1
|
Im SS, Seo J, You JE, Bang HW, Kim Y, Kweon J, Kim Y, Shin DM, Son J. BIX01294 suppresses PDAC growth through inhibition of glutaminase-mediated glutathione dynamics. Mol Metab 2025; 94:102113. [PMID: 39961401 PMCID: PMC11905835 DOI: 10.1016/j.molmet.2025.102113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
OBJECTIVES Increased expression of glutaminase (GLS) has been found to correlate with more aggressive disease and poorer prognosis in patients with several types of cancer, including breast, lung, and pancreatic cancer. G9a histone methyltransferase inhibitors may have anticancer activity. The present study assessed whether BIX01294 (BIX), a G9a histone methyltransferase inhibitor, can inhibit glutaminase (GLS) in pancreatic ductal adenocarcinoma (PDAC) cells. METHODS The effects of BIX on mitochondrial metabolism in PDAC cells were evaluated by targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomic analysis. To assess the impact of BIX on glutathione dynamics, real-time changes in glutathione levels were monitored by FreSHtracer-based GSH assays. RESULTS BIX significantly inhibited the growth of PDAC cells, both in vitro and in vivo, and robustly induced apoptotic cell death. BIX significantly increased the cellular NADP+/NADPH ratio and decreased the ratio of reduced-to-oxidized glutathione (GSH:GSSG). In addition, BIX decreased GSH levels and increased ROS levels. N-acetyl-l-cysteine (NAC) supplementation dramatically rescued PDAC cells from BIX-induced apoptosis. Furthermore, BIX inhibited the transcription of GLS by inhibiting Jumonji-domain histone demethylases but not G9a histone methyltransferase. One Jumonji-domain histone demethylase, KDM6B, epigenetically regulated GLS expression by binding to the GLS gene promoter. CONCLUSIONS Collectively, these findings suggest that BIX could be a potent therapeutic agent in patients with PDAC through its inhibition of GLS-mediated cellular redox balance.
Collapse
Affiliation(s)
- Se Seul Im
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Jihyeon Seo
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Ji Eun You
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Hye Won Bang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - YongHwan Kim
- Department of Cell and Genetic Engineering, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Jiyeon Kweon
- Department of Cell and Genetic Engineering, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Yongsub Kim
- Department of Cell and Genetic Engineering, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Dong-Myung Shin
- Department of Cell and Genetic Engineering, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Jaekyoung Son
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea.
| |
Collapse
|
2
|
Wang Y, Wang Z, Liu M, Chen C, Xi Q, Tang J, Yu Z, Wang S, Yu L, Yu M. Nutrient transporter-oriented nanoinhibitor counteracts intracellular metabolic reprogramming for RT-resistant HCC treatment. Mater Today Bio 2025; 31:101608. [PMID: 40104639 PMCID: PMC11914751 DOI: 10.1016/j.mtbio.2025.101608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Radiotherapy (RT) is the primary treatment modality for hepatocellular carcinoma (HCC). Inevitably, the X-ray exposure also increases the metabolic stress and energy demands in surviving tumor cells, which leads to metabolic reprogramming that reduces the sensitivity of HCC to clinical treatments including RT. Nevertheless, the current research in tumor metabolic therapy predominantly focuses on inhibiting glycolytic pathways, and the consequent metabolic compensation behavior of tumor cells exacerbates the risks of drug resistance and recurrence. To address this challenge, we innovatively proposed a tumor-specific multi-metabolic pathway regulation strategy navigated by tumor cell surface nutrient transporter (2-DG/BP MRs), which can be triggered by X-ray radiation to achieve dual blockade of glycolysis and glutamine metabolism pathways. Thus, this nanosystem reconfigured metabolic pathways within tumor cells to counteract RT-induced metabolic reprogramming through dual metabolic inhibition (glycolysis and glutamine metabolism). This approach disrupted the essential energy supply required for cancer cell proliferation without causing metabolic disorders in normal cells, thereby sensitizing HCC to RT. This tumor cell-specific metabolic intervention strategy provides a safe and effective approach for combination therapy in clinically RT-resistant tumors.
Collapse
Affiliation(s)
- Yuehua Wang
- Department of General Surgery, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Zhenjie Wang
- Office of Drug Clinical Trials, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Mengnan Liu
- Department of General Surgery, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China
| | - Chaojie Chen
- Department of General Surgery, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China
| | - Qiye Xi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jingwen Tang
- Department of General Surgery, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
| | - Shengtao Wang
- School of Biomedical Engineering and Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Ling Yu
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Meng Yu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
3
|
Yang H, Gao S, Lu G, He J, Dong J, Zhang X, Liu L, Zhong K, Zha G, Han L, Guo S, Li H, Wang Y. SIRT5-mediated GLS and GDH desuccinylation attenuates the autophagy of bovine mammary epithelial cells induced by ammonia. Cell Signal 2025; 127:111570. [PMID: 39694127 DOI: 10.1016/j.cellsig.2024.111570] [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: 09/09/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Sirtuin 5 (SIRT5) in mitochondria possesses a strong capacity for lysine desuccinylation, involving in various biological processes. Our previous research demonstrated that NH3 regulated autophagy dependent on SIRT5 in bovine mammary epithelial cells (bMECs). Interestingly, we discovered that SIRT5 reduced the content of NH3 and glutamate by inhibiting GLS activity in bMECs, the ratio of ADP/ATP also declined. In this study, we identified that SIRT5 interacted with endogenous GLS and GDH through Co-IP assay, but had no effect on endogenous GLS and GDH expression. SIRT5 made the succinylation levels of GLS and GDH significantly declined and resulted in the reduction of GLS and GDH activity. Next, the content of ammonia and glutamate, as well as the related autophagy markers were measured, we found that SIRT5 affected the glutamine metabolism, which attenuated ammonia release in MAC-T cells, accompanying with cellular autophagy decline, reducing the formation of autophagosome. Deletion of SIRT5 gene in MAC-T cells by means of CRISPR-cas9, we found the content of NH3 and glutamate increased, as well as autophagy promoted, which could be alleviated by SIRT5 overexpression. SIRT5 KO also resulted in increase of succinylation of GLS and GDH, as well as autophagy response in bMECs. Furthermore, SIRT5 promoted the maintenance of mitochondria homeostasis. Mechanistically, SIRT5 reduced ammonia release by modulating the succinylation levels and enzymatic activities of GLS and GDH in mitochondria and promoted the maintenance of mitochondria homeostasis, as well as further attenuated ammonia-stimulated autophagy in bovine mammary epithelial cells.
Collapse
Affiliation(s)
- Hanlin Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Shikai Gao
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Guangyang Lu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Junhui He
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Jinru Dong
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Xinyi Zhang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Luya Liu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Kai Zhong
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Guangming Zha
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Liqiang Han
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Shuang Guo
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Heping Li
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China.
| | - Yueying Wang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China.
| |
Collapse
|
4
|
Zhu X, Zhou X, Li S, Liu Z, Yu S, Shi H, Zhu L, Song B, Si Z, Sun M, Zhu W. PFKFB3 decreases α-ketoglutarate production while partial PFKFB3 knockdown in macrophages ameliorates arthritis in tumor necrosis factor-transgenic mice. Int Immunopharmacol 2025; 148:114102. [PMID: 39870011 DOI: 10.1016/j.intimp.2025.114102] [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: 11/11/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025]
Abstract
OBJECTIVE Aberrant 6-phosphofructo-2kinase/fructose-2,6-bisphoshatase 3 (PFKFB3) expression is tightly correlated with multiple steps of tumorigenesis; however, the pathological significance of PFKFB3 in macrophages in patients with rheumatoid arthritis (RA) remains obscure. In this study, we examined whether PFKFB3 modulates macrophage activation and promotes RA development. METHOD Peripheral blood mononuclear cells (PBMCs) from patients with RA, THP-1 cells, and bone marrow-derived macrophages from conditional PFKFB3-knockout mice were used to investigate the mechanism underlying PFKFB3-induced macrophage regulation of RA. RESULT We demonstrated that patients with RA have higher PFKFB3 levels than healthy volunteers. PFKFB3 silencing suppressed M1 macrophage polarization and downregulated IL-1β, CD80, IFIT1, CCL8, and CXCL10 in macrophages of patients with RA. PFKFB3 overexpression markedly upregulated IRF5, HIF1α, IL-1β, CD80, IFI27, IFI44, IFIT1, IFIT3, CCL2, CCL8, CXCL10, CXCL11, and MMP13 in phorbol 12-myristate 13-acetate-induced THP-1 cells, although these changes were partially reversed by PFK15, an inhibitor of PFKFB3 enzyme activity. Co-immunoprecipitation assays revealed that PFKFB3 interacted with GLUD1 and decreased glutamate dehydrogenase (GDH) activity and α-ketoglutarate production. PFKFB3, TNFα, IL-6, IFNγ, CXCL9, CXCL10, CXCL11, MMP13, and MMP19 were downregulated in bone marrow-derived macrophages of conditional PFKFB3-knockout mice relative to those of wild-type mice. Partial PFKFB3 knockdown in macrophages ameliorated the clinical signs of arthritis and bone destruction, inhibited proinflammatory factor expression, and promoted GDH activity and α-ketoglutarate production in tumor necrosis factor-transgenic mice. Single-cell sequencing revealed that macrophages were the most abundant cells in the ankles of arthritic mice, and partial PFKFB3 knockdown promoted M2-like polarization and was correlated with TREM2, SPP1, APOE, and C1Q expression. CONCLUSION PFKFB3 is upregulated in macrophages in patients with RA. PFKFB3 aggravates arthritis by modulating macrophage activity, which may be related to decreased α-ketoglutarate production.
Collapse
Affiliation(s)
- Xiaodong Zhu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Xiaohui Zhou
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Shuaiyi Li
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Zenghui Liu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Shidi Yu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Hong Shi
- Department of Rheumatology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - LingLing Zhu
- Department of Rheumatology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Baohui Song
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Zihou Si
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Mingshuang Sun
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Wei Zhu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China.
| |
Collapse
|
5
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
6
|
Liang Z, Zhao S, Liu Y, Cheng C. The promise of mitochondria in the treatment of glioblastoma: a brief review. Discov Oncol 2025; 16:142. [PMID: 39924629 PMCID: PMC11807951 DOI: 10.1007/s12672-025-01891-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
Glioblastoma (GBM) is a prevalent and refractory type of brain tumor. Over the past two decades, there have been minimal advancements in GBM therapy. The current standard treatment involves surgical excision followed by radiation and chemotherapy. Compared to other tumors, GBM is more challenging to treat due to the presence of glioma stem-like cells (GSCs) and the blood-brain barrier, resulting in an extremely low survival rate. Mitochondria play a critical role in tumor respiration, metabolism, and multiple signaling pathways involved in tumor formation, progression, and cell apoptosis. Consequently, mitochondria represent promising targets for developing novel anticancer agents, including those targeting oxidative phosphorylation, reactive oxygen species (ROS), mitochondrial transfer, and mitophagy. This review outlines the mitochondrial-related therapeutic targets in GBM, highlighting the potential of mitochondria as a target for GBM treatment.
