1
|
Wang X, Wang Z, Liu Z, Huang F, Pan Z, Zhang Z, Liu T. Nutritional strategies in oncology: The role of dietary patterns in modulating tumor progression and treatment response. Biochim Biophys Acta Rev Cancer 2025; 1880:189322. [PMID: 40228747 DOI: 10.1016/j.bbcan.2025.189322] [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: 02/10/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Dietary interventions can influence tumor growth by restricting tumor-specific nutritional requirements, altering the nutrient availability in the tumor microenvironment, or enhancing the cytotoxicity of anticancer drugs. Metabolic reprogramming of tumor cells, as a significant hallmark of tumor progression, has a profound impact on immune regulation, severely hindering tumor eradication. Dietary interventions can modify tumor metabolic processes to some extent, thereby further improving the efficacy of tumor treatment. In this review, we emphasize the impact of dietary patterns on tumor progression. By exploring the metabolic differences of nutrients in normal cells versus cancer cells, we further clarify how dietary patterns influence cancer treatment. We also discuss the effects of dietary patterns on traditional treatments such as immunotherapy, chemotherapy, radiotherapy, and the gut microbiome, thereby underscoring the importance of precision nutrition.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zeyao Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zihan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Fanxuan Huang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Zhiren Zhang
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China; Departments of Cardiology and Pharmacy and Breast Cancer surgery, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, China.
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China; Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.
| |
Collapse
|
2
|
Li C, Lin Y, Zheng H, Zeng H, Xu L, Wu D, Lao J, Cai P, Liang S, Wang C, Lin T, Zhong W. Glutamate transporter SLC1A6 promotes resistance to immunotherapy in cancer. Cancer Immunol Immunother 2025; 74:240. [PMID: 40481876 PMCID: PMC12145344 DOI: 10.1007/s00262-025-04074-4] [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: 03/05/2025] [Accepted: 04/30/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Resistance to immune checkpoint inhibitors remains a significant challenge in the treatment of cancer. Emerging evidence suggests that metabolic reprogramming plays a crucial role in tumor metabolism and progression. Our study strived to investigate the role and underlying mechanisms of the glutamate transporter SLC1A6 in resistance to immunotherapy of cancer. METHODS Single-cell RNA sequencing was performed on bladder cancer patients receiving neoadjuvant immunotherapy to identify the expression of SLC1A6 in treatment-resistant cases. The clinical prognostic value of SLC1A6 in cancer was validated using publicly available lung cancer single-cell datasets, as well as transcriptomic data from both bladder and lung cancer cohorts. Flow cytometry was employed to assess the impact of SLC1A6 knockdown on the effector function of CD8⁺ T cell. In vivo tumor models were used to evaluate the role of SLC1A6 in immunotherapy resistance, with immunofluorescence staining performed to examine GZMB⁺ CD8⁺ T cell infiltration. RESULTS SLC1A6 was highly expressed in bladder cancer patients resistant to neoadjuvant immunotherapy, and its expression was associated with disease progression, poor prognosis, and low immune infiltration. Knockdown of SLC1A6 in tumor cells enhanced CD8⁺ T cell effector function. SLC1A6 knockdown also improved the efficacy of immunotherapy and increased the infiltration of GZMB⁺ CD8⁺ T cells within the tumor microenvironment. CONCLUSIONS SLC1A6 plays a critical role in resistance to immunotherapy in cancer. Targeting SLC1A6 may provide a promising therapeutic strategy for improving responses to neoadjuvant immunotherapy and advancing combination treatment approaches.
Collapse
Affiliation(s)
- Chenchen Li
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yi Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Haoran Zheng
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Hengda Zeng
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| | - Longhao Xu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Daqin Wu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jianwen Lao
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Peicong Cai
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Shuai Liang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Chunhui Wang
- Department of Urology, Yan'an Hospital Affiliated With Kunming Medical University, Kunming, People's Republic of China
| | - Tianxin Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China.
