemical tools to pharmacologically validate and build upon these data are lacking. We used AtomNet®, a deep convolutional neural network bioactivity predictor, to identify compound 7 as a potential inhibitor of PDHK1. During the process of hit validation, the active species was determined to be isomeric compound 10. Structure-activity studies based on 10 identified 17 as a low μM inhibitor of PDHK1 (IC50 = 1.5 ± 0.3 μM) that is selective against the other PDHK isoforms in both biochemical and cell-based assays. In A549 epithelial lung carcinoma cells, compound 17 inhibits phosphorylation of PDC E1α Ser232, a site that is specifically phosphorylated only by PDHK1, while minimally suppressing phosphorylation of Ser293, a site that is phosphorylated by all four PDHK isoforms. Altogether, these data identify 17 as a selective PDHK1 chemical probe useful for biochemical and cell-based studies.
Collapse
Affiliation(s)
- Mason A Baber
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mya D Gough
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Larisa Yeomans
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | - Chih-Jung Chen
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Zahra Assar
- Cayman Chemical Company, Inc., Ann Arbor, MI, 48108, USA
| | - Peter L Toogood
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
33
|
Cho WC, Wong CF. Potential benefits of combined treatment with Hsp90 inhibitor AUY922 and cisplatin for overcoming drug resistance in nasopharyngeal carcinoma. Am J Cancer Res 2025; 15:533-545. [PMID: 40084353 PMCID: PMC11897633 DOI: 10.62347/osgo7209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/16/2025] [Indexed: 03/16/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) initially responds well to platinum-based therapy but often develops resistance. Combining therapies may offer a viable approach to address this resistance. Heat shock protein 90 (Hsp90) has shown promising anticancer activity in various cancer types. This study aimed to investigate the efficacy of an Hsp90 inhibitor, luminespib (AUY922), and evaluate the synergistic effect of combining AUY922 with cisplatin on two cisplatin-resistant human NPC cell lines. The response of cisplatin-resistant NPC cells to AUY922 and/or cisplatin was assessed through proliferation assay, cell cycle analysis, Annexin V apoptosis detection, Western blot analysis, in vivo investigation, and histological analysis. Our results indicated that AUY922/cisplatin combination significantly inhibited the proliferation of both non-resistant and resistant NPC cells. Moreover, Annexin V analysis indicated apoptosis when AUY922 was administered alone or in combination with cisplatin. Consistently, Western blot analysis revealed increased cleavage of PARP. Most importantly, the combination treatment demonstrated enhanced tumor growth inhibition in nude mice xenograft models, without notable adverse effects. These findings highlight the antiproliferative effects and anticancer activity of the AUY922/cisplatin combination in cisplatin-resistant NPC cells. The combination treatment of AUY922 and cisplatin holds promise as a strategy to overcome drug resistance in NPC patients.
Collapse
Affiliation(s)
- William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital Hong Kong SAR, China
| | - Chi F Wong
- Department of Clinical Oncology, Queen Elizabeth Hospital Hong Kong SAR, China
| |
Collapse
|
34
|
Cai A, Ye H, Lin Y, Li J, Fang D, Pan Z, Li Z, Luo G, Huang Y, Lai C. Circular RNA pappalysin-1 enhances glycolysis via microRNA-656-3p targeting G-protein subunit gamma-5 to promote colon cancer progression. Clinics (Sao Paulo) 2025; 80:100594. [PMID: 39951875 PMCID: PMC11874721 DOI: 10.1016/j.clinsp.2025.100594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/13/2024] [Accepted: 01/22/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Colon Cancer (CC) is a common malignant tumor. The aim of this study was to investigate the role and regulatory mechanism of circular RNA pappalysin-1 (circ-PAPPA; hsa_circ_0088233) in CC. METHODS In cancer tissues from CC patients, circ-PAPPA expression was measured and its relationship with patients' clinical features was analyzed. Plasmid vectors or oligonucleotides interfering with the expression of circ-PAPPA, microRNA (miR)-656-3p or G-protein subunit Gamma-5 (GNG5) were transfected into CC cells. Cell viability was detected by MTT and colony formation assay; apoptosis was detected by flow cytometry; and cell migration and invasion were detected by wound healing assay and Transwell. Glycolytic capacity of CC cells was assessed by measuring glucose uptake and lactate production using commercial kits. The targeting relationship between miR-656-3p and circ-PAPPA or GNG5 was verified by bioinformatics website starBase and dual luciferase reporter gene assay assays. RESULTS Circ-PAPPA was upregulated in CC and was negatively correlated with benign pathological features and 5-year survival rates of CC patients. Circ-PAPPA silencing inhibited the growth and glycolysis of CC cells through upregulating miR-656-3p. GNG5, a target of miR-656-3p, could reverse the impacts of silencing circ-PAPPA on CC cells. CONCLUSION Circ-PAPPA may play an oncogenic role in CC by promoting cell growth and glycolysis through the miR-656-3p/GNG5 axis.
