1
|
Lu L, Tao R. Prognostic implications of glucose metabolism pathways in colon adenocarcinoma: a comprehensive outlook on the molecular landscape and immunotherapy. Biochem Biophys Res Commun 2025; 768:151961. [PMID: 40345006 DOI: 10.1016/j.bbrc.2025.151961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/30/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Colon adenocarcinoma (COAD) is a common and aggressive cancer characterized by significant metabolic alterations, particularly in glucose metabolism. Identifying key genes and pathways involved in glucose metabolism could provide valuable prognostic biomarkers and therapeutic targets. METHODS Clinical and transcriptomic data for patients with COAD were obtained from TCGA and validated using external datasets (GSE17536 and GSE39582). Seventeen glucose metabolism-related pathways were selected from the MSigDB and analysed using ssGSEA. WGCNA was used to identify key gene modules. Prognostic genes were selected via univariate Cox regression, Lasso-Cox regression, and multivariate Cox regression. Model validation was conducted using independent datasets. Immunotherapy prediction and immune infiltration analyses were also performed. A-NEK9 knockdown cell line was established using SW1116 and SW480 cell lines. The effect of NEK9 on COAD was evaluated in vivo and in vitro. The effects of NEK9 on glucose uptake and lactate production were also assessed. RESULTS A prognostic model based on five glucose metabolism-related genes (NEK9, HS2ST1, AC016394.3, H2BC21, and MIR23A) was developed. The model demonstrated strong predictive value, with high-risk patients showing poorer survival outcomes in both the TCGA and external validation cohorts. Additionally, lower risk scores were associated with better responses to immunotherapy, as indicated by TIDE and SubMap analyses. These findings were further validated through ROC analysis, which revealed robust predictive performance for immunotherapy response across multiple cohorts. NEK9 promoted the proliferation and tumour angiogenesis of SW1116 and SW480 cells, inhibited apoptosis, and enhanced glucose uptake and lactate production in tumour cells. NEK9 knockdown significantly inhibited the tumorigenic ability of COAD in mice. CONCLUSIONS This study highlights the role of glucose metabolism in COAD and presents a novel prognostic model based on glucose metabolism-related genes. The model has potential clinical applications for predicting survival and guiding immunotherapy decisions in patients with COAD.
Collapse
Affiliation(s)
- Ling Lu
- Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Center for Rehabilitation Medicine Center, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Ran Tao
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
2
|
Kim Y, Nickles TM, Lee PM, Bok RA, Gordon JW, Larson PEZ, Vigneron DB, von Morze C, Ohliger MA. A data-driven approach for improved quantification of in vivo metabolic conversion rates of hyperpolarized [1- 13C]pyruvate. Magn Reson Med 2025; 93:2346-2356. [PMID: 39963732 PMCID: PMC11971488 DOI: 10.1002/mrm.30445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/01/2024] [Accepted: 01/13/2025] [Indexed: 04/06/2025]
Abstract
PURPOSE Accurate quantification of metabolism in hyperpolarized (HP) 13C MRI is essential for clinical applications. However, kinetic model parameters are often confounded by uncertainties in radiofrequency flip angles and other model parameters. METHODS A data-driven kinetic fitting approach for HP 13C-pyruvate MRI was proposed that compensates for uncertainties in the B1 + field. We hypothesized that introducing a scaling factor to the flip angle to minimize fit residuals would allow more accurate determination of the pyruvate-to-lactate conversion rate (kPL). Numerical simulations were performed under different conditions (flip angle, kPL, and T1 relaxation), with further testing using HP 13C-pyruvate MRI of rat liver and kidneys. RESULTS Simulations showed that the proposed method reduced kPL error from 60% to 1% when the prescribed and actual flip angles differed by 60%. The method also showed robustness to T1 uncertainties, achieving median kPL errors within ±3% even when the assumed T1 was incorrect by up to a factor of 2. In rat studies, better-quality fitting for lactate signals (a 1.4-fold decrease in root mean square error [RMSE] for lactate fit) and tighter kPL distributions (an average of 3.1-fold decrease in kPL standard deviation) were achieved using the proposed method compared with when no correction was applied. CONCLUSION The proposed data-driven kinetic fitting approach provided a method to accurately quantify HP 13C-pyruvate metabolism in the presence of B1 + inhomogeneity. This model may also be used to correct for other error sources, such as T1 relaxation and flow, and may prove to be clinically valuable in improving tumor staging or assessing treatment response.
Collapse
Affiliation(s)
- Yaewon Kim
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Tanner M. Nickles
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Philip M. Lee
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Robert A. Bok
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Jeremy W. Gordon
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Peder E. Z. Larson
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Neurological SurgeryUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Daniel B. Vigneron
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Neurological SurgeryUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Cornelius von Morze
- Mallinckrodt Institute of RadiologyWashington UniversitySt. LouisMissouriUSA
| | - Michael A. Ohliger
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| |
Collapse
|
3
|
Hu C, Chen L, Ding Y, Ye M, Tang Q. Metabolic changes in neuroendocrine neoplasms. Cell Mol Life Sci 2025; 82:205. [PMID: 40377669 DOI: 10.1007/s00018-025-05656-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/05/2025] [Accepted: 03/11/2025] [Indexed: 05/18/2025]
Abstract
Neuroendocrine neoplasms (NENs) are a group of highly heterogeneous neoplasms originating from neuroendocrine cells with a gradually increased incidence. Metabolic change is one of the recognized markers of tumor progression, which has been extensively and systematically studied in other malignant tumors. However, metabolic change in NENs has been relatively poorly studied, and systematic reviews are lacking. We reviewed the relationship between metabolic changes and NENs from the aspects of glucose metabolism, lipid metabolism, metabolic syndrome, amino acid metabolism and metabolomics, and discussed the potential therapeutic strategies of metabolic changes for NENs.
Collapse
Affiliation(s)
- Chunhua Hu
- Shanghai Key Laboratory of Gut Microecology and Associated Major Diseases Research, Digestive Disease Research and Clinical Translation Center, Department of Gastroenterology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Lingyi Chen
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yi Ding
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Mujie Ye
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Qiyun Tang
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Lin S, Yan R, Zhu J, Li B, Zhong Y, Han S, Wang H, Wu J, Chen Z, Jiang Y, Pan A, Huang X, Chen X, Zhu P, Cao S, Liang W, Ye P, Gao Y. Rapid and Noninvasive Early Detection of Lung Cancer by Integration of Machine Learning and Salivary Metabolic Fingerprints Using MS LOC Platform: A Large-Scale Multicenter Study. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416719. [PMID: 40365752 DOI: 10.1002/advs.202416719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/09/2025] [Indexed: 05/15/2025]
Abstract
Most lung cancer (LC) patients are diagnosed at advanced stages due to the lack of effective screening tools. This multicenter study analyzes 1043 saliva samples (334 LC cases and 709 non-LC cases) using a novel high-throughput platform for metabolic fingerprint acquisition. Machine learning identifies 35 metabolic features distinguishing LC from non-LC subjects, enabling the development of a classification model named SalivaMLD. In the validation set and test set, SalivaMLD demonstrates strong diagnostic performance, achieving an area under the curve of 0.849-0.850, a sensitivity of 81.69-83.33%, and a specificity of 74.23-74.39%, outperforming conventional tumor biomarkers. Notably, SalivaMLD exhibits superior accuracy in distinguishing early stage LC patients. Hence, this rapid and noninvasive screening method may be widely applied in clinical practice for LC detection.
Collapse
Affiliation(s)
- Shuang Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Runlan Yan
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Junqi Zhu
- Respiratory and Critical Care Medicine Department, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Bei Li
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Yinyan Zhong
- Pengbu Subdistrict Community Healthcare Center, Shangcheng District, Hangzhou, 310000, China
| | - Shuang Han
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Huiting Wang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Jianmin Wu
- Lab of Nanomedicine and Omic-based Diagnosis, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Zhao Chen
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Yuyue Jiang
- Respiratory and Critical Care Medicine Department, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Aiwu Pan
- Department of Internal Medicine, the Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xuqing Huang
- Respiratory and Critical Care Medicine Department, Affiliated Hospital of Hangzhou Normal University, Hangzhou, 310000, China
| | - Xiaoming Chen
- Well-healthcare Technologies, Co., Ltd., Hangzhou, 310012, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Pingya Zhu
- Well-healthcare Technologies, Co., Ltd., Hangzhou, 310012, China
| | - Sheng Cao
- Well-healthcare Technologies, Co., Ltd., Hangzhou, 310012, China
| | - Wenhua Liang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Peng Ye
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Yue Gao
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| |
Collapse
|
5
|
Dong H, Zhang C, Wang H, Dai Y. Causal links between plasma lipidome and ovarian cancer risk: evidence from Mendelian randomization. Discov Oncol 2025; 16:745. [PMID: 40355763 PMCID: PMC12069180 DOI: 10.1007/s12672-025-02541-z] [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: 01/22/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
S. Plasma lipids in circulation are integral to the physiopathological processes of the ovary and may impact the development of various ovarian conditions, including ovarian cancer (OC). This study utilized a two-sample Mendelian randomization method to examine the causal link between changes in 179 plasma lipid groups and ovarian cancer (OC) to gain deeper insights into this association. We used the inverse variance weighted (IVW) method as the main tool for analysis. We utilized statistical data from plasma lipidomics involving 7,174 Finnish individuals and OC data from the FinnGen consortium, including 2,339 European OC patients and 222,078 European healthy controls. Our analysis revealed that elevated levels of four plasma lipids-Phosphatidylcholine (14:0_16:0, O-18:2_18:2, 16:0_20:4)-are linked to an increased risk of OC, while Sphingomyelin (d34:2) seems to act as a protective factor(all P < 0.05). We also conducted tests for heterogeneity and pleiotropy in the MR results. Additionally, reverse MR analysis indicated that OC does not affect plasma levels of these lipids. To determine whether the observed significant plasma lipids influence OC through common risk factors, we selected BMI as a confounder for multivariable Mendelian randomization (MVMR) analysis. The results showed that Sphingomyelin (d34:2) levels remained significantly associated with OC even after including BMI as an exposure factor. Furthermore, we investigated whether these four lipids mediated the effect of BMI on OC but found no evidence supporting their mediating role. In summary, our findings confirm a causal link between certain plasma lipid species and OC, providing fresh perspectives for risk evaluation and potential therapeutic strategies.
Collapse
Affiliation(s)
- Huke Dong
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chen Zhang
- Lu'an Hospital of Traditional Chinese Medicine, Lu'an, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Ying Dai
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
6
|
Jiang S, Li W, Zhang Y, Lin J, Huang P. Oxygen-regulated enzymatic nanoplatform for synchronous intervention in glycolysis and oxidative phosphorylation to augment antitumor therapy. J Control Release 2025; 381:113594. [PMID: 40024344 DOI: 10.1016/j.jconrel.2025.113594] [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: 12/21/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Tumor cells typically undergo metabolic reprogramming to obtain substantial energy via glycolysis and oxidative phosphorylation (OXPHOS). Intervening in this reprogramming is expected to have therapeutic effects, but simultaneous intervention in these two metabolic pathways is challenging. Herein, we developed an "open-source and throttling" oxygen (O₂) modulation strategy by which we can simultaneously intervene in these two metabolic pathways. Our O₂ modulation nanoplatform (denoted as OAGO) is fabricated via the self-assembly of glucose oxidase (GOx) and oligomycin A (OA) and is coated with bacterial outer membrane vesicles (OMVs). OAGO elicits simultaneous GOx-mediated inhibition of glycolysis and OA-induced inhibition of OXPHOS. The resulting production of GOx-catalyzed hydrogen peroxide leads to oxidative stress, which exacerbates the inhibition of mitochondrial function. Meanwhile, OA reduces intratumoral O₂ consumption (i.e., the "throttling" strategy), and OMVs increase the tumor blood O₂ level (i.e., the "open-source" strategy). This results in an increase in O₂ levels for GOx catalysis, thereby exacerbating energy consumption. In addition, OMVs increase intratumoral OAGO accumulation and enable photothermal therapy in the 4T1 mouse model, which also raises the tumor blood O₂ level and benefits GOx catalysis. This synchronous intervention in two metabolic pathways alongside O₂ modulation constitutes a promising approach for efficient antitumor therapy.
