1
|
Ye J, Hu P, Zhang R, Zhou L, Luo Z, Chen Y, Ruan S, Zhu M, Ding H, Qian Y, Xing Y, Meng T, Wang C, Song D. Targeting Hyperglycemic Bone Pre-Metastatic Niche for Breast Cancer Bone Metastasis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e04924. [PMID: 40491413 DOI: 10.1002/advs.202504924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/26/2025] [Indexed: 06/11/2025]
Abstract
Bone is the most common site of breast cancer metastasis, yet understanding the intricate mechanisms and potential therapeutic targets remains nascent. Here it is reported that breast cancer establishes a hyperglycemic bone pre-metastatic niche before migrating to bone tissue and further enhances glucose metabolism following metastatic colonization. An intervention strategy is subsequently proposed targeting glucose metabolism utilizing a biomimetic-engineered enzyme-based nanoplatform. This platform's membrane shielding reduces the interaction between engineered glucose oxidase and circulating glucose, while the engineered enzyme specifically targets glucose metabolism, enabling self-amplifying starvation combined with selective chemotherapy. Such precision can precisely inhibit breast cancer bone metastases and block distal tumor dissemination. This study provides novel insights into the role of glucose metabolism in the pre-metastatic niche and presents a proof-of-concept for metabolic-targeted strategies in breast cancer bone metastasis treatment. This approach holds significant promise for improving therapeutic outcomes in metastatic breast cancer by targeting the metabolic vulnerabilities of the bone microenvironment and halting systemic tumor spread.
Collapse
Affiliation(s)
- Jianxin Ye
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Peng Hu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200080, China
| | - Rui Zhang
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Lei Zhou
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zonghua Luo
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, 200080, China
| | - Yanan Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200080, China
| | - Shengzhe Ruan
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Mengyi Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200080, China
| | - Huaze Ding
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yike Qian
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Tong Meng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Changping Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
2
|
Huang X, Hou S, Li Y, Xu G, Xia N, Duan Z, Luo K, Tian B. Targeting lipid metabolism via nanomedicine: A prospective strategy for cancer therapy. Biomaterials 2025; 317:123022. [PMID: 39754967 DOI: 10.1016/j.biomaterials.2024.123022] [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: 11/20/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025]
Abstract
Lipid metabolism has been increasingly recognized to play an influencing role in tumor initiation, progression, metastasis, and therapeutic drug resistance. Targeting lipid metabolic reprogramming represents a promising therapeutic strategy. Despite their structural complexity and poor targeting efficacy, lipid-metabolizing drugs, either used alone or in combination with chemotherapeutic agents, have been employed in clinical practice. The advent of nanotechnology offers new approaches to enhancing therapeutic effects, includingthe targeted delivery and integration of lipid metabolic reprogramming with chemotherapy, photodynamic therapy (PDT), and immunotherapy. The integrated nanoformulation, nanomedicine, could significantly advance the field of lipid metabolism therapy. In this review, we will briefly introduce the concept of cancer lipid metabolism reprogramming, then elaborate the latest advances in engineered nanomedicine for targeting lipid metabolism during cancer treatment, and finally provide our insights into future perspectives of nanomedicine for interference with lipid metabolism in the tumor microenvironment.
Collapse
Affiliation(s)
- Xing Huang
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shengzhong Hou
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinggang Li
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Xu
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Ning Xia
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| | - Kui Luo
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| | - Bole Tian
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Zhang S, Li Z, Li B, Li Z, Peng H, Shen L, Zhu L, Mo T, Peng J, Zhuo L, Wang Z, Jiang W. Novel Carbamate-Based o-aminobenzamide Derivatives as Potent Antigastric Carcinoma Agents via Disrupting NAD + Salvage Synthesis. J Med Chem 2025. [PMID: 40448666 DOI: 10.1021/acs.jmedchem.4c02686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Blocking NAD+ biosynthesis presents an appealing strategy for antitumor therapies. This study developed a series of o-aminobenzamide derivatives with substantial antitumor efficacy against gastric cancer. Notably, compound 9a demonstrated exceptional antitumor activity against undifferentiated gastric cancer HGC27 cells (IC50 = 0.049 μM), and significant inhibitory effects on cellular proliferation, self-renewal, invasion, and migration. Mechanistic investigations revealed that 9a could damage mitochondria, arrest the cell cycle, promote apoptosis, and alter cellular metabolism. Furthermore, the rate-limiting enzyme NAMPT in the NAD+ salvage synthetic pathway was identified as a primary target of 9a. By inhibiting NAMPT, 9a reduced intracellular levels of NAD+ and ATP, while NMN, a natural product of NAMPT, counteracts its antimetabolic and cytotoxic effects. Overall, this study highlights 9a as a promising NAMPT inhibitor with significant activity against undifferentiated gastric cancer, laying the groundwork for developing novel antigastric cancer agents through inhibiting NAD+ biosynthesis.
Collapse
Affiliation(s)
- Siyi Zhang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Zhen Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Bo Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Zhiyi Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Huiqian Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lixian Shen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- School of Basic Medicine, The First Affiliated Hospital, The Second Affiliated Hospital, MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lejing Zhu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Tong Mo
- School of Basic Medicine, The First Affiliated Hospital, The Second Affiliated Hospital, MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Jialiang Peng
- School of Basic Medicine, The First Affiliated Hospital, The Second Affiliated Hospital, MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Linsheng Zhuo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- School of Basic Medicine, The First Affiliated Hospital, The Second Affiliated Hospital, MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, Qinghai, China
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410008, Hunan, China
| | - Weifan Jiang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
4
|
Yuan SSF, Vadhan A, Nguyen HDH, Chen PY, Tseng CH, Wu CH, Chen YC, Wu YC, Hu SCS, Lo S, Hou MF, Wang YY. Oncogenic role of fumarate hydratase in breast cancer: metabolic reprogramming and mechanistic insights. Cancer Metab 2025; 13:26. [PMID: 40437625 PMCID: PMC12121060 DOI: 10.1186/s40170-025-00397-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 05/24/2025] [Indexed: 06/01/2025] Open
Abstract
Breast cancer remains the most prevalent malignancy among women globally, with its complexity linked to genetic variations and metabolic alterations within tumor cells. This study investigates the role of fumarate hydratase (FH), a key enzyme in the tricarboxylic acid (TCA) cycle, in breast cancer progression. Our findings reveal that FH mRNA and protein levels are significantly upregulated in breast cancer tissues and correlate with poor patient prognosis and aggressive tumor characteristics. Using in vitro and in vivo models, we demonstrate that FH overexpression enhances breast cancer cell proliferation, migration, and invasion through metabolic reprogramming and by increasing reactive oxygen species (ROS) production. Furthermore, we identify matrix metalloproteinase 1 (MMP1) as a downstream effector of FH, linked to p21 downregulation, elucidating a novel regulatory pathway influencing tumor behavior. Interestingly, unlike its tumor-suppressing role in other cancer types, this study highlights FH's oncogenic potential in breast cancer. Our results suggest that FH enhances cancer cell viability and aggressiveness via both catalytic and non-catalytic mechanisms. This work not only underscores the metabolic adaptations of breast cancer cells but also proposes FH as a potential biomarker and therapeutic target for breast cancer management.
Collapse
Affiliation(s)
- Shyng-Shiou F Yuan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, Center for Intelligent Drug Systems and Smart Biodevices (IDS2B), National Yang Ming Chiao Tung University, 75 Bo-Ai Street, Hsinchu, 300, Taiwan.
| | - Anupama Vadhan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, No. 8, Xuefu W. RdYunlin County 632, Huwei Township, Taiwan
| | - Hieu D H Nguyen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Pang-Yu Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Huang Tseng
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Division of Oral Pathology & Maxillofacial Radiology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Ching-Hu Wu
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Yu-Chieh Chen
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Yi-Chia Wu
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Stephen Chu-Sung Hu
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Steven Lo
- Canniesburn Regional Plastic Surgery and Burns Unit, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G4 0SFG12 8QQ, UK
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Yen-Yun Wang
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
5
|
Geng X, Li M, Zhang L, Cai Y, Chen X, Mu X, Wang J, Liu B. P5CS deacetylation mediated by SIRT2 facilitates tumor growth by enhancing mitochondrial respiration in hepatocellular carcinoma. Oncogene 2025:10.1038/s41388-025-03456-3. [PMID: 40425834 DOI: 10.1038/s41388-025-03456-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 05/05/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Cancer cells typically exhibit enhanced mitochondrial metabolism to fulfill their energy and biosynthetic demands for growth. The mitochondrial response to fluctuations in cellular energy demand is essential for cellular adaptation and proper organ function. The mitochondrial delta-1-pyrroline-5-carboxylate synthase (P5CS) encoded by the ALDH18A1 gene, the key enzyme for proline synthesis, is frequently up-regulated during tumor development. However, the regulatory mechanisms governing P5CS activity in the occurrence and development of hepatocellular carcinoma (HCC) remain largely unknown. In this study, we observe that P5CS is highly expressed in HCC tissues, and elevated levels of P5CS expression are associated with poor prognosis in HCC patients. Notably, the knockdown of P5CS inhibits the proliferation, migratory and invasive capabilities of HCC cells by reducing mitochondrial respiration. Furthermore, we demonstrate that SIRT2 interacts with P5CS and mediates the deacetylation of P5CS at lysines K311 and K347, thereby activating its enzymatic activity. Activated P5CS significantly enhances mitochondrial respiration, which supports the proliferation and tumorigenesis of HCC cells. In addition, SIRT2 knockdown inhibits the proliferation, migratory and invasive capabilities of HCC cells. These observations suggest that SIRT2-mediated P5CS deacetylation is a crucial signaling event through which cancer cells sustain mitochondrial respiration and promote HCC progression. This finding offers the potential for targeting SIRT2-mediated P5CS deacetylation as a therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Xiaofang Geng
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
| | - Mengyao Li
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Lu Zhang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Yihan Cai
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Xin Chen
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Xiayue Mu
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Jie Wang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
| | - Bowen Liu
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
| |
Collapse
|
6
|
Gong Y, Chen L, Wang H, Zheng D, Li F, Wu C, Li Y, Deng Y, He Z, Yu C. ANXA1 promotes intrahepatic cholangiocarcinoma proliferation and growth by regulating glutamine metabolism through GOT1 stabilization. J Exp Clin Cancer Res 2025; 44:151. [PMID: 40390008 PMCID: PMC12087091 DOI: 10.1186/s13046-025-03400-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is a malignant tumor with a poor prognosis, marked by a postoperative recurrence rate of 50-60% and a 5-year survival rate of 8-30%. Abnormal tumor metabolism, particularly, amino acid metabolism, plays a key role in malignant progression. However, the molecular mechanisms linking amino acid metabolism to ICC progression remain unclear. METHODS Bioinformatics was used to identity the key amino acid metabolism related gene in ICC, qRT-PCR, western blotting and immunohistochemical (IHC) were used to detect the expression of ANXA1 in normal tissues or ICC tissues and cells at mRNA and protein levels. The effects of ANXA1 on the proliferation ability of ICC in vitro and in vivo were investigated using CCK8, cloning formation experiment, EdU, IHC, nude mice subcutaneous tumorigenesis model. Immunoprecipitation, mass spectrometry analysis, protein ubiquitin level detection test, immunofluorescence co-localization, and redox stress metabolite detection test were used to explore the metabolism-related regulatory mechanism of ANXA1. RESULTS we employed bioinformatics analysis to classify ICC into metabolic subgroups with distinct prognoses and identified the associated biomarker Annexin A1(ANXA1), whose high expression is correlated with poor prognosis and promotes ICC development. Mass spectrometry analysis revealed that ANXA1 interacts with the key enzyme in glutamine metabolism, glutamic-oxaloacetic transaminase 1(GOT1). Through in vitro and in vivo experiments, overexpressed ANXA1 stabilizes GOT1 by recruiting the deubiquitinase USP5. This stabilization enhances glutamine uptake, as well as the production of aspartate and glutamate, which in turn reduces oxidative stress, thereby promoting tumor cell growth. Moreover, knockdown of ANXA1 combined with glutamine uptake inhibition significantly suppressed ICC cell proliferation and Inhibited subcutaneous tumor formation and growth. CONCLUSIONS These results suggest that the ANXA1/USP5/GOT1 axis promotes glutamine metabolism and ICC proliferation and growth. Inhibiting ANXA1 alongside glutamine uptake inhibition offers a promising strategy for treating ICC.
Collapse
Affiliation(s)
- Yanyu Gong
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Liwen Chen
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Hao Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Dijie Zheng
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Futang Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Changhao Wu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Yongning Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Yazhu Deng
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
- Key Laboratory of Hepatobiliary and Pancreatic Diseases Treatment and Bioinformatics Research, Guizhou Medical University, Guiyang, 550001, China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China.
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China.
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China.
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China.
- Key Laboratory of Hepatobiliary and Pancreatic Diseases Treatment and Bioinformatics Research, Guizhou Medical University, Guiyang, 550001, China.