Collapse
Affiliation(s)
- AiYuan Cai
- Department of Paediatrics, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City 518034, Guangdong Province, PR China
| | - HuiShi Ye
- Department of Paediatrics, Dongguan Hospital of Guangzhou University of Chinese Medicine, Dongguan City, Guangdong Province, PR China
| | - YuanHong Lin
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province, PR China
| | - JinYun Li
- Acupuncture Rehabilitation Clinical College, Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province, PR China
| | - DongSheng Fang
- Acupuncture Rehabilitation Clinical College, Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province, PR China
| | - ZhongBin Pan
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province, PR China
| | - ZhiWei Li
- Xi 'an Jiaotong University, Xi 'an City, Shaanxi Province, PR China
| | - GuangLiang Luo
- Department of Paediatrics, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City 518034, Guangdong Province, PR China
| | - YanFang Huang
- Department of Paediatrics, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City 518034, Guangdong Province, PR China
| | - CiAi Lai
- Department of Paediatrics, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City 518034, Guangdong Province, PR China.
| |
Collapse
|
35
|
Chen J, Huang Z, Chen Y, Tian H, Chai P, Shen Y, Yao Y, Xu S, Ge S, Jia R. Lactate and lactylation in cancer. Signal Transduct Target Ther 2025; 10:38. [PMID: 39934144 DOI: 10.1038/s41392-024-02082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Accumulated evidence has implicated the diverse and substantial influence of lactate on cellular differentiation and fate regulation in physiological and pathological settings, particularly in intricate conditions such as cancer. Specifically, lactate has been demonstrated to be pivotal in molding the tumor microenvironment (TME) through its effects on different cell populations. Within tumor cells, lactate impacts cell signaling pathways, augments the lactate shuttle process, boosts resistance to oxidative stress, and contributes to lactylation. In various cellular populations, the interplay between lactate and immune cells governs processes such as cell differentiation, immune response, immune surveillance, and treatment effectiveness. Furthermore, communication between lactate and stromal/endothelial cells supports basal membrane (BM) remodeling, epithelial-mesenchymal transitions (EMT), metabolic reprogramming, angiogenesis, and drug resistance. Focusing on lactate production and transport, specifically through lactate dehydrogenase (LDH) and monocarboxylate transporters (MCT), has shown promise in the treatment of cancer. Inhibitors targeting LDH and MCT act as both tumor suppressors and enhancers of immunotherapy, leading to a synergistic therapeutic effect when combined with immunotherapy. The review underscores the importance of lactate in tumor progression and provides valuable perspectives on potential therapeutic approaches that target the vulnerability of lactate metabolism, highlighting the Heel of Achilles for cancer treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ziyue Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ya Chen
- Department of Radiology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Hao Tian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Yongning Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| |
Collapse
|
36
|
Ferdousmakan S, Mansourian D, Seyedi Asl FS, Fathi Z, Maleki-Sheikhabadi F, Afjadi MN, Zalpoor H. Autophagy induced by metabolic processes leads to solid tumor cell metastatic dormancy and recurrence. Med Oncol 2025; 42:62. [PMID: 39899220 DOI: 10.1007/s12032-025-02607-6] [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: 10/22/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
A crucial cellular mechanism that has a complex impact on the biology of cancer, particularly in solid tumors, is autophagy. This review explores how metabolic processes trigger autophagy, which helps metastatic tumor cells go dormant and recur. During metastasis, tumor cells frequently encounter severe stressors, such as low oxygen levels and nutritional deprivation, which causes them to activate autophagy as a survival tactic. This process allows cancer stem cells (CSCs) to withstand severe conditions while also preserving their features. After years of dormancy, dormant disseminated tumor cells (DTCs) may reappear as aggressive metastatic cancers. The capacity of autophagy to promote resistance to treatments and avoid immune detection is intimately related to this phenomenon. According to recent research, autophagy promotes processes, such as the epithelial-to-mesenchymal transition (EMT) and helps build a pre-metastatic niche, which makes treatment strategies more challenging. Autophagy may be a promising therapeutic target because of its dual function as a tumor suppressor in early-stage cancer and a survival promoter in advanced stages. To effectively treat metastatic diseases, it is crucial to comprehend how metabolic processes interact with autophagy and affect tumor behavior. In order to find novel therapeutic approaches that can interfere with these processes and improve patient outcomes, this study highlights the critical need for additional investigation into the mechanisms by which autophagy controls tumor dormancy and recurrence.
Collapse
Affiliation(s)
- Saeid Ferdousmakan
- Department of Pharmacy Practice, Nargund College of Pharmacy, Bangalore, 560085, India
| | - Dorrin Mansourian
- Faculty of Pharmacy, Eastern Mediterranean University, Gazimagusa TRNC via Mersin 10, Mersin, Turkey
| | | | - Zeinab Fathi
- Medical School, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
37
|
Liu R, Fu M, Chen P, Liu Y, Huang W, Sun X, Zhu P, Wen Z, Cheng Y. Emerging roles of angiopoietin‑like 4 in human tumors (Review). Int J Oncol 2025; 66:9. [PMID: 39704206 PMCID: PMC11753769 DOI: 10.3892/ijo.2024.5715] [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: 09/30/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024] Open
Abstract
Angiopoietin‑like 4 (ANGPTL4), a member of the angiopoietin family, plays critical roles in angiogenesis, lipid metabolism and inflammation. It has been demonstrated that ANGPTL4 has significant influence on various diseases. Accumulating evidence has highlighted the impacts of ANGPTL4 on human malignancies. ANGPTL4 is commonly overexpressed in various types of cancer, such as breast, non‑small cell lung, gastric and colorectal cancer. Its upregulation promotes tumor growth, invasion, metastasis and angiogenesis, as well as metabolic reprogramming and resistance to programmed cell death, radiotherapy and chemotherapy. However, ANGPTL4 has also exhibited antitumor effects under certain conditions, indicating its complex roles in tumor biology. The transcriptional regulation of ANGPTL4 is influenced by multiple factors, such as HIF‑1, PPARs, TGF‑β and long non‑coding RNAs. In terms of signaling pathways, STATs, PI3K/AKT and COX-2/PGE2 are important in regulating cellular processes. The present review summarizes the biological functions of ANGPTL4 in tumors and its association with patient prognosis. Furthermore, the key molecular mechanisms and potential reasons for its dual roles in cancer are also discussed. In conclusion, ANGPTL4 is a valuable diagnostic biomarker and a potential therapeutic target for human cancers.