Collapse
Affiliation(s)
- Shanshan Jiang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China; Nanophotonics Research Center, Shenzhen Key Laboratory of Micro-Scale Optical Information Technology, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China
| | - Wanyu Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Yifan Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China.
| |
Collapse
|
7
|
Wang JD, Wang JX, Lin ZL, Xu N, Zhang L, Liu JJ, Gao R, Long ZJ. GPX1 confers resistance to metabolic stress in BCR/ABL-T315I mutant chronic myeloid leukemia cells. Cell Death Discov 2025; 11:229. [PMID: 40346054 PMCID: PMC12064725 DOI: 10.1038/s41420-025-02502-z] [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: 09/18/2024] [Revised: 03/26/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
Chronic myeloid leukemia (CML) harboring BCR/ABL-T315I mutation has been a challenging obstacle for targeted therapy due to the acquired resistance to tyrosine kinase inhibitor (TKI)-based therapy. Thus, it is especially urgent to investigate more effective therapeutic targets to overcome T315I-induced resistance. Here, we reported that BCR/ABL-T315I mutant CML cells possessed a long-term proliferative capacity and tolerance to metabolic stress. Importantly, we also found that selenoamino acid metabolism was increased in the bone marrows of BCR/ABL-T315I patients compared with non-T315I patients by GSEA from RNA-Seq data. Indeed, GPX1 was highly expressed in T315I mutant cells, while knockout of GPX1 significantly suppressed cell proliferation and triggered apoptosis under glucose-deprived condition. GPX1 knockout showed decreased cell metabolism signaling as well as mitochondrial gene expression by RNA-Seq. Mechanistically, GPX1 maintained mitochondrial activity and oxygen consumption rate (OCR), retaining mitochondrial redox homeostasis and oxidative phosphorylation (OXPHOS). Additionally, mercaptosuccinic acid (MSA), a GPX inhibitor, inhibited CML colony formation and induced cell apoptosis under glucose-free condition. Therefore, GPX1 is a promising therapeutic target to overcome drug resistance induced by the T315I mutation, which provides a novel approach for BCR/ABL-T315I CML treatment by disturbing mitochondrial OXPHOS.
Collapse
Affiliation(s)
- Jun-Dan Wang
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Jin-Xing Wang
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China
- Department of Pathology Technique, Guangdong Medical University, Dongguan, China
| | - Zhi-Li Lin
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ling Zhang
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Jia-Jun Liu
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Rui Gao
- Department of Oncology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zi-Jie Long
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
8
|
Oh S, Sim HB, Kim H, Mun SK, Ji M, Choi B, Kim DY, Kim JJ, Paik MJ. Cellular metabolomics study in colorectal cancer cells and media following treatment with 5-fluorouracil by gas chromatography-tandem mass spectrometry. Metabolomics 2025; 21:62. [PMID: 40335841 DOI: 10.1007/s11306-025-02263-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 04/18/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Metabolic reprogramming is a distinctive characteristic of colorectal cancer (CRC) which provides energy and nutrients for rapid proliferation. Although numerous studies have explored the rewired metabolism of CRC, the metabolic alterations occurring in CRC when the cell cycle is arrested by treatment with 5-fluorouracil (5-FU), an anticancer drug that arrests the S phase, remain unclear. METHODS A systematic profiling analysis was conducted as ethoxycarbonyl/methoxime/tert-butyldimethylsilyl derivatives using gas chromatography-tandem mass spectrometry in HT29 cells and media following 5-FU treatment in a concentration- and time-dependent manner. RESULTS In HT29 cells of 24 h after 5-FU treatment (3-100 μM) and 48 h after 5-FU treatment (1-10 μM), six amino acids, including valine, leucine, isoleucine, serine, glycine, and alanine and two organic acids, including pyruvic acid and lactic acid, were significantly increased compared to the DMSO-treated group. However, 48 h after 5-FU treatment (30-100 μM) in HT29 cells, the levels of these metabolites decreased along with an approximately 50% reduction in viability, an increase in the level of reactive oxygen species, induction of cycle arrest in the G1 phase, and the induction of apoptosis. On the other hand, the levels of fatty acids showed a continuous increase in HT29 cells 48 h after 5-FU treatment (1-100 μM). In the media, the decreased availabilities in the cellular uptake of nutrient metabolites, including valine, leucine, isoleucine, serine, and glutamine, were observed at 48 h after 5-FU treatment in a dose-dependent manner. CONCLUSION It is assumed that there is a possible shift in energy dependence from the tricarboxylic acid cycle to fatty acid metabolism. Thus, metabolic profiling analysis revealed altered energy metabolism in CRC cells following 5-FU treatment.
Collapse
Affiliation(s)
- Songjin Oh
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea
| | - Hyun Bo Sim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea
| | - Hyeongyeong Kim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea
| | - Seul-Ki Mun
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea
| | - Moongi Ji
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea
| | - Byeongchan Choi
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea
| | - Doo-Young Kim
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea
- New Drug Discovery Lab, Hyundai Pharm, Yongin, 17089, Republic of Korea
| | - Jong-Jin Kim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea.
| | - Man-Jeong Paik
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon, 57922, Republic of Korea.
| |
Collapse
|
9
|
Chen J, Trindl CA, Ye H, Huang D, Ooi A, Garcia JGN, Chapman E, Zhang DD. CYP4F11, an NRF2 Target Gene, Promotes Hepatocellular Carcinoma Cell Growth. Mol Carcinog 2025. [PMID: 40329467 DOI: 10.1002/mc.23925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/27/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025]
Abstract
Hepatocellular carcinoma (HCC), the most common form of primary liver cancer, is the third leading cause of cancer-related mortality globally. Current systemic therapies for HCC are limited and often exhibit unsatisfactory efficacy, underscoring the need for novel therapeutic approaches. Nuclear factor erythroid 2-related factor-2 (NRF2), a master transcription factor regulating cellular redox and metabolic homeostasis, is frequently overexpressed in HCC due to mutations in NFE2L2/NRF2 or its negative regulator Kelch-like ECH-associated protein 1 (KEAP1), contributing to tumor progression. In this study, we identify CYP4F11, a member of the Cytochrome P450 family, as a direct target gene of NRF2. CYP4F11, primarily expressed in the liver, is crucial in fatty acid oxidation and eicosanoid metabolism. We demonstrate that CYP4F11 expression is driven by NRF2 and is significantly elevated in HCC patients harboring NFE2L2 gain of function or KEAP1 loss of function mutations. Functionally, CYP4F11 promotes HCC cell growth, and reduced expression of CYP4F11 not only suppresses HCC cell proliferation but also enhances sorafenib-induced HCC cell death. Further, NRF2 inhibition sensitizes HCC to sorafenib through downregulation of CYP4F11. These findings position CYP4F11 as a novel contributor to HCC progression and highlight the potential of targeting the NRF2-CYP4F11 axis for HCC treatment.
Collapse
Affiliation(s)
- Jinjing Chen
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Carlee A Trindl
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Haofeng Ye
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Dichun Huang
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Aikseng Ooi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Joe G N Garcia
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Eli Chapman
- Department of Pharmacology and Therapeutics, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, University of Florida, College of Medicine, Jupiter, Florida, USA
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Donna D Zhang
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
10
|
Jing T, Tang D. Intratumoral microbiota: a new force in the development and treatment of esophageal cancer. Clin Transl Oncol 2025; 27:1921-1932. [PMID: 39455494 DOI: 10.1007/s12094-024-03757-1] [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: 04/14/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Esophageal cancer (EC) ranks among the most prevalent cancers worldwide, with a particularly high incidence in the Asian population. Due to the inconspicuous nature of early symptoms, patients with esophageal cancer are typically diagnosed in the middle to late stages, resulting in suboptimal overall treatment outcomes. Consequently, there is an urgent need to explore and refine therapeutic strategies. Microorganisms have been identified in numerous tumor tissues, including EC, and these microorganisms are referred to as the intratumoral microbiome. Intratumoral microbiota and their metabolic byproducts can influence the progression and treatment of esophageal cancer through various mechanisms, such as modulating tumor cell metabolism and local immune responses. Therefore, the intratumoral microbiota may potentially serve as a target for the treatment of esophageal cancer. This review delineates the composition, origin, and diagnostic significance of intratumoral microbiota in esophageal cancer tissue, and discusses the mechanisms by which intratumoral microbiota contribute to the onset of esophageal cancer. In addition, the impact of intratumoral microbiota on the treatment of esophageal cancer and its intervention measures are also addressed.
Collapse
Affiliation(s)
- Tianyang Jing
- Clinical Medical College, Yangzhou University, Yangzhou, 22500, Jiangsu Province, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
11
|
Guo S, Zhang L, Ren J, Lu Z, Ma X, Liu X, Jin H, Li J. The roles of enhancer, especially super-enhancer-driven genes in tumor metabolism and immunity. Int J Biol Macromol 2025; 308:142414. [PMID: 40132720 DOI: 10.1016/j.ijbiomac.2025.142414] [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: 10/16/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Abnormal metabolism is a characteristic of malignant tumors. Numerous factors play roles in the regulation of tumor metabolism. As epigenetic regulators, enhancers, especially the super-enhancers (SEs), serve as platforms for transcription factors that regulate the expression of metabolism-related enzymes or transporters at the gene level. In this study, we review the effects of enhancer/ SE-driven genes on tumor metabolism and immunity. Enhancers/SEs play regulatory roles in glucose metabolism (glycolysis, gluconeogenesis, tricarboxylic acid (TCA) cycle, pyruvate, and pentose phosphate pathway, lipid metabolism (cholesterol, fatty acid, phosphatide, and sphingolipid), and amino acid metabolism (glutamine, tryptophan, arginine, and cystine). By regulating tumor metabolism, enhancers and SEs can reprogram tumor microenvironment, especially the status of various immune cells. Therefore, interfering enhancers/SEs that regulate the tumor metabolism is likely to enhance the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Songyue Guo
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Lu Zhang
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Jiao Ren
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Zhong Lu
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Xiaolin Ma
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Xinling Liu
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China.
| | - Hongchuan Jin
- Department of Medical Oncology, Cancer Center of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China.
| | - Jiaqiu Li
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China.
| |
Collapse
|
12
|
Yuan F, Jia G, Wen W, Xu S, Gunchick V, Deng K, Long J, Yu D, Shu XO, Zheng W. Blood metabolic biomarkers and colorectal cancer risk: results from large prospective cohort and Mendelian randomisation analyses. Br J Cancer 2025:10.1038/s41416-025-02997-4. [PMID: 40307439 DOI: 10.1038/s41416-025-02997-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Emerging evidence suggests metabolic dysregulation may contribute to colorectal cancer (CRC) aetiology. We aimed to identify pre-diagnostic metabolic biomarkers for CRC risk in 230,420 UK Biobank participants. METHODS Nuclear magnetic resonance spectroscopy was used to quantify 249 metabolic biomarkers in plasma samples collected at baseline. Cox proportional hazards models were used to estimate hazard ratios and 95% confidence intervals (CIs) for associations of metabolic biomarkers with CRC risk after adjusting for potential confounders. To infer the potential causality of biomarkers that were associated with CRC independent of the others, we performed genome-wide association analyses among 199,732 UK Biobank participants of European ancestry to identify biomarker-associated genetic variants, followed by two-sample Mendelian randomization (MR) analyses using summary statistics of 78,473 CRC cases and 107,143 controls of European ancestry. RESULTS During a median follow-up time of 9.7 years, 2,410 incident primary CRC cases were identified. Among 43 CRC-associated (P-value < 0.001) metabolic biomarkers, ten biomarkers including fatty acids (FAs), inflammation, ketone bodies, and lipoprotein lipids were associated with CRC risk after mutual adjustment. MR analyses provided strong evidence for potential causal associations of CRC risk with percentages of linolic acid [odds ratio (OR) = 0.89, 95% CI = 0.83-0.96, P-value = 3 × 10-3] and saturated FAs (OR = 1.14, 95% CI = 1.03-1.25, P-value = 9 × 10-3) to total FAs. CONCLUSIONS We identified multiple CRC-associated metabolic biomarkers. Perturbed lipid and lipoprotein metabolism may promote colorectal carcinogenesis.
Collapse
Affiliation(s)
- Fangcheng Yuan
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Guochong Jia
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shuai Xu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Valerie Gunchick
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kui Deng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Danxia Yu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
13
|
Ping P, Ma Y, Xu X, Li J. Reprogramming of fatty acid metabolism in thyroid cancer: Potential targets and mechanisms. Chin J Cancer Res 2025; 37:227-249. [PMID: 40353071 PMCID: PMC12062987 DOI: 10.21147/j.issn.1000-9604.2025.02.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Thyroid cancer (TC) is one of the most common endocrine system tumors, and its incidence continues to increase worldwide. Although most TC patients have a good prognosis, especially with continuous advancements in surgery, radioactive iodine therapy, chemotherapy, endocrine therapy and targeted therapy, the effectiveness of disease treatment has significantly improved. However, there are still some cases with a higher risk of death and greater aggressiveness. In these more challenging advanced or highly aggressive cases, tyrosine kinase inhibitors appear to be an effective treatment option. Unfortunately, these drugs are less than ideal in terms of efficacy because of their toxicity and potential for intrinsic or acquired resistance. Therefore, exploring new strategies targeting the metabolic characteristics of TC cells and overcoming drug resistance barriers in existing treatments have become key topics in the current field of TC research. In recent years, lipid metabolic reprogramming has gained attention as an important aspect of cancer development. Lipid metabolic reprogramming not only participates in the formation of the cell membrane structure, but also plays an important role in signal transduction and promoting cell proliferation. In particular, fatty acid (FA) metabolic reprogramming has attracted widespread attention and plays an important role in multiple aspects such as tumor growth, metastasis, enhanced invasive ability, immune escape, and drug resistance. Although TC is considered a disease that is highly dependent on specific types of metabolic activities, a comprehensive understanding of the specific mechanism of action of FA metabolic reprogramming in this process is lacking. This article aims to review how FA metabolic reprogramming participates in the occurrence and development of TC, focusing on the impact of abnormal FA metabolic pathways and changes in the expression and regulation of related genes over the course of this disease. By examining the complex interactions between FA metabolic disorders and carcinogenic signaling pathways in depth, we aim to identify new therapeutic targets and develop more precise and effective treatments for TC.