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen, 518052, China.
| | - Chao Yu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China.
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China.
- Key Laboratory of Liver, Pancreas and Spleen of Guizhou Medical University, Gallbladder, Guiyang, 550001, China.
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China.
- Key Laboratory of Hepatobiliary and Pancreatic Diseases Treatment and Bioinformatics Research, Guizhou Medical University, Guiyang, 550001, China.
| |
Collapse
|
7
|
Cao Y, Qian R, Yao R, Zheng Q, Yang C, Yang X, Ji S, Zhang L, Zhan S, Wang Y, Wang T, Wang H, Wong CM, Yuan S, Heeschen C, Gao Q, Bernards R, Qin W, Wang C. DYRK1A-TGF-β signaling axis determines sensitivity to OXPHOS inhibition in hepatocellular carcinoma. Dev Cell 2025; 60:1483-1497.e7. [PMID: 39798576 DOI: 10.1016/j.devcel.2024.12.035] [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: 04/05/2024] [Revised: 09/14/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025]
Abstract
Intervening in mitochondrial oxidative phosphorylation (OXPHOS) has emerged as a potential therapeutic strategy for certain types of cancers. Employing kinome-based CRISPR screen, we find that knockout of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) synergizes with OXPHOS inhibitor IACS-010759 in liver cancer cells. Targeting DYRK1A combined with OXPHOS inhibitors activates TGF-β signaling, which is crucial for OXPHOS-inhibition-triggered cell death. Mechanistically, upregulation of glutamine transporter solute carrier family 1 member 5 (SLC1A5) transcription compensates for the increased glutamine requirement upon OXPHOS inhibition. DYRK1A directly phosphorylates SMAD3 Thr132, thereby suppressing the negative impact of TGF-β signaling on transcription of SLC1A5, leading to intrinsic resistance of liver cancer cells to OXPHOS inhibition. Moreover, we demonstrate the therapeutic efficacy of IACS-010759 in combination with DYRK1A inhibition in multiple liver cancer models, including xenografts, patient-derived xenografts, and spontaneous tumor model. Our study elucidates how the DYRK1A-TGF-β signaling axis controls the response of tumor cells to OXPHOS inhibition and provides valuable insights into targeting OXPHOS for liver cancer therapy.
Collapse
Affiliation(s)
- Ying Cao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruolan Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruilian Yao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Quan Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xupeng Yang
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuyi Ji
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linmen Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujie Zhan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiping Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianshi Wang
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Ming Wong
- State Key Laboratory for Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Shengxian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Christopher Heeschen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Liang MZ, Huang XF, Zhu JC, Bao JX, Chen CL, Wang XW, Lou YW, Pan YT, Dai YW. A machine learning-based glycolysis and fatty acid metabolism-related prognostic signature is constructed and identified ACSL5 as a novel marker inhibiting the proliferation of breast cancer. Comput Biol Chem 2025; 119:108507. [PMID: 40403353 DOI: 10.1016/j.compbiolchem.2025.108507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/27/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025]
Abstract
INTRODUCTION A new perspective on cancer metabolism suggests that it varies by context and is diverse. Cancer metabolism reprogramming can create a heterogeneous microenvironment that affects immune cell infiltration and function, complicating the selection of treatment methods. However, the specifics of this relationship remain unclear in breast cancer. This research aims to explore how glycolysis and fatty acid metabolism (GF) influence the immune microenvironment and their predictive capabilities for immunotherapy responses and overall survival. METHODS We at first time identified 602 GF-related genes. Utilizing multiple datasets from various centers and employing 10 different machine learning algorithms, we developed a GF-related signature called GFSscore, driven by artificial intelligence. RESULTS The GFSscore served as an independent prognostic indicator and demonstrated greater robustness than other models. Its validity was validated through multiple databases. Our study found that breast cancer patients with a high GFSscore, indicative of a greater tendency towards glycolytic activity, experienced poorer prognosis due to immunosuppression from distinct immune evasion mechanisms. Conversely, those with a low GFSscore, more inclined towards fatty acid metabolism, had better outcomes. Additionally, the GFSscore has the potential to forecast how well a patient might respond to immunotherapy and their susceptibility to chemotherapy medications. Moreover, we found that the overexpressed ACSL5 gene inhibits the proliferation of BRCA through experiments. CONCLUSIONS The GFSscore may offer patients personalized therapy by identifying new therapeutic targets for tumors. By understanding the relationship between cancer metabolism and the immune microenvironment, we can better tailor treatments to individual patients.
Collapse
Affiliation(s)
- Mei-Zhen Liang
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xian-Feng Huang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Jun-Chang Zhu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Jing-Xia Bao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Cheng-Liang Chen
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xiao-Wu Wang
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yun-Wei Lou
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Ya-Ting Pan
- Yongkang First People's Hospital Medical Group, Jinhua, Zhejiang, China.
| | - Yin-Wei Dai
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
9
|
Tang S, Wang Q, Wang Z, Cai L, Pan D, Li J, Chen Q, Zhou Y, Shen YQ. NSD1 mutation status determines metabolic inhibitor sensitivity in head and neck squamous cell carcinomas by regulating mitochondrial respiration. J Pathol 2025. [PMID: 40371884 DOI: 10.1002/path.6430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/20/2025] [Accepted: 03/24/2025] [Indexed: 05/16/2025]
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are the most common malignant tumors in the head and neck region, characterized by a high recurrence rate and early metastasis. Despite advances in treatment, patient outcomes and prognosis remain poor, highlighting the urgent need for new therapeutic strategies. Recent research has increasingly focused on targeting glucose metabolism as a therapeutic strategy for cancer, revealing multiple promising targets and potential drugs. However, the metabolic heterogeneity among tumors leads to variable sensitivity to metabolic inhibitors in different patients, limiting their clinical utility. In this study, we employed bioinformatics analysis, cell experiments, animal models, and multi-omics approaches to reveal differences in glucose metabolism phenotypes among HNSCC patients and elucidated the underlying molecular mechanisms driving these differences. Our findings showed that NSD1 mutation status affects the glucose metabolism phenotype in HNSCC, with NSD1 wild-type HNSCC exhibiting higher mitochondrial respiration and NSD1 mutant HNSCC showing weaker mitochondrial respiration but enhanced glycolysis. We further demonstrated that NSD1 regulates mitochondrial respiration in HNSCC via epigenetic modulation of the TGFB2/PPARGC1A signaling axis. Additionally, we found that NSD1 wild-type HNSCC is more sensitive to mitochondrial respiration inhibitors, whereas NSD1 mutant HNSCC shows increased sensitivity to glycolysis inhibitors. In summary, we found that NSD1 can epigenetically regulate the TGFB2/PPARGC1A axis to modulate mitochondrial respiration and sensitivity to metabolic inhibitors in HNSCC. These findings suggest a novel strategy for selecting metabolic inhibitors for HNSCC based on the NSD1 gene status of patients. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Shouyi Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, PR China
| | - Qing Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Zhen Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Luyao Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Dan Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Yu Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| |
Collapse
|
10
|
Girolimetti G, Gagliardi S, Cordella P, Bramato G, Di Corato R, Romano R, Guerra F, Bucci C. Induced mitochondrial deficit by NDUFS3 transient silencing reduces RAB7 expression and causes lysosomal dysfunction in pancreatic cancer cells. Cell Commun Signal 2025; 23:224. [PMID: 40369571 PMCID: PMC12079996 DOI: 10.1186/s12964-025-02214-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/23/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND RAB7 is a small GTPase with multiple cellular roles, regulating late endocytic trafficking and lysosomal biogenesis, influencing mitochondria-lysosome crosstalk, and contributing to many mitochondrial processes. Mitochondrial dysfunctions are widely reported in cancer and the development of cancer therapeutic strategies targeting mitochondria gained momentum in recent years. Mitochondrial impairment can cause alterations of mitochondria-lysosome crosstalk and can influence lysosomal function. Here, we used cell models of pancreatic cancer, one of the deadliest cancers worldwide, to cause a transient mild mitochondrial deficit lowering NDUFS3 protein levels in order to investigate the consequences on RAB7 and on the late endocytic pathway and, thus, the contribution of the mitochondria-lysosomes communication alterations to cancer progression. METHODS NDUFS3 and RAB7 downregulation was obtained by RNA interference (RNAi). Seahorse assays, Western blot analysis, mitochondrial staining, and Transmission Electron Microscopy (TEM) were used to assess silencing effects on mitochondrial structure and functioning. Western blotting was used to investigate expression of late endocytic pathway proteins and of the invasion marker vimentin. Confocal microscopy was used to analyze the mitochondrial network and lysosomal assessment. Zymography was performed to evaluate the ability to digest the extracellular matrix linked to cancer migration. SRB and colony assays were performed to assess cancer viability and proliferation. Wound healing assay and FluoroBlok membranes were used to determine migration and invasiveness. RESULTS In pancreatic cancer cells, transient silencing of the NDUFS3 protein caused mitochondrial deficit, slower oxidative metabolism, and mitochondrial morphology alterations. In this context, we observed RAB7 downregulation and impairment of the late endocytic pathway. In addition, NDUFS3-silenced RAB7-downregulated cells showed less invasive tumorigenic potential revealed by reduced levels of vimentin and other Epithelial-to-Mesenchymal Transition proteins, decreased viability, migration and invasiveness. Moreover, we found that modulation of RAB7 expression may regulate vimentin levels and influence mitochondrial morphology and levels of mitochondrial proteins. CONCLUSIONS Overall, our data show that mitochondrial deficit determines alterations of the crosstalk with lysosomes, leading to dysfunctions, and that this process is regulated by RAB7 acting as an oncogene. This highlights the synergic role of RAB7 and mitochondrial dysfunction, focusing on a cellular mechanism that may boost the effect of mitochondrial dysfunction in the cells, leading to the reduction of the tumorigenic potential.
Collapse
Affiliation(s)
- Giulia Girolimetti
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Sinforosa Gagliardi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Paola Cordella
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Grazia Bramato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Riccardo Di Corato
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Provinciale Lecce-Monteroni, Lecce, 73100, Italy
- Center for Biomolecular Nanotechnologies, Istituto Italiano di Tecnologia, Arnesano, 73010, Italy
| | - Roberta Romano
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Cecilia Bucci
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy.
| |
Collapse
|
11
|
Zirpoli S, Copperman N, Patel S, Forrest A, Hou Z, Matherly LH, Loeb DM, Di Cristofano A. Inhibition Of One-Carbon Metabolism In Ewing Sarcoma Results In Profound And Prolonged Growth Suppression Associated With Purine Depletion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.13.647987. [PMID: 40376089 PMCID: PMC12080949 DOI: 10.1101/2025.04.13.647987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Ewing sarcoma (EWS) is the second most common primary bone malignancy in adolescents and young adults. Patients who present with localized disease have experienced a steadily improving survival rate over the years, whereas those who present with metastatic disease have the same dismal prognosis as 30 years ago, with long term survival rates less than 20%, despite maximal intensification of chemotherapy. Thus, novel treatment approaches are a significant unmet clinical need. Targeting metabolic differences between EWS and normal cells offers a promising approach to improve outcomes for these patients. One-carbon metabolism utilizes serine and folate to generate glycine and tetrahydrofolate (THF)-bound one-carbon units required for de novo nucleotide biosynthesis. Elevated expression of several one-carbon metabolism genes is significantly associated with reduced survival in EWS patients. We show that both genetic and pharmacological inhibition of a key enzyme of the mitochondrial arm of the one-carbon metabolic pathway, serine hydroxymethyltransferase 2 (SHMT2), leads to substantial inhibition of EWS cell proliferation and colony-forming ability, and that this effect is primarily caused by depletion of glycine and one-carbon units required for synthesis of purine nucleotides. Inhibition of one-carbon metabolism at a different node, using the clinically relevant dihydrofolate reductase inhibitor Pralatrexate, similarly yields a profound growth inhibition, with depletion of thymidylate and purine nucleotides. Genetic depletion of SHMT2 dramatically impairs tumor growth in a xenograft model of EWS. Together, these data establish the upregulation of the one-carbon metabolism as a novel and targetable vulnerability of EWS cells, which can be exploited for therapy. Statement of Significance Using both genetic and pharmacologic approaches, this study identifies Ewing sarcoma's dependence on the mitochondrial arm, but not the cytoplasmic arm, of one-carbon metabolism as a targetable vulnerability that can be effectively harnessed for therapy.