Collapse
Affiliation(s)
- Ruyi Liu
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Miaomiao Fu
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yuchen Liu
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Weicheng Huang
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xing Sun
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Pengfei Zhu
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhihua Wen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
38
|
Wang P, Wang G, Li H, Yuan Y, Chen H, Wang S, Sun Z, Meng F, Li Y, Yang F, Wang J, Chen K, Qiu M. Nicotinamide N-methyltransferase negatively regulates metastasis-promoting property of cancer-associated fibroblasts in lung adenocarcinoma. Cancer Commun (Lond) 2025; 45:110-137. [PMID: 39623600 PMCID: PMC11833673 DOI: 10.1002/cac2.12633] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 02/19/2025] Open
Abstract
BACKGROUND Recurrence and metastasis remain significant challenges in lung adenocarcinoma (LUAD) after radical resection. The mechanisms behind the recurrence and metastasis of LUAD remain elusive, and deregulated cellular metabolism is suspected to play a significant role. This study explores the metabolic and epigenetic regulation mediated by nicotinamide N-methyl transferase (NNMT) in LUAD. METHODS Untargeted metabolomic analyses were performed to detect metabolism irregularities. Single-cell RNA sequencing (RNA-seq) databases and multiplex immunofluorescence analysis were used to identify the location of NNMT within the tumor microenvironment. The biological functions of NNMT were investigated both in vitro and in vivo, with RNA-seq and chromatin immunoprecipitation-PCR providing insights into underlying mechanisms. Finally, single-cell RNA-seq data and immunohistochemistry of primary tumors were analyzed to validate the main findings. RESULTS Untargeted metabolomic analyses revealed metabolic aberrations in amino acids, organic acids, lipids, and nicotinamide pathways, which are linked to metastasis of non-small cell lung cancer. NNMT is a key enzyme in nicotinamide metabolism, and we found the bulk tissue mRNA level of NNMT gene was inversely associated with LUAD metastasis. NNMT was proved to be predominantly expressed in cancer-associated fibroblasts (CAFs) within the stromal regions of LUAD, and a low stromal NNMT expression was identified as a predictor of poor disease-free survival following radical resection of LUAD. The isolation and primary culture of CAFs from LUAD enabled in vitro and in vivo experiments, which confirmed that NNMT negatively regulated the metastasis-promoting properties of CAFs in LUAD. Mechanistically, the downregulation of NNMT led to an increase in intracellular methyl groups by reducing the activity of the methionine cycle, resulting in heightened methylation at H3K4me3. This alteration triggered the upregulation of genes involved in extracellular matrix remodeling in CAFs, including those encoding collagens, integrins, laminins, and matrix metalloproteinases, thereby facilitating cancer cell invasion and metastasis. Reanalysis of single-cell RNA-seq data and immunohistochemistry assays of primary LUAD tissues substantiated NNMT's negative regulation of these genes in CAFs. CONCLUSIONS This study provides novel insights into the metabolic and epigenetic regulatory functions of NNMT in CAFs, expanding the current understanding of LUAD metastasis regulation and suggesting potential avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Peiyu Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
- Department of Thoracic SurgeryThe First Affiliated Hospital of Zhengzhou University, ZhengzhouHenanP. R. China
| | - Guangxi Wang
- Department of Pathology, School of Basic Medical SciencesInstitute of Systems Biomedicine, Peking‐Tsinghua Center for Life Sciences, Peking University Health Science CenterBeijingP. R. China
| | - Haoran Li
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Yuyao Yuan
- Department of Pathology, School of Basic Medical SciencesInstitute of Systems Biomedicine, Peking‐Tsinghua Center for Life Sciences, Peking University Health Science CenterBeijingP. R. China
| | - Haiming Chen
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Shaodong Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Zewen Sun
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Fanjie Meng
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang HospitalCapital Medical UniversityBeijingP. R. China
| | - Yun Li
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Fan Yang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Jun Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Kezhong Chen
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Mantang Qiu
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| |
Collapse
|
39
|
Zapatería B, Arias E. Aging, cancer, and autophagy: connections and therapeutic perspectives. Front Mol Biosci 2025; 11:1516789. [PMID: 39935707 PMCID: PMC11811537 DOI: 10.3389/fmolb.2024.1516789] [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: 10/24/2024] [Accepted: 12/24/2024] [Indexed: 02/13/2025] Open
Abstract
Aging and cancer are intricately linked through shared molecular processes that influence both the onset of malignancy and the progression of age-related decline. As organisms age, cellular stress, genomic instability, and an accumulation of senescent cells create a pro-inflammatory environment conducive to cancer development. Autophagy, a cellular process responsible for degrading and recycling damaged components, plays a pivotal role in this relationship. While autophagy acts as a tumor-suppressive mechanism by preventing the accumulation of damaged organelles and proteins, cancer cells often exploit it to survive under conditions of metabolic stress and treatment resistance. The interplay between aging, cancer, and autophagy reveals key insights into tumorigenesis, cellular senescence, and proteostasis dysfunction. This review explores the molecular connections between these processes, emphasizing the potential for autophagy-targeted therapies as strategies that could be further explored in both aging and cancer treatment. Understanding the dual roles of autophagy in suppressing and promoting cancer offers promising avenues for therapeutic interventions aimed at improving outcomes for elderly cancer patients while addressing age-related deterioration.