Collapse
Affiliation(s)
- Pengbin Ping
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
- Department of Radiation Therapy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Yuhong Ma
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Xiaoying Xu
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Juan Li
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
14
|
Zhang X, Lin Z, Xu B, Ma C, Jiang B, Geng Y, Sheng Y, Dai Y, Xue Y, Ren Y. Unveiling gastric precancerous stages: metabolomic insights for early detection and intervention. BMC Gastroenterol 2025; 25:318. [PMID: 40301782 PMCID: PMC12042622 DOI: 10.1186/s12876-025-03898-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Gastric precancerous lesions (GPL) represent a heterogeneous, multi-stage process that involves transition from a benign to a malignant state. To optimize prevention and intervention strategies, accurate methods must clearly distinguish between precancerous stages and predict progression risks at early stages. METHODS The metabolomic profiles of 188 GPL tissues and matched normal tissues were characterized using ultra-high-performance liquid chromatography-tandem mass spectrometry. Both multivariate and univariate statistical analyses were used to identify metabolomic features differentiating normal, atrophic, and intestinal metaplasia states in the stomach, followed by preliminary functional validation. RESULTS From experiments conducted on two cohorts, we established a reliable clinical gastric tissue metabolomic map, which clearly distinguished between normal, atrophic, and intestinalized gastric tissues. We then identified metabolic biomarkers that differentiated various GPL stages. Furthermore, key metabolites were validated in in vitro studies. Relative acyl group and glycerophospholipid abundance was higher in normal gastric tissue when compared to GPL, whereas organic acids were more prevalent in precancerous tissues than in normal tissues. A combination of glycerophosphocholine, tiglylcarnitine, malate, sphingosine, and γ-glutamylglutamic acid may serve as powerful biomarkers to distinguish normal tissue from GPL. CONCLUSION We used ultra-high-performance liquid chromatography with tandem mass spectrometry to effectively characterize metabolomic profiles in clinical gastric tissue samples. Key metabolites were identified and validated using targeted metabolomics. This study identified the metabolomic profiles of gastric tissues with atrophy and intestinal metaplasia of the gastric mucosa, uncovering and preliminarily validating key metabolites that may be used to assess high-risk populations and diagnose GPL, potentially advancing targeted gastric cancer prevention and treatment efforts.
Collapse
Affiliation(s)
- Xiaoyue Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Ziming Lin
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Boyan Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Chenyu Ma
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Bowen Jiang
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yan Geng
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Yingyue Sheng
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Yuanyuan Dai
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China.
| | - Yilin Ren
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China.
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
15
|
Zhang L, Zhao X, Hu J, Li T, Chen HZ, Zhang A, Wang H, Yu J, Zhang L. PRPS2 enhances RNA m 6A methylation by stimulating SAM synthesis through enzyme-dependent and independent mechanisms. Nat Commun 2025; 16:3966. [PMID: 40295500 PMCID: PMC12037730 DOI: 10.1038/s41467-025-59119-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Cancer cells exploit altered metabolic pathways to dynamically regulate epigenetic methylation and thus promote tumorigenesis and metastasis. In various human cancers, such as lung adenocarcinoma, the level of a key cellular metabolite, S-adenosylmethionine (SAM), is prominently upregulated for RNA hypermethylation as the methyl donor. However, the specific mechanisms by which cancer cells produce SAM to sustain RNA methylation remain elusive. Here, we demonstrate that PRPS2, a phosphoribosyl pyrophosphate synthetase isoform involved in the first and rate-limiting step of the purine biosynthesis pathway, exhibits distinct oncogenic functionality in regulating RNA methylation, unlike its homolog PRPS1. PRPS2 utilizes four non-conserved key residues to bypass the typical ADP/GDP allosteric feedback inhibition, enabling sustained excess production of newly synthesized ATP. Moreover, PRPS2 stabilizes methionine adenosyltransferase 2 A (MAT2A) through direct interactions to positively stimulate ATP utilization and SAM synthesis for RNA m6A specific methylation via the WTAP/METTL3/METTL14 methyltransferase complex, thereby promoting lung tumorigenesis. Our study links nucleotide biosynthesis with RNA epigenetics in cancer progression through the PRPS2-MAT2A-WTAP/METTL3/METTL14 axis, and elucidates both enzyme-dependent and independent functions of PRPS2. These findings have significant implications for developing targeted therapies for cancers associated with PRPS2 abnormalities.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Xian Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jingyan Hu
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Tingting Li
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Hong-Zhuan Chen
- Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ao Zhang
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hao Wang
- The Division of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Liang Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
- Department of Chemical Biology, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
16
|
Bakke KM, Bousquet PA, Meltzer S, Bjørnetrø T, Rise F, Wilkins AL, Redalen KR, Ree AH. Serum metabolite levels identify incipient metastatic progression of rectal cancer. COMMUNICATIONS MEDICINE 2025; 5:142. [PMID: 40289234 PMCID: PMC12034819 DOI: 10.1038/s43856-025-00868-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The cellular metabolism undergoes reprogramming during the metastatic process. We hypothesised that serum metabolites at the time of primary tumour diagnosis might identify rectal cancer patients prone to metastatic progression. METHODS One hundred twenty-three rectal cancer patients from a prospective observational biomarker study were followed up to 5 years after study entry. We have assessed metabolites in serum sampled at the time of diagnosis by 1H-nuclear magnetic resonance spectroscopy, using the internal reference trimethylsilylpropanoic acid for quantification. RESULTS Here we show that patients who develop overt metastatic disease more than 6 months after the primary tumour diagnosis have elevated serum levels (Kruskal-Wallis test) of alanine (P = 0.005), lactate (P = 0.023), pyruvate (P = 0.041) and citrate (P = 0.007) compared to those without metastases at the 5-year follow-up or with metastases already 6 months or sooner after the cancer diagnosis. Patients with serum citrate above 0.24 mmol/L have poorer progression-free survival compared to those with levels below (P < 0.001; log-rank test). CONCLUSIONS We observe a distinct serum metabolite profile, in particular involving citrate to the best of our knowledge shown for the first time clinically, in rectal cancer patients at heightened risk of metastasis already when the primary tumour is diagnosed, offering insights into the metabolism of metastatic progression.
Collapse
Affiliation(s)
- Kine M Bakke
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.
- Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway.
| | - Paula A Bousquet
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Sebastian Meltzer
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Tonje Bjørnetrø
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Frode Rise
- Department of Chemistry, University of Oslo, Oslo, Norway
| | - Alistair L Wilkins
- School of Science and Engineering, University of Waikato, Hamilton, New Zealand
| | - Kathrine Røe Redalen
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Wei J, Mo CD, Zhu GY, Qiu FY, Qin SP, Hou QY, Zhou Y, Wei WS, Huang Z, Yang JR. The relationship between infectious pathogen antibodies, plasma metabolites, and breast cancer: A Mendelian randomization study with mediation analysis. Medicine (Baltimore) 2025; 104:e42283. [PMID: 40295253 PMCID: PMC12040016 DOI: 10.1097/md.0000000000042283] [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: 11/26/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Breast cancer (BC) has the second highest incidence rate among women worldwide. Although there are various treatment methods, the prognosis is poor once metastasis occurs. However, the extent to which pathogens of infectious diseases influence the risk of BC remains unclear. The goal of this study is to determine if these pathogens are causally related to BC development. A Mendelian randomization (MR) analysis is used to assess the causal relationship between infectious pathogen antibodies and the risk of BC, as well as their potential intermediary factors. Two-sample MR analysis using inverse variance weighting (IVW) is conducted to determine the causal relationship between infectious pathogen antibodies and the risk of BC. Additionally, the mediating role of 1400 metabolites between infectious pathogen antibodies and the risk of BC is analyzed. There were 5 infectious pathogen antibodies and 86 metabolites associated with BC (P < .05). There were 14 metabolites that mediated the pathway between infectious pathogen antibodies and BC. X-07765 levels showed a significant negative mediating effect on the relationship between Anti-human herpes virus 6 IgG seropositivity and BC (beta = -0.0025, 95% CI [-0.0046, -0.0003], P = .0236), accounting for 14.8% of the effect (95% CI: 27.7-1.99). This study provides a thorough evaluation of the causal relationships among infectious pathogen antibodies, plasma metabolites, and BC. Our research has identified 5 infectious pathogen antibodies that exhibit a causal relationship with BC, mediated through 86 distinct metabolites.
Collapse
Affiliation(s)
- Jing Wei
- Guangxi Medical University, Nanning, PR China
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Chong-de Mo
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Gui-yue Zhu
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Fang-yu Qiu
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Sheng-peng Qin
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Qi-yan Hou
- Graduate School of Guangxi University of Chinese Medicine,Nanning, PR China
| | - Ying Zhou
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Wen-song Wei
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Zhen Huang
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Jian-rong Yang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| |
Collapse
|
18
|
Qi Y, Wang A, Chen S, Chen W. NCAPH promotes glucose metabolism reprogramming and cell stemness in ovarian cancer cells through the MEK/ERK/PD-L1 pathway. J Ovarian Res 2025; 18:81. [PMID: 40259316 PMCID: PMC12010566 DOI: 10.1186/s13048-025-01659-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 04/01/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUNDS Ovarian cancer is a prevalent malignant tumor that affects the female reproductive system with the characteristic of high heterogeneity. Non-structural maintenance of chromosomes condensin I complex subunit H (NCAPH) has been implicated in a variety of cancers. METHODS The expression of NCAPH before and after transfection were assessed using RT-qPCR and western blot analysis. Cell stemness was evaluated through spheroid formation assay. The extracellular acidification rate (ECAR) of ovarian cancer cells was measured utilizing Seahorse Glycolysis Stress Test Assay while oxygen consumption rate (OCR) was estimated with Seahorse Mito Stress Test Assay. Lactate production and glucose consumption were quantified using corresponding assay kits. Western blot was employed to analyze the expression of stem cell markers, glycolysis- and MEK/ERK/PD-L1 signaling pathway-related proteins. In vivo, tumor size and weight were recorded, and immunohistochemical staining was used to assess MEK/ERK/PD-L1 and KI67 expression in tumor tissues from nude mice. RESULTS It was observed that NCAPH expression is upregulated in ovarian cancer cells. Silencing NCAPH led to repression of both stemness characteristics and glucose metabolism reprogramming. Furthermore, knockdown of NCAPH inhibited the MEK/ERK/PD-L1 signaling pathway both in vitro and in vivo, resulting in suppressed tumor growth in mouse models. CONCLUSION Collectively, silencing NCAPH impedes malignant progression of ovarian cancer through modulation of the MEK/ERK/PD-L1 pathway. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Yingying Qi
- Department of Gynecology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aiping Wang
- Department of Gynecology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Silin Chen
- Department of Gynecology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Chen
- Department of Gynecology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
19
|
Addanki S, Kim L, Stevens A. Understanding and Targeting Metabolic Vulnerabilities in Acute Myeloid Leukemia: An Updated Comprehensive Review. Cancers (Basel) 2025; 17:1355. [PMID: 40282531 PMCID: PMC12025543 DOI: 10.3390/cancers17081355] [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: 02/21/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Acute Myeloid Leukemia (AML) is characterized by aggressive proliferation and metabolic reprogramming that support its survival and resistance to therapy. This review explores the metabolic distinctions between AML cells and normal hematopoietic stem cells (HSCs), emphasizing the role of altered mitochondrial function, oxidative phosphorylation (OXPHOS), and biosynthetic pathways in leukemic progression. AML cells exhibit distinct metabolic vulnerabilities, including increased mitochondrial biogenesis, reliance on glycolysis and amino acid metabolism, and unique signaling interactions that sustain leukemic stem cells (LSCs). These dependencies provide potential therapeutic targets, as metabolic inhibitors have demonstrated efficacy in disrupting AML cell survival while sparing normal hematopoietic cells. We examine current and emerging metabolic therapies, such as inhibitors targeting glycolysis, amino acid metabolism, and lipid biosynthesis, highlighting their potential in overcoming drug resistance. However, challenges remain in translating these strategies into clinical practice due to AML's heterogeneity and adaptability. Further research into AML's metabolic plasticity and precision medicine approaches is crucial for improving treatment outcomes. Understanding and exploiting AML's metabolic vulnerabilities could pave the way for novel, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Sridevi Addanki
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Alexandra Stevens
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
20
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
21
|
Guo Y, Lin Z, Zhou Z, Zhang W, Mao S, Shan Z, Wu P, Yao X. Oncogenic and immunological functions of USP35 in pan-cancer and its potential as a biomarker in kidney clear cell carcinoma. BMC Cancer 2025; 25:617. [PMID: 40188027 PMCID: PMC11972461 DOI: 10.1186/s12885-025-13964-w] [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: 11/01/2024] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Ubiquitin-specific protease 35 (USP35) has gained attention as a regulator in cancer progression. However, its specific role in kidney clear cell carcinoma (KIRC) remains unclear. METHODS USP35 expression in KIRC tumor and normal tissues was evaluated using TCGA data. Correlations between USP35 expression, clinical parameters, and survival outcomes were examined. Functional enrichment analyses were performed to explore the pathways associated with USP35 expression. Immune-related analyses were conducted to assess the effect of USP35 on immune cell recruitment and neoantigen presentation. Drug sensitivity analyses were used to identify potential therapeutic agents targeting USP35. RESULTS USP35 was significantly overexpressed in KIRC tumor tissues compared to normal tissues, and its high expression correlated with advanced disease stages and poor survival outcomes. Gene set enrichment analysis revealed that high USP35 expression was associated with oncogenic pathways, including glycerophospholipid and linoleic acid metabolism, while low expression linked to nitrogen and purine metabolism. USP35 also modulated immune responses, affecting immune cell recruitment and neoantigen presentation, suggesting a role in immune evasion. Drug sensitivity analysis showed that high USP35 expression correlated with increased sensitivity to paclitaxel, bosutinib, and lapatinib. In vitro knockdown of USP35 significantly reduced KIRC cell proliferation, migration, and epithelial-mesenchymal transition (EMT), further supporting its role in tumor progression. CONCLUSION USP35 is overexpressed in KIRC and associated with poor prognosis, likely promoting tumor progression through oncogenic pathways and immune modulation. Its correlation with drug sensitivity positions USP35 as a potential therapeutic target, warranting further investigation into its mechanistic functions and therapeutic applications.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/mortality
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Kidney Neoplasms/immunology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/mortality
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/drug therapy
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Ubiquitin-Specific Proteases/genetics
- Ubiquitin-Specific Proteases/metabolism
- Gene Expression Regulation, Neoplastic
- Prognosis
- Cell Line, Tumor
- Cell Proliferation
- Epithelial-Mesenchymal Transition
Collapse
Affiliation(s)
- Yadong Guo
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Ziyou Lin
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zijing Zhou
- Laboratory of Ruijin Hospitalaffiliated to, Wuxi Branchaq, Shanghai Jiaotong University School of Medicine, Wuxi, Jiangsu, China
| | - Wentao Zhang
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shiyu Mao
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Zezhi Shan
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Pengfei Wu
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| | - Xudong Yao
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Markov N, Sabirova S, Sharapova G, Gomzikova M, Brichkina A, Barlev NA, Egger M, Rizvanov A, Simon HU. Mitochondrial, metabolic and bioenergetic adaptations drive plasticity of colorectal cancer cells and shape their chemosensitivity. Cell Death Dis 2025; 16:253. [PMID: 40185729 PMCID: PMC11971274 DOI: 10.1038/s41419-025-07596-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
The extent of mitochondrial heterogeneity and the presence of mitochondrial archetypes in cancer remain unknown. Mitochondria play a central role in the metabolic reprogramming that occurs in cancer cells. This process adjusts the activity of metabolic pathways to support growth, proliferation, and survival of cancer cells. Using a panel of colorectal cancer (CRC) cell lines, we revealed extensive differences in their mitochondrial composition, suggesting functional specialisation of these organelles. We differentiated bioenergetic and mitochondrial phenotypes, which point to different strategies used by CRC cells to maintain their sustainability. Moreover, the efficacy of various treatments targeting metabolic pathways was dependent on the respiration and glycolysis levels of cancer cells. Furthermore, we identified metabolites associated with both bioenergetic profiles and cell responses to treatments. The levels of these molecules can be used to predict the therapeutic efficacy of anti-cancer drugs and identify metabolic vulnerabilities of CRC. Our study indicates that the efficacy of CRC therapies is closely linked to mitochondrial status and cellular bioenergetics.