Collapse
Affiliation(s)
- Zhuo Liang
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Songyun Zhao
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yuankun Liu
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Chao Cheng
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
| |
Collapse
|
7
|
Zuo CY, Zhang CS, Zhang HX, Gou CY, Lei H, Tian FW, Wang ZX, Yin HY, Yu SG. Moxibustion Alleviates Inflammation via SIRT5-mediated Post-translational Modification and Macrophage Polarization. Inflammation 2025:10.1007/s10753-025-02239-y. [PMID: 39899130 DOI: 10.1007/s10753-025-02239-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/30/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025]
Abstract
Macrophage polarisation is influenced by Sirtuin5 (SIRT5), which is crucial for regulating anti-inflammatory processes. Moxibustion, a traditional Chinese medicine therapy, exerts anti-inflammatory effects by altering the succinate/α-ketoglutarate (α-KG) ratio, an indicator of the M1 to M2 macrophage shift. Glutamate dehydrogenase 1 (GLUD1), a key enzyme involved in α-KG production, is desuccinylated by SIRT5. Currently, the potential influence of moxibustion on SIRT5-GLUD1-α-KG-mediated macrophage polarization in inflammatory diseases remains unexplored. C57BL/6 J and Sirt5 knockout mice were used as complete Freund's adjuvant (CFA)-induced adjuvant arthritis models. Moxibustion and acupoint injections of MC3482 were administered. Paw capacity asssays and ELISA were performed to quantify inflammatory effects and the expression of succinate, and α-KG expressions. Flow cytometry (FCM) and immunofluorescence were used to assesss the expression of M1- and M2-like macrophages. LC-MS/MS-based proteomic analysis was performed, and GLUD1 was identified desuccinylated protein associated with SIRT5. Western blotting and immunoprecipitation (IP) were used to detect SIRT5, GLUD1, and succinylated GLUD1expressions. Moxibustion and the SIRT5-mediated desuccinylation inhibitor MC3482 decreased inflammation by increasing the number of M2 macrophages and reducing the number of M1 macrophage in the CFA model. The potential mechanism may be related to the effects of moxibustion and SIRT5 inhibition, which inverted succinate and α-KG levels in the CFA group, resulting in low succinate, high α-KG, and increased GLUD1 succinylation after treatment. These findings suggest that the anti-inflammatory effects moxibustion are related to the impact of macrophage conversion after SIRT5-mediated post-translational modification.
Collapse
Affiliation(s)
- Chuan-Yi Zuo
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Cheng-Shun Zhang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Han-Xiao Zhang
- Faculty of Medicine, Université Paris-Saclay, 94800, Villejuif, France
| | - Chun-Yan Gou
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Hong Lei
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Feng-Wei Tian
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Zhu-Xing Wang
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Hai-Yan Yin
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| | - Shu-Guang Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| |
Collapse
|
8
|
Glorieux C, Enríquez C, Buc Calderon P. The complex interplay between redox dysregulation and mTOR signaling pathway in cancer: A rationale for cancer treatment. Biochem Pharmacol 2025; 232:116729. [PMID: 39709038 DOI: 10.1016/j.bcp.2024.116729] [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: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
The mechanistic target of rapamycin (mTOR) is a highly conserved serine/threonine kinase that plays a critical role in regulating cellular processes such as growth, proliferation, and metabolism in healthy cells. Dysregulation of mTOR signaling and oxidative stress have been implicated in various diseases including cancer. This review aims to provide an overview of the current understanding of mTOR and its involvement in cell survival and the regulation of cancer cell metabolism as well as its complex interplay with reactive oxygen species (ROS). On the one hand, ROS can inhibit or activate mTOR pathway in cancer cells through various mechanisms. Conversely, mTOR signaling can induce oxidative stress in tumor cells notably due to the inhibition in the expression of antioxidant enzyme genes. Since mTOR is often activated and plays crucial role in cancer cell survival, the use of mTOR inhibitors, which often induce ROS accumulation, could be an interesting approach for cancer treatment. This review will address the advantages, disadvantages, combination strategies, and limitations associated with therapeutic modulation of mTOR signaling pathway in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, China.
| | - Cinthya Enríquez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile; Programa de Doctorado en Química Medicinal, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile; Instituto de Química Medicinal, Universidad Arturo Prat, 1100000 Iquique, Chile; Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Brussels, Belgium.
| |
Collapse
|
9
|
Liu B, Zhao Z, Wang P, Aihemaiti K, Zhu L, Wei Q, Li W, Yuan X, Wu J, Jiang C, Hao M, Wang J. GlutaR: A High-Performance Fluorescent Protein-Based Sensor for Spatiotemporal Monitoring of Glutamine Dynamics In Vivo. Angew Chem Int Ed Engl 2025; 64:e202416608. [PMID: 39539096 DOI: 10.1002/anie.202416608] [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: 08/29/2024] [Revised: 11/03/2024] [Accepted: 11/12/2024] [Indexed: 11/16/2024]
Abstract
Glutamine is the most abundant amino acid in human blood and muscle, and is integral to a wide variety of functions in cancer cells. However, the inability to monitor the subcellular distribution of glutamine in real-time has obscured understanding of glutamine metabolism under physiological and pathological conditions. Here, we report the development of a genetically encoded fluorescent sensor and demonstrate how this GlnBP-cpYFP fusion "GlutaR sensor" undergoes glutamine-induced conformational changes reflected in detectable fluorescence responses. Obtained after iterative screening of approximately 1,600 variants, GlutaR exhibits a ratiometric readout, fast response kinetics, and high responsivity (R488/405 of ~1000 %), and we demonstrate its selectivity for monitoring glutamine fluctuations in multiple cell types. Additionally, using digitonin permeabilization of GlutaR HeLa cells, we generated a calibration curve and performed in situ titration to quantify free glutamine concentrations in subcellular compartments (cytosol, nucleus, mitochondria). Subsequently, we applied GlutaR to investigate how chemical and genetic inhibition of glutamine synthetase (GS) and glutaminase (GLS) differentially alter glutamine levels in subcellular compartments. Finally, we demonstrate GlutaR's ability to monitor dynamic glutamine levels in muscle and liver tissues of diabetic mice in vivo. These findings collectively demonstrate GlutaR as a versatile tool for the spatiotemporal characterization of glutamine metabolism in living cells and tissues.
Collapse
Affiliation(s)
- Bingjie Liu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zijie Zhao
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Pengcheng Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Peking University Third Hospital, Beijing, 100191, China
| | - Kamiran Aihemaiti
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Fudan University, Shanghai, 200433, China
| | - Lixin Zhu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qingpeng Wei
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jing Wu
- Medical and Health Analysis Center, Biological Imaging and Analysis Laboratory, Peking University, Beijing, 100191, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Min Hao
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- National Biomedical Imaging Center, Peking University, Beijing, 100191, China
| |
Collapse
|
10
|
Yi Y, Wang G, Zhang W, Yu S, Fei J, An T, Yi J, Li F, Huang T, Yang J, Niu M, Wang Y, Xu C, Xiao ZXJ. Mitochondrial-cytochrome c oxidase II promotes glutaminolysis to sustain tumor cell survival upon glucose deprivation. Nat Commun 2025; 16:212. [PMID: 39747079 PMCID: PMC11695821 DOI: 10.1038/s41467-024-55768-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Glucose deprivation, a hallmark of the tumor microenvironment, compels tumor cells to seek alternative energy sources for survival and growth. Here, we show that glucose deprivation upregulates the expression of mitochondrial-cytochrome c oxidase II (MT-CO2), a subunit essential for the respiratory chain complex IV, in facilitating glutaminolysis and sustaining tumor cell survival. Mechanistically, glucose deprivation activates Ras signaling to enhance MT-CO2 transcription and inhibits IGF2BP3, an RNA-binding protein, to stabilize MT-CO2 mRNA. Elevated MT-CO2 increases flavin adenosine dinucleotide (FAD) levels in activating lysine-specific demethylase 1 (LSD1) to epigenetically upregulate JUN transcription, consequently promoting glutaminase-1 (GLS1) and glutaminolysis for tumor cell survival. Furthermore, MT-CO2 is indispensable for oncogenic Ras-induced glutaminolysis and tumor growth, and elevated expression of MT-CO2 is associated with poor prognosis in lung cancer patients. Together, these findings reveal a role for MT-CO2 in adapting to metabolic stress and highlight MT-CO2 as a putative therapeutic target for Ras-driven cancers.
Collapse
Affiliation(s)
- Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| | - Guoqiang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Wenhua Zhang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Shuhan Yu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Department of Oncology & Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Junjie Fei
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Tingting An
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jianqiao Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Fengtian Li
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, China
| | - Ting Huang
- Department of Oncology & Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jian Yang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengmeng Niu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
- Department of Oncology & Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Cortellino S, D'Angelo M, Quintiliani M, Giordano A. Cancer knocks you out by fasting: Cachexia as a consequence of metabolic alterations in cancer. J Cell Physiol 2025; 240:e31417. [PMID: 39245862 DOI: 10.1002/jcp.31417] [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: 04/28/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024]
Abstract
Neoplastic transformation reprograms tumor and surrounding host cell metabolism, increasing nutrient consumption and depletion in the tumor microenvironment. Tumors uptake nutrients from neighboring normal tissues or the bloodstream to meet energy and anabolic demands. Tumor-induced chronic inflammation, a high-energy process, also consumes nutrients to sustain its dysfunctional activities. These tumor-related metabolic and physiological changes, including chronic inflammation, negatively impact systemic metabolism and physiology. Furthermore, the adverse effects of antitumor therapy and tumor obstruction impair the endocrine, neural, and gastrointestinal systems, thereby confounding the systemic status of patients. These alterations result in decreased appetite, impaired nutrient absorption, inflammation, and shift from anabolic to catabolic metabolism. Consequently, cancer patients often suffer from malnutrition, which worsens prognosis and increases susceptibility to secondary adverse events. This review explores how neoplastic transformation affects tumor and microenvironment metabolism and inflammation, leading to poor prognosis, and discusses potential strategies and clinical interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), School for Advanced Studies, Federico II University, Naples, Italy
- SHRO Italia Foundation ETS, Candiolo, Turin, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
12
|
Li W, Chen J, Guo Z. Targeting metabolic pathway enhance CAR-T potency for solid tumor. Int Immunopharmacol 2024; 143:113412. [PMID: 39454410 DOI: 10.1016/j.intimp.2024.113412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/01/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have great potential in cancer therapy, particularly in treating hematologic malignancies. However, their efficacy in solid tumors remains limited, with a significant proportion of patients failing to achieve long-term complete remission. One major challenge is the premature exhaustion of CAR-T cells, often due to insufficient metabolic energy. The survival, function and metabolic adaptation of CAR-T cells are key determinants of their therapeutic efficacy. We explore how targeting metabolic pathways in the tumor microenvironment can enhance CAR-T cell therapy by addressing metabolic competition and immunosuppression that impair CAR-T cell function. Tumors undergo metabolically reprogrammed to meet their rapid proliferation, thereby modulating metabolic pathways in immune cells to promote immunosuppression. The distinct metabolic requirements of tumors and T cells create a competitive environment, affecting the efficacy of CAR-T cell therapy. Recent research on glucose, lipid and amino acid metabolism, along with the interactions between tumor and immune cell metabolism, has revealed that targeting these metabolic processes can enhance antitumor immune responses. Combining metabolic interventions with existing antitumor therapies can fulfill the metabolic demands of immune cells, providing new ideas for tumor immunometabolic therapies. This review discusses the latest advances in the immunometabolic mechanisms underlying tumor immunosuppression, their implications for immunotherapy, and summarizes potential metabolic targets to improve the efficacy of CAR-T therapy.
Collapse
Affiliation(s)
- Wenying Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Jiannan Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
13
|
Mao Y, Xia Z, Xia W, Jiang P. Metabolic reprogramming, sensing, and cancer therapy. Cell Rep 2024; 43:115064. [PMID: 39671294 DOI: 10.1016/j.celrep.2024.115064] [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: 07/04/2024] [Revised: 10/30/2024] [Accepted: 11/21/2024] [Indexed: 12/15/2024] Open
Abstract
The metabolic reprogramming of tumor cells is a crucial strategy for their survival and proliferation, involving tissue- and condition-dependent remodeling of certain metabolic pathways. While it has become increasingly clear that tumor cells integrate extracellular and intracellular signals to adapt and proliferate, nutrient and metabolite sensing also exert direct or indirect influences, although the underlying mechanisms remain incompletely understood. Furthermore, metabolic changes not only support the rapid growth and dissemination of tumor cells but also promote immune evasion by metabolically "educating" immune cells in the tumor microenvironment (TME). Recent studies have highlighted the profound impact of metabolic reprogramming on the TME and the potential of targeting metabolic pathways as a therapeutic strategy, with several enzyme inhibitors showing promising results in clinical trials. Thus, understanding how tumor cells alter their metabolic pathways and metabolically remodel the TME to support their survival and proliferation may offer new strategies for metabolic therapy and immunotherapy.