Collapse
Affiliation(s)
- Begoña Zapatería
- Department of Medicine (Marion Bessin Liver Research Center), Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Esperanza Arias
- Department of Medicine (Marion Bessin Liver Research Center), Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
- Einstein Aging Research Center, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
40
|
Liu X, Sun X, Mu W, Li Y, Bu W, Yang T, Zhang J, Liu R, Ren J, Zhou J, Li P, Shi Y, Shao C. Autophagic flux-lipid droplet biogenesis cascade sustains mitochondrial fitness in colorectal cancer cells adapted to acidosis. Cell Death Discov 2025; 11:21. [PMID: 39856069 PMCID: PMC11761495 DOI: 10.1038/s41420-025-02301-6] [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: 07/25/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer development is associated with adaptation to various stressful conditions, such as extracellular acidosis. The adverse tumor microenvironment also selects for increased malignancy. Mitochondria are integral in stress sensing to allow for tumor cells to adapt to stressful conditions. Here, we show that colorectal cancer cells adapted to acidic microenvironment (CRC-AA) are more reliant on oxidative phosphorylation than their parental cells, and the acetyl-CoA in CRC-AA cells are generated from fatty acids and glutamine, but not from glucose. Consistently, CRC-AA cells exhibit increased mitochondrial mass and fitness that depends on an upregulated autophagic flux-lipid droplet axis. Lipid droplets (LDs) function as a buffering system to store the fatty acids derived from autophagy and to protect mitochondria from lipotoxicity in CRC-AA cells. Blockade of LD biogenesis causes mitochondrial dysfunction that can be rescued by inhibiting carnitine palmitoyltransferase 1 α (CPT1α). High level of mitochondrial superoxide is essential for the AMPK activation, resistance to apoptosis, high autophagic flux and mitochondrial function in CRC-AA cells. Thus, our results demonstrate that the cascade of autophagic flux and LD formation plays an essential role in sustaining mitochondrial fitness to promote cancer cell survival under chronic acidosis. Our findings provide insight into the pro-survival metabolic plasticity in cancer cells under microenvironmental or therapeutic stress and imply that this pro-survival cascade may potentially be targeted in cancer therapy.
Collapse
Affiliation(s)
- Xiaojie Liu
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
- Biochip Laboratory, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, China
| | - Xue Sun
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Wenqing Mu
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Yanan Li
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Wenqing Bu
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Tingting Yang
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Jia Zhang
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Rui Liu
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Jiayu Ren
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Peishan Li
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Yufang Shi
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Changshun Shao
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China.
| |
Collapse
|
41
|
Liu J, Shu T, Mu Y, Zheng W, Lu X, Tao H. Curdione combined with borneol treats bacterial mixed HPV infection by regulating the crosstalk among immune cells. Front Immunol 2025; 16:1503355. [PMID: 39911394 PMCID: PMC11794296 DOI: 10.3389/fimmu.2025.1503355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Background Human papillomavirus (HPV) infection is a worldwide reproductive system disease. Baofukang suppository, a traditional herbal preparation that includes curdione and borneol, has been reported to treat bacterial vaginosis (BV) and HPV infection in China. However, the therapeutic mechanism is still unknown. This study aims to explore the molecular mechanisms of curdione and borneol in treating HPV infection. Methods We conducted a retrospective cohort analysis of medical records from a single-center study involving 205 HPV patients, focusing on the correlation between HPV clearance and co-infection with other pathogens, confirming the efficacy of Baofukang suppository. Bioinformatics and network pharmacology approaches were employed to identify therapeutic targets of Baofukang suppository for BV/HPV co-infections. qRT-PCR, Western blot, immunofluorescence staining, and flow cytometry were utilized to validate the therapeutic targets of curdione and borneol, along with the associated immune molecular changes. Finally, the molecular mechanisms and therapeutic efficacy of curdione and borneol were confirmed in vivo using an LPS/TC-1 cervical orthotopic injection model. Results Curdione and borneol selectively inhibit the secretion of interleukin-6 (IL-6) and interleukin-1β (IL-1β) by macrophages. The reduction in IL-6 and IL-1β levels effectively inhibits the expression of CD274 (Programmed death ligand 1, PD-L1) in infected epithelial cells by inhibiting STAT3 phosphorylation, thereby suppressing their immune evasion capabilities. Furthermore, curdione and borneol enhance the expression of tumor necrosis factor α (TNF-α) and caspase 1 (CASP1) in macrophages, as well as the expression of interleukin 12 (IL-12) and interleukin 23 (IL-23) in dendritic cells (DCs). The expression of these inflammatory factors effectively promotes the migration and differentiation of T cells to the site of infection, completing the clearance of infected epithelial cells. Conclusion The main components of Baofukang suppository, curdione and borneol, inhibit the progression of HPV infection and the occurrence of cervical cancer by modulating the communication between innate and adaptive immunity, promoting the recruitment and recognition of CD8+ T cells to eliminate HPV-infected epithelial cells.