Collapse
Affiliation(s)
- Nikita Markov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Sirina Sabirova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Laboratory of Intercellular Communication, Kazan Federal University, Kazan, Russia
| | - Gulnaz Sharapova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Marina Gomzikova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Laboratory of Intercellular Communication, Kazan Federal University, Kazan, Russia
| | - Anna Brichkina
- Institute of Systems Immunology, Center for Tumor Biology and Immunology, Philipps University of Marburg, Marburg, Germany
| | - Nick A Barlev
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Marcel Egger
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Albert Rizvanov
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany.
| |
Collapse
|
23
|
Zeng B, Shi H, Liu T, Tang J, Lin J, Lin X, Zeng T. The influence of homologous recombination repair on temozolomide chemosensitivity in gliomas. Carcinogenesis 2025; 46:bgaf017. [PMID: 40120126 DOI: 10.1093/carcin/bgaf017] [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: 11/18/2024] [Revised: 02/22/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025] Open
Abstract
Gliomas represent a prevalent form of primary brain tumors, with temozolomide (TMZ) serving as the established first-line therapeutic option. Nevertheless, the effectiveness of TMZ is hindered by the development of chemoresistance. Recent investigations have underscored the correlation of homologous recombination repair (HRR), a pivotal mechanism responsible for mending DNA double-strand breaks, with TMZ resistance in glioma treatment. This review centers on elucidating the significance of HRR in the management of gliomas, with a particular emphasis on pivotal molecules implicated in the HRR process, including RAD51, ATM, ATR, and newly identified small molecules that impact HRR. Modulating the expression of these genes can effectively restrain pathways such as ATM/CHK2, ATR/CHK1, and PI3K/AKT, subsequently augmenting the sensitivity of gliomas to TMZ. Noteworthy efforts have been directed towards exploring inhibitors of these pathways in recent research endeavors, culminating in encouraging outcomes. In conclusion, the involvement of HRR in glioma resistance unveils novel therapeutic avenues, with targeting crucial molecules in the HRR pathway, holding promise for enhancing the effectiveness of TMZ therapy.
Collapse
Affiliation(s)
- Biyun Zeng
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P. R. China
- School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
| | - Hansen Shi
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P. R. China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Jinjing Tang
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P. R. China
| | - Juncheng Lin
- School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
| | - Xiaocong Lin
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P. R. China
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Tao Zeng
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P. R. China
| |
Collapse
|
24
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
25
|
Barnabas GD, Bhat TA, Goebeler V, Leclair P, Azzam N, Melong N, Anderson C, Gom A, An S, Ergin EK, Shen Y, Conrrero A, Mungall AJ, Mungall KL, Maxwell CA, Reid GSD, Hirst M, Jones S, Chan JA, Senger DL, Berman JN, Parker SJ, Bush JW, Strahlendorf C, Deyell RJ, Lim CJ, Lange PF. Proteomics and personalized PDX models identify treatment for a progressive malignancy within an actionable timeframe. EMBO Mol Med 2025; 17:625-644. [PMID: 40204966 PMCID: PMC11982353 DOI: 10.1038/s44321-025-00212-8] [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: 08/27/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 04/11/2025] Open
Abstract
Genomics has transformed the diagnostic landscape of pediatric malignancies by identifying and integrating actionable features that refine diagnosis, classification, and treatment. Yet, translating precision oncology data into effective therapies for hard-to-cure childhood, adolescent, and young adult malignancies remains a significant challenge. We present the case for combining proteomics with patient-derived xenograft models to identify personalized treatment for an adolescent with primary and metastatic spindle epithelial tumor with thymus-like elements (SETTLE). Within two weeks of biopsy, proteomics identified elevated SHMT2 as a target for therapy with the anti-depressant sertraline. Drug response was confirmed within two months using a personalized chicken chorioallantoic membrane model of the patient's SETTLE tumor. Following failure of cytotoxic chemotherapy and second-line therapy, the patient received sertraline treatment and showed decreased tumor growth rates, albeit with clinically progressive disease. We demonstrate that proteomics and fast-track xenograft models provide supportive pre-clinical data in a clinically meaningful timeframe to impact clinical practice. By this, we show that proteome-guided and functional precision oncology are feasible and valuable complements to the current genome-driven precision oncology practices.
Collapse
Affiliation(s)
- Georgina D Barnabas
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Tariq A Bhat
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Verena Goebeler
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Pascal Leclair
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Nadine Azzam
- Children's Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
| | - Nicole Melong
- Children's Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
| | - Colleen Anderson
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Pathology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Alexis Gom
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Seohee An
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Enes K Ergin
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Yaoqing Shen
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, BC, Canada
| | - Agustina Conrrero
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Andrew J Mungall
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, BC, Canada
| | - Karen L Mungall
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, BC, Canada
| | - Christopher A Maxwell
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Gregor S D Reid
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Martin Hirst
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Steven Jones
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer A Chan
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Pathology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Donna L Senger
- Department of Medicine, McGill, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Jason N Berman
- Children's Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Seth J Parker
- Department of Biochemistry, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Jonathan W Bush
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Caron Strahlendorf
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Rebecca J Deyell
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.
| | - C James Lim
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.
| | - Philipp F Lange
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada.
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.
| |
Collapse
|
26
|
Guo Z, Hui Y, Sun S, Kong F. KAT3B Promotes the Glycolysis and Malignant Progression of Lung Cancer by Mediating the Succinylation Modification of PKM2. J Biochem Mol Toxicol 2025; 39:e70259. [PMID: 40226997 DOI: 10.1002/jbt.70259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/29/2024] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Lysine succinyltransferase KAT3B plays a critical role in the progression of various cancers by modulating key metabolic pathways, including glycolysis. However, the function and underlying mechanism of KAT3B in glycolysis and lung cancer (LC) progression remain to be further studied. We determined mRNA expression levels of lysine succinyl-modifying enzymes through qRT-PCR. Protein expression and succinylation status of glycolysis-related proteins PKM2, LDHA, and ENO1 were analyzed via Western blot. Co-immunoprecipitation and immunofluorescence microscopy were employed to verify the interaction between KAT3B and PKM2. Bioinformatics analysis predicted succinylation sites on PKM2, which were subsequently validated through site-directed mutagenesis. The effects of KAT3B and PKM2 on LC cell malignancy and glycolysis were evaluated using CCK-8, transwell migration, glucose uptake, lactate production, ECAR, and OCR assays. A xenograft tumor model was utilized to assess the impact of KAT3B on LC tumor growth. We confirmed the augmentation of KAT3B in LC, which also was correlated with advanced TNM stages and elevated T stages of LC patients. Conversely, KAT3B knockdown suppressed the growth, metastasis, and glycolytic activity of LC cells in vitro, while also inhibiting tumor growth in vivo. KAT3B mediated succinylation at PKM2 K298, and the suppression of LC cell malignancy and glycolysis upon KAT3B downregulation was largely reversed by upregulation of PKM2. The KAT3B/PKM2 axis may be a novel target for LC therapy.
Collapse
Affiliation(s)
- Zhifeng Guo
- Department of Oncology, Section II, Chifeng Municipal Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Yan Hui
- Department of Oncology, Section II, Chifeng Municipal Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Siqi Sun
- Department of Oncology, Section II, Chifeng Municipal Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Fanlong Kong
- Department of Oncology, Section II, Chifeng Municipal Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| |
Collapse
|
27
|
Lv Y, Pu L, Ran B, Xiang B. Targeting tumor angiogenesis and metabolism with photodynamic nanomedicine. Front Cell Dev Biol 2025; 13:1558393. [PMID: 40235732 PMCID: PMC11996804 DOI: 10.3389/fcell.2025.1558393] [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: 01/10/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
Photodynamic therapy (PDT) holds considerable promise as a tumor treatment modality, characterized by its targeted action, compatibility with other therapeutic approaches, and non - invasive features. PDT can achieve remarkable spatiotemporal precision in tumor ablation through the generation of reactive oxygen species (ROS). Nevertheless, despite its potential in tumor treatment, PDT encounters multiple challenges in practical applications. PDT is highly oxygen - dependent, and thus the effectiveness of PDT can be markedly influenced by tumor hypoxia. The co-existence of abnormal vasculature and metabolic deregulation gives rise to a hypoxic microenvironment, which not only sustains tumor survival but also undermines the therapeutic efficacy of PDT. Consequently, targeting tumor angiogenesis and metabolism is essential for revitalizing PDT. This review emphasizes the mechanisms and strategies for revitalizing PDT in tumor treatment, predominantly concentrating on interfering with tumor angiogenesis and reprogramming tumor cell metabolism. Lastly, the outlining future perspectives and current limitations of PDT are also summarized. This could provide new insights and methodologies for overcoming the challenges associated with PDT in tumor treatment, ultimately advancing the field of PDT.