Collapse
Affiliation(s)
- Youxiang Mao
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ziyan Xia
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Wenjun Xia
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Peng Jiang
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
14
|
Ponton-Almodovar A, Sanderson S, Rattan R, Bernard JJ, Horibata S. Ovarian tumor microenvironment contributes to tumor progression and chemoresistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:53. [PMID: 39802952 PMCID: PMC11724355 DOI: 10.20517/cdr.2024.111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
Ovarian cancer is one of the deadliest gynecologic cancers affecting the female reproductive tract. This is largely attributed to frequent recurrence and development of resistance to the platinum-based drugs cisplatin and carboplatin. One of the major contributing factors to increased cancer progression and resistance to chemotherapy is the tumor microenvironment (TME). Extracellular signaling from cells within the microenvironment heavily influences progression and drug resistance in ovarian cancer. This is frequently done through metabolic reprogramming, the process where cancer cells switch between biochemical pathways to increase their chances of survival and proliferation. Here, we focus on how crosstalk between components of the TME and the tumor promotes resistance to platinum-based chemotherapy. We highlight the role of cancer-associated fibroblasts, immune cells, adipocytes, and endothelial cells in ovarian tumor progression, invasion, metastasis, and chemoresistance. We also highlight recent advancements in targeting components of the TME as a novel therapeutic avenue to combat chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Adriana Ponton-Almodovar
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Authors contributed equally
| | - Samuel Sanderson
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA
- Authors contributed equally
| | - Ramandeep Rattan
- Department of Women’s Health Services, Henry Ford Health System, Detroit, MI 48202, USA
| | - Jamie J. Bernard
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Sachi Horibata
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Cell and Molecular Biology Program, College of Natural Science, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
15
|
Shi J, Lin Z, Zheng Z, Chen M, Huang X, Wang J, Li M, Shao J. Glutamine metabolism promotes human trophoblast cell invasion via COL1A1 mediated by PI3K-AKT pathway. J Reprod Immunol 2024; 166:104321. [PMID: 39243705 DOI: 10.1016/j.jri.2024.104321] [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: 03/21/2024] [Revised: 07/13/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024]
Abstract
Abnormal trophoblast invasion function is an important cause of recurrent spontaneous abortion (RSA). Recent research has revealed a connection between glutamine metabolism and RSA. However, the interplay between these three factors and their related mechanisms remains unclear. To address this issue, we collected villus tissues from 10 healthy women with induced abortion and from 10 women with RSA to detect glutamine metabolism. Then, the trophoblast cell line HTR-8/SVneo was used in vitro to explore the effect of glutamine metabolism on trophoblast cells invasion, which was tested by transwell assay. We found that the concentration of glutamine in the villi of the normal pregnancy group was significantly higher than that in the RSA group. Correspondingly, the expression levels of key enzymes involved in glutamine synthesis and catabolism, including glutamine synthetase and glutaminase, were significantly higher in the villi of the normal pregnancy group. Regarding trophoblast cells, glutamine markedly enhanced the proliferative and invasive abilities of HTR-8/SVneo cells. Additionally, collagen type I alpha 1 (COL1A1) was confirmed to be a downstream target of glutamine, and glutamine also activated the PI3K-AKT pathway in HTR-8/SVneo cells. These findings indicate that glutamine metabolism facilitates the invasion of trophoblasts by up-regulating COL1A1 expression through the activation of the PI3K-AKT pathway, but the specific mechanism of COL1A1 requires further study.
Collapse
Affiliation(s)
- Jialu Shi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200090, China
| | - Zhi Lin
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200090, China
| | - Zimeng Zheng
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
| | - Min Chen
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province 310000, China
| | - Xu Huang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
| | - Jiarui Wang
- Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Mingqing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China.
| | - Jun Shao
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200090, China.
| |
Collapse
|
16
|
Zhang Y, Wei M, Wang X, Xu Y, Zong R, Lin X, Li S, Chen W, Liu Z, Chen Q. Dipeptide alanine-glutamine ameliorates retinal neurodegeneration in an STZ-induced rat model. Front Pharmacol 2024; 15:1490443. [PMID: 39629074 PMCID: PMC11611560 DOI: 10.3389/fphar.2024.1490443] [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: 09/03/2024] [Accepted: 11/06/2024] [Indexed: 12/06/2024] Open
Abstract
Introduction Diabetic retinopathy (DR) is a common complication of diabetes. Retinal neuronal degeneration is an early event in DR, indicated by the declined electroretinogram (ERG). Dipeptide alanine-glutamine (Ala-Gln) is widely used as a nutritional supplement in the clinic and has anti-inflammatory effects on the gastrointestinal system. Studies also reported that glutamine has beneficial effects on diabetes. This study aimed to investigate the possible therapeutic effects of Ala-Gln in diabetic retinal neurodegeneration and to delineate its mechanism of action. Methods The Streptozotocin (STZ)-induced rat model was used as a DR model. ERG was used to measure the neuronal function of the retina. Western blot analysis was performed to test the expression of proteins. Immunofluorescence staining was used for the detection and localization of proteins. Results In diabetic rats, the amplitudes of ERG were declined, while Ala-Gln restored the declined ERG. Retinal levels of inflammatory factors were significantly decreased in Ala-Gln-treated diabetic rats. Ala-Gln mitigated the declined levels of glutamine synthetase and ameliorated the upregulated levels of glial fibrillary acidic protein (GFAP) in diabetic retinas. Moreover, Ala-Gln upregulated the glycolytic enzymes pyruvate kinase isozymes 2 (PKM2), lactate dehydrogenase A (LDHA) and LDHB and stimulated the mTOR signaling pathway in diabetic retinas. The mitochondrial function was improved after the treatment of Ala-Gln in diabetic retinas. Discussion Ala-Gln ameliorates retinal neurodegeneration by reducing inflammation and enhancing glucose metabolism and mitochondrial function in DR. Therefore, manipulation of metabolism by Ala-Gln may be a novel therapeutic avenue for retinal neurodegeneration in DR.
Collapse
Affiliation(s)
- Yuhan Zhang
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Mingyan Wei
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xin Wang
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuan Xu
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Rongrong Zong
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiang Lin
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Shiying Li
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Wensheng Chen
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Zuguo Liu
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Qian Chen
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
17
|
Zhu B, Xiang K, Li T, Li X, Shi F. The signature of extracellular vesicles in hypoxic breast cancer and their therapeutic engineering. Cell Commun Signal 2024; 22:512. [PMID: 39434182 PMCID: PMC11492701 DOI: 10.1186/s12964-024-01870-w] [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: 07/03/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024] Open
Abstract
Breast cancer (BC) currently ranks second in the global cancer incidence rate. Hypoxia is a common phenomenon in BC. Under hypoxic conditions, cells in the tumor microenvironment (TME) secrete numerous extracellular vesicles (EVs) to achieve intercellular communication and alter the metabolism of primary and metastatic tumors that shape the TME. In addition, emerging studies have indicated that hypoxia can promote resistance to tumor treatment. Engineered EVs are expected to become carriers for cancer treatment due to their high biocompatibility, low immunogenicity, high drug delivery efficiency, and ease of modification. In this review, we summarize the mechanisms of EVs in the primary TME and distant metastasis of BC under hypoxic conditions. Additionally, we highlight the potential applications of engineered EVs in mitigating the malignant phenotypes of BC cells under hypoxia.
Collapse
Affiliation(s)
- Baiheng Zhu
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kehao Xiang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Tanghua Li
- The First Clinical Medical School, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin Li
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Fujun Shi
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
18
|
Park M, Jin J, An DY, Kim DH, Lee J, Yun JW, Hwang I, Park JS, Kim MK, Lee YM, Byun JK, Choi YK, Park KG. Targeting YAP Activity and Glutamine Metabolism Cooperatively Suppresses Tumor Progression by Preventing Extracellular Matrix Accumulation. Cancer Res 2024; 84:3388-3401. [PMID: 39073839 DOI: 10.1158/0008-5472.can-23-3933] [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: 12/12/2023] [Revised: 05/17/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Cancer cells use multiple mechanisms to evade the effects of glutamine metabolism inhibitors. The pathways that govern responses to alterations in glutamine availability within the tumor may represent therapeutic targets for combinatorial strategies with these inhibitors. Here, we showed that targeting glutamine utilization stimulated Yes-associated protein (YAP) signaling in cancer cells by reducing cyclic adenosine monophosphate/protein kinase A (PKA)-dependent phosphorylation of large tumor suppressor (LATS). Elevated YAP activation induced extracellular matrix (ECM) deposition by increasing the secretion of connective tissue growth factor that promoted the production of fibronectin and collagen by surrounding fibroblasts. Consequently, inhibiting YAP synergized with inhibition of glutamine utilization to effectively suppress tumor growth in vivo, along with a concurrent decrease in ECM deposition. Blocking ECM remodeling also augmented the tumor suppressive effects of the glutamine utilization inhibitor. Collectively, these data reveal mechanisms by which targeting glutamine utilization increases ECM accumulation and identify potential strategies to reduce ECM levels and increase the efficacy of glutamine metabolism inhibitors. Significance: Blocking glutamine utilization activates YAP to promote ECM deposition by fibroblasts, highlighting the potential of YAP inhibitors and antifibrotic strategies as promising approaches for effective combination metabolic therapies in cancer.
Collapse
Affiliation(s)
- Mihyang Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
| | - Jonghwa Jin
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Da Young An
- Department of Biomedical Science, Kyungpook National University, Daegu, South Korea
| | - Dong-Ho Kim
- Department of Biomedical Science, Kyungpook National University, Daegu, South Korea
| | - Jaebon Lee
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, South Korea
| | - Jae Won Yun
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, South Korea
| | - Ilseon Hwang
- Department of Pathology, Keimyung University School of Medicine, Daegu, South Korea
| | - Jae Seok Park
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea
| | - Mi Kyung Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea
| | - You Mie Lee
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, Republic of Korea
- Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, South Korea
| | - Jun-Kyu Byun
- Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, South Korea
| | - Yeon-Kyung Choi
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
- Department of Biomedical Science, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
19
|
Taneja N, Chauhan A, Kulshreshtha R, Singh S. HIF-1 mediated metabolic reprogramming in cancer: Mechanisms and therapeutic implications. Life Sci 2024; 352:122890. [PMID: 38971364 DOI: 10.1016/j.lfs.2024.122890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Cancer cells undergo metabolic reprogramming to survive in hypoxic conditions and meet the elevated energy demands of the cancer microenvironment. This metabolic alteration is orchestrated by hypoxia-inducible factor 1 (HIF-1), regulating various processes within cancer cells. The intricate metabolic modifications induced by hypoxia underscore the significance of HIF-1-induced metabolic reprogramming in promoting each aspect of cancer progression. The complex interactions between HIF-1 signalling and cellular metabolic processes in response to hypoxia are examined in this study, focusing on the metabolism of carbohydrates, nucleotides, lipids, and amino acids. Comprehending the various regulatory mechanisms controlled by HIF-1 in cellular metabolism sheds light on the intricate biology of cancer growth and offers useful insights for developing targeted treatments.