Collapse
|
12
|
Zheng P, Hu Z, Shen Y, Gu L, Ouyang Y, Duan Y, Ji G, Dong B, Lin Y, Wen T, Tian Q, Hou Y, Zhou Q, Sun X, Chen X, Wang KL, Luo S, Wu S, Sun Y, Li M, Xiao L, Wu Q, Meng Y, Liu G, Wang Z, Bai X, Duan S, Ding Y, Bi Y, Wang Y, Li G, Liu X, Lu Z, Wu X, Tang Z, Xu D. PSAT1 impairs ferroptosis and reduces immunotherapy efficacy via GPX4 hydroxylation. Nat Chem Biol 2025:10.1038/s41589-025-01887-3. [PMID: 40281343 DOI: 10.1038/s41589-025-01887-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/14/2025] [Indexed: 04/29/2025]
Abstract
Tumor cells adapt to the inflammatory tumor microenvironment (TME) and develop resistance to immunotherapy, with ferroptosis being a major form of tumor cell death. However, the mechanisms by which tumor cells coordinate TME stimuli and their unique metabolic traits to evade ferroptosis and develop resistance to immunotherapy remain unclear. Here we showed that interferon-γ (IFNγ)-activated calcium/calmodulin-dependent protein kinase II phosphorylates phosphoserine aminotransferase 1 (PSAT1) at serine 337 (S337), allowing it to interact with glutathione peroxidase 4 (GPX4) and stabilize the protein, counteracting ferroptosis. PSAT1 elevates GPX4 stability by promoting α-ketoglutarate-dependent PHD3-mediated GPX4 proline 159 (P159) hydroxylation, disrupting its binding to HSC70 and inhibiting autophagy-mediated degradation. In mice, reconstitution of PSAT1 S337A or GPX4 P159A promotes ferroptosis and suppresses triple-negative breast cancer (TNBC) progression. Blocking PSAT1 pS337 with CPP elevates IFNγ-induced ferroptosis and enhances the efficacy of programmed cell death protein 1 (PD-1) antibodies in TNBC. Additionally, PSAT1-mediated GPX4 hydroxylation correlates with poor immunotherapy outcomes in patients with TNBC, highlighting PSAT1's noncanonical role in suppressing ferroptosis and immunotherapy sensitivity.
Collapse
Affiliation(s)
- Peixiang Zheng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zhiqiang Hu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuli Shen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Lina Gu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuan Ouyang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yuran Duan
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Guimei Ji
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Bofei Dong
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yanni Lin
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ting Wen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qi Tian
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yueru Hou
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Sun
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaohan Chen
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | | | - Shudi Luo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Shiqi Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuening Sun
- Department of Pharmacy, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Min Li
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Liwei Xiao
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ying Meng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Guijun Liu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zheng Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shengzhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanli Bi
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Yuhao Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Gaopeng Li
- Department of Colorectal Surgery and Oncology of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoguang Liu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiaohong Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China.
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Daqian Xu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China.
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China.
| |
Collapse
|
13
|
Zhou ZY, Bai N, Zheng WJ, Ni SJ. MultiOmics analysis of metabolic dysregulation and immune features in breast cancer. Int Immunopharmacol 2025; 152:114376. [PMID: 40054322 DOI: 10.1016/j.intimp.2025.114376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 02/09/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025]
Abstract
Metabolic disorders and diminished immune response are hallmark characteristics of tumors. However, limited studies have comprehensively integrated metabolic and immunological factors to evaluate or predict the prognosis of cancer patients. In this study, we utilized 72 metabolic pathway gene sets from the MsigDB database to conduct GSVA, univariate regression, and prognostic analyses on 247 breast cancer samples sourced from the TCGA and GEO databases. Consequently, five metabolic pathways with significant research value were identified. Based on these findings, unsupervised clustering was performed on the breast cancer samples to compare differences in gene expression, clinicopathological features, immune infiltration levels, and prognosis across different clusters. This process led to the identification of nine metabolism-related characteristic genes. Additionally, single-cell sequencing analysis was employed to assess the spatial expression patterns of these characteristic genes, revealing significantly higher expression indices in tumor cells compared to non-tumor cells. Subsequently, machine learning algorithms were applied to reconstruct metabolic risk models for evaluating the prognosis of breast cancer patients. The results indicated that the high metabolic risk group exhibited higher gene mutation scores, a greater proportion of unfavorable clinicopathological parameters, and lower chemokine and immune scores compared to the low-risk group. In conclusion, the metabolic risk model constructed using metabolism-related characteristic genes can accurately distinguish and predict the survival prognosis and immunotherapy outcomes of breast cancer patients, offering novel targets and insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Zuo-Yuan Zhou
- Department of Oncology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Nan Bai
- Medical school of Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Wen-Jie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| | - Su-Jie Ni
- Department of Oncology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
14
|
Jiang Y, Qian Z, Wang C, Wu D, Liu L, Ning X, You Y, Mei J, Zhao X, Zhang Y. Targeting B7-H3 inhibition-induced activation of fatty acid synthesis boosts anti-B7-H3 immunotherapy in triple-negative breast cancer. J Immunother Cancer 2025; 13:e010924. [PMID: 40221152 PMCID: PMC11997833 DOI: 10.1136/jitc-2024-010924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most malignant breast cancer, highlighting the need for effective immunotherapeutic targets. The immune checkpoint molecule B7-H3 has recently gained attention as a promising therapeutic target due to its pivotal role in promoting tumorigenesis and cancer progression. However, the therapeutic impact of B7-H3 inhibitors (B7-H3i) remains unclear. METHODS Transcriptomic and metabolomic analyses were conducted to explore the underlying mechanisms of B7-H3 inhibition in TNBC. The therapeutic efficacy of the combined treatment strategy was substantiated through comprehensive phenotypic assays conducted in vitro and validated in vivo using animal models. RESULTS B7-H3 blockade induces a "primed for death" stress state in cancer cells, leading to distinct alterations in metabolic pathways. Specifically, B7-H3 knockdown activated the AKT signaling pathway and upregulated sterol regulatory element-binding protein 1 (SREBP1), which in turn elevated FASN expression. The simultaneous inhibition of both B7-H3 and FASN more effectively attenuated the malignant progression of TNBC. CONCLUSIONS Our findings propose an "immune attack-metabolic compensation" dynamic model and suggest the feasibility of a dual-targeting strategy that concurrently inhibits both B7-H3 and FASN to enhance therapeutic efficacy in TNBC patients.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhiwen Qian
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cenzhu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Danping Wu
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Lu Liu
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xin Ning
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yilan You
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Mei
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoqian Zhao
- Department of Breast Surgery, Women's Hospital of Jiangnan University, Wuxi, China
| | - Yan Zhang
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
15
|
Kim DH, Kim DJ, Park SJ, Jang WJ, Jeong CH. Inhibition of GLS1 and ASCT2 Synergistically Enhances the Anticancer Effects in Pancreatic Cancer Cells. J Microbiol Biotechnol 2025; 35:e2412032. [PMID: 40223274 PMCID: PMC12010092 DOI: 10.4014/jmb.2412.12032] [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: 12/18/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 04/15/2025]
Abstract
Pancreatic cancer, a leading cause of cancer-related deaths, is characterized by increased dependence on glutamine metabolism. Telaglenastat (CB-839), a glutaminase (GLS) inhibitor targets glutamine metabolism; however, its efficacy as monotherapy is limited owing to metabolic adaptations. In this study, we demonstrated that CB-839 effectively inhibited cell growth in pancreatic cancer cells, but activated the general control nonderepressible 2 (GCN2)-activating transcription factor 4 (ATF4) signaling pathway. ATF4 knockdown reduced glutamine transporter alanine, serine, and cysteine transporter 2 (ASCT2) expression, glutamine uptake, and cell viability under glutamine deprivation-recovery conditions, confirming its protective role in mitigating glutamine-related metabolic stress. Notably, the combination of CB-839 and the ASCT2 inhibitor V-9302 demonstrated a synergistic effect, significantly suppressing pancreatic cancer cell survival. These findings highlight ATF4 and ASCT2 as crucial therapeutic targets and indicate that dual inhibition of GLS and ASCT2 may enhance treatment outcomes for pancreatic cancer.
Collapse
Affiliation(s)
- Dong-Hwan Kim
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Dong Joon Kim
- Department of Microbiology, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jun Park
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Won-Jun Jang
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
16
|
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
|
17
|
Yan W, Wang S, Zhu L, Yu X, Li J. Targeted editing of CCL5 with CRISPR-Cas9 nanoparticles enhances breast cancer immunotherapy. Apoptosis 2025; 30:912-935. [PMID: 39870938 PMCID: PMC11947030 DOI: 10.1007/s10495-024-02032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2024] [Indexed: 01/29/2025]
Abstract
Breast cancer remains one of the leading causes of cancer-related mortality among women worldwide. Immunotherapy, a promising therapeutic approach, often faces challenges due to the immunosuppressive tumor microenvironment. This study explores the innovative use of CRISPR-Cas9 technology in conjunction with FCPCV nanoparticles to target and edit the C-C Motif Chemokine Ligand 5 (CCL5) gene, aiming to improve the efficacy of breast cancer immunotherapy. Single-cell RNA sequencing (scRNA-seq) and TCGA-BRCA data identified CCL5 as a key immune-related gene in breast cancer. Using CRISPR-Cas9, sgRNA targeting CCL5 was designed and delivered to breast cancer cells and humanized mouse models via FCPCV nanoparticles. In vitro experiments demonstrated that FCPCV nanoparticles effectively silenced CCL5, enhanced CD8+ T cell activity, and increased the production of cytokines such as IFN-γ, TNF-α, and GZMB. In vivo studies revealed significant tumor suppression, improved immune microenvironment, and increased CD8+/CD4+ ratios in treated mice, without notable toxic side effects. These findings highlight the potential of CRISPR-Cas9 nanoparticle-mediated gene editing as a novel strategy for enhancing breast cancer immunotherapy, providing a new direction for personalized and effective cancer treatment.
Collapse
Affiliation(s)
- Wei Yan
- Department of Thoracic Oncology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, Nanchang, 330029, China
| | - Shuo Wang
- Department of Thoracic Oncology, Ganzhou Cancer Hospital, Ganzhou Institute for Cancer Research, The Affiliated Cancer Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Lihui Zhu
- Department of Endoscopy Center, Jiangxi Provincial Children's Hospital, Nanchang, 330006, China
| | - Xinlin Yu
- Department of Medical Laboratory, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, Nanchang, Jiangxi, 330029, China.
| | - Jianglong Li
- Department of Breast Cancer Surgery, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, Nanchang, Jiangxi, 330029, China.
| |
Collapse
|
18
|
Ren X, Wu Y, Song T, Yang Q, Zhou Q, Lin J, Xu L, Xiang B, Chen Z, Zhang Y. Clonorchis sinensis Promotes Intrahepatic Cholangiocarcinoma Progression by Activating FASN-Mediated Fatty Acid Metabolism. J Gastroenterol Hepatol 2025; 40:1004-1015. [PMID: 39806791 DOI: 10.1111/jgh.16879] [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: 10/28/2024] [Revised: 11/27/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Clonorchis sinensis infection is an important risk factor for intrahepatic cholangiocarcinoma (ICC). C. sinensis positive (C.s+) ICC patients had much shorter overall survival (OS) compared with C. sinensis negative (C.s-) group. This study aims to explore the impact and underlying mechanism of C. sinensis infection on ICC progression. METHODS In this study, ICC patients underwent surgery from two medical centers enrolled. RNA sequencing was used to determine the downstream activated pathways and genes. Furthermore, we demonstrated the potential mechanism of C. sinensis infection in promoting ICC progression through in vitro co culture systems and two animal models. RESULTS Through RNA sequencing, we found fatty acid metabolism and the expression of fatty acid synthase (FASN), a key enzyme catalyzing long-chain fatty acid synthesis, were significantly elevated in C.s+ ICCs. Then, we found excretory/secretory products (ESPs) secreted by C. sinensis could significantly upregulate the expression of transcription factor E2F1, thereby promoting FASN expression and fatty acid synthesis in tumor cells, which ultimately accelerating tumor progression. However, the promotive effect disappeared when FASN was knocked down. Meanwhile, ESPs could promote tumor growth, increasing FASN expression and free fatty acid level in both subcutaneous and orthotopic mouse models. CONCLUSION This study indicates that C. sinensis infection could upregulate the level of FASN and activate fatty acid synthesis pathway, thereby accelerating ICC progression. This provides a new insight for the clinical treatment of ICC with C. sinensis infection.