Collapse
Affiliation(s)
- Yong Lv
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lihui Pu
- Department of Critical Care, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Ran
- School of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Bo Xiang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Zhu L, Ding J, Xue W, Zhou S, Wang L, Jiang A, Zhao M, He Q, Ren A. Manganese Peroxidase Participates in the Liquid-Solid-Gas Triphase Regulation on Microbial Degradation of Lignocellulose in Solid-State Fermentation. Biotechnol Bioeng 2025; 122:908-921. [PMID: 39810331 DOI: 10.1002/bit.28927] [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: 10/03/2024] [Revised: 12/30/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025]
Abstract
The three-phase structure of solid-state fermentation (SSF) directly affects substrate degradation and fermentation efficiency. However, the mechanism of three-phase regulation on lignocellulose utilization and microbial metabolism is still unclear. Based on comparative transcriptome analysis, a lignocellulose degrading enzyme, manganese peroxidase (GlMnP), which was significantly affected by water stress meanwhile related to triphase utilization, was screened to reveal the mechanism using Ganoderma lucidum as the reference strain. The results showed that GlMnP directly participates in lignocellulose degradation by positively regulating the activity of carboxymethylcellulase (CMCase), filter paper (FPAse), and laccase (LACase) enzymes, and indirectly participates in lignocellulose degradation by negatively regulating the redox levels in microorganisms. In addition, GlMnP can also control microbial glycolysis rate to enhance lignocellulose utilization. The results indicated that GlMnP participates in the liquid-solid-gas triphase regulation on lignocellulose degradation by G. lucidum in SSF.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Juan Ding
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Wenjing Xue
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Shu Zhou
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Longyu Wang
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Ailiang Jiang
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Mingwen Zhao
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Qin He
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Ang Ren
- Department of Microbiology, Sanya Institute of Nanjing Agricultural University, Key Lab of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
29
|
Zeng Q, Li L, Chang T, Sun Y, Zheng B, Xue L, Liu C, Li X, Huang R, Gu J, An Z, Yao H, Zhou D, Fan J, Dai Y. Phosphorylation of POU3F3 Mediated Nuclear Translocation Promotes Proliferation in Non-Small Cell Lung Cancer through Accelerating ATP5PF Transcription and ATP Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411503. [PMID: 39932442 PMCID: PMC11967767 DOI: 10.1002/advs.202411503] [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: 09/18/2024] [Revised: 01/09/2025] [Indexed: 04/05/2025]
Abstract
Targeting oxidative phosphorylation (OXPHOS) through inhibiting the electron transport chain (ETC) has shown promising pre-clinical efficacy in cancer therapy. Although aerobic glycolysis is a hallmark of cancer, emerging evidence suggest OXPHOS is frequently enhanced, providing metabolic advantages for cell proliferation, metastasis, and drug resistance in a variety of aggressive cancer types including non-small cell lung cancer (NSCLC), yet the underlying molecular mechanisms remain elusive. Here it is reported that POU-domain containing family protein POU3F3 is translocated into the nuclei of NSCLC cell lines harboring mutant RAS, where it activates transcription of ATP5PF, an essential component of mitochondrial ATP synthase and consequent ATP production, leading to enhanced NSCLC proliferation and migration. Moreover, it is further found out that ERK1 phosphorylates POU3F3 at the S393 site in the cytoplasm and promotes the nuclear translocation of POU3F3 via receptor importin β1 in RAS mutant NSCLC cells. Mechanistically, RNA sequencing analysis combined with chromatin immunoprecipitation (ChIP) assay revealed that POU3F3 binds to the promoter of ATP5PF, leading to enhanced ATP5PF transcription and ATP production. Together, this study uncovers a novel RAS-POU3F3-ATP5PF axis in facilitating NSCLC progression, providing a new perspective on the understanding of molecular mechanisms for NSCLC progression.
Collapse
Affiliation(s)
- Qi‐Gang Zeng
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Le Li
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Tao Chang
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Yong Sun
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Bin Zheng
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Ling‐Na Xue
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Chao‐Ling Liu
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Xia‐Qing Li
- Institute of Nephrology and Blood PurificationThe First Affiliated HospitalJinan UniversityGuangdong510632China
- Nephrology departmentThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityGuangdong517000China
| | - Ruo‐Tong Huang
- Department of Metabolism, Digestion, and ReproductionFaculty of MedicineImperial College LondonLondonW12 0NNUK
| | - Jia‐Xin Gu
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Zhao‐Rong An
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Hao‐Tao Yao
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Dan‐Yang Zhou
- Department of RespiratoryNanjing First HospitalNanjing Medical UniversityJiangsu210012China
| | - Jun Fan
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Yong Dai
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| |
Collapse
|
30
|
Gore M, Kabekkodu SP, Chakrabarty S. Exploring the metabolic alterations in cervical cancer induced by HPV oncoproteins: From mechanisms to therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189292. [PMID: 40037419 DOI: 10.1016/j.bbcan.2025.189292] [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: 09/12/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Collapse
Affiliation(s)
- Mrudula Gore
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
31
|
Liu C, Pan S, Pan X, Yang J, Yao H, Yang Z, Hao S, Liu Y, Liu P, Zhang S. High-throughput single-cell metabolites profiling reveals metabolic reprogramming confers cisplatin resistance in lung cancer. Talanta 2025; 285:127355. [PMID: 39673986 DOI: 10.1016/j.talanta.2024.127355] [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: 10/10/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
Lung cancer is the most common cause of cancer-related deaths worldwide. Platinum-containing two-drug regimens are the standard first-line chemotherapeutic regimen, but acquired resistance remains a major challenge. Cancer cells can evolve and adapt to therapeutic stress by reprogramming their metabolism and passing on drug resistance to neighboring drug-sensitive cancer cells through cell-to-cell interactions. Here, we have developed a method to study the interactions between cells. Using human lung cancer A549 cells, we constructed a drug-sensitive cell line expressing red fluorescent protein and a cisplatin-resistant cell line. Employing label-free mass cytometry, we acquired metabolites information at the single-cell level. Through pseudotime analysis, we identified two most important clusters of metabolites. We discovered that phosphatidylcholines are strongly associated with drug resistance. Through unsupervised learning, we observed that drug-sensitive cells in co-culture transform into a novel cell state after cisplatin treatment. This method offers a novel tool for investigating the mechanisms underlying the development of cancer cell drug resistance.
Collapse
Affiliation(s)
- Changyi Liu
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, PR China
| | - Siyuan Pan
- Department of Chemistry, Tsinghua University, Beijing, 100084, PR China
| | - Xingyu Pan
- Department of Chemistry, Tsinghua University, Beijing, 100084, PR China
| | - Jinlei Yang
- Department of Chemistry, Tsinghua University, Beijing, 100084, PR China
| | - Huan Yao
- Division of Chemical Metrology and Analytical Science, National Institute of Metrology, Beijing, 100029, PR China
| | - Zhenli Yang
- Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100730, PR China
| | - Sijia Hao
- Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100730, PR China
| | - Yuqin Liu
- Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100730, PR China
| | - Peng Liu
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, PR China.
| | - Sichun Zhang
- Department of Chemistry, Tsinghua University, Beijing, 100084, PR China.
| |
Collapse
|
32
|
O’Keefe S, Wang Q. ACAT1 regulates tertiary lymphoid structures: A target for enhancing immunotherapy in non-small cell lung cancer. J Clin Invest 2025; 135:e191094. [PMID: 40166937 PMCID: PMC11957684 DOI: 10.1172/jci191094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Non-small cell lung cancer (NSCLC), the most common type of lung cancer, remains a leading cause of cancer-related mortality worldwide. Immune checkpoint inhibitors (ICIs) have emerged as a promising therapy for NSCLC but only benefit a subset of patients. In this issue of the JCI, Jiao et al. revealed that acetyl-CoA acetyltransferase 1 (ACAT1) limited the efficacy of ICIs in NSCLC by impeding tertiary lymphoid structures (TLS) in the tumor microenvironment (TME). Targeting ACAT1 in tumor cells reduced mitochondrial hypersuccinylation and oxidative stress, enhancing TLS abundance and improving the efficacy of ICIs in preclinical murine models of NSCLC.
Collapse
Affiliation(s)
- Sophie O’Keefe
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- University of Virginia Comprehensive Cancer Center, Charlottesville, Virginia, USA
| | - Qiwei Wang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- University of Virginia Comprehensive Cancer Center, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
33
|
Millman SE, Chaves-Perez A, Janaki-Raman S, Ho YJ, Morris JP, Narendra V, Chen CC, Jackson BT, Yashinskie JJ, Mezzadra R, Devine TI, Barthet VJA, Saoi M, Baslan T, Tian S, Sachs Z, Finley LWS, Cross JR, Lowe SW. α-Ketoglutarate dehydrogenase is a therapeutic vulnerability in acute myeloid leukemia. Blood 2025; 145:1422-1436. [PMID: 39791576 PMCID: PMC11969269 DOI: 10.1182/blood.2024025245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/22/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Perturbations in intermediary metabolism contribute to the pathogenesis of acute myeloid leukemia (AML) and can produce therapeutically actionable dependencies. Here, we probed whether α-ketoglutarate (αKG) metabolism represents a specific vulnerability in AML. Using functional genomics, metabolomics, and mouse models, we identified the αKG dehydrogenase complex, which catalyzes the conversion of αKG to succinyl coenzyme A, as a molecular dependency across multiple models of adverse-risk AML. Inhibition of 2-oxoglutarate dehydrogenase (OGDH), the E1 subunit of the αKG dehydrogenase complex, impaired AML progression and drove differentiation. Mechanistically, hindrance of αKG flux through the tricarboxylic acid (TCA) cycle resulted in rapid exhaustion of aspartate pools and blockade of de novo nucleotide biosynthesis, whereas cellular bioenergetics was largely preserved. Additionally, increased αKG levels after OGDH inhibition affected the biosynthesis of other critical amino acids. Thus, this work has identified a previously undescribed, functional link between certain TCA cycle components and nucleotide biosynthesis enzymes across AML. This metabolic node may serve as a cancer-specific vulnerability, amenable to therapeutic targeting in AML and perhaps in other cancers with similar metabolic wiring.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors
- Ketoglutarate Dehydrogenase Complex/metabolism
- Ketoglutarate Dehydrogenase Complex/genetics
- Animals
- Mice
- Humans
- Ketoglutaric Acids/metabolism
- Citric Acid Cycle
Collapse
Affiliation(s)
- Scott E. Millman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Almudena Chaves-Perez
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sudha Janaki-Raman
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yu-Jui Ho
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John P. Morris
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Varun Narendra
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Chi-Chao Chen
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | - Riccardo Mezzadra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Tessa I. Devine
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Valentin J. A. Barthet
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michelle Saoi
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sha Tian
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Zohar Sachs
- Division of Hematology, Oncology, and Transplantation, Department of Medicine and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Lydia W. S. Finley
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Justin R. Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Scott W. Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
- Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
34
|
Liu X, Liu B, Wang J, Liu H, Wu J, Qi Y, Liu Y, Zhu H, Li C, Yang L, Song J, Yao G, Tian W, Zhao K, Han L, Shu K, Zhang S, Man J, You C, Huang H, Li R. PHGDH activation fuels glioblastoma progression and radioresistance via serine synthesis pathway. J Exp Clin Cancer Res 2025; 44:99. [PMID: 40102981 PMCID: PMC11921657 DOI: 10.1186/s13046-025-03361-3] [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: 01/12/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Glioma stem-like cells (GSCs) are key drivers of treatment resistance and recurrence in glioblastoma (GBM). Phosphoglycerate dehydrogenase (PHGDH), a crucial enzyme in the de novo serine synthesis pathway (SSP), is implicated in tumorigenesis and therapy resistance across various cancers. However, its specific role in GBM, particularly in radioresistance, remains poorly understood. METHODS In silico analysis of GBM patient data assessed SSP enrichment and PHGDH expression linked with tumor stemness. Comparative gene expression analysis focused on PHGDH in paired GBM specimens and GSCs. Genetic and pharmacological loss-of-function assays were performed in vitro and in vivo to evaluate PHGDH's impact on GSC self-renewal and malignant progression. Comprehensive transcriptomic and metabolomic analyses, along with chromatin immunoprecipitation, mass spectrometry, and various other biochemical assays, were used to elucidate PHGDH-mediated mechanisms in GBM progression and radioresistance. RESULTS PHGDH expression is significantly elevated in GSCs, associated with aggressive glioma progression and poor clinical outcomes. PHGDH activation enhances GSC self-renewal by regulating redox homeostasis, facilitating one-carbon metabolism, and promoting DNA damage response via SSP activation. Importantly, MYC was identified as a crucial transcriptional regulator of PHGDH expression. Furthermore, genetic ablation or pharmacological inhibition of PHGDH markedly reduced tumor growth and increased tumor sensitivity to radiotherapy, thereby improving survival outcomes in orthotopic GSC-derived and patient-derived GBM xenograft models. CONCLUSIONS This study underscores the pivotal role of MYC-mediated PHGDH activation in driving GSC malignant progression and radioresistance in GBM. Targeting PHGDH presents a promising approach to enhance radiotherapy efficacy in GBM patients.