Collapse
Affiliation(s)
- Nikita Taneja
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India
| | - Akansha Chauhan
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| | - Sandhya Singh
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India.
| |
Collapse
|
20
|
Allevato MM, Trinh S, Koshizuka K, Nachmanson D, Nguyen TTC, Yokoyama Y, Wu X, Andres A, Wang Z, Watrous J, Molinolo AA, Mali P, Harismendy O, Jain M, Wild R, Gutkind JS. A genome-wide CRISPR screen reveals that antagonism of glutamine metabolism sensitizes head and neck squamous cell carcinoma to ferroptotic cell death. Cancer Lett 2024; 598:217089. [PMID: 38964731 DOI: 10.1016/j.canlet.2024.217089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024]
Abstract
Glutamine is a conditionally essential amino acid for the growth and survival of rapidly proliferating cancer cells. Many cancers are addicted to glutamine, and as a result, targeting glutamine metabolism has been explored clinically as a therapeutic approach. Glutamine-catalyzing enzymes are highly expressed in primary and metastatic head and neck squamous cell carcinoma (HNSCC). However, the nature of the glutamine-associated pathways in this aggressive cancer type has not been elucidated. Here, we explored the therapeutic potential of a broad glutamine antagonist, DRP-104 (sirpiglenastat), in HNSCC tumors and aimed at shedding light on glutamine-dependent pathways in this disease. We observed a potent antitumoral effect of sirpiglenastat in HPV- and HPV + HNSCC xenografts. We conducted a whole-genome CRISPR screen and metabolomics analyses to identify mechanisms of sensitivity and resistance to glutamine metabolism blockade. These approaches revealed that glutamine metabolism blockade results in the rapid buildup of polyunsaturated fatty acids (PUFAs) via autophagy nutrient-sensing pathways. Finally, our analysis demonstrated that GPX4 mediates the protection of HNSCC cells from accumulating toxic lipid peroxides; hence, glutamine blockade sensitizes HNSCC cells to ferroptosis cell death upon GPX4 inhibition. These findings demonstrate the therapeutic potential of sirpiglenastat in HNSCC and establish a novel link between glutamine metabolism and ferroptosis, which may be uniquely translated into targeted glutamine-ferroptosis combination therapies.
Collapse
Affiliation(s)
- Michael M Allevato
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Sally Trinh
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Keiichi Koshizuka
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Daniela Nachmanson
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Thien-Tu C Nguyen
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Yumi Yokoyama
- Dracen Pharmaceuticals Inc., 9276 Scranton Rd. Suite 200, San Diego, CA, USA
| | - Xingyu Wu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Allen Andres
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Zhiyong Wang
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Jeramie Watrous
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Alfredo A Molinolo
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Olivier Harismendy
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Mohit Jain
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Robert Wild
- Dracen Pharmaceuticals Inc., 9276 Scranton Rd. Suite 200, San Diego, CA, USA
| | - J Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
21
|
Yu S, Chen M, Zhu X, Chen C, Liang J, Wang H, Lu J, Ding Y, Kong M, Teng L, Zhou D. The combination of exon sequencing and metabolomics to establish a molecular typing system for gastric cancer. Heliyon 2024; 10:e34317. [PMID: 39170180 PMCID: PMC11336309 DOI: 10.1016/j.heliyon.2024.e34317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024] Open
Abstract
Background Gastric cancer (GC) is one of the most common malignant tumors in the world. It has become increasingly difficult to meet the needs of precision therapy using the existing molecular typing system. Therefore, developing a more effective molecular typing system for GC is urgent. Methods In this study, 100 Chinese GC patients were included. Whole-exome sequencing (WES) and metabolomics analysis were performed to reveal the characteristics of genomic and metabolic changes. Results In WES, nonsynonymous mutations accounted for the majority. Based on metabolomics, GC has been divided into three subtypes with distinct metabolic features. Importantly, we ultimately divided GC into four subtypes with different metabolic characteristics, genomic alterations, and clinical prognoses by incorporating biomics analysis. Conclusions Integrating biological features, we constructed a novel molecular system for GC that was closely related to genetics and metabolism, providing new insights for further understanding the heterogeneity and formulating precise treatment strategies.
Collapse
Affiliation(s)
- Shanshan Yu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Ming Chen
- Department of Surgical Oncology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Xiaohua Zhu
- Department of Medical Oncology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Cheng Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinxiao Liang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Haiyong Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jun Lu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Mei Kong
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Donghui Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
22
|
Yin Y, Feng W, Chen J, Chen X, Wang G, Wang S, Xu X, Nie Y, Fan D, Wu K, Xia L. Immunosuppressive tumor microenvironment in the progression, metastasis, and therapy of hepatocellular carcinoma: from bench to bedside. Exp Hematol Oncol 2024; 13:72. [PMID: 39085965 PMCID: PMC11292955 DOI: 10.1186/s40164-024-00539-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy with high incidence, recurrence, and metastasis rates. The emergence of immunotherapy has improved the treatment of advanced HCC, but problems such as drug resistance and immune-related adverse events still exist in clinical practice. The immunosuppressive tumor microenvironment (TME) of HCC restricts the efficacy of immunotherapy and is essential for HCC progression and metastasis. Therefore, it is necessary to elucidate the mechanisms behind immunosuppressive TME to develop and apply immunotherapy. This review systematically summarizes the pathogenesis of HCC, the formation of the highly heterogeneous TME, and the mechanisms by which the immunosuppressive TME accelerates HCC progression and metastasis. We also review the status of HCC immunotherapy and further discuss the existing challenges and potential therapeutic strategies targeting immunosuppressive TME. We hope to inspire optimizing and innovating immunotherapeutic strategies by comprehensively understanding the structure and function of immunosuppressive TME in HCC.
Collapse
Affiliation(s)
- Yue Yin
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Weibo Feng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Jie Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Xilang Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Guodong Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiao Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Daiming Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Kaichun Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Limin Xia
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
23
|
Kim M, Hwang S, Jeong SM. Targeting cellular adaptive responses to glutaminolysis perturbation for cancer therapy. Mol Cells 2024; 47:100096. [PMID: 39038517 PMCID: PMC11342766 DOI: 10.1016/j.mocell.2024.100096] [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: 04/15/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
Metabolic aberrations, notably deviations in glutamine metabolism, are crucial in the oncogenic process, offering vital resources for the unlimited proliferation and enhanced survival capabilities of cancer cells. The dependency of malignant cells on glutamine metabolism has led to the proposition of targeted therapeutic strategies. However, the capability of cancer cells to initiate adaptive responses undermines the efficacy of these therapeutic interventions. This review meticulously examines the multifaceted adaptive mechanisms that cancer cells deploy to sustain survival and growth following the disruption of glutamine metabolism. Emphasis is placed on the roles of transcription factors, alterations in metabolic pathways, the mechanistic target of rapamycin complex 1 signaling axis, autophagy, macropinocytosis, nucleotide biosynthesis, and the scavenging of ROS. Thus, the delineation and subsequent targeting of these adaptive responses in the context of therapies aimed at glutamine metabolism offer a promising avenue for circumventing drug resistance in cancer treatment.
Collapse
Affiliation(s)
- Minjoong Kim
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Sunsook Hwang
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Seung Min Jeong
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| |
Collapse
|
24
|
Meng T, He D, Han Z, Shi R, Wang Y, Ren B, Zhang C, Mao Z, Luo G, Deng J. Nanomaterial-Based Repurposing of Macrophage Metabolism and Its Applications. NANO-MICRO LETTERS 2024; 16:246. [PMID: 39007981 PMCID: PMC11250772 DOI: 10.1007/s40820-024-01455-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024]
Abstract
Macrophage immunotherapy represents an emerging therapeutic approach aimed at modulating the immune response to alleviate disease symptoms. Nanomaterials (NMs) have been engineered to monitor macrophage metabolism, enabling the evaluation of disease progression and the replication of intricate physiological signal patterns. They achieve this either directly or by delivering regulatory signals, thereby mapping phenotype to effector functions through metabolic repurposing to customize macrophage fate for therapy. However, a comprehensive summary regarding NM-mediated macrophage visualization and coordinated metabolic rewiring to maintain phenotypic equilibrium is currently lacking. This review aims to address this gap by outlining recent advancements in NM-based metabolic immunotherapy. We initially explore the relationship between metabolism, polarization, and disease, before delving into recent NM innovations that visualize macrophage activity to elucidate disease onset and fine-tune its fate through metabolic remodeling for macrophage-centered immunotherapy. Finally, we discuss the prospects and challenges of NM-mediated metabolic immunotherapy, aiming to accelerate clinical translation. We anticipate that this review will serve as a valuable reference for researchers seeking to leverage novel metabolic intervention-matched immunomodulators in macrophages or other fields of immune engineering.
Collapse
Affiliation(s)
- Tingting Meng
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Danfeng He
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Zhuolei Han
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Rong Shi
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
- Department of Breast Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730030, People's Republic of China
| | - Yuhan Wang
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Bibo Ren
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Cheng Zhang
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Zhengwei Mao
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China.
| | - Gaoxing Luo
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China.
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
25
|
Sharma A, Debik J, Naume B, Ohnstad HO, Bathen TF, Giskeødegård GF. Comprehensive multi-omics analysis of breast cancer reveals distinct long-term prognostic subtypes. Oncogenesis 2024; 13:22. [PMID: 38871719 DOI: 10.1038/s41389-024-00521-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024] Open
Abstract
Breast cancer (BC) is a leading cause of cancer-related death worldwide. The diverse nature and heterogeneous biology of BC pose challenges for survival prediction, as patients with similar diagnoses often respond differently to treatment. Clinically relevant BC intrinsic subtypes have been established through gene expression profiling and are implemented in the clinic. While these intrinsic subtypes show a significant association with clinical outcomes, their long-term survival prediction beyond 5 years often deviates from expected clinical outcomes. This study aimed to identify naturally occurring long-term prognostic subgroups of BC based on an integrated multi-omics analysis. This study incorporates a clinical cohort of 335 untreated BC patients from the Oslo2 study with long-term follow-up (>12 years). Multi-Omics Factor Analysis (MOFA+) was employed to integrate transcriptomic, proteomic, and metabolomic data obtained from the tumor tissues. Our analysis revealed three prominent multi-omics clusters of BC patients with significantly different long-term prognoses (p = 0.005). The multi-omics clusters were validated in two independent large cohorts, METABRIC and TCGA. Importantly, a lack of prognostic association to long-term follow-up above 12 years in the previously established intrinsic subtypes was shown for these cohorts. Through a systems-biology approach, we identified varying enrichment levels of cell-cycle and immune-related pathways among the prognostic clusters. Integrated multi-omics analysis of BC revealed three distinct clusters with unique clinical and biological characteristics. Notably, these multi-omics clusters displayed robust associations with long-term survival, outperforming the established intrinsic subtypes.
Collapse
Affiliation(s)
- Abhibhav Sharma
- Department of Public Health and Nursing (ISM), Norwegian University of Science and Technology- NTNU, Trondheim, Norway.
| | - Julia Debik
- Department of Public Health and Nursing (ISM), Norwegian University of Science and Technology- NTNU, Trondheim, Norway
- Department of Circulation and Medical Imaging, NTNU, Trondheim, Norway
| | - Bjørn Naume
- Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hege Oma Ohnstad
- Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, NTNU, Trondheim, Norway
| | - Guro F Giskeødegård
- Department of Public Health and Nursing (ISM), Norwegian University of Science and Technology- NTNU, Trondheim, Norway.
| |
Collapse
|
26
|
Lu J, Gui W, Tang S, Shi Q, Wang X, Huang L, Shen Y, Yang S, Xiang J, Yuan J, Mo J, Kong X, Huang M, Li X, Lu C. Nicotinamide N-Methyltransferase (NNMT) is Involved in Gastric Adenocarcinoma Immune Infiltration by Driving Amino Acid Metabolism. Nutr Cancer 2024; 76:745-759. [PMID: 38855943 DOI: 10.1080/01635581.2024.2359741] [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: 11/08/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
Objectives: This study investigates the role of Nicotinamide N-methyltransferase (NNMT) in immune infiltration modulation through amino acid metabolism in gastric adenocarcinoma (STAD). Methods: Utilizing data from The Cancer Genome Atlas (TCGA) and validated with clinical samples, we analyzed NNMT expression and its prognostic implications in STAD. Differential amino acid profiles between cancerous and adjacent normal tissues were assessed, along with their associations with NNMT. Results: NNMT exhibits heightened expression in STAD cancer tissues, positively correlating with tumor immune infiltration. Additionally, twenty-eight amino acids display differential expression in gastric tissue, with their metabolic enzymes showing connections to NNMT. Conclusions: Elevated NNMT expression in STAD tissues potentially influences amino acid metabolism, thereby affecting immune infiltration dynamics and tumorigenesis in gastric adenocarcinoma.