Collapse
Affiliation(s)
- Xiaoxue Ren
- Department of Oncology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yanqing Wu
- Department of Gastroenterology and Hepatology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Tongtong Song
- Center of Hepato-Pancreato-Biliary Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qingxia Yang
- Department of Oncology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qianying Zhou
- Department of Oncology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jie Lin
- Second Department of General Surgery, Shunde Hospital, Southern Medical University, Foshan, China
| | - Lixia Xu
- Department of Oncology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bangde Xiang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zebin Chen
- Center of Hepato-Pancreato-Biliary Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ying Zhang
- Department of Oncology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
19
|
Richiardone E, Giolito MV, Al Roumi R, Ambroise J, Boidot R, Drotleff B, Ghesquière B, Lupo B, Trusolino L, Bardelli A, Arena S, Feron O, Corbet C. Acidosis overrides molecular heterogeneity to shape therapeutically targetable metabolic phenotypes in colon cancers. Cancer Lett 2025; 613:217512. [PMID: 39900217 DOI: 10.1016/j.canlet.2025.217512] [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: 11/04/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
Colorectal cancer (CRC) represents a prototypical example of a cancer type for which inter- and intra-tumor heterogeneities remain major challenges for the clinical management of patients. Besides genotype-mediated phenotypic alterations, tumor microenvironment (TME) conditions are increasingly recognized to promote intrinsic diversity and phenotypic plasticity and sustain disease progression. In particular, acidosis is a common hallmark of solid tumors, including CRC, and it is known to induce aggressive cancer cell phenotypes. In this study, we report that long-term adaptation to acidic pH conditions is associated with common metabolic alterations, including a glycolysis-to-respiration switch and a higher reliance on the activity of phosphoglycerate dehydrogenase (PHGDH), in CRC cells initially displaying molecularly heterogeneous backgrounds. Pharmacological inhibition of PHGDH activity or mitochondrial respiration induces greater growth-inhibitory effects in acidosis-exposed CRC cells in 2D and 3D culture conditions, and in patient-derived CRC organoids. These data pave the way for drugs targeting the acidic tumor compartment as a "one-size-fits-all" therapeutic approach to delay CRC progression.
Collapse
Affiliation(s)
- Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Maria Virginia Giolito
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Rim Al Roumi
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Jérôme Ambroise
- Centre des Technologies Moléculaires Appliquées (CTMA), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 54, B-1200, Brussels, Belgium
| | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-François Leclerc Cancer Center-UNICANCER, 21079, Dijon, France
| | | | - Bart Ghesquière
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Barbara Lupo
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Alberto Bardelli
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Torino, Italy; IFOM ETS, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Sabrina Arena
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium; WEL Research Institute, Avenue Pasteur 6, B-1300, Wavre, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium; WEL Research Institute, Avenue Pasteur 6, B-1300, Wavre, Belgium.
| |
Collapse
|
20
|
Liang Z, Zhang T, Huang J, Huang Z, Zhao Z, Cai S, Ma J. A comprehensive prognostic and immunological analysis of hexokinase domain containing protein-1 (HKDC1) in pan-cancer. PeerJ 2025; 13:e19083. [PMID: 40124623 PMCID: PMC11929506 DOI: 10.7717/peerj.19083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Background Currently, research on the role of hexokinase domain-containing protein-1 (HKDC1) in neoplasm metabolism remains sparse. This study seeks to conduct a thorough investigation of HKDC1's potential functions across thirty-three different tumor types, utilizing data obtained from The Cancer Genome Atlas (TCGA). Method We conducted a thorough data extraction from the TCGA database, subsequently employing R (version 4.2.2) and its associated software packages for detailed analysis. Our investigation centered on evaluating the differential expression and prognostic significance of HKDC1, while also examining its connections to tumor heterogeneity, mutation profiles, and RNA modifications. Furthermore, we analyzed the relationship between HKDC1 expression and tumor immunity utilizing the TIMER analysis approach. Results A comprehensive analysis of various tumor types has revealed that HKDC1 is significantly upregulated in many malignant tumors. Importantly, patients with elevated HKDC1 levels in their tumor tissues often experience poorer prognoses. The association between HKDC1 expression, immune cell infiltration, and the existence of immune checkpoints suggests a possible connection between the tumor microenvironment and HKDC1, alongside tumor advancement. Gene set enrichment analysis (GSEA) further substantiates the idea that HKDC1 may play a role in several critical pathways and biological processes associated with neoplasm. Additionally, the overexpression of HKDC1 is influenced by promoter methylation and alterations in DNA copy number amplification. Furthermore, in vitro experiments demonstrated that silencing HKDC1 resulted in a marked reduction in the proliferation, migration, and invasion capabilities of neoplasm cells. Conclusion Our initial pan-cancer analysis provided a comprehensive understanding of the oncogenic roles of HKDC1 across diverse cancer types. Moreover, HKDC1 has the potential to serve as a significant prognostic biomarker.
Collapse
Affiliation(s)
- Zhi Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Tianhao Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jiajia Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhixin Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zeyu Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shirong Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jinping Ma
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
21
|
Li G, Wang Z, Guo Y, Ni C, Gao Y, Xu K, Xiao T, Shi X, Shen M. Copper-doped layered double hydroxides co-deliver proteins/drugs for cascaded chemodynamic/immunotherapy via dual regulation of tumor metabolism. Acta Biomater 2025; 195:350-362. [PMID: 39921184 DOI: 10.1016/j.actbio.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
We report here a Cu2+-doped layered double hydroxide (LDH) nanoplatform to load both monocarboxylate transporter inhibitor diclofenac (DC) and lactate oxidase (LOX) for dual modulation of tumor lactate and redox metabolisms to activate immunotherapy through enhanced Cu-mediated chemodynamic therapy (CDT) of tumors. The formed LDH-DC-LOX nanoparticles with a diameter of 55 nm are stable, can be effectively taken up by cancer cells to regulate lactate through both LOX-mediated catalytic conversion and DC-enabled inhibition of extracellular efflux of lactate, and can exert redox metabolism through CDT via Cu2+-mediated glutathione (GSH) depletion and Fenton-like reaction with hydrogen peroxide (H2O2) that can be further accumulated via LOX-mediated catalysis. The major advantage of the developed LDH-DC-LOX nanoparticles lies in the therapeutic synergy and cascade that can be achieved through the loaded DC and LOX for enhanced tumor lactate metabolism regulation, for enhanced Cu-mediated CDT and redox metabolism regulation, and for activation of immunotherapy that can further enhances the Cu2+-mediated CDT effect. The developed LDH nanoplatform demonstrated here for effective murine breast tumor treatment provides a new paradigm for dual regulation of lactate and redox metabolisms that may enable synergistic and cascaded combination therapy of different cancer types. STATEMENT OF SIGNIFICANCE: Targeting the tumor microenvironment (TME) to alter tumor metabolic pathways represents a promising strategy for next-generation cancer therapy. Herein, a copper-doped layered double hydroxide (LDH) nanoplatform is developed to co-deliver both diclofenac (DC) and lactate oxidase (LOX) to tumor cells for efficient dual regulation of tumor lactate and redox metabolisms, resulting in synergistic and cascaded chemodynamic therapy/immunotherapy of breast tumors. The developed LDH-DC-LOX nanoparticles can release Cu2+ and DC under an acidic TME, and can act in synergy to reduce TME lactate and generate H2O2, thus modulating redox metabolism and activating anticancer immunotherapy. The secreted cytokine IFN-γ after activation of antitumor immune responses can further mediate enhanced chemodynamic therapy effect through downregulation of cystine/glutamate transporter SLC7A11 to suppress GSH synthesis.
Collapse
Affiliation(s)
- Gaoming Li
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Zhiqiang Wang
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yunqi Guo
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Cheng Ni
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yue Gao
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Kaibing Xu
- Research Center for Analysis and Measurement, Donghua University, Shanghai 201620, China
| | - Tingting Xiao
- Institute of Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Xiangyang Shi
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, Funchal 9020-105, Portugal.
| | - Mingwu Shen
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
22
|
Fan S, Wang W, Che W, Xu Y, Jin C, Dong L, Xia Q. Nanomedicines Targeting Metabolic Pathways in the Tumor Microenvironment: Future Perspectives and the Role of AI. Metabolites 2025; 15:201. [PMID: 40137165 PMCID: PMC11943624 DOI: 10.3390/metabo15030201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Tumor cells engage in continuous self-replication by utilizing a large number of resources and capabilities, typically within an aberrant metabolic regulatory network to meet their own demands. This metabolic dysregulation leads to the formation of the tumor microenvironment (TME) in most solid tumors. Nanomedicines, due to their unique physicochemical properties, can achieve passive targeting in certain solid tumors through the enhanced permeability and retention (EPR) effect, or active targeting through deliberate design optimization, resulting in accumulation within the TME. The use of nanomedicines to target critical metabolic pathways in tumors holds significant promise. However, the design of nanomedicines requires the careful selection of relevant drugs and materials, taking into account multiple factors. The traditional trial-and-error process is relatively inefficient. Artificial intelligence (AI) can integrate big data to evaluate the accumulation and delivery efficiency of nanomedicines, thereby assisting in the design of nanodrugs. Methods: We have conducted a detailed review of key papers from databases, such as ScienceDirect, Scopus, Wiley, Web of Science, and PubMed, focusing on tumor metabolic reprogramming, the mechanisms of action of nanomedicines, the development of nanomedicines targeting tumor metabolism, and the application of AI in empowering nanomedicines. We have integrated the relevant content to present the current status of research on nanomedicines targeting tumor metabolism and potential future directions in this field. Results: Nanomedicines possess excellent TME targeting properties, which can be utilized to disrupt key metabolic pathways in tumor cells, including glycolysis, lipid metabolism, amino acid metabolism, and nucleotide metabolism. This disruption leads to the selective killing of tumor cells and disturbance of the TME. Extensive research has demonstrated that AI-driven methodologies have revolutionized nanomedicine development, while concurrently enabling the precise identification of critical molecular regulators involved in oncogenic metabolic reprogramming pathways, thereby catalyzing transformative innovations in targeted cancer therapeutics. Conclusions: The development of nanomedicines targeting tumor metabolic pathways holds great promise. Additionally, AI will accelerate the discovery of metabolism-related targets, empower the design and optimization of nanomedicines, and help minimize their toxicity, thereby providing a new paradigm for future nanomedicine development.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei Dong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| | - Qin Xia
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| |
Collapse
|
23
|
Chen Z, Fang Y, Zhang J. RBIS as a prognostic biomarker for progression-free survival in prostate cancer: insights from bioinformatics and immune infiltration analysis. Discov Oncol 2025; 16:297. [PMID: 40069535 PMCID: PMC11896948 DOI: 10.1007/s12672-025-02083-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/05/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a highly prevalent malignancy among men, with significant regional variations in incidence and poor survival rates in advanced stages. Ribosome biogenesis (RB) is pivotal for cancer cell proliferation, yet the specific role of the ribosomal biogenesis factor (RBIS) gene in PCa remains underexplored. This study aims to elucidate the biological and clinical relevance of RBIS in PCa progression. METHODS We analyzed RNA-sequencing data from the TCGA database and three GEO datasets to compare RBIS expression in normal versus tumor tissues. The relationship between RBIS expression and clinicopathological features, including tumor stage, Gleason score, and progression-free survival (PFS), was assessed. Co-expression and functional enrichment analyses identified key biological processes involving RBIS, while genetic alterations, immune infiltration, and drug sensitivity were also evaluated. RESULTS RBIS expression was significantly higher in PCa tissues and correlated with advanced tumor stage, higher Gleason scores, and poorer PFS. Multivariate analysis confirmed RBIS as an independent prognostic marker. Functional analysis implicated RBIS in energy metabolism and protein synthesis. Moreover, RBIS expression was associated with immune cell infiltration and drug sensitivity, highlighting its potential as a therapeutic target. CONCLUSION RBIS represents a promising biomarker for PCa diagnosis, prognosis, and therapy. Further research is required to validate these findings and advance RBIS's clinical application, offering novel insights into PCa management.
Collapse
Affiliation(s)
- Zexiao Chen
- Department of Breast Surgery, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, China
| | - Yutong Fang
- Department of Breast Surgery, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, China.
| | - Jianhua Zhang
- Department of Minimally Invasive Interventional, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, China.
| |
Collapse
|
24
|
Kohli M, Poulogiannis G. Harnessing the Power of Metabolomics for Precision Oncology: Current Advances and Future Directions. Cells 2025; 14:402. [PMID: 40136651 PMCID: PMC11940876 DOI: 10.3390/cells14060402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/24/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic reprogramming is a hallmark of cancer, with cancer cells acquiring many unique metabolic traits to support malignant growth, and extensive intra- and inter-tumour metabolic heterogeneity. Understanding these metabolic characteristics presents opportunities in precision medicine for both diagnosis and therapy. However, despite its potential, metabolic phenotyping has lagged behind genetic, transcriptomic, and immunohistochemical profiling in clinical applications. This is partly due to the lack of a single experimental technique capable of profiling the entire metabolome, necessitating the use of multiple technologies and approaches to capture the full range of cancer metabolic plasticity. This review examines the repertoire of tools available for profiling cancer metabolism, demonstrating their applications in preclinical and clinical settings. It also presents case studies illustrating how metabolomic profiling has been integrated with other omics technologies to gain insights into tumour biology and guide treatment strategies. This information aims to assist researchers in selecting the most effective tools for their studies and highlights the importance of combining different metabolic profiling techniques to comprehensively understand tumour metabolism.