Collapse
Affiliation(s)
- Jingwei Liu
- Department of Gynecology, Wuhu Maternal and Child Health (MCH) Center, Wuhu, China
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Tong Shu
- Graduate School, Wannan Medical College, Wuhu, China
| | - Yiheng Mu
- Graduate School, Wannan Medical College, Wuhu, China
| | - Wanlin Zheng
- Graduate School, Wannan Medical College, Wuhu, China
| | - Xiaohuan Lu
- Department of Plastic Surgery, The Second Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, The Second Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hong Tao
- Department of Gynecology, Wuhu Maternal and Child Health (MCH) Center, Wuhu, China
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| |
Collapse
|
42
|
Zhang Y, Yang E, Chen M, Zhang J, Liu Q, Lei Z, Xu T, Cai X, Feng C. Quality diversity of three calcium-rich Primulina vegetables: A comprehensive analysis of calcium content, metabolite profiles, taste characteristics, and medicinal potential. Food Chem 2025; 463:141538. [PMID: 39388873 DOI: 10.1016/j.foodchem.2024.141538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/12/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Primulina plants native to karst regions are exceptionally rich in calcium and have been developed into high‑calcium leafy vegetables. However, limited knowledge of their metabolites, taste characteristics, and potential medicinal value restricts further genetic improvements. This study conducted a comprehensive analysis on three breeding species of Primulina vegetables. Common garden experiment demonstrated significant calcium enrichment capability, with calcium content ranging from 204.45 to 391.52 mg/100 g. Through widely-targeted metabolomics, 1121 metabolites were identified within these Primulina vegetables. Furthermore, comparative analysis identified 976 differentially accumulated metabolites across nine comparison groups, driven mainly by flavonoids, phenolic acids, and lipids. Integration of electronic tongue analysis and metabolomics revealed taste profiles and identified 17 key candidate compounds related to taste. Based on network pharmacology analysis, 32 active ingredients were found in Primulina vegetables, which highlighted potential medicinal value. These findings provide a data-driven foundation for breeding programs aimed at enhancing nutritional and flavor traits.
Collapse
Affiliation(s)
- Yi Zhang
- Jiangxi Provincial Key Laboratory of ex situ Plant Conservation and Utilization, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; College of Life Science, Nanchang University, Nanchang, China.
| | - Endian Yang
- Jiangxi Provincial Key Laboratory of ex situ Plant Conservation and Utilization, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; College of Life Science, Nanchang University, Nanchang, China.
| | - Mingjie Chen
- College of Life Sciences, Henan Provincial Key Laboratory of Tea Plant Biology, Xinyang Normal University, Xinyang 464000, China.
| | - Jie Zhang
- Jiangxi Provincial Key Laboratory of ex situ Plant Conservation and Utilization, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China.
| | - Qin Liu
- Jiangxi Provincial Key Laboratory of ex situ Plant Conservation and Utilization, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; College of Life Science, Nanchang University, Nanchang, China
| | - Ziyi Lei
- Jiangxi Provincial Key Laboratory of ex situ Plant Conservation and Utilization, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; College of Life Science, Nanchang University, Nanchang, China
| | - Tingting Xu
- Jiangxi Provincial Key Laboratory of ex situ Plant Conservation and Utilization, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China
| | - Xinxia Cai
- Jiangxi Provincial Key Laboratory of ex situ Plant Conservation and Utilization, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China
| | - Chen Feng
- Jiangxi Provincial Key Laboratory of ex situ Plant Conservation and Utilization, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China.
| |
Collapse
|
43
|
Chu WK, Huang SC, Chang CF, Wu JL, Gong HY. Migration of primordial germ cells and their relationship of PGCs with sex development in transgenic germline-specific fluorescent freshwater angelfish (Pterophyllum scalare). Sci Rep 2025; 15:1308. [PMID: 39779963 PMCID: PMC11711190 DOI: 10.1038/s41598-025-85480-7] [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: 08/21/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Primordial germ cells (PGCs), the progenitors of gametes, are essential for teleost reproduction. While their formation is conserved across teleosts, the activation, migration routes, and localization periods vary among species. In this study, we developed a novel transgenic line, Tg(ddx4:TcCFP13-nanos3), based on the Nile tilapia genome, to label PGCs with clear fluorescent signals in the freshwater angelfish (Pterophyllum scalare). Our findings reveal a complex, multistage PGCs migration process in angelfish, with a significantly extended localization period (168 hpf) compared to zebrafish (24 hpf). Notably, individual differences in PGCs abundance were observed during early somite development. Analysis of PGCs counts and subsequent sexual maturation demonstrate a potential correlation between PGCs abundance and sex determination: 90% of PGCs-reduced individuals developed as males, while 83% of PGCs-rich individuals developed as females. This study provides a foundation for understanding PGCs migration and sex development in freshwater angelfish, offering valuable insights into reproductive biology and ornamental fish species. Furthermore, this in vivo PGCs tracking system for Cichlids provides a versatile tool for advancing research and applications in germ cell biology.
Collapse
Affiliation(s)
- Wai-Kwan Chu
- Marine Molecular Genetics & Biotechnology Laboratory, Department of Aquaculture, National Taiwan Ocean University, Keelung, 202301, Taiwan
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, 202301, Taiwan
| | - Shih-Chin Huang
- Marine Molecular Genetics & Biotechnology Laboratory, Department of Aquaculture, National Taiwan Ocean University, Keelung, 202301, Taiwan
- Fisheries Research Institute, Kinmen County, 893, Taiwan
| | - Ching-Fong Chang
- Marine Molecular Genetics & Biotechnology Laboratory, Department of Aquaculture, National Taiwan Ocean University, Keelung, 202301, Taiwan
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, 202301, Taiwan
| | - Jen-Leih Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
- College of Life Sciences, National Taiwan Ocean University, Keelung, 202301, Taiwan
| | - Hong-Yi Gong
- Marine Molecular Genetics & Biotechnology Laboratory, Department of Aquaculture, National Taiwan Ocean University, Keelung, 202301, Taiwan.