Collapse
Affiliation(s)
- Xiaojin Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bangxin Liu
- Department of Neurosurgery, General Hospital of Central Theatre Command of People's Liberation Army, Wuhan, Hubei, China
| | - Junwen Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongbin Liu
- Department of Neurosurgery, General Hospital of Central Theatre Command of People's Liberation Army, Wuhan, Hubei, China
- Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jiasheng Wu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yiwei Qi
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuan Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chaoxi Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liu Yang
- Department of Neurosurgery, General Hospital of Central Theatre Command of People's Liberation Army, Wuhan, Hubei, China
| | - Jian Song
- Department of Neurosurgery, General Hospital of Central Theatre Command of People's Liberation Army, Wuhan, Hubei, China
| | - Guojie Yao
- Department of Neurosurgery, General Hospital of Central Theatre Command of People's Liberation Army, Wuhan, Hubei, China
| | - Weidong Tian
- Department of Neurosurgery, The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Kai Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lin Han
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Suojun Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Chao You
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Haohao Huang
- Department of Neurosurgery, General Hospital of Central Theatre Command of People's Liberation Army, Wuhan, Hubei, China.
- General Hospital of Central Theater Command and Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei, China.
| | - Ran Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
35
|
Cui B, Tu S, Li H, Zeng Z, Xiao R, Guo J, Liang X, Liu C, Pan L, Chen W, Ge M, Zhong X, Ye L, Chen H, Zhang Q, Xu Y. METTL3 knockout accelerates hepatocarcinogenesis via inhibiting endoplasmic reticulum stress response. FEBS Open Bio 2025. [PMID: 40103332 DOI: 10.1002/2211-5463.70023] [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: 09/21/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is among the most common causes of cancer-related deaths worldwide. Previous studies showed that N6-methyladenosine (m6A), the most abundant chemical modification in eukaryotic RNAs, is implicated in HCC progression. Using liver-specific conditional knockout mice, we found that the loss of METTL3, the core catalytic subunit of m6A methyltransferase, significantly promoted hepatic tumor initiation under various oncogenic challenges, contrary to the previously reported oncogenic role of METTL3 in liver cancer cell lines or xenograft models. Mechanistically, we hypothesized that METTL3 deficiency accelerated HCC initiation by inhibiting m6A deposition on MANF transcripts, impairing nuclear export and thus MANF protein levels, which led to insufficient endoplasmic reticulum (ER) stress response pathway activation. Our findings suggest a tumor-suppressive role for METTL3 in the early stages of HCC, emphasizing the importance of understanding the dynamic role of epigenetic regulation in tumorigenesis and targeted therapy.
Collapse
Affiliation(s)
- Bo Cui
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Silin Tu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haibo Li
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhancheng Zeng
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruiqi Xiao
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Guo
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoqi Liang
- Cell-Gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chang Liu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lijie Pan
- Laboratory Animal Center, Sun Yat-sen University, Guangzhou, China
| | - Wenjie Chen
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Cell-Gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mian Ge
- Department of Anesthesiology, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofen Zhong
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linsen Ye
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huaxin Chen
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qi Zhang
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Cell-Gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Xu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
36
|
Wang M, Xu S, Xu J, Wei J, Wu Y. WTAP contributes to platinum resistance in high-grade serous ovarian cancer by up-regulating malic acid: insights from liquid chromatography and mass spectrometry analysis. Cancer Metab 2025; 13:14. [PMID: 40098185 PMCID: PMC11916999 DOI: 10.1186/s40170-025-00383-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
High-grade serous cancer (HGSC) is the most prevalent and aggressive subtype of ovarian cancer. In this study, we utilized liquid chromatography and mass spectrometry analysis to investigate metabolic alterations in HGSC. Among the 1353 metabolites identified, 140 were significantly differed between HGSC and normal ovarian tissue. KEGG pathway enrichment analysis revealed 23 distinct metabolic pathways, including the alanine/aspartate/glutamate metabolism, pyruvate metabolism, biosynthesis of amino acids, and citrate cycle, etc. Of the significantly differentiated metabolites, malic acid, fumarate, and phosphoenolpyruvate were found in the citrate cycle and glycolysis. In further analysis, 22 differentially expressed genes (DEGs) of glucose metabolism were found between HGSC and normal controls. Multivariate Cox analysis of the 22 DEGs showed that ME1, ALDOC, and RANBP2 were associated with overall survival in the TCGA cohort.Bioinformatic analysis indicated WTAP is strongly correlated to the expression of ME1, which is a rate-limiting enzyme that regulates the shuttle of malic acid in mitochondria and cytoplasm. After the knockdown of WTAP in A2780 and OVCAR-3 cells, the activity of the malic enzyme decreased which led to the accumulation of malic acid and citric acid, and the reduction of pyruvate and lactic acid. In A2780 and OVCAR-3 cells, the IC50 to platinum was increased after the knockdown of WTAP. After the knockdown of WTAP, the expression of ME1 was down-regulated and the m6A modification was down-regulated in ovarian cell lines. On the SRAMP website, there were two binding sites with high m6A scores at the 5 '-UTR 177 and 970 of ME1 mRNA. WTAP contributes to the platinum resistance through regulating the conversion from aerobic glycolysis to OXPHOS by upregulating the expression of ME1.
Collapse
Affiliation(s)
- Ming Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Shuiqing Xu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Jianqing Xu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Jiahui Wei
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Yumei Wu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China.
| |
Collapse
|
37
|
Li S, Lu Z, Jiang W, Xu Y, Chen R, Wang J, Jiao B, Lu X. Chaetocin, a Natural Inhibitor of Transketolase, Suppresses the Non-Oxidative Pentose Phosphate Pathway and Inhibits the Growth of Drug-Resistant Non-Small Cell Lung Cancer. Antioxidants (Basel) 2025; 14:330. [PMID: 40227333 PMCID: PMC11939327 DOI: 10.3390/antiox14030330] [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: 12/26/2024] [Revised: 02/16/2025] [Accepted: 03/09/2025] [Indexed: 04/15/2025] Open
Abstract
Worldwide, lung cancer is the most common cause of cancer-related death, which is made worse by the development of drug resistance during treatment. It is urgent to develop new therapeutic methods and small molecule drugs for tumor resistance. Chaetocin, extracted from Chaetomium minutum, is a natural compound with good antitumor activity. However, there are few studies on its tumor resistance. In this paper, firstly, chaetotocin significantly inhibited the viability and migration of cisplatin-resistant non-small cell lung cancer (NSCLC) cells and inhibited the xenograft growth of nude mice. Chaetocin at 4 mg/kg significantly inhibited A549/DDP xenograft growth with an inhibition rate of 70.43%. Subsequently, the underlying mechanism behind the actions of chaetocin was explored. It was discovered that chaetocin can inhibit transketolase (TKT), thereby inhibiting the growth of NSCLC cells and inducing cell death. Compared with cisplatin-sensitive cells, a lower concentration of chaetocin can inhibit cisplatin-resistance cell viability and migration. Mechanistically, TKT was identified as a potential target for chaetocin. The KD value of the interaction between chaetocin and TKT was 63.2 μM. An amount of 0.2 μM chaetocin may suppress the enzyme activity and expression level of TKT. We found the TKT expression is higher in cisplatin-resistant cells, which further explains why these cells were more vulnerable to chaetocin in terms of cell phenotype. Additionally, the muti-omics analysis and RNA interference suggested that chaetocin can inhibit the PI3K/Akt signaling pathway through TKT. In conclusion, chaetocin could directly bind to TKT, inhibiting its enzyme activity and expression, which interfered with intracellular metabolism and oxidation-reduction balance, and then regulated the PI3K/Akt signaling pathway to inhibit the growth of NSCLC and induce apoptosis.
Collapse
Affiliation(s)
- Song Li
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Zhanying Lu
- Experimental Training Center of Basic Medical Science, Naval Medical University, Shanghai 200433, China;
| | - Wenli Jiang
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Yao Xu
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Ran Chen
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Jie Wang
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Binghua Jiao
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Xiaoling Lu
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| |
Collapse
|
38
|
Moi D, Carradori S, Gallorini M, Mencarelli N, Deplano A, Angeli A, Vittorio S, Supuran CT, Onnis V. Investigation on Human Carbonic Anhydrase IX and XII Inhibitory Activity and A549 Antiproliferative Activity of a New Class of Coumarinamides. Pharmaceuticals (Basel) 2025; 18:372. [PMID: 40143148 PMCID: PMC11944513 DOI: 10.3390/ph18030372] [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: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
Background-Aggressive solid tumors are commonly characterized by both basic intracellular pH and acidic extracellular pH, which increase cell survival and proliferation. As carbonic anhydrases IX/XII are involved in this pH regulation, their inhibition is an appealing approach in cancer therapy, avoiding cancer cell survival and proliferation. Substituted coumarins are selective non-classical CA IX and CA XII inhibitors. Methods-In this study, new 7-hydroxycoumarinamides were synthesized and assayed for CA inhibition and antiproliferative activity. Results-All of the coumarinamides showed human CA IX and CA XII selective inhibition over the off-target CA I and CA II isoforms. Coumarin acts as a suicide inhibitor because its heterocyclic ring can be hydrolyzed by CA esterase activity to give the corresponding 2-hydroxycinnamic acid derivative which blocks the entrance of the active site. The 2-hydroxycinnamic acid derivatives deriving from the most potent and selective coumarinamides were docked into CA IX and XII to better understand the activity and selectivity against the two CA isoforms. The most active coumarinamides also produced a decrease of A549 cell proliferation and were able to arrest cells at the G1/S checkpoint. Conclusions-These results may open new perspectives for developing coumarin-based CA IX/XII inhibitors.
Collapse
Affiliation(s)
- Davide Moi
- Department of Life and Environmental Sciences, Unit of Pharmaceuitical, Pharmacological and Nutraceutical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, I-09042 Monserrato, CA, Italy; (D.M.); (A.D.)
| | - Simone Carradori
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, I-66100 Chieti, CH, Italy; (S.C.); (M.G.); (N.M.)
| | - Marialucia Gallorini
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, I-66100 Chieti, CH, Italy; (S.C.); (M.G.); (N.M.)
| | - Noemi Mencarelli
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, I-66100 Chieti, CH, Italy; (S.C.); (M.G.); (N.M.)
| | - Alberto Deplano
- Department of Life and Environmental Sciences, Unit of Pharmaceuitical, Pharmacological and Nutraceutical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, I-09042 Monserrato, CA, Italy; (D.M.); (A.D.)
| | - Andrea Angeli
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, I-50019 Sesto Fiorentino, FI, Italy; (A.A.); (C.T.S.)
| | - Serena Vittorio
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, I-20133 Milano, MI, Italy;
| | - Claudiu T. Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, I-50019 Sesto Fiorentino, FI, Italy; (A.A.); (C.T.S.)
| | - Valentina Onnis
- Department of Life and Environmental Sciences, Unit of Pharmaceuitical, Pharmacological and Nutraceutical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, I-09042 Monserrato, CA, Italy; (D.M.); (A.D.)
| |
Collapse
|
39
|
Jacome MA, Wu Q, Chen J, Mohamed ZS, Mokhtari S, Piña Y, Etame AB. Molecular Underpinnings of Brain Metastases. Int J Mol Sci 2025; 26:2307. [PMID: 40076927 PMCID: PMC11900073 DOI: 10.3390/ijms26052307] [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: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Brain metastases are the most commonly diagnosed type of central nervous system tumor, yet the mechanisms of their occurrence are still widely unknown. Lung cancer, breast cancer, and melanoma are the most common etiologies, but renal and colorectal cancers have also been described as metastasizing to the brain. Regardless of their origin, there are common mechanisms for progression to all types of brain metastases, such as the creation of a suitable tumor microenvironment in the brain, priming of tumor cells, adaptations to survive spreading in lymphatic and blood vessels, and development of mechanisms to penetrate the blood-brain barrier. However, there are complex genetic and molecular interactions that are specific to every type of primary tumor, making the understanding of the metastatic progression of tumors to the brain a challenging field of study. In this review, we aim to summarize current knowledge on the pathophysiology of brain metastases, from specific genetic characteristics of commonly metastatic tumors to the molecular and cellular mechanisms involved in progression to the central nervous system. We also briefly discuss current challenges in targeted therapies for brain metastases and how there is still a gap in knowledge that needs to be overcome to improve patient outcomes.