Collapse
Affiliation(s)
| | - George Poulogiannis
- Signalling and Cancer Metabolism Laboratory, Division of Cell and Molecular Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK;
| |
Collapse
|
25
|
D’Aprile S, Denaro S, Gervasi A, Vicario N, Parenti R. Targeting metabolic reprogramming in glioblastoma as a new strategy to overcome therapy resistance. Front Cell Dev Biol 2025; 13:1535073. [PMID: 40078366 PMCID: PMC11897528 DOI: 10.3389/fcell.2025.1535073] [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: 11/26/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Glioblastoma (GBM) is one of the deadliest tumors due to its high aggressiveness and resistance to standard therapies, resulting in a dismal prognosis. This lethal tumor carries out metabolic reprogramming in order to modulate specific pathways, providing metabolites that promote GBM cells proliferation and limit the efficacy of standard treatments. Indeed, GBM remodels glucose metabolism and undergoes Warburg effect, fuelling glycolysis even when oxygen is available. Moreover, recent evidence revealed a rewiring in nucleotide, lipid and iron metabolism, resulting not only in an increased tumor growth, but also in radio- and chemo-resistance. Thus, while on the one hand metabolic reprogramming is an advantage for GBM, on the other hand it may represent an exploitable target to hamper GBM progression. Lately, a number of studies focused on drugs targeting metabolism to uncover their effects on tumor proliferation and therapy resistance, demonstrating that some of these are effective, in combination with conventional treatments, sensitizing GBM to radiotherapy and chemotherapy. However, GBM heterogeneity could lead to a plethora of metabolic alterations among subtypes, hence a metabolic treatment might be effective for proneural tumors but not for mesenchymal ones, which are more aggressive and resistant to conventional approaches. This review explores key mechanisms of GBM metabolic reprogramming and their involvement in therapy resistance, highlighting how metabolism acts as a double-edged sword for GBM, taking into account metabolic pathways that seem to offer promising treatment options for GBM.
Collapse
Affiliation(s)
| | | | | | | | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
26
|
Dong S, Li X, Huang Q, Li Y, Li J, Zhu X, Xue C, Chen R, Zeng Y, Wu J, Zhong Y, Hu S. Resistance to immunotherapy in non-small cell lung cancer: Unraveling causes, developing effective strategies, and exploring potential breakthroughs. Drug Resist Updat 2025; 81:101215. [PMID: 40081220 DOI: 10.1016/j.drup.2025.101215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Over the last two decades, advancements in deciphering the intricate interactions between oncology and immunity have fueled a meteoric rise in immunotherapy for non-small cell lung cancer, typified by an explosive growth of immune checkpoint inhibitors. However, resistance to immunotherapy remains inevitable. Herein we unravel the labyrinthine mechanisms of resistance to immunotherapy, characterized by their involvement of nearly all types of cells within the body, beyond the extrinsic cancer cells, and importantly, such cells are not only (inhibitory or excitatory, or both) signal recipients but also producers, acting in a context-dependent manner. At the molecular level, these mechanisms underlie genetic and epigenetic aberrations, which are regulated by or regulate various protein kinases, growth factors, and cytokines with inherently dynamic and spatially heterogeneous properties. Additionally, macroscopic factors such as nutrition, comorbidities, and the microbiome within and around organs or tumor cells are involved. Therefore, developing therapeutic strategies combined with distinct action informed by preclinical, clinical, and real-world evidence, such as radiotherapy, chemotherapy, targeted therapy, antibody-drug conjugates, oncolytic viruses, and cell-based therapies, may stand as a judicious reality, although the ideality is to overcome resistance point-by-point through a novel drug. Notably, we highlight a realignment of treatment aims, moving the primary focus from eliminating cancer cells -- such as through chemotherapy and radiotherapy -- to promoting immune modulation and underscore the value of regulating various components within the host macro- or micro-environment, as their effects, even if seemingly minimal, can cumulatively contribute to visible clinical benefit when applied in combination with ICIs. Lastly, this review also emphasizes the current hurdles scattered throughout preclinical and clinical studies, and explores evolving directions in the landscape of immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Shuang Dong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Xiaoyu Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Qing Huang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuanxiang Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | | | - Xianmin Zhu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Chang Xue
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Runzhi Chen
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuan Zeng
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Jingyi Wu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yi Zhong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| |
Collapse
|
27
|
Andryszkiewicz W, Gąsiorowska J, Kübler M, Kublińska K, Pałkiewicz A, Wiatkowski A, Szwedowicz U, Choromańska A. Glucose Metabolism and Tumor Microenvironment: Mechanistic Insights and Therapeutic Implications. Int J Mol Sci 2025; 26:1879. [PMID: 40076506 PMCID: PMC11900028 DOI: 10.3390/ijms26051879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Metabolic reprogramming in cancer cells involves changes in glucose metabolism, glutamine utilization, and lipid production, as well as promoting increased cell proliferation, survival, and immune resistance by altering the tumor microenvironment. Our study analyzes metabolic reprogramming in neoplastically transformed cells, focusing on changes in glucose metabolism, glutaminolysis, and lipid synthesis. Moreover, we discuss the therapeutic potential of targeting cancer metabolism, focusing on key enzymes involved in glycolysis, the pentose phosphate pathway, and amino acid metabolism, including lactate dehydrogenase A, hexokinase, phosphofructokinase and others. The review also highlights challenges such as metabolic heterogeneity, adaptability, and the need for personalized therapies to overcome resistance and minimize adverse effects in cancer treatment. This review underscores the significance of comprehending metabolic reprogramming in cancer cells to engineer targeted therapies, personalize treatment methodologies, and surmount challenges, including metabolic plasticity and therapeutic resistance.
Collapse
Affiliation(s)
- Wiktoria Andryszkiewicz
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Julia Gąsiorowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Maja Kübler
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Karolina Kublińska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Agata Pałkiewicz
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Adam Wiatkowski
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Urszula Szwedowicz
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
28
|
Xu Y, Jiang X, Hu Z. Synergizing metabolomics and artificial intelligence for advancing precision oncology. Trends Mol Med 2025:S1471-4914(25)00016-4. [PMID: 39956738 DOI: 10.1016/j.molmed.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/18/2025]
Abstract
Metabolomics has emerged as a transformative tool in precision oncology, with substantial potential for advancing biomarker discovery, monitoring treatment responses, and aiding drug development. Integrating artificial intelligence (AI) into metabolomics optimizes data acquisition and analysis, facilitating the interpretation of complex metabolic networks and enabling more effective multiomics integration. In this opinion, we explore recent advances in the application of metabolomics within precision oncology, emphasizing the unique advantages that AI-driven metabolomics offers. We propose that AI not only complements but also amplifies the potential of current platforms, accelerating research progress and ultimately improving patient outcomes. Finally, we discuss the opportunities and challenges involved in translating AI-driven metabolomics into clinical practice for precision oncology.
Collapse
Affiliation(s)
- Yipeng Xu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xiaojuan Jiang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
29
|
Cui Y, Cao X, Zhang Y, Fu C, Li D, Sun Y, Zhang Y, Xu T, Tsukamoto T, Cao D, Jiang J. Protein phosphatase 1 regulatory subunit 15 A (PPP1R15A) promoted the progression of gastric cancer by activating cell autophagy under energy stress. J Exp Clin Cancer Res 2025; 44:52. [PMID: 39948597 PMCID: PMC11823012 DOI: 10.1186/s13046-025-03320-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Glucose metabolism plays a critical role in tumor progression. When glucose intake is insufficient and the tumor's growth rate exceeds its energy supply, tumor cells typically adapt and overcome the energy stress through compensatory mechanisms to maintain the survival of tumor cells, which may also be related to tumor recurrence or metastasis. METHODS Different concentrations of glucose were selected as the basis for the energy stress model of gastric cancer. Then CCK-8 and flow cytometry were used to detect its effects on cell proliferation, apoptosis, and cell cycle. Differentially expressed genes (DEGs) were screened by RNA sequencing and the regulated pathways were identified by gene set enrichment analysis. The regulatory relationship between the gene PPP1R15A and its transcription factor JUN was proved by ChIP-qPCR and dual-luciferase reporter assay. The gain and loss of function assays were conducted to examine the effects of PPP1R15A under energy stress in vivo and in vitro. Potential regulatory mechanisms of PPP1R15A were further analyzed through a combination of online databases, RNA sequencing, and metabolite sequencing. The regulation of PPP1R15A on cell autophagy under energy stress was detected by western blot, transmission electron microscope, mRFP-GFP-LC3 adenovirus and laser scanning confocal microscopy. RESULTS PPP1R15A and the transcription factor JUN were significantly upregulated by glucose deprivation (0 mM vs. 25 mM), JUN combined with the promoter of PPP1R15A and activated its expression. Both PPP1R15A and JUN were highly expressed in gastric cancer tissues and were independent risk factors for prognosis in the gastric cancer cohort. Overexpression of PPP1R15A promoted cell proliferation, inhibited apoptosis, and was involved in cell cycle arrest. Further RNA and metabolite sequencing suggested that PPP1R15A was associated with cell autophagy. In vitro experiments confirmed that both glucose deprivation and overexpression of PPP1R15A promoted the biosynthesis of autolysosome and autophagosome, and activated the cleavage of LC3 complex in gastric cancer cells. Moreover, PPP1R15A knockdown inhibited cell autophagy induced by glucose deprivation. CONCLUSIONS PPP1R15A sustained the survival of gastric cancer cells by regulating autophagy under energy stress to resist or adapt to harsh environments.