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, 202301, Taiwan.
| |
Collapse
|
44
|
Chen Z, Xu J, Fang K, Jiang H, Leng Z, Wu H, Zhang Z, Wang Z, Li Z, Sun M, Zhao Z, Feng A, Zhang S, Chu Y, Ye L, Xu M, He L, Chen T. FOXC1-mediated serine metabolism reprogramming enhances colorectal cancer growth and 5-FU resistance under serine restriction. Cell Commun Signal 2025; 23:13. [PMID: 39773485 PMCID: PMC11708197 DOI: 10.1186/s12964-024-02016-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
Colorectal cancer (CRC) is the most common gastrointestinal malignancy, and 5-Fluorouracil (5-FU) is the principal chemotherapeutic drug used for its treatment. However, 5-FU resistance remains a significant challenge. Under stress conditions, tumor metabolic reprogramming influences 5-FU resistance. Serine metabolism plasticity is one of the crucial metabolic pathways influencing 5-FU resistance in CRC. However, the mechanisms by which CRC modulates serine metabolic reprogramming under serine-deprived conditions remain unknown. We found that exogenous serine deprivation enhanced the expression of serine synthesis pathway (SSP) genes, which in turn supported CRC cell growth and 5-FU resistance. Serine deprivation activate the ERK1/2-p-ELK1 signaling axis, leading to upregulated FOXC1 expression in CRC cells. Elevated FOXC1 emerged as a critical element, promoting the transcription of serine metabolism enzymes PHGDH, PSAT1, and PSPH, which in turn facilitated serine production, supporting CRC growth. Furthermore, through serine metabolism, FOXC1 influenced purine metabolism and DNA damage repair, thereby increasing 5-FU resistance. Consequently, combining dietary serine restriction with targeted therapy against the ERK1/2-pELK1-FOXC1 axis could be a highly effective strategy for treating CRC, enhancing the efficacy of 5-FU.
Collapse
Affiliation(s)
- Zhukai Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiacheng Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kang Fang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hanyu Jiang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhuyun Leng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Wu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zehua Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zeyu Wang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhaoxing Li
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mingchuang Sun
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziying Zhao
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anqi Feng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shihan Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan Chu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lechi Ye
- Department of Colorectal Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Meidong Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Lingnan He
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
45
|
Zhang J, Argueta D, Tong X, Vinters HV, Mathern GW, Cepeda C. Iconography of abnormal non-neuronal cells in pediatric focal cortical dysplasia type IIb and tuberous sclerosis complex. Front Cell Neurosci 2025; 18:1486315. [PMID: 39835291 PMCID: PMC11743721 DOI: 10.3389/fncel.2024.1486315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Once believed to be the culprits of epileptogenic activity, the functional properties of balloon/giant cells (BC/GC), commonly found in some malformations of cortical development including focal cortical dysplasia type IIb (FCDIIb) and tuberous sclerosis complex (TSC), are beginning to be unraveled. These abnormal cells emerge during early brain development as a result of a hyperactive mTOR pathway and may express both neuronal and glial markers. A paradigm shift occurred when our group demonstrated that BC/GC in pediatric cases of FCDIIb and TSC are unable to generate action potentials and lack synaptic inputs. Hence, their role in epileptogenesis remained obscure. In this review, we provide a detailed characterization of abnormal non-neuronal cells including BC/GC, intermediate cells, and dysmorphic/reactive astrocytes found in FCDIIb and TSC cases, with special emphasis on electrophysiological and morphological assessments. Regardless of pathology, the electrophysiological properties of abnormal cells appear more glial-like, while others appear more neuronal-like. Their morphology also differs in terms of somatic size, shape, and dendritic elaboration. A common feature of these types of non-neuronal cells is their inability to generate action potentials. Thus, despite their distinct properties and etiologies, they share a common functional feature. We hypothesize that, although the exact role of abnormal non-neuronal cells in FCDIIb and TSC remains mysterious, it can be suggested that cells displaying more glial-like properties function in a similar way as astrocytes do, i.e., to buffer K+ ions and neurotransmitters, while those with more neuronal properties, may represent a metabolic burden due to high energy demands but inability to receive or transmit electric signals. In addition, due to the heterogeneity of these cells, a new classification scheme based on morphological, electrophysiological, and gene/protein expression in FCDIIb and TSC cases seems warranted.
Collapse
Affiliation(s)
- Joyce Zhang
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California - Los Angeles, Los Angeles, CA, United States
| | - Deneen Argueta
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California - Los Angeles, Los Angeles, CA, United States
| | - Xiaoping Tong
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Harry V. Vinters
- Department of Pathology and Laboratory Medicine, University of California - Los Angeles, Los Angeles, CA, United States
| | - Gary W. Mathern
- Department of Neurosurgery, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA, United States
| | - Carlos Cepeda
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California - Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
46
|
Tanaka R, Sugiura K, Osabe K, Hattori M, Nagai T. Genetically encoded bioluminescent glucose indicator for biological research. Biochem Biophys Res Commun 2025; 742:151092. [PMID: 39626367 DOI: 10.1016/j.bbrc.2024.151092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024]
Abstract
Glucose is an essential energy source in living cells and is involved in various phenomena. To understand the roles of glucose, measuring cellular glucose levels is important. Here, we developed a bioluminescent glucose indicator called LOTUS-Glc. Unlike fluorescence, bioluminescence doesn't require excitation light when imaging. Using LOTUS-Glc, we demonstrated drug effect evaluation, concurrent use with the optogenetic tool in HEK293T cells, and the measurement of light-dependent glucose fluctuations in plant-derived protoplasts. LOTUS-Glc would be a useful tool for understanding the roles of glucose in living organisms.