Collapse
Affiliation(s)
- Maria A. Jacome
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Jianan Chen
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | | | - Sepideh Mokhtari
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Yolanda Piña
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| |
Collapse
|
40
|
Lian C, He M, Zhao C, Wang T, Tong F, Chen J, Ju R. PLIN2: a potential prognostic markers of early-stage atypical endometrial hyperplasia. J Gynecol Oncol 2025; 36:36.e84. [PMID: 40114556 DOI: 10.3802/jgo.2025.36.e84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/04/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE In the background of endometrial hyperplasia triggered by obesity and estrogen, could the perilipin 2 (PLIN2) serve as a possible prognostic marker for early atypical endometrial hyperplasia (AEH)? METHODS A retrospective study examined blood lipid levels in endometrial cancer (EC) or AEH patients. An AEH mice model was established administrating with estradiol and/or high-fat (HF) diet. Hematoxylin and eosin staining were employed to assess pathological changes in the endometrium. Immunohistochemical staining were employed to evaluate the expression of adipose metabolism and endometrial hyperplasia proteins. The Cell Counting Kit-8 assay, cell colony-forming assays, and western blotting were utilized to verify the impact of oleic acid (OA) on HEC-1A cells. RESULTS The retrospective study revealed elevated blood lipid levels among patients with EC or AEH. Prolonged HF diet stimulated the endometrium to exhibit features of complex atypical hyperplasia. In the early stage, PLIN2 (p=0.006) expression significantly increased with endometrial glandular hyperplasia. Both PLIN2 (p=0.008) and progesterone receptor (PR; p=0.019) exhibited elevated expression concurrent with simple endometrial hyperplasia. When AEH occurred, there were notable rise in the expression of PLIN2 (p<0.001), PR (p=0.044), and estrogen receptor (p=0.045). The atypical hyperplasia glands demonstrated notably elevated PLIN2 expression in comparison to surrounding normal glands in AEH lesions. OA was found to enhance the proliferation and clonal formation of HEC-1A cells. HEC-1A cells induced by OA demonstrated elevated autophagy levels accompanied by enhanced expression of PLIN2. CONCLUSION PLIN2 may potentially serve as a biomarker for early development of AEH and EC, facilitating diagnosis and intervention and contributing to the determination of prognosis.
Collapse
Affiliation(s)
- Chao Lian
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Minling He
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Chengcheng Zhao
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Tianming Wang
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Fang Tong
- Department of Pathology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China.
| | - Rong Ju
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
41
|
Xu A, Li X, Cai Q, Yang C, Yang M, Gao H, Cheng M, Chen X, Ji F, Tang H, Wang K. CircXPO6 promotes breast cancer progression through competitively inhibiting the ubiquitination degradation of c-Myc. Mol Cell Biochem 2025; 480:1731-1745. [PMID: 39179754 DOI: 10.1007/s11010-024-05093-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: 03/28/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024]
Abstract
The number of breast cancer (BC) patients is increasing year by year, which is severely endangering to human life and health. c-Myc is a transcription factor, studies have shown that it is a very significant factor in tumor progression, but how it is regulated in BC is still not well understood. Here, we used the RIP microarray sequencing to confirm circXPO6, which had a high affinity with c-Myc and highly expressed in triple-negative breast cancer (TNBC) tissues and cells. CircXPO6 overexpression promoted tumor growth in vivo and in vitro. Furthermore, circXPO6 largely promoted the expression of genes related to glucose metabolism, such as GLUT1, HK2, and MCT4 in TNBC cells. Finally, high levels of circXPO6 expression were found to be closely associated with malignant pathological factors, such as tumor size, lymph node metastasis, TNM staging, and histopathological grading of TNBC. Mechanistically, circXPO6 interacted with c-Myc to prevent speckle-type POZ-mediated c-Myc ubiquitination and degradation, thus promoting TNBC progression. Through the regulation of c-Myc-mediated signal transduction, circXPO6 plays a key role in TNBC progresses. This discovery can provide new ideas for TNBC molecular targeted therapy.
Collapse
Affiliation(s)
- Aiqi Xu
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Xi Li
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, Guangzhou, 510180, People's Republic of China
| | - Qiaoting Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ciqiu Yang
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Mei Yang
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Hongfei Gao
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Minyi Cheng
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Xianzhe Chen
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Fei Ji
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, People's Republic of China.
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Kun Wang
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
42
|
Seidel LM, Thudium J, Smith C, Sapehia V, Sommer N, Wujak M, Weissmann N, Seeger W, Schermuly RT, Novoyatleva T. Death-associated protein kinase 1 prevents hypoxia-induced metabolic shift and pulmonary arterial smooth muscle cell proliferation in PAH. Cell Signal 2025; 127:111527. [PMID: 39622428 DOI: 10.1016/j.cellsig.2024.111527] [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: 03/22/2024] [Revised: 10/31/2024] [Accepted: 11/20/2024] [Indexed: 12/08/2024]
Abstract
Pulmonary hypertension (PH) is a general term used to describe high blood pressure in the lungs from any cause. Pulmonary arterial hypertension (PAH) is a progressive, and fatal disease that causes the walls of the pulmonary arteries to tighten and stiffen. One of the major characteristics of PAH is the hyperproliferation and resistance to apoptosis of vascular cells, which trigger excessive pulmonary vascular remodeling and vasoconstriction. The death-associated protein DAP-kinase (DAPK) is a tumor suppressor and Ser/Thr protein kinase, which was previously shown to regulate the hypoxia inducible factor (HIF)-1α. Against this background, we now show that DAPK1 regulates human pulmonary arterial smooth muscle cell (hPASMC) proliferation and energy metabolism in a HIF-dependent manner. DAPK1 expression is downregulated in pulmonary vessels and PASMCs of human and experimental PH lungs. Reduced expression of DAPK1 in hypoxia and non-hypoxia PAH-PASMCs correlates with increased expression of HIF-1/2α. RNA interference-mediated depletion of DAPK1 leads to fundamental metabolic changes, including a significantly decreased rate of oxidative phosphorylation associated with enhanced expression of both HIF-1α and HIF-2α and glycolytic enzymes, as hexokinase 2 (HK2), lactate dehydrogenase A (LDHA), and an integrator between the glycolysis and citric acid cycle, pyruvate dehydrogenase kinase 1 (PDK1). DAPK1 ablation in healthy donor hPASMCs leads to an increase in proliferation, while its overexpression provides the opposite effects. Together our data indicate that DAPK1 serves as a new inhibitor of the pro-proliferative and glycolytic phenotype of PH in PASMCs acting via HIF-signaling pathway.
Collapse
MESH Headings
- Death-Associated Protein Kinases/metabolism
- Death-Associated Protein Kinases/genetics
- Death-Associated Protein Kinases/antagonists & inhibitors
- Humans
- Cell Proliferation
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Cell Hypoxia
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/metabolism
- Glycolysis
- Cells, Cultured
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Hexokinase/metabolism
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase
Collapse
Affiliation(s)
- Laura-Marie Seidel
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Jana Thudium
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Caroline Smith
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Vandna Sapehia
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Natascha Sommer
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Magdalena Wujak
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany; Department of Medicinal Chemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Norbert Weissmann
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Werner Seeger
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute for Lung Health, Giessen, Germany
| | - Ralph T Schermuly
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Tatyana Novoyatleva
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany.
| |
Collapse
|
43
|
Lin DW, Carranza FG, Borrego S, Lauinger L, Dantas de Paula L, Pulipelli HR, Andronicos A, Hertel KJ, Kaiser P. Nutrient control of splice site selection contributes to methionine addiction of cancer. Mol Metab 2025; 93:102103. [PMID: 39862967 PMCID: PMC11834112 DOI: 10.1016/j.molmet.2025.102103] [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: 10/08/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Many cancer cells depend on exogenous methionine for proliferation, whereas non-tumorigenic cells can divide in media supplemented with the metabolic precursor homocysteine. This phenomenon is known as methionine dependence of cancer or methionine addiction. The underlying mechanisms driving this cancer-specific metabolic addiction are poorly understood. Here we find that methionine dependence is associated with severe dysregulation of pre-mRNA splicing. METHODS We used triple-negative breast cancer cells and their methionine-independent derivatives R8 to compare RNA expression profiles in methionine and homocysteine growth media. The data set was also analyzed for alternative splicing. RESULTS When tumorigenic cells were cultured in homocysteine medium, cancer cells failed to efficiently methylate the spliceosomal snRNP component SmD1, which resulted in reduced binding to the Survival-of-Motor-Neuron protein SMN leading to aberrant splicing. These effects were specific for cancer cells as neither Sm protein methylation nor splicing fidelity was affected when non-tumorigenic cells were cultured in homocysteine medium. Sm protein methylation is catalyzed by Protein Arginine Methyl Transferase 5 (Prmt5). Reducing methionine concentrations in the culture medium sensitized cancer cells to Prmt5 inhibition supporting a mechanistic link between methionine dependence of cancer and splicing. CONCLUSIONS Our results link nutritional demands to splicing changes and thereby provide a link between the cancer-specific metabolic phenomenon, described as methionine addiction over 40 years ago, with a defined cellular pathway that contributes to cancer cell proliferation.
Collapse
Affiliation(s)
- Da-Wei Lin
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Francisco G Carranza
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, USA
| | - Stacey Borrego
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Linda Lauinger
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Lucas Dantas de Paula
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Harika R Pulipelli
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, USA
| | - Anna Andronicos
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Klemens J Hertel
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, USA.
| | - Peter Kaiser
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA.
| |
Collapse
|
44
|
Li X, Zhu XH, Li Y, Wang T, Zhang G, Wiesner HM, Liang ZP, Chen W. Quantitative mapping of key glucose metabolic rates in the human brain using dynamic deuterium magnetic resonance spectroscopic imaging. PNAS NEXUS 2025; 4:pgaf072. [PMID: 40109558 PMCID: PMC11922071 DOI: 10.1093/pnasnexus/pgaf072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
Deuterium (2H) magnetic resonance spectroscopic imaging (DMRSI) is a newly developed technology for assessing glucose metabolism by simultaneously measuring deuterium-labeled glucose and its downstream metabolites (1) and has a potential to provide a powerful neurometabolic imaging tool for quantitative studies of cerebral glucose metabolism involving multiple metabolic pathways in the human brain. In this work, we developed a dynamic DMRSI method that combines advanced radiofrequency coil and postprocessing techniques to substantially improve the imaging signal-to-noise ratio for detecting deuterated metabolites and enable robust dynamic DMRSI of the human brain at 7 T with very high resolution (HR; 0.7 cc nominal voxel and 2.5 min/image) and whole-brain coverage. Utilizing this capability, we were able to map and differentiate metabolite contents and dynamics throughout the human brain following oral administration of deuterated glucose. Furthermore, by introducing a sophisticated kinetic model, we demonstrated that three key cerebral metabolic rates of glucose consumption (CMRGlc), lactate production (CMRLac), and tricarboxylic acid (TCA) cycle (V TCA), as well as the maximum apparent rate of forward glucose transport (T max) can be simultaneously imaged in the human brain through a single dynamic DMRSI measurement. The results clearly show that the glucose transport, neurotransmitter turnover, CMRGlc, and V TCA are significantly higher in gray matter than in white matter in the human brain; and the mean metabolic rates and their ratios measured in this study are consistent with the values reported in the literature. The HR dynamic DMRSI methodology presented herein is of great significance and value for the quantitative assessment of human brain glucose metabolism, aerobic glycolysis, and metabolic reprogramming under physiopathological conditions.
Collapse
Affiliation(s)
- Xin Li
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Xiao-Hong Zhu
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Yudu Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tao Wang
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Guangle Zhang
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Hannes M Wiesner
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Zhi-Pei Liang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Wei Chen
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
45
|
Li J, Huang K, Thakur M, McBride F, Sadagopan A, Gallant DS, Khanna P, Laimon YN, Li B, Mohanna R, Ge M, Weiss CN, Achom M, Xu Q, Matar S, Lee GSM, Huang K, Gui M, Wu CL, Cornejo KM, Choueiri TK, Ryback BA, Signoretti S, Bar-Peled L, Viswanathan SR. Oncogenic TFE3 fusions drive OXPHOS and confer metabolic vulnerabilities in translocation renal cell carcinoma. Nat Metab 2025; 7:478-492. [PMID: 39915638 DOI: 10.1038/s42255-025-01218-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/09/2025] [Indexed: 02/12/2025]
Abstract
Translocation renal cell carcinoma (tRCC) is an aggressive subtype of kidney cancer driven by TFE3 gene fusions, which act via poorly characterized downstream mechanisms. Here we report that TFE3 fusions transcriptionally rewire tRCCs toward oxidative phosphorylation (OXPHOS), contrasting with the highly glycolytic nature of most other renal cancers. Reliance on this TFE3 fusion-driven OXPHOS programme renders tRCCs vulnerable to NADH reductive stress, a metabolic stress induced by an imbalance of reducing equivalents. Genome-scale CRISPR screening identifies tRCC-selective vulnerabilities linked to this metabolic state, including EGLN1, which hydroxylates HIF-1α and targets it for proteolysis. Inhibition of EGLN1 compromises tRCC cell growth by stabilizing HIF-1α and promoting metabolic reprogramming away from OXPHOS, thus representing a vulnerability for OXPHOS-dependent tRCC cells. Our study defines tRCC as being dependent on a mitochondria-centred metabolic programme driven by TFE3 fusions and nominates EGLN1 inhibition as a therapeutic strategy in this cancer.