Collapse
Affiliation(s)
- Yingnan Cui
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Doctor of excellence program (DEP), The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yangyu Zhang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chenhao Fu
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dongming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuanlin Sun
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuzheng Zhang
- Department of Hospital Infection Management, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Tingshuang Xu
- Core facility of The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology I, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Donghui Cao
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Jing Jiang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
30
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
31
|
Cheng Q, Chen Y, Zou D, Li Q, Shi X, Qin Q, Liu M, Wang L, Wang Z. Targeting Metabolic Adaptation of Colorectal Cancer with Vanadium-Doped Nanosystem to Enhance Chemotherapy and Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409329. [PMID: 39739629 PMCID: PMC11831457 DOI: 10.1002/advs.202409329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/16/2024] [Indexed: 01/02/2025]
Abstract
The anti-tumor efficacy of current pharmacotherapy is severely hampered due to the adaptive evolution of tumors, urgently needing effective therapeutic strategies capable of breaking such adaptability. Metabolic reprogramming, as an adaptive survival mechanism, is closely related to therapy resistance of tumors. Colorectal cancer (CRC) cells exhibit a high energy dependency that is sustained by an adaptive metabolic conversion between glucose and glutamine, helping tumor cells to withstand nutrient-deficient microenvironments and various treatments. We discover that transition metal vanadium (V) effectively inhibits glucose metabolism in CRC and synergizes with glutaminase inhibitors (BPTES) to disrupt CRC's energy dependency. Thus, a dual energy metabolism suppression nanosystem (VSi-BP@HA) is engineered by loading BPTES into V-doped hollow mesoporous silica nanoparticles. This nanosystem effectively dampens CRC energy metabolism, eradicating 33% of tumors in mice. Strikingly, the cell biological and preclinical model datasets provide compelling evidence showing that VSi-BP@HA not only reverses CRC cells chemo-resistance but also drastically potentiates anti-PD1 immunotherapy. Therefore, this nanosystem provides not only a promising approach to suppress CRC, but also a potential adjunct tool for enhancing chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Qian Cheng
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart EquipmentWuhan430022China
| | - Yuzhe Chen
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart EquipmentWuhan430022China
| | - Danyi Zou
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
| | - Qilin Li
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart EquipmentWuhan430022China
| | - Xiaolei Shi
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart EquipmentWuhan430022China
| | - Qushuhua Qin
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart EquipmentWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Miaodeng Liu
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart EquipmentWuhan430022China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart EquipmentWuhan430022China
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart EquipmentWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
32
|
Hu X, Ling D. Artificial metabzyme-driven metabolic reprogramming and precision oncology. Clin Transl Med 2025; 15:e70215. [PMID: 39888283 PMCID: PMC11782831 DOI: 10.1002/ctm2.70215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 02/01/2025] Open
Affiliation(s)
- Xi Hu
- School of PharmacyAnhui University of Chinese MedicineHefeiChina
- Institute of PharmaceuticsAnhui Academy of Chinese MedicineHefeiChina
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and ApplicationAnhui University of Chinese MedicineHefeiChina
| | - Daishun Ling
- Frontiers Science Center for Transformative MoleculesSchool of Chemistry and Chemical EngineeringNational Center for Translational Medicine, National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and TherapyShanghai Jiao Tong UniversityShanghaiChina
- WLA LaboratoriesShanghaiChina
| |
Collapse
|
33
|
Wang H, Chu D, Zhang M, Huang X, Shi Y, Zhao Y, Qu H, Li D, Xu Z, Gao L, Zhang X, Wang W. Manganese-doped carbon dots with catalase-like activity enable MRI-guided enhanced photodynamic therapy. Colloids Surf B Biointerfaces 2025; 246:114398. [PMID: 39608308 DOI: 10.1016/j.colsurfb.2024.114398] [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: 10/27/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
The tumor microenvironment (TME) exhibits characteristics such as hypoxia, weak acidity, and enrichment of glutathione and hydrogen peroxide (H2O2), which greatly limits the effectiveness of tumor magnetic resonance imaging (MRI) and photodynamic therapy (PDT). Carbon dots (CDs) nanozymes are excellent candidate materials with both diagnostic and therapeutic potential. However, CDs nanozymes with both ultra-high relaxation rate and good therapeutic effect are still to be developed. Herein, novel carbon dots (MPC-CDs) were synthesized from polyethyleneimine (PEI), the photosensitizer hexahydroporphyrin (Ce6) and manganese. The Ce6 enabled the MPC-CDs to exhibit excellent PDT therapeutic ability, with a singlet oxygen yield as high as 1.52. The doping of the metal manganese gave the complexes CAT-like activity, and the singlet oxygen rate was further increased in the presence of H2O2, up to 1.97. In addition, manganese endowed the CDs with better MRI capabilities, and the r1 and r2 relaxation rates were significantly improved by 7.8-fold and 4.6-fold under acidic and H2O2 conditions. The in vitro and in vivo results showed that MPC-CDs could achieve TME-responsive MR imaging and synergistic anti-tumor effects, providing an effective strategy to further enhance the effectiveness of tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Huihui Wang
- Department of Radiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; Jiangsu Key laboratory of integrated traditional Chinese and Western Medicine for prevention and treatment of Senile Diseases, Yangzhou University, Yangzhou 225001, China
| | - Dongchuan Chu
- Department of Radiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China
| | - Maolin Zhang
- Department of Radiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China
| | - Xueping Huang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Yu Shi
- Department of Radiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Yi Zhao
- Department of Radiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China
| | - Hang Qu
- Department of Radiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China
| | - Dandan Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Zhuobin Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Xing Zhang
- Department of spinal surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
| | - Wei Wang
- Department of Radiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; Jiangsu Key laboratory of integrated traditional Chinese and Western Medicine for prevention and treatment of Senile Diseases, Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
34
|
Zabielska J, Stelmanska E, Szrok-Jurga S, Kobiela J, Czumaj A. Lipids Metabolism Inhibition Antiproliferative Synergy with 5-Fluorouracil in Human Colorectal Cancer Model. Int J Mol Sci 2025; 26:1186. [PMID: 39940954 PMCID: PMC11818398 DOI: 10.3390/ijms26031186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Colorectal cancer (CRC) is recognized as the third most lethal cancer worldwide. While existing treatment options demonstrate considerable efficacy, they are often constrained by non-selectivity and substantial side effects. Recent studies indicate that lipid metabolism significantly influences carcinogenesis, highlighting it as a promising avenue for developing targeted anticancer therapies. The purpose of the study was to see if acyl-coenzyme A: cholesterol acyltransferase 1 (ACAT1), 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), and stearoyl-CoA 9-desaturase (SCD1) are good metabolic targets and whether the use of inhibitors of these enzymes together with 5-fluorouracil (5-FU) would have a synergistic effect on CRC cell viability. To confirm that the correct lipid targets were chosen, the expression levels of ACAT1, HMGCR, and SCD1 were examined in CRC patients and cell models. At first, each compound (Avasimibe, Lovastatin, MF-438, and 5-FU was tested separately, and then each inhibitor was paired with 5-FU to assess the synergistic effect on cell viability. Gene expression of selected enzymes significantly increased in tissue samples obtained from CRC patients and cancer cell lines (HT-29). Inhibition of any of the selected enzymes reduced CRC cell growth in a dose-dependent manner. More importantly, the combination of 5-FU + Avasimibe (an ACAT1 inhibitor) and 5-FU + MF-438 (an SCD1 inhibitor) produced a stronger antiproliferative effect than the inhibitors alone. 5-FU combined either with Avasimibe or MF-438 showed a synergistic effect with an HSA score of 47.00 at a dose of 0.3 + 30 µM, respectively (2.66% viability rate vs. 46%; p < 0.001), and 39.34 at a dose of 0.3 + 0.06 µM (46% vs. 10.33%; p < 0.001), respectively. The association of 5-FU with Lovastatin (HMGCR inhibitor) did not significantly impact CRC cell viability in a synergistic manner. Inhibition of lipid metabolism combined with standard chemotherapy is a promising strategy that reduces CRC cell viability and allows for the use of a lower drug dose. The combination of 5-FU and Avasimibe has the greatest therapeutic potential among studied compounds.
Collapse
Affiliation(s)
- Judyta Zabielska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (J.Z.); (E.S.); (S.S.-J.)
| | - Ewa Stelmanska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (J.Z.); (E.S.); (S.S.-J.)
| | - Sylwia Szrok-Jurga
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (J.Z.); (E.S.); (S.S.-J.)
| | - Jarosław Kobiela
- Department of Surgical Oncology, Transplant Surgery and General Surgery, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland
| |
Collapse
|
35
|
Cheng C, Zha Q, Sun L, Cui T, Guo X, Xing C, Chen Z, Ji C, Liang S, Tao S, Chu J, Wu C, Chu Q, Gu X, Zhang N, Fu Y, Deng S, Zhu Y, Wang J, Liu Y, Liu L. VCP downstream metabolite glycerol-3-phosphate (G3P) inhibits CD8 +T cells function in the HCC microenvironment. Signal Transduct Target Ther 2025; 10:26. [PMID: 39848960 PMCID: PMC11758394 DOI: 10.1038/s41392-024-02120-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
CD8+T cells within the tumor microenvironment (TME) are often functionally impaired, which limits their ability to mount effective anti-tumor responses. However, the molecular mechanisms behind this dysfunction remain incompletely understood. Here, we identified valosin-containing protein (VCP) as a key regulator of CD8+T cells suppression in hepatocellular carcinoma (HCC). Our findings reveal that VCP suppresses the activation, expansion, and cytotoxic capacity of CD8+T cells both in vitro and in vivo, significantly contributing to the immunosuppressive nature of the TME. Mechanistically, VCP stabilizes the expression of glycerol-3-phosphate dehydrogenase 1-like protein (GPD1L), leading to the accumulation of glycerol-3-phosphate (G3P), a downstream metabolite of GPD1L. The accumulated G3P diffuses into the TME and directly interacts with SRC-family tyrosine kinase LCK, a critical component of the T-cell receptor (TCR) signaling pathway in CD8+T cells. This interaction heightens the phosphorylation of Tyr505, a key inhibitory residue, ultimately reducing LCK activity and impairing downstream TCR signaling. Consequently, CD8+T cells lose their functional capacity, diminishing their ability to fight against HCC. Importantly, we demonstrated that targeting VCP in combination with anti-PD1 therapy significantly suppresses HCC tumor growth and restores the anti-tumor function of CD8+T cells, suggesting synergistic therapeutic potential. These findings highlight a previously unrecognized mechanism involving VCP and G3P in suppressing T-cell-mediated immunity in the TME, positioning VCP as a promising upstream target for enhancing immunotherapy in HCC.
Collapse
Affiliation(s)
- Cheng Cheng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Qingrui Zha
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Linmao Sun
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Tianming Cui
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xinyu Guo
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Changjian Xing
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Zhengxiang Chen
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Changyong Ji
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shuhang Liang
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
- Department of Gastrointestinal Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Shengwei Tao
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Junhui Chu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Chenghui Wu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Qi Chu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xuetian Gu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Ning Zhang
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yumin Fu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shumin Deng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yitong Zhu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Yao Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| |
Collapse
|
36
|
Liao Y, Chen J, Yao H, Zheng T, Tu J, Chen W, Guo Z, Zou Y, Wen L, Xie X. Single-cell profiling of SLC family transporters: uncovering the role of SLC7A1 in osteosarcoma. J Transl Med 2025; 23:103. [PMID: 39844299 PMCID: PMC11752724 DOI: 10.1186/s12967-025-06086-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Osteosarcoma is the most common malignant bone tumor in children and adolescents, characterized by high disability and mortality rates. Over the past three decades, therapeutic outcomes have plateaued, underscoring the critical need for innovative therapeutic targets. Solute carrier (SLC) family transporters have been implicated in the malignant progression of a variety of tumors, however, their specific role in osteosarcoma remains poorly understood. METHODS The single-cell sequencing data from GSE152048 and GSE162454, along with RNA-seq from the TARGET and GSE21257 cohorts, were utilized for the analysis in this study. LASSO regression analysis was conducted to identify prognostic genes and construct an SLC-related prognostic signature. Survival analysis and ROC analysis evaluated the validity of the prognostic signature. The ESTIMATE and CIBERSORT Packages were utilized to assess the immune infiltration status. Pseudotime and CellChat analyses were performed to investigate the relationship between SLC7A1, malignant phenotypes, and the immune microenvironment. CCK8 assays, EdU staining, colony formation assays, Transwell assays, and co-culture systems were used to assess the effects of SLC7A1 on cell proliferation, metastasis, and macrophage polarization. Finally, virtual docking identified potential drugs targeting SLC7A1. RESULTS SLCs displayed distinct expression patterns across various cell types within the osteosarcoma microenvironment, with myeloid cells exhibiting a preference for amino acid uptake. A prognostic model comprising nine genes was constructed via LASSO regression, with SLC7A1 showing the highest hazard ratio. Multiple analytical algorithms indicated that SLCs were associated with immune cell infiltration and immune checkpoint gene expression. Single-cell analysis indicated that SLC7A1 was predominantly expressed in osteosarcoma cells and correlated with various malignant tumor characteristics. SLC7A1 also regulate interactions between tumor cells and macrophages, as well as modulate macrophage function through multiple pathways. In vitro assays and survival analysis demonstrated that inhibition of SLC7A1 suppressed the malignant phenotype of osteosarcoma cells, with SLC7A1 expression correlating with poor prognosis. Co-culture models confirmed the involvement of SLC7A1 in macrophage polarization. Finally, virtual screening and CETSA identified Cepharanthine as potential inhibitors of SLC7A1. CONCLUSION SLC-related prognostic signatures can be utilized for the prognostic evaluation of osteosarcoma. Pharmacological inhibition of SLC7A1 may be a feasible therapeutic approach for osteosarcoma.
Collapse
Affiliation(s)
- Yan Liao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Junkai Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Hao Yao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Ting Zheng
- The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, 510620, China
| | - Jian Tu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Weidong Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - ZeHao Guo
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Yutong Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| | - Lili Wen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
37
|
Zeng J, Nie Z, Shang Y, Mai J, Zhang Y, Yang Y, Xu C, Zhao J, Fan Z, Xiao J. CancerSCEM 2.0: an updated data resource of single-cell expression map across various human cancers. Nucleic Acids Res 2025; 53:D1278-D1286. [PMID: 39460627 PMCID: PMC11701606 DOI: 10.1093/nar/gkae954] [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: 09/10/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The field of single-cell RNA sequencing (scRNA-seq) has advanced rapidly in the past decade, generating significant amounts of valuable data for researchers to study complex tumor profiles. This data is crucial for gaining innovative insights into cancer biology. CancerSCEM (https://ngdc.cncb.ac.cn/cancerscem) is a public resource that integrates, analyzes and visualizes scRNA-seq data related to cancer, and it provides invaluable support to numerous cancer-related studies. With CancerSCEM 2.0, scRNA-seq data have increased from 208 to 1466 datasets, covering tumor, matching-normal and peripheral blood samples across 127 research projects and 74 cancer types. The new version of this resource enhances transcriptome analysis by adding copy number variation evaluation, transcription factor enrichment, pseudotime trajectory construction, and diverse biological feature scoring. It also introduces a new cancer metabolic map at the single-cell level, providing an intuitive understanding of metabolic regulation across different cancer types. CancerSCEM 2.0 has a more interactive analysis platform, including four modules and 14 analytical functions, allowing researchers to perform cancer scRNA-seq data analyses in various dimensions. These enhancements are expected to expand the usability of CancerSCEM 2.0 to a broader range of cancer research and clinical applications, potentially revolutionizing our understanding of cancer mechanisms and treatments.