Collapse
Affiliation(s)
- Jianyong Lu
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Wenliang Gui
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Shen Tang
- School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Qianqian Shi
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Xinhang Wang
- School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Liyuan Huang
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Yinghui Shen
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Shuting Yang
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Junni Xiang
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Jianglang Yuan
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Jiao Mo
- School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xingxing Kong
- School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Mingwei Huang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiyi Li
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Nanning, China
| | - Cailing Lu
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Nanning, China
| |
Collapse
|
27
|
Zou J, Mai C, Lin Z, Zhou J, Lai G. Targeting metabolism of breast cancer and its implications in T cell immunotherapy. Front Immunol 2024; 15:1381970. [PMID: 38680483 PMCID: PMC11045902 DOI: 10.3389/fimmu.2024.1381970] [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: 02/05/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
Breast cancer is a prominent health issue amongst women around the world. Immunotherapies including tumor targeted antibodies, adoptive T cell therapy, vaccines, and immune checkpoint blockers have rejuvenated the clinical management of breast cancer, but the prognosis of patients remains dismal. Metabolic reprogramming and immune escape are two important mechanisms supporting the progression of breast cancer. The deprivation uptake of nutrients (such as glucose, amino acid, and lipid) by breast cancer cells has a significant impact on tumor growth and microenvironment remodeling. In recent years, in-depth researches on the mechanism of metabolic reprogramming and immune escape have been extensively conducted, and targeting metabolic reprogramming has been proposed as a new therapeutic strategy for breast cancer. This article reviews the abnormal metabolism of breast cancer cells and its impact on the anti-tumor activity of T cells, and further explores the possibility of targeting metabolism as a therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Jialuo Zou
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Cunjun Mai
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhiqin Lin
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jian Zhou
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Guie Lai
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
28
|
Alhozeel B, Pandey SK, Shteinfer-Kuzmine A, Santhanam M, Shoshan-Barmatz V. Silencing the Mitochondrial Gatekeeper VDAC1 as a Potential Treatment for Bladder Cancer. Cells 2024; 13:627. [PMID: 38607066 PMCID: PMC11012128 DOI: 10.3390/cells13070627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 04/13/2024] Open
Abstract
The strategy for treating bladder cancer (BC) depends on whether there is muscle invasion or not, with the latter mostly treated with intravesical therapy, such as with bacillus Calmette-Guérin (BCG). However, BCG treatment is unsuccessful in 70% of patients, who are then subjected to radical cystectomy. Although immune-checkpoint inhibitors have been approved as a second-line therapy for a subset of BC patients, these have failed to meet primary endpoints in clinical trials. Thus, it is crucial to find a new treatment. The mitochondrial gatekeeper protein, the voltage-dependent anion channel 1 (VDAC1), mediates metabolic crosstalk between the mitochondria and cytosol and is involved in apoptosis. It is overexpressed in many cancer types, as shown here for BC, pointing to its significance in high-energy-demanding cancer cells. The BC cell lines UM-UC3 and HTB-5 express high VDAC1 levels compared to other cancer cell lines. VDAC1 silencing in these cells using siRNA that recognizes both human and mouse VDAC1 (si-m/hVDAC1-B) reduces cell viability, mitochondria membrane potential, and cellular ATP levels. Here, we used two BC mouse models: subcutaneous UM-UC3 cells and chemically induced BC using the carcinogen N-Butyl-N-(4-hydroxybutyl) nitrosamine (BBN). Subcutaneous UM-UC3-derived tumors treated with si-m/hVDAC1 showed inhibited tumor growth and reprogrammed metabolism, as reflected in the reduced expression of metabolism-related proteins, including Glut1, hexokinase, citrate synthase, complex-IV, and ATP synthase, suggesting reduced metabolic activity. Furthermore, si-m/hVDAC1-B reduced the expression levels of cancer-stem-cell-related proteins (cytokeratin-14, ALDH1a), modifying the tumor microenvironment, including decreased angiogenesis, extracellular matrix, tumor-associated macrophages, and inhibited epithelial-mesenchymal transition. The BBN-induced BC mouse model showed a clear carcinoma, with damaged bladder morphology and muscle-invasive tumors. Treatment with si-m/hVDAC1-B encapsulated in PLGA-PEI nanoparticles that were administered intravesically directly to the bladder showed a decreased tumor area and less bladder morphology destruction and muscle invasion. Overall, the obtained results point to the potential of si-m/hVDAC1-B as a possible therapeutic tool for treating bladder cancer.
Collapse
Affiliation(s)
- Belal Alhozeel
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.A.); (S.K.P.); (M.S.)
| | - Swaroop Kumar Pandey
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.A.); (S.K.P.); (M.S.)
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Manikandan Santhanam
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.A.); (S.K.P.); (M.S.)
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.A.); (S.K.P.); (M.S.)
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| |
Collapse
|
29
|
Gao L, Wei Z, Ying F, Huang L, Zhang J, Sun S, Wang Z, Cai J, Zhang Y. Glutamine metabolism prognostic index predicts tumour microenvironment characteristics and therapeutic efficacy in ovarian cancer. J Cell Mol Med 2024; 28:e18198. [PMID: 38506093 PMCID: PMC10951877 DOI: 10.1111/jcmm.18198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/17/2024] [Accepted: 02/09/2024] [Indexed: 03/21/2024] Open
Abstract
Mounting evidence has highlighted the multifunctional characteristics of glutamine metabolism (GM) in cancer initiation, progression and therapeutic regimens. However, the overall role of GM in the tumour microenvironment (TME), clinical stratification and therapeutic efficacy in patients with ovarian cancer (OC) has not been fully elucidated. Here, three distinct GM clusters were identified and exhibited different prognostic values, biological functions and immune infiltration in TME. Subsequently, glutamine metabolism prognostic index (GMPI) was constructed as a new scoring model to quantify the GM subtypes and was verified as an independent predictor of OC. Patients with low-GMPI exhibited favourable survival outcomes, lower enrichment of several oncogenic pathways, less immunosuppressive cell infiltration and better immunotherapy responses. Single-cell sequencing analysis revealed a unique evolutionary trajectory of OC cells from high-GMPI to low-GMPI, and OC cells with different GMPI might communicate with distinct cell populations through ligand-receptor interactions. Critically, the therapeutic efficacy of several drug candidates was validated based on patient-derived organoids (PDOs). The proposed GMPI could serve as a reliable signature for predicting patient prognosis and contribute to optimising therapeutic strategies for OC.
Collapse
Affiliation(s)
- Lingling Gao
- Department of Obstetrics and Gynecology, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zheng Wei
- Department of Obstetrics and GynecologyThird Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalTaiyuanChina
| | - Feiquan Ying
- Department of Obstetrics and Gynecology, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Lin Huang
- Department of Obstetrics and Gynecology, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jingni Zhang
- Department of Obstetrics and Gynecology, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Si Sun
- Department of Obstetrics and Gynecology, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
30
|
Wang B, Pei J, Xu S, Liu J, Yu J. A glutamine tug-of-war between cancer and immune cells: recent advances in unraveling the ongoing battle. J Exp Clin Cancer Res 2024; 43:74. [PMID: 38459595 PMCID: PMC10921613 DOI: 10.1186/s13046-024-02994-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/22/2024] [Indexed: 03/10/2024] Open
Abstract
Glutamine metabolism plays a pivotal role in cancer progression, immune cell function, and the modulation of the tumor microenvironment. Dysregulated glutamine metabolism has been implicated in cancer development and immune responses, supported by mounting evidence. Cancer cells heavily rely on glutamine as a critical nutrient for survival and proliferation, while immune cells require glutamine for activation and proliferation during immune reactions. This metabolic competition creates a dynamic tug-of-war between cancer and immune cells. Targeting glutamine transporters and downstream enzymes involved in glutamine metabolism holds significant promise in enhancing anti-tumor immunity. A comprehensive understanding of the intricate molecular mechanisms underlying this interplay is crucial for developing innovative therapeutic approaches that improve anti-tumor immunity and patient outcomes. In this review, we provide a comprehensive overview of recent advances in unraveling the tug-of-war of glutamine metabolism between cancer and immune cells and explore potential applications of basic science discoveries in the clinical setting. Further investigations into the regulation of glutamine metabolism in cancer and immune cells are expected to yield valuable insights, paving the way for future therapeutic interventions.
Collapse
Affiliation(s)
- Bolin Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinli Pei
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Shengnan Xu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
31
|
Fan Y, Xue H, Li Z, Huo M, Gao H, Guan X. Exploiting the Achilles' heel of cancer: disrupting glutamine metabolism for effective cancer treatment. Front Pharmacol 2024; 15:1345522. [PMID: 38510646 PMCID: PMC10952006 DOI: 10.3389/fphar.2024.1345522] [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: 11/28/2023] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
Cancer cells have adapted to rapid tumor growth and evade immune attack by reprogramming their metabolic pathways. Glutamine is an important nitrogen resource for synthesizing amino acids and nucleotides and an important carbon source in the tricarboxylic acid (TCA) cycle and lipid biosynthesis pathway. In this review, we summarize the significant role of glutamine metabolism in tumor development and highlight the vulnerabilities of targeting glutamine metabolism for effective therapy. In particular, we review the reported drugs targeting glutaminase and glutamine uptake for efficient cancer treatment. Moreover, we discuss the current clinical test about targeting glutamine metabolism and the prospective direction of drug development.
Collapse
Affiliation(s)
- Yuxin Fan
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| | - Han Xue
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| | - Zhimin Li
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| | - Mingge Huo
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| | - Hongxia Gao
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
| | - Xingang Guan
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| |
Collapse
|
32
|
Muranaka H, Akinsola R, Billet S, Pandol SJ, Hendifar AE, Bhowmick NA, Gong J. Glutamine Supplementation as an Anticancer Strategy: A Potential Therapeutic Alternative to the Convention. Cancers (Basel) 2024; 16:1057. [PMID: 38473414 PMCID: PMC10930819 DOI: 10.3390/cancers16051057] [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: 02/09/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Glutamine, a multifaceted nonessential/conditionally essential amino acid integral to cellular metabolism and immune function, holds pivotal importance in the landscape of cancer therapy. This review delves into the intricate dynamics surrounding both glutamine antagonism strategies and glutamine supplementation within the context of cancer treatment, emphasizing the critical role of glutamine metabolism in cancer progression and therapy. Glutamine antagonism, aiming to disrupt tumor growth by targeting critical metabolic pathways, is challenged by the adaptive nature of cancer cells and the complex metabolic microenvironment, potentially compromising its therapeutic efficacy. In contrast, glutamine supplementation supports immune function, improves gut integrity, alleviates treatment-related toxicities, and improves patient well-being. Moreover, recent studies highlighted its contributions to epigenetic regulation within cancer cells and its potential to bolster anti-cancer immune functions. However, glutamine implementation necessitates careful consideration of potential interactions with ongoing treatment regimens and the delicate equilibrium between supporting normal cellular function and promoting tumorigenesis. By critically assessing the implications of both glutamine antagonism strategies and glutamine supplementation, this review aims to offer comprehensive insights into potential therapeutic strategies targeting glutamine metabolism for effective cancer management.
Collapse
Affiliation(s)
- Hayato Muranaka
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rasaq Akinsola
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sandrine Billet
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew E. Hendifar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Neil A. Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Research, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Jun Gong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (R.A.); (S.B.); (S.J.P.); (A.E.H.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
33
|
Feng S, Aplin C, Nguyen TTT, Milano SK, Cerione RA. Filament formation drives catalysis by glutaminase enzymes important in cancer progression. Nat Commun 2024; 15:1971. [PMID: 38438397 PMCID: PMC10912226 DOI: 10.1038/s41467-024-46351-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/22/2024] [Indexed: 03/06/2024] Open
Abstract
The glutaminase enzymes GAC and GLS2 catalyze the hydrolysis of glutamine to glutamate, satisfying the 'glutamine addiction' of cancer cells. They are the targets of anti-cancer drugs; however, their mechanisms of activation and catalytic activity have been unclear. Here we demonstrate that the ability of GAC and GLS2 to form filaments is directly coupled to their catalytic activity and present their cryo-EM structures which provide a view of the conformational states essential for catalysis. Filament formation guides an 'activation loop' to assume a specific conformation that works together with a 'lid' to close over the active site and position glutamine for nucleophilic attack by an essential serine. Our findings highlight how ankyrin repeats on GLS2 regulate enzymatic activity, while allosteric activators stabilize, and clinically relevant inhibitors block, filament formation that enables glutaminases to catalyze glutaminolysis and support cancer progression.