Collapse
Affiliation(s)
- Rikuto Tanaka
- Graduate School of Frontier Biosciences, The University of Osaka, Suita, Osaka, 565-0871, Japan
| | - Kazunori Sugiura
- SANKEN, The University of Osaka, Ibaraki, Osaka, 567-0047, Japan
| | - Kenji Osabe
- SANKEN, The University of Osaka, Ibaraki, Osaka, 567-0047, Japan
| | - Mitsuru Hattori
- SANKEN, The University of Osaka, Ibaraki, Osaka, 567-0047, Japan
| | - Takeharu Nagai
- Graduate School of Frontier Biosciences, The University of Osaka, Suita, Osaka, 565-0871, Japan; SANKEN, The University of Osaka, Ibaraki, Osaka, 567-0047, Japan; Research Institute for Electronic Science, Hokkaido University, Sapporo, Hokkaido, 001-0021, Japan.
| |
Collapse
|
47
|
Ellingson BM, Okobi Q, Chong R, Plawat R, Zhao E, Gafita A, Sonni I, Chun S, Filka E, Yao J, Telesca D, Li S, Li G, Lai A, Nghiemphu P, Czernin J, Nathanson DA, Cloughesy TF. A comparative study of preclinical and clinical molecular imaging response to EGFR inhibition using osimertinib in glioblastoma. Neurooncol Adv 2025; 7:vdaf022. [PMID: 40051661 PMCID: PMC11883343 DOI: 10.1093/noajnl/vdaf022] [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] [Indexed: 03/09/2025] Open
Abstract
Background To demonstrate the potential value of 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) as a rapid, non-invasive metabolic imaging surrogate for pharmacological modulation of EGFR signaling in EGFR-driven GBM, we synchronously conducted a preclinical imaging study using patient-derived orthotopic xenograft (PDOX) models and validated it in a phase II molecular imaging study in recurrent GBM (rGBM) patients using osimertinib. Methods A GBM PDOX mouse model study was performed concurrently with an open-label, single-arm, single-center, phase II study of osimertinib (NCT03732352) that enrolled 12 patients with rGBM with EGFR alterations. Patients received osimertinib daily and 3 18F-FDG PET scans: two 24 h apart prior to dosing, and one 48 h after dosing. Results GBM PDOX models suggest osimertinib has limited impact on both 18F-FDG uptake (+ 9.8%-+25.9%) and survival (+ 15.5%; P = .01), which may be explained by insufficient exposure in the brain (Kpuu: 0.30) required to robustly inhibit the EGFR alterations found in GBM. Treatment with osimertinib had subtle, but measurable decreases in the linear rate of change of 18F-FDG nSUV growth rate averaging -4.5% per day (P = .01) and change in 18F-FDG uptake was correlated with change in tumor growth rate (R2 = 0.4719, P = .0195). No metabolic (PERCIST) or radiographic (RANO) responses were seen, and no improvements in PFS or OS were observed. Conclusions This study demonstrated the feasibility of using FDG PET as a clinically reliable imaging biomarker for assessing EGFR inhibition in GBM, while revealing osimertinib's limited impact on both metabolic activity and tumor growth in GBM, findings that were concordant between preclinical and clinical observations.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- UCLA Brain Tumor Imaging Laboratory (BTIL), Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Qunicy Okobi
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Robert Chong
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Rhea Plawat
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Eva Zhao
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Andrei Gafita
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Ida Sonni
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Saewon Chun
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Emese Filka
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Donatello Telesca
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Shanpeng Li
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Gang Li
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Albert Lai
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Phioanh Nghiemphu
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Johannes Czernin
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - David A Nathanson
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Timothy F Cloughesy
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
48
|
Huang P, Gao W, Fu C, Wang M, Li Y, Chu B, He A, Li Y, Deng X, Zhang Y, Kong Q, Yuan J, Wang H, Shi Y, Gao D, Qin R, Hunter T, Tian R. Clinical functional proteomics of intercellular signalling in pancreatic cancer. Nature 2025; 637:726-735. [PMID: 39537929 DOI: 10.1038/s41586-024-08225-y] [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: 07/21/2023] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an atypical, highly stromal tumour microenvironment (TME) that profoundly contributes to its poor prognosis1. Here, to better understand the intercellular signalling between cancer and stromal cells directly in PDAC tumours, we developed a multidimensional proteomic strategy called TMEPro. We applied TMEPro to profile the glycosylated secreted and plasma membrane proteome of 100 human pancreatic tissue samples to a great depth, define cell type origins and identify potential paracrine cross-talk, especially that mediated through tyrosine phosphorylation. Temporal dynamics during pancreatic tumour progression were investigated in a genetically engineered PDAC mouse model. Functionally, we revealed reciprocal signalling between stromal cells and cancer cells mediated by the stromal PDGFR-PTPN11-FOS signalling axis. Furthermore, we examined the generic shedding mechanism of plasma membrane proteins in PDAC tumours and revealed that matrix-metalloprotease-mediated shedding of the AXL receptor tyrosine kinase ectodomain provides an additional dimension of intercellular signalling regulation in the PDAC TME. Importantly, the level of shed AXL has a potential correlation with lymph node metastasis, and inhibition of AXL shedding and its kinase activity showed a substantial synergistic effect in inhibiting cancer cell growth. In summary, we provide TMEPro, a generically applicable clinical functional proteomic strategy, and a comprehensive resource for better understanding the PDAC TME and facilitating the discovery of new diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Peiwu Huang
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Weina Gao
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Changying Fu
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunguang Li
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Bizhu Chu
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - An He
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yuan Li
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Xiaomei Deng
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yehan Zhang
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Qian Kong
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Jingxiong Yuan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hebin Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Shi
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
- Bristol Myers Squibb, San Diego, CA, USA.