Collapse
Affiliation(s)
- Jingyao Zeng
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhi Nie
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunfei Shang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jialin Mai
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yadong Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuntian Yang
- Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chenle Xu
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Zhao
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuojing Fan
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingfa Xiao
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
38
|
Yan S, Xue P, Sun Y, Bai T, Shao S, Zeng X. Cupric Doping Hollow Prussian Blue Nanoplatform for Enhanced Cholesterol Depletion: a Promising Strategy for Breast Cancer Therapy and Metastasis Inhibition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409967. [PMID: 39606805 PMCID: PMC11744725 DOI: 10.1002/advs.202409967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/19/2024] [Indexed: 11/29/2024]
Abstract
The dysregulated cholesterol metabolism in breast cancer cells drives malignancy, invasion, and metastasis, emphasizing the significance of reducing abnormal cholesterol accumulation for effective cancer treatment and metastasis inhibition. Despite its promise, cholesterol oxidase (ChOx) encounters challenge due to limited catalytic efficiency and susceptibility to harsh conditions. To overcome these hurdles, biocompatible nanoplatforms (Cu-HPB/C) tailored for efficient cholesterol depletion are introduced. Cu2+-doped hollow Prussian blue (Cu-HPB) acts as a carrier, shelter, and enhancer for ChOx, bolstering tumor-targeting ability, stability, and enzymatic activity. Tumor-responsive released Cu2+ notably augments ChOx activity, facilitating cholesterol depletion and disrupting lipid rafts, thereby impeding cell invasion and migration. Additionally, H2O2 generated from the oxidase reaction enhances Cu-HPB's chemo dynamic therapeutic efficacy. Transcriptomic analysis validates Cu-HPB/C's impact on cholesterol homeostasis and reveals cell death mechanisms including oxidative stress, ferroptosis, cuproptosis, and apoptosis. Demonstrating therapeutic efficacy in both 4T1 tumor subcutaneous and metastasis mouse models, the study presents a direct and effective strategy for tumor therapy and metastasis inhibition through enhanced cholesterol depletion.
Collapse
Affiliation(s)
- Shuangqian Yan
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province UniversityBiomedical Research Center of South ChinaCollege of Life SciencesFujian Normal University1 Keji RoadFuzhou350117P. R. China
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies)Straits Laboratory of Flexible Electronics (SLoFE) Fujian Normal UniversityFuzhouFujian350117P. R. China
| | - Panpan Xue
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies)Straits Laboratory of Flexible Electronics (SLoFE) Fujian Normal UniversityFuzhouFujian350117P. R. China
| | - Ying Sun
- Department of GastroenterologyFuzhou No. 1 Hospital Affiliated with Fujian Medical UniversityFuzhouFujian350009P. R. China
| | - Tingjie Bai
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province UniversityBiomedical Research Center of South ChinaCollege of Life SciencesFujian Normal University1 Keji RoadFuzhou350117P. R. China
| | - Sijie Shao
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies)Straits Laboratory of Flexible Electronics (SLoFE) Fujian Normal UniversityFuzhouFujian350117P. R. China
| | - Xuemei Zeng
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province UniversityBiomedical Research Center of South ChinaCollege of Life SciencesFujian Normal University1 Keji RoadFuzhou350117P. R. China
| |
Collapse
|
39
|
Wang C, Ma X. The role of acetylation and deacetylation in cancer metabolism. Clin Transl Med 2025; 15:e70145. [PMID: 39778006 PMCID: PMC11706801 DOI: 10.1002/ctm2.70145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
As a hallmark of cancer, metabolic reprogramming adjusts macromolecular synthesis, energy metabolism and redox homeostasis processes to adapt to and promote the complex biological processes of abnormal growth and proliferation. The complexity of metabolic reprogramming lies in its precise regulation by multiple levels and factors, including the interplay of multiple signalling pathways, precise regulation of transcription factors and dynamic adjustments in metabolic enzyme activity. In this complex regulatory network, acetylation and deacetylation, which are important post-translational modifications, regulate key molecules and processes related to metabolic reprogramming by affecting protein function and stability. Dysregulation of acetylation and deacetylation may alter cancer cell metabolic patterns by affecting signalling pathways, transcription factors and metabolic enzyme activity related to metabolic reprogramming, increasing the susceptibility to rapid proliferation and survival. In this review, we focus on discussing how acetylation and deacetylation regulate cancer metabolism, thereby highlighting the central role of these post-translational modifications in metabolic reprogramming, and hoping to provide strong support for the development of novel cancer treatment strategies. KEY POINTS: Protein acetylation and deacetylation are key regulators of metabolic reprogramming in tumour cells. These modifications influence signalling pathways critical for tumour metabolism. They modulate the activity of transcription factors that drive gene expression changes. Metabolic enzymes are also affected, altering cellular metabolism to support tumour growth.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyang CityLiaoning ProvinceChina
- Key Laboratory of Gynecological Oncology of Liaoning ProvinceDepartment of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaoxin Ma
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyang CityLiaoning ProvinceChina
- Key Laboratory of Gynecological Oncology of Liaoning ProvinceDepartment of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
40
|
Sharma NK, Sarode SC, Sekar G, Sonawane K, Bomle D. Challenges in Metabolite Biomarkers as Avenues of Diagnosis and Prognosis of Cancer. J Cancer Prev 2024; 29:105-112. [PMID: 39790219 PMCID: PMC11706722 DOI: 10.15430/jcp.24.015] [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: 08/03/2024] [Revised: 09/09/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025] Open
Abstract
Given the evolutionary nature of tumor complexities and heterogeneity, the early diagnosis of cancer encounters various challenges. Complexities at the level of metabolite reprogramming are compelling in the background of invasiveness, metastasis, drug- and radiation-induced metabolic alterations, immunotherapy-influenced changes, and pro-tumor niche including microbiome. Therefore, it is crucial to examine both current and future obstacles associated with early cancer detection specifically in the context of tumor metabolite biomarkers at preclinical and clinical levels. In conclusion, the significance of tumor metabolite biomarkers must be aligned with a comprehensive approach to achbieve diagnosis and prognosis of cancer patients by securing solutions to formidable challenges.
Collapse
Affiliation(s)
- Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| | - Sachin C. Sarode
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, India
| | - Gopinath Sekar
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur, India
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| | - Dhanashree Bomle
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| |
Collapse
|
41
|
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
|
42
|
Wang J, Zhao F, Zhang Q, Sun Z, Xiahou Z, Wang C, Liu Y, Yu Z. Unveiling the NEFH+ malignant cell subtype: Insights from single-cell RNA sequencing in prostate cancer progression and tumor microenvironment interactions. Front Immunol 2024; 15:1517679. [PMID: 39759507 PMCID: PMC11695424 DOI: 10.3389/fimmu.2024.1517679] [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: 10/26/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background Prostate cancer (PCa) is a multifactorial and heterogeneous disease, ranking among the most prevalent malignancies in men. In 2020, there were 1,414,259 new cases of PCa worldwide, accounting for 7.3% of all malignant tumors. The incidence rate of PCa ranks third, following breast cancer and lung cancer. Patients diagnosed with high-grade PCa frequently present with existing or developing metastases, complicating their treatment and resulting in poorer prognoses, particularly for those with bone metastases. Utilizing single-cell RNA sequencing (scRNA-seq), we identified specific malignant cell subtypes that are closely linked to high-grade PCa. By investigating the mechanisms that govern interactions within the tumor microenvironment (TME), we aim to offer new theoretical insights that can enhance the prevention, diagnosis, and treatment of PCa, ultimately striving to improve patient outcomes and quality of life. Methods Data on scRNA-seq was obtained from the GEO database. The gene ontology and gene set enrichment analysis were employed to analyze differential expression genes. Using inferCNV analysis to identify malignant epithelial cells. We subsequently employed Monocle, Cytotrace, and Slingshot packages to infer subtype differentiation trajectories. The cellular communication between malignant cell subtypes and other cells was predicted using the CellChat package. Furthermore, we employed pySCENIC to analyze and identify the regulatory networks of transcription factors (TFs) in malignant cell subtypes. The MDA PCa 2b and VCap cell lines were employed to validate the analysis results through cellular functional experiments. In addition, a risk scoring model was developed to assess the variation in clinical characteristics, prognosis, immune infiltration, immune checkpoint, and drug sensitivity. Results A malignant cell subtype in PCa with high expression of NEFH was identified through scRNA-seq analysis. This subtype was situated at the differentiation terminal, exhibited a higher level of malignancy, and exhibited characteristics that were more prone to advanced tumor lesions. In addition, our research underscored the intricate interactions that exist within the TME, particularly the interaction between PTN secreted by this subtype and fibroblasts via the NCL receptor. This interaction may be closely associated with cancer-associated fibroblasts and tumor progression. Subsequently, we determined that the NEFH+ malignant cell subtype was significantly correlated with the TF IRX4. This TF is linked to a worse prognosis in PCa and may affect disease progression by regulating gene transcription. Our conclusions were additionally verified through cellular experiments. Furthermore, the prognostic model we developed demonstrated satisfactory predictive performance, with gene sets from the high NmRS group facilitating tumor progression and deterioration. The analysis of immune infiltration was instrumental in the development of clinical intervention strategies and patient prognosis. Conclusion By examining the cellular heterogeneity of a unique NEFH+ malignant cell subtype within the PCa microenvironment, we were able to disclose their reciprocal interaction with disease progression. This offers a novel viewpoint on the diagnosis and treatment of PCa.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, The Second People’s Hospital of Meishan City, Meishan, Sichuan, China
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Fu Zhao
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiang Zhang
- Department of Urology, The Second People’s Hospital of Meishan City, Meishan, Sichuan, China
| | - Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Changzhong Wang
- Department of Urology, The First People’s Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Yan Liu
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Zongze Yu
- Department of Urology, The Second People’s Hospital of Meishan City, Meishan, Sichuan, China
| |
Collapse
|
43
|
Shen Y, Qiu A, Huang X, Wen X, Shehzadi S, He Y, Hu Q, Zhang J, Luo D, Yang S. AKR1B10 and digestive tumors development: a review. Front Immunol 2024; 15:1462174. [PMID: 39737179 PMCID: PMC11682995 DOI: 10.3389/fimmu.2024.1462174] [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: 07/09/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Aldo-keto reductase family 1 member B10 (AKR1B10) is a member of the AKR1B subfamily. It is mainly found in cytoplasm, and it is typically expressed in the stomach and intestines. Given that its expression is low or absent in other tissues, AKR1B10 is a potential diagnostic and therapeutic biomarker for various digestive system diseases. Here, we review recent research progress on AKR1B10 in digestive system tumors such as hepatocellular carcinoma, gastric carcinoma, colorectal carcinoma, pancreatic carcinoma, oral squamous cell carcinoma, laryngeal squamous cell carcinoma, cholangiocarcinoma, and nasopharyngeal carcinoma, over the last 5 years. We also discuss the current trends and future research directions for AKR1B10 in both oncological and non-oncological diseases to provide a scientific reference for further exploration of this gene.
Collapse
Affiliation(s)
- Yao Shen
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ailin Qiu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin Huang
- Laboratory Medicine Center, Shenzhen Luohu Hospital Group, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
- First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaosha Wen
- Laboratory Medicine Center, Shenzhen Luohu Hospital Group, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
| | - Sundar Shehzadi
- Laboratory Medicine Center, Shenzhen Luohu Hospital Group, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
| | - Yan He
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qian Hu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jian Zhang
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Dixian Luo
- Laboratory Medicine Center, Shenzhen Luohu Hospital Group, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
| | - Shenghui Yang
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Preventive Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
44
|
Wu R, Zhu H, He Q, Yuan T, Yang B. Metabolic reprogramming in KRAS-mutant cancers: Proven targetable vulnerabilities and potential therapeutic strategies. Drug Discov Today 2024; 29:104220. [PMID: 39481592 DOI: 10.1016/j.drudis.2024.104220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Kras (Ki-ras2 Kirsten rat sarcoma viral oncogene homolog), one of the most frequently mutated oncogenes in the human genome, is considered 'untargetable'. Although specific KRASG12C inhibitors have been developed, their overall impact is limited, highlighting the need for further research on targeting KRAS-mutant cancers. Metabolic abnormalities are key hallmarks of cancer, with KRAS-driven tumors exhibiting traits like glycolysis upregulation, glutamine addiction, lipid droplet accumulation, highly active macropinocytosis, and metabolic reprogramming-associated tumor microenvironment remodeling. Targeting these unique metabolic characteristics offers a promising strategy for new cancer treatments. This review summarizes recent advances in our understanding of the metabolic network in KRAS-mutated tumor cells, discusses potential targetable vulnerabilities, and outlines clinical developments in relevant therapies, while also addressing challenges to improve strategies against these aggressive cancers.
Collapse
Affiliation(s)
- Ruilin Wu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China.