Collapse
Affiliation(s)
- Shi Feng
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Cody Aplin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Thuy-Tien T Nguyen
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Shawn K Milano
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Richard A Cerione
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA.
- Department of Molecular Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
34
|
Zhang L, Li X, Gao H, Li P. The Role of Circular RNA Variants Generated from the NFIX Gene in Different Diseases. Mol Pharm 2024; 21:1027-1037. [PMID: 38315004 DOI: 10.1021/acs.molpharmaceut.3c00933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Circular RNAs (circRNAs) have been identified as important regulators in different developmental processes and disease pathogenesis. The loop structure of circRNAs makes them very stable in different conditions and microenvironments. circRNAs can affect microRNA (miRNA) and RNA binding protein (RBP) activity, encode functional proteins and regulate gene transcription. Recently, two circNFIX variants derived from the same gene, the Nuclear Factor I X (NFIX) gene, were determined as participants in the pathological processes of various diseases such as heart diseases and cancers. Both circNFIX variants are exonic circular RNAs and mainly function by sponging miRNAs. In this review, we summarize the current knowledge on circRNAs, elucidate the origins and properties of two circNFIX variants, explore the roles of two circNFIX variants in different diseases, and present clinical perspectives.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, No. 38 DengZhou Road, Qingdao 266021, China
| | - Xin Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, No. 38 DengZhou Road, Qingdao 266021, China
| | - Huijuan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, No. 38 DengZhou Road, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, No. 38 DengZhou Road, Qingdao 266021, China
| |
Collapse
|
35
|
Meng L, Yang J, Gao Y, Cao Q, Jiang S, Xiao Y, Wang H, Liu W, Yuan A, Li Y, Huang H. Biomimetic Nanomedicine Targeting Orchestrated Metabolism Coupled with Regulatory Factors to Disrupt the Metabolic Plasticity of Breast Cancer. ACS NANO 2024; 18:4360-4375. [PMID: 38277483 DOI: 10.1021/acsnano.3c10129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
Targeting nutrient metabolism has been proposed as an effective therapeutic strategy to combat breast cancer because of its high nutrient requirements. However, metabolic plasticity enables breast cancer cells to survive under unfavorable starvation conditions. The key mammalian target regulators rapamycin (mTOR) and hypoxia-inducible-factor-1 (HIF-1) tightly link the dynamic metabolism of glutamine and glucose to maintain nutrient flux. Blocking nutrient flow also induces autophagy to recycle nutrients in the autophagosome, which exacerbates metastasis and tumor progression. Compared to other common cancers, breast cancer is even more dependent on mTOR and HIF-1 to orchestrate the metabolic network. Therefore, we develop a cascade-boosting integrated nanomedicine to reprogram complementary metabolism coupled with regulators in breast cancer. Glucose oxidase efficiently consumes glucose, while the delivery of rapamycin inside limits the metabolic flux of glutamine and uncouples the feedback regulation of mTOR and HIF-1. The hydroxyl radical generated in a cascade blocks the later phase of autophagy without nutrient recycling. This nanomedicine targeting orchestrated metabolism can disrupt the coordination of glucose, amino acids, nucleotides, lipids, and other metabolic pathways in breast cancer tissues, effectively improving the durable antitumor effect and prognosis of breast cancer. Overall, the cascade-boosting integrated system provides a viable strategy to address cellular plasticity and efficient enzyme delivery.
Collapse
Affiliation(s)
- Lingtong Meng
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Jingpeng Yang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yang Gao
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Qinyan Cao
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Shunjie Jiang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yuyang Xiao
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Haoran Wang
- Medical School, Nanjing University, Nanjing, 210093, China
| | - Wenzheng Liu
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Ahu Yuan
- Medical School, Nanjing University, Nanjing, 210093, China
| | - Yanan Li
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - He Huang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
36
|
Teng M, Li Z, Gu Y, Fan Y, Wang D, Liu M, Li Y, Wei G, Huang Y. Real-time monitoring of glucose metabolism and effects of metformin on HepG2 cells using 13C in-cell NMR spectroscopy. Biochem Biophys Res Commun 2024; 694:149383. [PMID: 38150918 DOI: 10.1016/j.bbrc.2023.149383] [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: 07/03/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
Metformin is currently a strong candidate antitumor agent for multiple cancers, and has the potential to inhibit cancer cell viability, growth, and proliferation. Metabolic reprogramming is a critical feature of cancer cells. However, the effects of metformin which targets glucose metabolism on HepG2 cancer cells remain unclear. In this study, to explore the effects of metformin on glucose metabolism in HepG2 cells, we conducted real-time metabolomic monitoring of live HepG2 cells treated with metformin using 13C in-cell NMR spectroscopy. Metabolic tracing with U-13C6-glucose revealed that metformin significantly increased the production of 13C-G3P and 13C-glycerol, which were reported to attenuate liver cancer development, but decreased the production of potential oncogenesis-supportive metabolites, including 13C-lactate, 13C-alanine, 13C-glycine, and 13C-glutamate. Moreover, the expression levels of enzymes associated with the measured metabolites were carried out. The results showed that the levels of ALT1, MCT4, GPD2 and MPC1 were greatly reduced, which were consistent with the changes of measured metabolites in 13C in-cell NMR spectroscopy. Overall, our approach directly provides fundamental insights into the effects of metformin on glucose metabolism in live HepG2 cells, and highlights the potential mechanism of metformin, including the increase in production of G3P and glycerol derived from glucose, as well as the inhibition of glucose incorporation into lactate, alanine, glutamate, and glycine.
Collapse
Affiliation(s)
- Muzhou Teng
- Key Laboratory of the Digestive System Tumors of Gansu Province, The Second Clinical Medical College of Lanzhou University, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Zhijia Li
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Yanmei Gu
- Key Laboratory of the Digestive System Tumors of Gansu Province, The Second Clinical Medical College of Lanzhou University, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Yitao Fan
- Key Laboratory of the Digestive System Tumors of Gansu Province, The Second Clinical Medical College of Lanzhou University, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Daijun Wang
- Key Laboratory of the Digestive System Tumors of Gansu Province, The Second Clinical Medical College of Lanzhou University, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Meiyu Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Yumin Li
- Key Laboratory of the Digestive System Tumors of Gansu Province, The Second Clinical Medical College of Lanzhou University, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.
| | - Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Yanjie Huang
- Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China; Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials in Henan Province, Zhengzhou, Henan, 450046, China.
| |
Collapse
|
37
|
Chen J, Lu H, Cao D, Sun J, Qi F, Liu X, Liu J, Yang J, Yu M, Zhou H, Cheng N, Wang J, Zhang Y, Peng P, Wang T, Shen K, Sun W. Urine and serum metabolomic analysis of endometrial cancer diagnosis and classification based on ultra-performance liquid chromatography mass spectrometry. Metabolomics 2024; 20:18. [PMID: 38281200 DOI: 10.1007/s11306-023-02085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
OBJECTIVE This study aimed to reveal the urinary and serum metabolic pattern of endometrial cancer (EC) and establish diagnostic models to identify EC from controls, high-risk from low-risk EC, and type II from type I EC. METHOD This study included 146 EC patients (comprising 79 low-risk and 67 high-risk patients, including 124 type I and 22 type II) and 59 controls. The serum and urine samples were analyzed using ultraperformance liquid chromatography mass spectrometry. Analysis was used to elucidate the distinct metabolites and altered metabolic pathways. Receiver operating characteristic (ROC) analyses were employed to discover and validate the potential biomarker models. RESULTS Serum and urine metabolomes displayed significant differences between EC and controls, with metabolites related to amino acid and nicotinamide metabolisms. The serum and urine panels distinguished these two groups with Area Under the Curve (AUC) of 0.821 and 0.902, respectively. The panel consisting of serum and urine metabolites demonstrated the best predictive ability (AUC = 0.953 and 0.976 in discovering and validation group). In comparing high-risk and low risk EC, differential metabolites were enriched in purine and glutamine metabolism. The AUC values for serum and urine panels were 0.818, and 0.843, respectively. The combined panel exhibited better predictive accuracy (0.881 in discovering group and 0.936 in external validation). In the comparison between type I and type II group, altered folic acid metabolism was identified. The serum, urine and combined panels discriminated these two groups with the AUC of 0.829, 0.913 and 0.922, respectively. CONCLUSION The combined urine and serum metabolome effectively revealed the metabolic patterns in EC patients, offering valuable diagnostic models for EC diagnosis and classification.
Collapse
Affiliation(s)
- Junyu Chen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hezhen Lu
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Dongyan Cao
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Jiameng Sun
- Core Facility of Instrument, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Feng Qi
- Core Facility of Instrument, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyan Liu
- Core Facility of Instrument, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaqi Liu
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mei Yu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Huimei Zhou
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ninghai Cheng
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinhui Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ying Zhang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Peng Peng
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tao Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Keng Shen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Sun
- China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
38
|
Rong H, Wang D, Wang Y, Dong C, Wang G. YTHDF1 in Tumor Cell Metabolism: An Updated Review. Molecules 2023; 29:140. [PMID: 38202722 PMCID: PMC10779796 DOI: 10.3390/molecules29010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
With the advancement of research on m6A-related mechanisms in recent years, the YTHDF protein family within m6A readers has garnered significant attention. Among them, YTHDF1 serves as a pivotal member, playing a crucial role in protein translation, tumor proliferation, metabolic reprogramming of various tumor cells, and immune evasion. In addition, YTHDF1 also exerts regulatory effects on tumors through multiple signaling pathways, and numerous studies have confirmed its ability to assist in the reprogramming of the tumor cell-related metabolic processes. The focus of research on YTHDF1 has shifted in recent years from its m6A-recognition and -modification function to the molecular mechanisms by which it regulates tumor progression, particularly by exploring the regulatory factors that interact with YTHDF1 upstream and downstream. In this review, we elucidate the latest signaling pathway mechanisms of YTHDF1 in various tumor cells, with a special emphasis on its distinctive characteristics in tumor cell metabolic reprogramming. Furthermore, we summarize the latest pathological and physiological processes involving YTHDF1 in tumor cells, and analyze potential therapeutic approaches that utilize YTHDF1. We believe that YTHDF1 represents a highly promising target for future tumor treatments and a novel tumor biomarker.
Collapse
Affiliation(s)
| | | | | | | | - Guiling Wang
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, China; (H.R.); (D.W.); (Y.W.); (C.D.)
| |
Collapse
|
39
|
Che J, Zhao Y, Gu B, Li S, Li Y, Pan K, Sun T, Han X, Lv J, Zhang S, Fan B, Li C, Wang C, Wang J, Zhang T. Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with the progression of gastroesophageal cancer. BMC Cancer 2023; 23:1238. [PMID: 38102546 PMCID: PMC10724912 DOI: 10.1186/s12885-023-11744-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Previous metabolic studies in upper digestive cancer have mostly been limited to cross-sectional study designs, which hinders the ability to effectively predict outcomes in the early stage of cancer. This study aims to identify key metabolites and metabolic pathways associated with the multistage progression of epithelial cancer and to explore their predictive value for gastroesophageal cancer (GEC) formation and for the early screening of esophageal squamous cell carcinoma (ESCC). METHODS A case-cohort study within the 7-year prospective Esophageal Cancer Screening Cohort of Shandong Province included 77 GEC cases and 77 sub-cohort individuals. Untargeted metabolic analysis was performed in serum samples. Metabolites, with FDR q value < 0.05 and variable importance in projection (VIP) > 1, were selected as differential metabolites to predict GEC formation using Random Forest (RF) models. Subsequently, we evaluated the predictive performance of these differential metabolites for the early screening of ESCC. RESULTS We found a distinct metabolic profile alteration in GEC cases compared to the sub-cohort, and identified eight differential metabolites. Pathway analyses showed dysregulation in D-glutamine and D-glutamate metabolism, nitrogen metabolism, primary bile acid biosynthesis, and steroid hormone biosynthesis in GEC patients. A panel of eight differential metabolites showed good predictive performance for GEC formation, with an area under the receiver operating characteristic curve (AUC) of 0.893 (95% CI = 0.816-0.951). Furthermore, four of the GEC pathological progression-related metabolites were validated in the early screening of ESCC, with an AUC of 0.761 (95% CI = 0.716-0.805). CONCLUSIONS These findings indicated a panel of metabolites might be an alternative approach to predict GEC formation, and therefore have the potential to mitigate the risk of cancer progression at the early stage of GEC.