Collapse
Affiliation(s)
- Jiao Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kaimeng Huang
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Meha Thakur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fiona McBride
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ananthan Sadagopan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Daniel S Gallant
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Prateek Khanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Bingchen Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Razan Mohanna
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Maolin Ge
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Cary N Weiss
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mingkee Achom
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Qingru Xu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sayed Matar
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Gwo-Shu Mary Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kun Huang
- Molecular Imaging Core and Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Miao Gui
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine and Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Kristine M Cornejo
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Birgitta A Ryback
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Liron Bar-Peled
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
| | - Srinivas R Viswanathan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
46
|
Nair R, Vu AH, Freer AK, Bhatia KS, Wang D, Savani MR, Matulis SM, Lonial S, Jaye DL, Boise LH, Seo SY, Corson TW, Nooka AK, Bhatt S, McBrayer SK, Gupta VA, Hu X, Barwick BG, Reddi AR, Shanmugam M. Heme promotes venetoclax resistance in multiple myeloma through MEK-ERK signaling and purine biosynthesis. Blood 2025; 145:732-747. [PMID: 39693611 PMCID: PMC12060166 DOI: 10.1182/blood.2024025690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/28/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
ABSTRACT We previously demonstrated that reduced intrinsic electron transport chain (ETC) activity predicts and promotes sensitivity to the B-cell lymphoma 2 (BCL-2) antagonist, venetoclax (Ven), in multiple myeloma (MM). Heme, an iron-containing prosthetic group and metabolite, is fundamental to maintaining ETC activity. Interrogation of the cyclin D1 group 2 subgroup of MM from the Relating Clinical Outcomes in MM to Personal Assessment of Genetic Profile (CoMMpass) trial (NCT01454297), which can be used as a proxy for Ven-sensitive MM (VS MM), shows reduced expression of the conserved heme biosynthesis pathway gene signature. Consistent with this, we identified that VS MM exhibits reduced heme biosynthesis and curiously elevated hemin (oxidized heme) uptake. Supplementation with hemin or protoporphyrin IX (heme lacking iron) promotes Ven resistance, whereas targeting ferrochetalase, the penultimate enzyme involved in heme biosynthesis, increases Ven sensitivity in cell lines and primary MM cells. Mechanistically, heme-mediated activation of prosurvival rapidly accelerated fibrosarcoma-rat sarcoma virus-mitogen-activated protein kinase (MEK) signaling and metabolic rewiring, increasing de novo purine synthesis, were found to contribute to heme-induced Ven resistance. Cotargeting BCL-2 and myeloid cell leukemia-1 suppresses heme-induced Ven resistance. Interrogation of the Multiple Myeloma Research Foundation CoMMpass study of patients shows increased purine and pyrimidine biosynthesis to corelate with poor progression-free survival and overall survival. Elevated heme and purine biosynthesis gene signatures were also observed in matched relapse refractory MM, underscoring the relevance of heme metabolism in therapy-refractory MM. Overall, our findings reveal, for the first time, a role for extrinsic heme, a physiologically relevant metabolite, in modulating proximity to the apoptotic threshold with translational implications for BCL-2 antagonism in MM therapy.
Collapse
Affiliation(s)
- Remya Nair
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - An H. Vu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Abigail K. Freer
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA
- Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Karanpreet S. Bhatia
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Dongxue Wang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Milan R. Savani
- Children's Medical Center Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
- Medical Scientist Training Program, University of Texas Southwestern Medical Center, Dallas, TX
| | - Shannon M. Matulis
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - David L. Jaye
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Timothy W. Corson
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Ajay K. Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore
| | - Samuel K. McBrayer
- Children's Medical Center Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
- Medical Scientist Training Program, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Vikas A. Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Xin Hu
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Amit R. Reddi
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA
- Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| |
Collapse
|
47
|
Xiong Y, Weng Y, Zhu S, Qin J, Feng J, Jing X, Luo C, Gong W, Sun R, Peng M. NOX4 modulates breast cancer progression through cancer cell metabolic reprogramming and CD8 + T cell antitumor activity. Front Immunol 2025; 16:1534936. [PMID: 39991149 PMCID: PMC11842241 DOI: 10.3389/fimmu.2025.1534936] [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: 11/26/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Breast cancer is the most frequently diagnosed malignancy and a leading cause of cancer-related mortality among women worldwide. Although NADPH oxidase 4 (NOX4) has been implicated in various oncogenic processes, its exact function in breast cancer progression, metabolic reprogramming, and immune modulation remains unclear. Methods We used murine 4T1 and EO771 breast cancer models to generate NOX4 knockout (KO) cell lines via CRISPR/Cas9. In vitro assays (cell proliferation, colony formation, wound healing, and Seahorse metabolic analyses) and in vivo orthotopic tumor studies assessed the impact of NOX4 loss. Transcriptomic changes were identified through RNA sequencing and gene set enrichment analysis. We performed MYC knockdown in NOX4 KO cells to investigate its mechanistic role. Flow cytometry characterized tumor-infiltrating immune cells. Finally, NOX4-overexpressing cells were tested for survival benefit and response to dual-checkpoint immunotherapy (anti-PD-1/anti-CTLA-4). Results NOX4 deletion accelerated tumor growth in vivo and enhanced proliferation, colony formation, and migratory capacity in vitro. Metabolic profiling showed that NOX4 KO cells had elevated glycolysis and fatty acid oxidation, along with increased mitochondrial mass. Transcriptomic and enrichment analyses revealed MYC pathway activation in NOX4 KO cells; suppressing MYC reversed these hyperproliferative and metabolic changes. Immunologically, NOX4 KO reduced CD8+ T cell infiltration and function, partially due to lowered CCL11/CCL5 levels, while PD-L1 expression was upregulated. In contrast, NOX4 overexpression improved survival in mice and synergized with checkpoint blockade, demonstrating a positive effect on anti-tumor immunity. Discussion These findings show that NOX4 constrains breast cancer aggressiveness by limiting MYC-driven metabolic adaptations and supporting CD8+ T cell-mediated immunity. Loss of NOX4 promotes a more malignant phenotype and dampens T cell responses, whereas its overexpression prolongs survival and enhances checkpoint inhibitor efficacy. Therapeutically targeting the NOX4-MYC axis and leveraging NOX4's immunomodulatory capacity could offer promising strategies for breast cancer management.
Collapse
Affiliation(s)
- Yingying Xiong
- Department of Clinical Laboratory, Wuhan Fourth Hospital, Wuhan, China
| | - Yiming Weng
- Cancer Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Shan Zhu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Qin
- Central Laboratory, Renmin Hospital, Wuhan University, Wuhan, China
| | - Jia Feng
- Cancer Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Xiaopeng Jing
- Department of Clinical Laboratory, Wuhan Fourth Hospital, Wuhan, China
| | - Chao Luo
- Department of Central Laboratory, The Affiliated Huaian No.1 People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Wei Gong
- Department of Oncology, XiangYang Central Hospital, Hubei University of Arts and Science, Xiangyang, China
| | - Rui Sun
- Cancer Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Min Peng
- Cancer Center, Renmin Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
48
|
Baumann AA, Buribayev Z, Wolkenhauer O, Salybekov AA, Wolfien M. Epigenomic Echoes-Decoding Genomic and Epigenetic Instability to Distinguish Lung Cancer Types and Predict Relapse. EPIGENOMES 2025; 9:5. [PMID: 39982247 PMCID: PMC11843950 DOI: 10.3390/epigenomes9010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025] Open
Abstract
Genomic and epigenomic instability are defining features of cancer, driving tumor progression, heterogeneity, and therapeutic resistance. Central to this process are epigenetic echoes, persistent and dynamic modifications in DNA methylation, histone modifications, non-coding RNA regulation, and chromatin remodeling that mirror underlying genomic chaos and actively influence cancer cell behavior. This review delves into the complex relationship between genomic instability and these epigenetic echoes, illustrating how they collectively shape the cancer genome, affect DNA repair mechanisms, and contribute to tumor evolution. However, the dynamic, context-dependent nature of epigenetic changes presents scientific and ethical challenges, particularly concerning privacy and clinical applicability. Focusing on lung cancer, we examine how specific epigenetic patterns function as biomarkers for distinguishing cancer subtypes and monitoring disease progression and relapse.
Collapse
Affiliation(s)
- Alexandra A. Baumann
- Department of Systems Biology and Bioinformatics, Institute of Computer Science, University of Rostock, 18051 Rostock, Germany; (A.A.B.)
- Faculty of Medicine Carl Gustav Carus, Institute for Medical Informatics and Biometry, TUD Dresden University of Technology, 01069 Dresden, Germany
| | - Zholdas Buribayev
- Department of Computer Science, Faculty of Information Technologies, Al-Farabi Kazakh National University, 050040 Almaty, Kazakhstan
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, Institute of Computer Science, University of Rostock, 18051 Rostock, Germany; (A.A.B.)
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, 80333 Freising, Germany
- Stellenbosch Institute of Advanced Study, Wallenberg Research Centre, Stellenbosch University, Stellenbosch 7535, South Africa
| | - Amankeldi A. Salybekov
- Regenerative Medicine Division, Cell and Gene Therapy Department, Qazaq Institute of Innovative Medicine, 010000 Astana, Kazakhstan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
| | - Markus Wolfien
- Faculty of Medicine Carl Gustav Carus, Institute for Medical Informatics and Biometry, TUD Dresden University of Technology, 01069 Dresden, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), 01069 Dresden, Germany
| |
Collapse
|
49
|
Oh W, Kim AMJ, Dhawan D, Knapp DW, Lim SO. Lactic acid inhibits the interaction between PD-L1 protein and PD-L1 antibody in the PD-1/PD-L1 blockade therapy-resistant tumor. Mol Ther 2025; 33:723-733. [PMID: 40308191 PMCID: PMC11852701 DOI: 10.1016/j.ymthe.2024.12.044] [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: 06/20/2024] [Revised: 11/15/2024] [Accepted: 12/27/2024] [Indexed: 05/02/2025] Open
Abstract
Immune checkpoint blockade therapy targeting the programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) axis has shown remarkable clinical impact in multiple cancer types. Nonetheless, despite the recent success of PD-1/PD-L1 blockade therapy, such response rates in cancer patients have been limited to tumors encompassing specific tumor microenvironment characteristics. The altered metabolic activity of cancer cells shapes the anti-tumor immune response by affecting the activity of immune cells. However, it remains mostly unknown how the altered metabolic activity of cancer cells impacts their resistance to PD-1/PD-L1 blockade therapy. Here, we found that tumor cell-derived lactic acid renders the immunosuppressive tumor microenvironment in the PD-1/PD-L1 blockade-resistant tumors by inhibiting the interaction between the PD-L1 protein and anti-PD-L1 antibody. Furthermore, we showed that the combination therapy of targeting PD-L1 with our PD-L1 antibody-drug conjugate (PD-L1-ADC) and reducing lactic acid with the monocarboxylate transporter 1 (MCT-1) inhibitor, AZD3965, can effectively treat the PD-1/PD-L1 blockade-resistant tumors. The findings of this study provide a new mechanism of how lactic acid induces an immunosuppressive tumor microenvironment and suggest a potential combination treatment to overcome the tumor resistance to PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Wonkyung Oh
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Alyssa Min Jung Kim
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Deepika Dhawan
- Department of Veterinary Clinical Science, Purdue University, West Lafayette, IN 47907, USA
| | - Deborah W Knapp
- Department of Veterinary Clinical Science, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Seung-Oe Lim
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
50
|
Liu R, Ren X, Park YE, Feng H, Sheng X, Song X, AminiTabrizi R, Shah H, Li L, Zhang Y, Abdullah KG, Dubois-Coyne S, Lin H, Cole PA, DeBerardinis RJ, McBrayer SK, Huang H, Zhao Y. Nuclear GTPSCS functions as a lactyl-CoA synthetase to promote histone lactylation and gliomagenesis. Cell Metab 2025; 37:377-394.e9. [PMID: 39642882 PMCID: PMC11798710 DOI: 10.1016/j.cmet.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/18/2024] [Accepted: 11/09/2024] [Indexed: 12/09/2024]
Abstract
Histone lysine lactylation is a physiologically and pathologically relevant epigenetic pathway that can be stimulated by the Warburg effect-associated L-lactate. Nevertheless, the mechanism by which cells use L-lactate to generate lactyl-coenzyme A (CoA) and how this process is regulated remains unknown. Here, we report the identification of guanosine triphosphate (GTP)-specific SCS (GTPSCS) as a lactyl-CoA synthetase in the nucleus. The mechanism was elucidated through the crystallographic structure of GTPSCS in complex with L-lactate, followed by mutagenesis experiments. GTPSCS translocates into the nucleus and interacts with p300 to elevate histone lactylation but not succinylation. This process depends on a nuclear localization signal in the GTPSCS G1 subunit and acetylation at G2 subunit residue K73, which mediates the interaction with p300. GTPSCS/p300 collaboration synergistically regulates histone H3K18la and GDF15 expression, promoting glioma proliferation and radioresistance. GTPSCS represents the inaugural enzyme to catalyze lactyl-CoA synthesis for epigenetic histone lactylation and regulate oncogenic gene expression in glioma.
Collapse
Affiliation(s)
- Ruilong Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Xuelian Ren
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yae Eun Park
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Huixu Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinlei Sheng
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaohan Song
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Roya AminiTabrizi
- Biological Science Division, Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Hardik Shah
- Biological Science Division, Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Lingting Li
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yu Zhang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Sarah Dubois-Coyne
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Hening Lin
- Howard Hughes Medical Institute, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Philip A Cole
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Ralph J DeBerardinis
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Samuel K McBrayer
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - He Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Yingming Zhao
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|