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ruijun Tian
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
49
|
Liao K, Liu K, Wang Z, Zhao K, Mei Y. TRIM2 promotes metabolic adaptation to glutamine deprivation via enhancement of CPT1A activity. FEBS J 2025; 292:275-293. [PMID: 38949993 DOI: 10.1111/febs.17218] [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: 01/06/2024] [Revised: 05/14/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Cancer cells undergo metabolic adaptation to promote their survival and growth under energy stress conditions, yet the underlying mechanisms remain largely unclear. Here, we report that tripartite motif-containing protein 2 (TRIM2) is upregulated in response to glutamine deprivation by the transcription factor cyclic AMP-dependent transcription factor (ATF4). TRIM2 is shown to specifically interact with carnitine O-palmitoyltransferase 1 (CPT1A), a rate-limiting enzyme of fatty acid oxidation. Via this interaction, TRIM2 enhances the enzymatic activity of CPT1A, thereby regulating intracellular lipid levels and protecting cells from glutamine deprivation-induced apoptosis. Furthermore, TRIM2 is able to promote both in vitro cell proliferation and in vivo xenograft tumor growth via CPT1A. Together, these findings establish TRIM2 as an important regulator of the metabolic adaptation of cancer cells to glutamine deprivation and implicate TRIM2 as a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Kaimin Liao
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiyue Liu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhongyu Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kailiang Zhao
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yide Mei
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
50
|
Hirai K, Saito H, Kato M, Kiyama M, Hanzawa H, Nakane A, Sekiya S, Yoshida K, Kishino A, Ikeda A, Kimura T, Takahashi J, Takeda S. Evaluation of induced pluripotent stem cell differentiation into neural progenitor cell using Raman spectra derived from extracellular vesicles in culture supernatants. J Biosci Bioeng 2025; 139:44-52. [PMID: 39419642 DOI: 10.1016/j.jbiosc.2024.09.004] [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: 04/16/2024] [Revised: 07/31/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Non-invasive cell culture monitoring technology is crucial to improve the manufacturing efficiency of cell products. We have found that extracellular vesicles (EVs) are secreted into the culture supernatants in the differentiation process from human induced pluripotent stem cells (iPSCs) to dopaminergic progenitor cells, and that the composition of EVs changes in accordance with the differentiation processes. In this study, we hypothesized that it is possible to evaluate the cultured cellular states by detecting compositional changes of EVs secreted from cultured cells with label-free Raman spectroscopy in a non-invasive manner. Therefore, Raman signal analysis derived from EV fractions isolated from culture supernatants throughout the differentiation process was conducted. iPSCs cultures were simultaneously implemented under a standard condition (control) and an artificial deviation condition inducing reductions in pluripotency by depleting FGF2 in culture medium (-FGF2), which is indispensable for maintaining the pluripotency. Subsequently, the differentiation step was conducted for each iPSCs culture under the same condition. As a result, it was found that under -FGF2, the expression level of the pluripotency marker NANOG decreased compared to that of the control and correlated with the identification results based on Raman signals with a correlation coefficient of 0.77. Lipid-derived Raman signals were extracted as identification factors, suggesting that changes in the lipid component of EV occur depending on the cellular states. From the above, we have found that the change in composition of EVs in the culture supernatant by detecting Raman signals would be a monitoring index of the cellular state of differentiation and pluripotency.
Collapse
Affiliation(s)
- Kakuro Hirai
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., Creative Lab for Innovation in Kobe 304, 6-3-7 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hikaru Saito
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., Creative Lab for Innovation in Kobe 304, 6-3-7 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Midori Kato
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., Creative Lab for Innovation in Kobe 304, 6-3-7 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Masaharu Kiyama
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., Creative Lab for Innovation in Kobe 304, 6-3-7 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hiroko Hanzawa
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., Creative Lab for Innovation in Kobe 304, 6-3-7 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Atsushi Nakane
- Regenerative and Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe KIMEC Center Building 5th Fl, 1-5-2 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Sayaka Sekiya
- Regenerative and Cellular Medicine Office, Sumitomo Pharma Co., Ltd., Tokyo Nihonbashi Tower, 2-7-1 Nihonbashi, Tokyo 103-6012, Japan
| | - Kenji Yoshida
- Regenerative and Cellular Medicine Office, Sumitomo Pharma Co., Ltd., Tokyo Nihonbashi Tower, 2-7-1 Nihonbashi, Tokyo 103-6012, Japan
| | - Akiyoshi Kishino
- Regenerative and Cellular Medicine Office, Sumitomo Pharma Co., Ltd., Tokyo Nihonbashi Tower, 2-7-1 Nihonbashi, Tokyo 103-6012, Japan
| | - Atsushi Ikeda
- Regenerative and Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe KIMEC Center Building 5th Fl, 1-5-2 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Toru Kimura
- Regenerative and Cellular Medicine Office, Sumitomo Pharma Co., Ltd., Tokyo Nihonbashi Tower, 2-7-1 Nihonbashi, Tokyo 103-6012, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shizu Takeda
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., Creative Lab for Innovation in Kobe 304, 6-3-7 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|