Collapse
Affiliation(s)
- Jiajing Che
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yongbin Zhao
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Bingbing Gu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuting Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yunfei Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Keyu Pan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Tiantian Sun
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xinyue Han
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jiali Lv
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuai Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Bingbing Fan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chunxia Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Cheng Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Jialin Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, China.
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
40
|
Bartman CR, Faubert B, Rabinowitz JD, DeBerardinis RJ. Metabolic pathway analysis using stable isotopes in patients with cancer. Nat Rev Cancer 2023; 23:863-878. [PMID: 37907620 PMCID: PMC11161207 DOI: 10.1038/s41568-023-00632-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 11/02/2023]
Abstract
Metabolic reprogramming is central to malignant transformation and cancer cell growth. How tumours use nutrients and the relative rates of reprogrammed pathways are areas of intense investigation. Tumour metabolism is determined by a complex and incompletely defined combination of factors intrinsic and extrinsic to cancer cells. This complexity increases the value of assessing cancer metabolism in disease-relevant microenvironments, including in patients with cancer. Stable-isotope tracing is an informative, versatile method for probing tumour metabolism in vivo. It has been used extensively in preclinical models of cancer and, with increasing frequency, in patients with cancer. In this Review, we describe approaches for using in vivo isotope tracing to define fuel preferences and pathway engagement in tumours, along with some of the principles that have emerged from this work. Stable-isotope infusions reported so far have revealed that in humans, tumours use a diverse set of nutrients to supply central metabolic pathways, including the tricarboxylic acid cycle and amino acid synthesis. Emerging data suggest that some activities detected by stable-isotope tracing correlate with poor clinical outcomes and may drive cancer progression. We also discuss current challenges in isotope tracing, including comparisons of in vivo and in vitro models, and opportunities for future discovery in tumour metabolism.
Collapse
Affiliation(s)
- Caroline R Bartman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Brandon Faubert
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| | - Ralph J DeBerardinis
- Howard Hughes Medical Institute and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
41
|
Gopal K, Abdualkader AM, Li X, Greenwell AA, Karwi QG, Altamimi TR, Saed C, Uddin GM, Darwesh AM, Jamieson KL, Kim R, Eaton F, Seubert JM, Lopaschuk GD, Ussher JR, Al Batran R. Loss of muscle PDH induces lactic acidosis and adaptive anaplerotic compensation via pyruvate-alanine cycling and glutaminolysis. J Biol Chem 2023; 299:105375. [PMID: 37865313 PMCID: PMC10692893 DOI: 10.1016/j.jbc.2023.105375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023] Open
Abstract
Pyruvate dehydrogenase (PDH) is the rate-limiting enzyme for glucose oxidation that links glycolysis-derived pyruvate with the tricarboxylic acid (TCA) cycle. Although skeletal muscle is a significant site for glucose oxidation and is closely linked with metabolic flexibility, the importance of muscle PDH during rest and exercise has yet to be fully elucidated. Here, we demonstrate that mice with muscle-specific deletion of PDH exhibit rapid weight loss and suffer from severe lactic acidosis, ultimately leading to early mortality under low-fat diet provision. Furthermore, loss of muscle PDH induces adaptive anaplerotic compensation by increasing pyruvate-alanine cycling and glutaminolysis. Interestingly, high-fat diet supplementation effectively abolishes early mortality and rescues the overt metabolic phenotype induced by muscle PDH deficiency. Despite increased reliance on fatty acid oxidation during high-fat diet provision, loss of muscle PDH worsens exercise performance and induces lactic acidosis. These observations illustrate the importance of muscle PDH in maintaining metabolic flexibility and preventing the development of metabolic disorders.
Collapse
Affiliation(s)
- Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Abdualrahman Mohammed Abdualkader
- Faculty of Pharmacy, Université de Montréal, Montréal, Quebec, Canada; Montreal Diabetes Research Center, Montréal, Quebec, Canada; Cardiometabolic Health, Diabetes and Obesity Research Network, Montréal, Quebec, Canada
| | - Xiaobei Li
- Faculty of Pharmacy, Université de Montréal, Montréal, Quebec, Canada; Montreal Diabetes Research Center, Montréal, Quebec, Canada; Cardiometabolic Health, Diabetes and Obesity Research Network, Montréal, Quebec, Canada
| | - Amanda A Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qutuba G Karwi
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada; Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Newfoundland and Labrador, Canada
| | - Tariq R Altamimi
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Christina Saed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Golam M Uddin
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - K Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ryekjang Kim
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Farah Eaton
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Rami Al Batran
- Faculty of Pharmacy, Université de Montréal, Montréal, Quebec, Canada; Montreal Diabetes Research Center, Montréal, Quebec, Canada; Cardiometabolic Health, Diabetes and Obesity Research Network, Montréal, Quebec, Canada.
| |
Collapse
|
42
|
Hu SS, Han Y, Tan TY, Chen H, Gao JW, Wang L, Yang MH, Zhao L, Wang YQ, Ding YQ, Wang S. SLC25A21 downregulation promotes KRAS-mutant colorectal cancer progression by increasing glutamine anaplerosis. JCI Insight 2023; 8:e167874. [PMID: 37937641 PMCID: PMC10721270 DOI: 10.1172/jci.insight.167874] [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: 12/09/2022] [Accepted: 09/20/2023] [Indexed: 11/09/2023] Open
Abstract
Emerging evidence shows that KRAS-mutant colorectal cancer (CRC) depends on glutamine (Gln) for survival and progression, indicating that targeting Gln metabolism may be a promising therapeutic strategy for KRAS-mutant CRC. However, the precise mechanism by which Gln metabolism reprogramming promotes and coordinates KRAS-mutant CRC progression remains to be fully investigated. Here, we discovered that solute carrier 25 member 21 (SLC25A21) expression was downregulated in KRAS-mutant CRC, and that SLC25A21 downregulation was correlated with poor survival of KRAS-mutant CRC patients. SLC25A21 depletion selectively accelerated the growth, invasion, migration, and metastasis of KRAS-mutant CRC cells in vitro and in vivo, and inhibited Gln-derived α-ketoglutarate (α-KG) efflux from mitochondria, thereby potentiating Gln replenishment, accompanied by increased GTP availability for persistent KRAS activation in KRAS-mutant CRC. The restoration of SLC25A21 expression impaired the KRAS-mutation-mediated resistance to cetuximab in KRAS-mutant CRC. Moreover, the arrested α-KG efflux that occurred in response to SLC25A21 depletion inhibited the activity of α-KG-dependent DNA demethylases, resulting in a further decrease in SLC25A21 expression. Our studies demonstrate that SLC25A21 plays a significant role as a tumor suppressor in KRAS-mutant CRC by antagonizing Gln-dependent anaplerosis to limit GTP availability for KRAS activation, which suggests potential alternative therapeutic strategies for KRAS-mutant CRC.
Collapse
Affiliation(s)
- Sha-Sha Hu
- Department of Pathology, Nanfang Hospital, and
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yue Han
- Department of Pathology, Nanfang Hospital, and
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tian-Yuan Tan
- Department of Pathology, Nanfang Hospital, and
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Chen
- Department of Pathology, Nanfang Hospital, and
| | - Jia-Wen Gao
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lan Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Min-Hui Yang
- Department of Pathology, Nanfang Hospital, and
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Li Zhao
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yi-Qing Wang
- Department of Pathology, Nanfang Hospital, and
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yan-Qing Ding
- Department of Pathology, Nanfang Hospital, and
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuang Wang
- Department of Pathology, Nanfang Hospital, and
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
Chen C, Naveed H, Chen K. Research progress on branched-chain amino acid aminotransferases. Front Genet 2023; 14:1233669. [PMID: 38028625 PMCID: PMC10658711 DOI: 10.3389/fgene.2023.1233669] [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: 06/09/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Branched-chain amino acid aminotransferases, widely present in natural organisms, catalyze bidirectional amino transfer between branched-chain amino acids and branched-chain α-ketoacids in cells. Branched-chain amino acid aminotransferases play an important role in the metabolism of branched-chain amino acids. In this paper, the interspecific evolution and biological characteristics of branched-chain amino acid aminotransferases are introduced, the related research of branched-chain amino acid aminotransferases in animals, plants, microorganisms and humans is summarized and the molecular mechanism of branched-chain amino acid aminotransferase is analyzed. It has been found that branched-chain amino acid metabolism disorders are closely related to various diseases in humans and animals and plants, such as diabetes, cardiovascular diseases, brain diseases, neurological diseases and cancer. In particular, branched-chain amino acid aminotransferases play an important role in the development of various tumors. Branched-chain amino acid aminotransferases have been used as potential targets for various cancers. This article reviews the research on branched-chain amino acid aminotransferases, aiming to provide a reference for clinical research on targeted therapy for various diseases and different cancers.
Collapse
Affiliation(s)
- Can Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Hassan Naveed
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Keping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
44
|
Murnan KM, Horbinski C, Stegh AH. Redox Homeostasis and Beyond: The Role of Wild-Type Isocitrate Dehydrogenases for the Pathogenesis of Glioblastoma. Antioxid Redox Signal 2023; 39:923-941. [PMID: 37132598 PMCID: PMC10654994 DOI: 10.1089/ars.2023.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 05/04/2023]
Abstract
Significance: Glioblastoma is an aggressive and devastating brain tumor characterized by a dismal prognosis and resistance to therapeutic intervention. To support catabolic processes critical for unabated cellular growth and defend against harmful reactive oxygen species, glioblastoma tumors upregulate the expression of wild-type isocitrate dehydrogenases (IDHs). IDH enzymes catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG), NAD(P)H, and CO2. On molecular levels, IDHs epigenetically control gene expression through effects on α-KG-dependent dioxygenases, maintain redox balance, and promote anaplerosis by providing cells with NADPH and precursor substrates for macromolecular synthesis. Recent Advances: While gain-of-function mutations in IDH1 and IDH2 represent one of the most comprehensively studied mechanisms of IDH pathogenic effects, recent studies identified wild-type IDHs as critical regulators of normal organ physiology and, when transcriptionally induced or down regulated, as contributing to glioblastoma progression. Critical Issues: Here, we will discuss molecular mechanisms of how wild-type IDHs control glioma pathogenesis, including the regulation of oxidative stress and de novo lipid biosynthesis, and provide an overview of current and future research directives that aim to fully characterize wild-type IDH-driven metabolic reprogramming and its contribution to the pathogenesis of glioblastoma. Future Directions: Future studies are required to further dissect mechanisms of metabolic and epigenomic reprogramming in tumors and the tumor microenvironment, and to develop pharmacological approaches to inhibit wild-type IDH function. Antioxid. Redox Signal. 39, 923-941.
Collapse
Affiliation(s)
- Kevin M. Murnan
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Malnati Brain Tumor Institute, Northwestern University, Chicago, Illinois, USA
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Alexander H. Stegh
- Department of Neurological Surgery, The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
45
|
Liu Q, Bode AM, Chen X, Luo X. Metabolic reprogramming in nasopharyngeal carcinoma: Mechanisms and therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2023; 1878:189023. [PMID: 37979733 DOI: 10.1016/j.bbcan.2023.189023] [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: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
The high prevalence of metabolic reprogramming in nasopharyngeal carcinoma (NPC) offers an abundance of potential therapeutic targets. This review delves into the distinct mechanisms underlying metabolic reprogramming in NPC, including enhanced glycolysis, nucleotide synthesis, and lipid metabolism. All of these changes are modulated by Epstein-Barr virus (EBV) infection, hypoxia, and tumor microenvironment. We highlight the role of metabolic reprogramming in the development of NPC resistance to standard therapies, which represents a challenging barrier in treating this malignancy. Furthermore, we dissect the state of the art in therapeutic strategies that target these metabolic changes, evaluating the successes and failures of clinical trials and the strategies to tackle resistance mechanisms. By providing a comprehensive overview of the current knowledge and future directions in this field, this review sets the stage for new therapeutic avenues in NPC.