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
45
|
Dai Y, Li J, Wang T, Zhang X, Du P, Dong Y, Jiao Z. Self-assembled metal-polyphenolic based multifunctional nanomedicine to improve therapy treatment of pancreatic cancer by inhibition of glutamine metabolism. Colloids Surf B Biointerfaces 2024; 244:114162. [PMID: 39178515 DOI: 10.1016/j.colsurfb.2024.114162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024]
Abstract
Cancer poses a significant threat to human health and life. Chemotherapy, immunotherapy and chemodynamic therapy (CDT) are effective treatments for cancer. However, the presence of metabolic reprogramming via glutamine in tumor cells limits their therapeutic effectiveness. Herein, we propose an effective assembly strategy to synthesize a novel metal-polyphenolic based multifunctional nanomedicine (Fe-DBEF) containing Pluronic F127 stable ferric ion crosslinked epigallocatechin gallate (EGCG) nanoparticles loaded with GLS1 inhibitor bis-2-(5-phenylacetamino-1,3,4-thiadiazole-2-yl) ethyl sulfide (BPTES) and chemotherapy drug doxorubicin (DOX). Our study demonstrates that Fe-DBEF nanomedicine exhibits high efficiency anti-proliferation properties in pancreatic cancer through a combination of in vitro cell experiments, human organoid experiments and KPC animal experiments. Notably, Fe-DBEF nanomedicine can reduce the production of glutathione (GSH) in tumor cells, thereby reducing their resistance to ROS therapy. Additionally, excessive ROS production also aggravates DNA damage caused by DOX, synergistically sensitizing chemotherapy and promoting apoptosis for efficient treatment of pancreatic cancer. Overall, our findings suggest that inhibiting glutamine metabolism to increase the sensitivity of chemotherapy/CDT using metal-polyphenolic based multifunctional nanomedicine provides a promising combination of multiple therapeutic means for treating pancreatic cancer.
Collapse
Affiliation(s)
- Yiwei Dai
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jieru Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Tao Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xinyu Zhang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Pengcheng Du
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Yuman Dong
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China; Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou, China.
| | - Zuoyi Jiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical School, Lanzhou University, Lanzhou, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China; Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
46
|
Cordani M, Michetti F, Zarrabi A, Zarepour A, Rumio C, Strippoli R, Marcucci F. The role of glycolysis in tumorigenesis: From biological aspects to therapeutic opportunities. Neoplasia 2024; 58:101076. [PMID: 39476482 PMCID: PMC11555605 DOI: 10.1016/j.neo.2024.101076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 11/11/2024]
Abstract
Glycolytic metabolism generates energy and intermediates for biomass production. Tumor-associated glycolysis is upregulated compared to normal tissues in response to tumor cell-autonomous or non-autonomous stimuli. The consequences of this upregulation are twofold. First, the metabolic effects of glycolysis become predominant over those mediated by oxidative metabolism. Second, overexpressed components of the glycolytic pathway (i.e. enzymes or metabolites) acquire new functions unrelated to their metabolic effects and which are referred to as "moonlighting" functions. These functions include induction of mutations and other tumor-initiating events, effects on cancer stem cells, induction of increased expression and/or activity of oncoproteins, epigenetic and transcriptional modifications, bypassing of senescence and induction of proliferation, promotion of DNA damage repair and prevention of DNA damage, antiapoptotic effects, inhibition of drug influx or increase of drug efflux. Upregulated metabolic functions and acquisition of new, non-metabolic functions lead to biological effects that support tumorigenesis: promotion of tumor initiation, stimulation of tumor cell proliferation and primary tumor growth, induction of epithelial-mesenchymal transition, autophagy and metastasis, immunosuppressive effects, induction of drug resistance and effects on tumor accessory cells. These effects have negative consequences on the prognosis of tumor patients. On these grounds, it does not come to surprise that tumor-associated glycolysis has become a target of interest in antitumor drug discovery. So far, however, clinical results with glycolysis inhibitors have fallen short of expectations. In this review we propose approaches that may allow to bypass some of the difficulties that have been encountered so far with the therapeutic use of glycolysis inhibitors.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Federica Michetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, Milan 20134, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy.
| | - Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, Milan 20134, Italy.
| |
Collapse
|
47
|
Jia X, Wang Y, Qiao Y, Jiang X, Li J. Nanomaterial-based regulation of redox metabolism for enhancing cancer therapy. Chem Soc Rev 2024; 53:11590-11656. [PMID: 39431683 DOI: 10.1039/d4cs00404c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Altered redox metabolism is one of the hallmarks of tumor cells, which not only contributes to tumor proliferation, metastasis, and immune evasion, but also has great relevance to therapeutic resistance. Therefore, regulation of redox metabolism of tumor cells has been proposed as an attractive therapeutic strategy to inhibit tumor growth and reverse therapeutic resistance. In this respect, nanomedicines have exhibited significant therapeutic advantages as intensively reported in recent studies. In this review, we would like to summarize the latest advances in nanomaterial-assisted strategies for redox metabolic regulation therapy, with a focus on the regulation of redox metabolism-related metabolite levels, enzyme activity, and signaling pathways. In the end, future expectations and challenges of such emerging strategies have been discussed, hoping to enlighten and promote their further development for meeting the various demands of advanced cancer therapies. It is highly expected that these therapeutic strategies based on redox metabolism regulation will play a more important role in the field of nanomedicine.
Collapse
Affiliation(s)
- Xiaodan Jia
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Wang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Qiao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiue Jiang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Jinghong Li
- Beijing Institute of Life Science and Technology, Beijing 102206, P. R. China
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
48
|
Su RY, Xu CH, Guo HJ, Meng LJ, Zhuo JY, Xu N, Li HG, He CY, Zhang XY, Lian ZX, Wang S, Cao C, Zhou R, Lu D, Zheng SS, Wei XY, Xu X. Oncogenic cholesterol rewires lipid metabolism in hepatocellular carcinoma via the CSNK2A1-IGF2R Ser2484 axis. J Adv Res 2024:S2090-1232(24)00540-X. [PMID: 39547439 DOI: 10.1016/j.jare.2024.11.021] [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: 06/02/2024] [Revised: 10/02/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Abstract
INTRODUCTION Alcohol consumption and hepatitis B virus (HBV) infection are common risk factors for hepatocellular carcinoma (HCC). However, few studies have focused on elucidating the mechanisms of HCC with combined alcohol and HBV etiology. OBJECTIVES We aimed to investigate the molecular features of alcohol and HBV on HCC and to seek out potential therapeutic strategies. METHODS Two independent cohorts of HCC patients (n = 539 and n = 140) were included to investigate HCC with synergetic alcohol and HBV (AB-HCC) background. Patient-derived cell lines, organoids, and xenografts were used to validate the metabolic fragile. High-throughput drug screening (1181 FDA-approved anticancer drugs) was leveraged to explore the potential therapeutic agents. RESULTS Here, we delineated AB-HCC as a distinctive metabolic subtype, hallmarked by oncogenic cholesterol, through the integration of clinical cohorts, proteomics, phosphoproteomics, and spatial transcriptome. Mechanistically, our findings revealed that cholesterol directly binds to CSNK2A1 (Casein Kinase 2 Alpha 1), augmenting its kinase activity and leading to phosphorylation of IGF2R (Insulin-Like Growth Factor 2 Receptor) at Ser2484. This cascade rewires lipid-driven mitochondrial oxidative phosphorylation, spawns reactive oxygen species measured by malondialdehyde assay, and perpetuates a positive feedback loop for cholesterol biosynthesis, ultimately culminating in tumorigenesis. Initial transcriptional activation of CSNK2A1 is driven by upregulation of RAD21 in AB-HCC. Our cholesterol profiling exposes AB-HCC's compensatory mechanism of AB-HCC, which capitalizes on both uptake and biosynthesis of cholesterol to confer survival edge. Moreover, high-throughput drug screening coupled with in vivo validation has uncovered the susceptibilities of AB-HCC, which can be effectively addressed by a combination of dietary cholesterol restriction and oral administration of Fostamatinib. The CSNK2A1-mediated cholesterol biosynthesis pathway has been implicated in various cancers characterized by cholesterol metabolism. CONCLUSION These findings not only pinpoint the oncogenic metabolite cholesterol as a hidden culprit in AB-HCC subtype, but also enlighten a novel combination strategy to rejuvenate tumor metabolism.
Collapse
Affiliation(s)
- Ren-Yi Su
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Chen-Hao Xu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hai-Jun Guo
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou 310006, China
| | - Li-Jun Meng
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Jian-Yong Zhuo
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou 310006, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Nan Xu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hui-Gang Li
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Chi-Yu He
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xuan-Yu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
| | - Zheng-Xin Lian
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou 310006, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Chenhao Cao
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, and College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Di Lu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310059, China.
| | - Shu-Sen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China; Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310022, China.
| | - Xu-Yong Wei
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou 310006, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China.
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310059, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China.
| |
Collapse
|
49
|
Wang C, Zhang R, Zhang H, Gao H, Zhu Y, Jiao L, Yi Z, Zhou M, Li X. Lipid metabolism-related gene signature predicts prognosis and unveils novel anti-tumor drugs in specific type of diffuse large B cell lymphoma. Mol Med 2024; 30:210. [PMID: 39538125 PMCID: PMC11559131 DOI: 10.1186/s10020-024-00988-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is the most common type of lymphoma which possess highly aggressive and heterogeneous. Despite advances in understanding heterogeneity and development of novel targeted agents, the prognosis of DLBCL patients remains unsatisfied. Lipids are crucial components of biological membranes and signal transduction while accumulating evidence has supported the vital roles of abnormal lipid metabolism in tumorigenesis. Furthermore, some related pathways could serve as prognostic biomarkers and potential therapeutic targets. However, the clinical significance of abnormal lipid metabolism reprogramming in DLBCL has not been investigated. In the current study, we developed a prognostic risk model for DLBCL based on the abnormal expressed lipid metabolism genes and moreover based on our risk model we classified patients with DLBCL into novel subtypes and identified potential drugs for DLBCL patients with certain lipid metabolism profiles. METHODS We utilized univariate Cox regression analysis to identify the prognosis-related lipid metabolism genes, and then performed LASSO Cox regression to identify prognostic related lipid metabolism related genes. Multivariate cox regression was used to establish the prognostic model. Patients were divided in to high and low risk groups based on the median risk score. Immune cell infiltration and GSEA were used to identify the pathways between high and low risk groups. Oncopredict algorithm was utilized to identify potential drug for high-risk patients. In vitro cell apoptosis and viability analysis were employed to verify the specific tumor inhibition effects of AZD5153. RESULTS Nineteen survival related lipid metabolism genes TMEM176B, LAYN, RAB6B, MMP9, ATAD3B, SLC2A11, CD3E, SLIT2, SLC2A13, SLC43A3, CD6, SIRPG, NEK6, LCP2, CTTN, CXCL2, SNX22, BCL6 and FABP4 were identified and subjected to build the prognostic model which was further verified in four external microarray cohorts and one RNA seq cohorts. Tumor immune microenvironment analysis and GSEA results showed that the activation of MYC targets genes rather than immunosuppression contribute to the poor survival outcome of patients in the high-risk group. AZD5153, a novel bivalent BET bromodomain inhibitor which could inhibit the transcription of MYC and E2F exhibited specific antitumor function for cells with high-risk score. CONCLUSIONS Our results provide the first lipid metabolism-based gene signature for predicting the survival of patients with DLBCL. Furthermore, by determining novel subtypes with our lipid metabolism prognostic model we illustrated that drugs that compromising MYC target genes rather than immune checkpoint inhibitors may be beneficial to DLBCL patients with certain lipid metabolism profiles.
Collapse
Affiliation(s)
- Cancan Wang
- Department of Pathology, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Ran Zhang
- Department of Pathology, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Huan Zhang
- Department of Pathology, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Haixia Gao
- Department of Pathology, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Yubing Zhu
- Department of Pathology, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Lichao Jiao
- Department of Pathology, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Zhiqiang Yi
- Department of Pathology, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Meiyu Zhou
- Chongqing University Fuling Hospital, Chongqing, China.
| | - Xinxia Li
- Department of Pathology, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China.
| |
Collapse
|
50
|
Feng Y, Ni Q, Wu N, Xie T, Yun F, Zhang X, Gao L, Gai Y, Li E, Yi X, Xie J, Zhang Q, Yang Z, Sai B, Kuang Y, Zhu Y. Molecular mechanisms of MAZ targeting up-regulation of NDUFS3 expression to promote malignant progression in melanoma. Commun Biol 2024; 7:1491. [PMID: 39532991 PMCID: PMC11557950 DOI: 10.1038/s42003-024-07209-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Myc-associated Zinc-finger Protein (MAZ) has been implicated in the malignant progression of various tumors. However, its expression and functional relationship of MAZ in melanoma have not been previously investigated. This study confirms elevated expression of MAZ in melanoma, correlating with poor patient prognosis. Furthermore, our findings demonstrate that MAZ enhances melanoma progression by promoting proliferation, migration and invasion. It is worth noting that we found that MAZ can target and regulate the transcription of NADH dehydrogenase [ubiquinone] iron-sulfur protein 3 (NDUFS3), a core subunit of mitochondrial complex I, to enhance mitochondrial metabolism and thus promote malignant progression of melanoma. Predictive modeling indicates that the co-expression of MAZ and NDUFS3 could serve as a potential prognostic marker for melanoma patients.
Collapse
Affiliation(s)
- Yu Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Qinxuan Ni
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Na Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Taiyu Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Fang Yun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Xuedan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Lingnan Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Yanlong Gai
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Enjiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Xiaojia Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Junlin Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Qiao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Zhe Yang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Buqing Sai
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Yingmin Kuang
- Department of Organ Transplantation, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Yuechun Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, China.
| |
Collapse
|