Collapse
Affiliation(s)
- Qian Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xue Chen
- Early Clinical Trial Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China.
| | - Xiangjian Luo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
46
|
Bao J, Yu Y. Identification of a prognostic evaluator from glutamine metabolic heterogeneity studies within and between tissues in hepatocellular carcinoma. Front Pharmacol 2023; 14:1241677. [PMID: 37954858 PMCID: PMC10637396 DOI: 10.3389/fphar.2023.1241677] [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: 06/17/2023] [Accepted: 09/21/2023] [Indexed: 11/14/2023] Open
Abstract
Background: The liver is the major metabolic organ of the human body, and abnormal metabolism is the main factor influencing hepatocellular carcinoma (HCC). This study was designed to determine the effect of glutamine metabolism on HCC heterogeneity and to develop a prognostic evaluator based on the heterogeneity study of glutamine metabolism within HCC tumors and between tissues. Methods: Single-cell transcriptome data were extracted from the GSE149614 dataset and processed using the Seurat package in R for quality control of these data. HCC subtypes in the Cancer Genome Atlas and the GSE14520 dataset were identified via consensus clustering based on glutamine family amino acid metabolism (GFAAM) process genes. The machine learning algorithms gradient boosting machine, support vector machine, random forest, eXtreme gradient boosting, decision trees, and least absolute shrinkage and selection operator were utilized to develop the prognosis model of differentially expressed genes among the molecular gene subtypes. Results: The samples in the GSE149614 dataset included 10 cell types, and there was no significant difference in the GFAAM pathway. HCC was classified into three molecular subtypes according to GFAAM process genes, showing molecular heterogeneity in prognosis, clinicopathological features, and immune cell infiltration. C1 showed the worst survival rate and the highest immune score and immune cell infiltration. A six-gene model for prognostic and immunotherapy responses was constructed among subtypes, and the calculated high-risk score was significantly correlated with poor prognosis, high immune abundance, and a low response rate of immunotherapy in HCC. Conclusion: Our discovery of GFAAM-associated marker genes may help to further decipher the role in HCC occurrence and progression. In particular, this six-gene prognostic model may serve as a predictor of treatment and prognosis in HCC patients.
Collapse
Affiliation(s)
- Jie Bao
- Digestive System Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Sun X, Chen H, Gao R, Huang Y, Qu Y, Yang H, Wei X, Hu S, Zhang J, Wang P, Zou Y, Hu K, Ge J, Sun A. Mitochondrial transplantation ameliorates doxorubicin-induced cardiac dysfunction via activating glutamine metabolism. iScience 2023; 26:107790. [PMID: 37731615 PMCID: PMC10507231 DOI: 10.1016/j.isci.2023.107790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 08/14/2023] [Accepted: 08/28/2023] [Indexed: 09/22/2023] Open
Abstract
Doxorubicin is a wildly used effective anticancer agent. However, doxorubicin use is also related to cardiotoxic side effect in some patients. Mitochondrial damage has been shown to be one of the pathogeneses of doxorubicin-induced myocardial injury. In this study, we demonstrated that mitochondrial transplantation could inhibit doxorubicin-induced cardiotoxicity by directly supplying functional mitochondria. Mitochondrial transplantation improved contractile function and respiratory capacity, reduced cellular apoptosis and oxidative stress in cardiomyocytes. Mitochondria isolated from various sources, including mouse hearts, mouse and human arterial blood, and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), all exerted similar cardioprotective effects. Mechanically, mitochondrial transplantation activates glutamine metabolism in doxorubicin-treated mice heart and blocking glutamine metabolism attenuated the cardioprotective effects of mitochondrial transplantation. Overall, our study demonstrates that mitochondria isolated from arterial blood could be used for mitochondrial transplantation, which might serve as a feasible promising therapeutic option for patients with doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
| | - Hang Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, P.R. China
| | - Rifeng Gao
- Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Ya Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
| | - Yanan Qu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
| | - Heng Yang
- The Second Affiliated Hospital of Nanchang University, Nanchang 330000, P.R. China
| | - Xiang Wei
- Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Shiyu Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
| | - Jian Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
| | - Peng Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
| | - Yunzeng Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
- Institute of Biomedical Science, Fudan University, Shanghai 200032, P.R. China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
- Institute of Biomedical Science, Fudan University, Shanghai 200032, P.R. China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai 200032, P.R. China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, P.R. China
- Institute of Biomedical Science, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
48
|
Chen X, Liu X, Du S. Unveiling the Role of Tumor-Infiltrating T Cells and Immunotherapy in Hepatocellular Carcinoma: A Comprehensive Review. Cancers (Basel) 2023; 15:5046. [PMID: 37894413 PMCID: PMC10605632 DOI: 10.3390/cancers15205046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a rapidly rising global health concern, ranking as the third-leading cause of cancer-related mortality. Despite medical advancements, the five-year survival rate remains a dismal 18%, with a daunting 70% recurrence rate within a five-year period. Current systematic treatments, including first-line sorafenib, yield an overall response rate (ORR) below 10%. In contrast, immunotherapies have shown promise by improving ORR to approximately 30%. The IMbravel150 clinical trial demonstrates that combining atezolizumab and bevacizumab surpasses sorafenib in terms of median progression-free survival (PFS) and overall survival (OS). However, the therapeutic efficacy for HCC patients remains unsatisfactory, highlighting the urgent need for a comprehensive understanding of antitumor responses and immune evasion mechanisms in HCC. In this context, understanding the immune landscape of HCC is of paramount importance. Tumor-infiltrating T cells, including cytotoxic T cells, regulatory T cells, and natural killer T cells, are key components in the antitumor immune response. This review aims to shed light on their intricate interactions within the immunosuppressive tumor microenvironment and explores potential strategies for revitalizing dysfunctional T cells. Additionally, current immune checkpoint inhibitor (ICI)-based trials, ICI-based combination therapies, and CAR-T- or TCR-T-cell therapies for HCC are summarized, which might further improve OS and transform the management of HCC in the future.
Collapse
Affiliation(s)
- Xiaokun Chen
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China; (X.C.); (X.L.)
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiao Liu
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China; (X.C.); (X.L.)
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China; (X.C.); (X.L.)
| |
Collapse
|
49
|
Wang B, Pei J, Xu S, Liu J, Yu J. System analysis based on glutamine catabolic-related enzymes identifies GPT2 as a novel immunotherapy target for lung adenocarcinoma. Comput Biol Med 2023; 165:107415. [PMID: 37657356 DOI: 10.1016/j.compbiomed.2023.107415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/31/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND In recent years, targeting glutamine metabolism has gained attention as a promising therapeutic approach. Glutamine catabolic-related enzymes play a crucial role in modulating glutamine metabolism and influencing immune responses in the tumor immune microenvironment (TME). However, current literature on the function of glutamine catabolic enzymes in lung adenocarcinoma (LUAD) is limited. METHODS We validated the glutamine dependency of LUAD cells in vitro, followed by transcriptome data to identify differentially expressed genes (DEGs), with transcriptome and single-cell data analysis utilized to explore the role of such genes within the tumor immune microenvironment. We performed employed subcutaneous injection of lewis lung carcinoma cells in C57BL/6 mice to confirm the role of candidate genes in tumor growth and anti-tumor immunity. RESULTS Our study revealed that glutamine is essential for the growth of LUAD cells. Subsequently, we identified four DEGs - glutamate pyruvate transaminase 1 (GPT1), glutamate pyruvate transaminase 2 (GPT2), glutamic-oxaloacetic transaminase 1 (GOT1), and glutamic-oxaloacetic transaminase 2 (GOT2) - in LUAD patients, which were highly expressed in tumor tissue and associated with an immunosuppressive TME. Single-cell sequencing analysis detected high expression levels of GOT1 and GOT2 in immune and stromal cell subpopulations, while GPT1 and GPT2 showed relatively lower expression. Based on the lower immune score and lower expression in immune and stromal cells, we validated the role of GPT2 in vivo for modulating the TME and tumor growth. Inhibition of GPT2 resulted in suppressed tumor growth and increased the expression of CD4 and CD8. Additionally, GPT2 inhibitors induced a stronger antitumor immunity when used in combination with anti-programmed cell death ligand 1. CONCLUSION This study is the first to show the critical role of glutamine catabolic-related enzymes in the TME, and identified GPT2 as a promising therapeutic target for inhibiting tumor growth and improving anti-tumour immune responses for LUAD. Additional studies will be required to define the roles glutamine catabolic-related enzymes play in LUAD.
Collapse
Affiliation(s)
- Bolin Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinli Pei
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Shengnan Xu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
50
|
Cheng L, Zhai H, Du J, Zhang G, Shi G. Lobetyolin inhibits cell proliferation and induces cell apoptosis by downregulating ASCT2 in gastric cancer. Cytotechnology 2023; 75:435-448. [PMID: 37655270 PMCID: PMC10465467 DOI: 10.1007/s10616-023-00588-w] [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: 03/27/2023] [Accepted: 06/29/2023] [Indexed: 09/02/2023] Open
Abstract
Gastric cancer (GC) is a heterogeneous disease and is the fifth most common cancer worldwide. Lobetyolin, as a bioactive ingredient extracted from Codonopsis pilosula (Franch.) Nannf., has been reported to exert anti-tumor effects in several cancer types. This study was aimed to investigate the role of lobetyolin in GC and the associated mechanism. MKN-45 and MKN-28 cells were incubated with concentrations of lobetyolin for 24 h. The viability and survival of GC cells were evaluated by performing MTT assay. Glutamine uptake, Adenosine Triphosphate, reactive oxygen species (ROS), and glutathione levels were measured by corresponding kits. Apoptosis and mitochondrial membrane potential of GC cells were determined by flow cytometry. Alanine, serine, cysteine-preferring transporter 2 (ASCT2) and the AKT/GSK3β/c-Myc pathway protein levels were examined by western blotting. Xenograft model and immunohistochemical staining were used to evaluate the pharmacological effects of lobetyolin in mice in vivo. We found that lobetyolin treatment suppressed the proliferative capacity of both MKN-45 and MKN-28 cells in a concentration-dependent manner. Lobetyolin reduced the uptake of glutamine and downregulated the expression levels of ASCT2 in GC cells and xenograft tumors. Lobetyolin effectively restrained the growth of tumors in vivo. In addition, lobetyolin induced the accumulation of ROS to attenuate mitochondria-mediated apoptosis via downregulation of ASCT2 expression. Lobetyolin promoted the phosphorylation of c-Myc and suppressed the phosphorylation of GSK3β and AKT in both MKN-45 and MKN-28 cells. The level of total Nrf2 protein was reduced after lobetyolin treatment. Overall, lobetyolin exerts anti-cancer effects by repressing cell proliferation and inducing cell apoptosis via downregulation of ASCT2 in GC.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Gastroenterology, The Central Hospital of Qianjiang, Yangtze University, Qianjiang, 433100 China
| | - Haoqing Zhai
- Department of Oncology, The Central Hospital of Qianjiang, Yangtze University, Qianjiang, 433100 China
| | - Juan Du
- Department of Internal Medicine, Hubei University Hospital, Wuhan, 430062 China
| | - Gang Zhang
- Department of Digestive 2, Wuhan Sixth Hospital, Wuhan, 430015 China
| | - Gan Shi
- Department of Gastroenterology, Wuhan Xinzhou District People’s Hospital, No.61, Xinzhou Street, Zhucheng Street, Xinzhou District, Wuhan, 430400 China
| |
Collapse
|