| | - Wenlong Zhong
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
3
|
Du Q, Li R, Wang J, Wang J, Jiang Y, Xu Q, Huang D, Qin T. RAN potentiates nuclear export of phosphorylated AMPK, reshaping lipid metabolism and impairing immune efficacy in lung adenocarcinoma. NPJ Precis Oncol 2025; 9:165. [PMID: 40481101 PMCID: PMC12144308 DOI: 10.1038/s41698-025-00977-8] [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: 12/06/2024] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
The therapeutic effectiveness of immune checkpoint inhibitors (ICIs) in lung cancer remains constrained and demonstrates substantial variability across different patients. Targeting the metabolism of tumors emerges an encouraging strategy to enhance the outcomes of tumor immunotherapy. We analyzed metabolic differences in lung cancer post-anti-PD-1 treatment using a single-cell RNA sequencing data (n = 15). Abnormal lipid metabolism is notable in patients with a non-major pathological response, and low RAN expression is linked to good immunotherapy response. RAN showed increased expression in lung adenocarcinoma (LUAD) versus normal lung tissues, correlating with worse prognosis, advanced staging, reduced immune cell activity, and greater sensitivity to common chemotherapeutic drugs. Knockdown of RAN caused G2/M phase arrest, inhibiting proliferation and clone formation in LUAD cells. RAN modifies lipid metabolism via nuclear p-AMPK output to aid tumor cells in resisting immunotherapy and reduces MHC-related molecule expression to evade CD8 + T cell detection. Combining Selinexor with immunotherapy might effectively counter immune tolerance and boost anti-tumor responses in LUAD.
Collapse
Affiliation(s)
- Qingwu Du
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, PR China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
- Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, PR China
| | - Rui Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, PR China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
- Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Jian Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, PR China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
- Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Jingya Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, PR China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
- Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, PR China
| | - Yantao Jiang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, PR China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
- Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, PR China
| | - Qi Xu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, PR China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
- Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, PR China
| | - Dingzhi Huang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, PR China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, PR China.
| | - Tingting Qin
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, PR China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
- Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, PR China.
| |
Collapse
|
4
|
Chen R, He L, Huang Z, Sun J, Wang Q. Causal relationship between the plasma lipidome and urological cancers: A two-sample Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42577. [PMID: 40489874 DOI: 10.1097/md.0000000000042577] [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] [Indexed: 06/11/2025] Open
Abstract
The plasma lipidome has been found to be closely related to inflammation, obesity, aging, and other diseases, including various cancers. However, the causal relationship between specific plasma lipid species and the risk of urological cancers remains unclear. This study aimed to investigate the causal relationship between 179 lipid species and 3 common types of urological cancers using a two-sample Mendelian randomization (MR) approach. Large-scale genome-wide association study datasets of 179 lipid species were analyzed to evaluate the association between the plasma lipidome and urological cancers, including bladder cancer, kidney cancer, and prostate cancer. The inverse variance weighted method was employed as the primary analysis approach, supplemented by MR-Egger regression, weighted median, weighted mode, and simple mode methods to ensure the reliability of the findings. Sensitivity analyses, including Cochran Q test, MR-Egger intercept test, MR Pleiotropy Residual Sum And Outlier, and leave-one-out analysis, were performed to assess the stability and robustness of the causal relationship. The inverse variance weighted analysis showed that higher levels of sterol ester (SE) (27:1/16:0) and other plasma lipidome were causally associated with increased bladder cancer risk. Similarly, elevated levels of phosphatidylcholine (16:0_22:6) and others were linked to increased kidney cancer risk. For prostate cancer, higher levels of SE (27:1/17:0) and others were associated with increased risk, while higher levels of SE (27:1/18:2) and others were associated with decreased risk. The study provides strong evidence for the causal relationships between plasma lipidome and bladder, kidney, and prostate cancers. These findings illuminate complex lipid metabolism pathways in urinary system cancer etiology and highlight specific lipid structures' differential impact on cancer risk. This research lays a foundation for further exploring biological mechanisms and developing early screening tools and targeted therapies for urological cancers.
Collapse
Affiliation(s)
- Rong Chen
- Department of Graduate, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Liujie He
- Naval Medical University, Shanghai, China
| | | | - Jie Sun
- Naval Medical University, Shanghai, China
| | - Qiang Wang
- Department of Graduate, Hebei North University, Zhangjiakou, Hebei Province, China
- Department of Urology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
5
|
Tang Y, Zhao B, Wang W, Chen H, Zhang J, Xie Y, Chen Y, Lin F, Li Y, Zhai X, Zhou W. The EGR1-mediated lncRNA TENM3-AS1 potentiates gastric cancer metastasis via reprogramming fatty acid metabolism. Mol Cancer 2025; 24:165. [PMID: 40481489 PMCID: PMC12142828 DOI: 10.1186/s12943-025-02341-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/22/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are essential modulators in tumor progression. While fatty acid (FA) metabolism can potentiate tumorigenesis, colonization, and metastasis, the roles of lncRNAs in reprograming FA metabolism and regulating gastric cancer (GC) metastasis remain elusive. METHODS Whole RNA-sequencing and in silico analyses were conducted to identify clinically significant lncRNAs involved in GC metastasis. Among the identified lncRNAs, we focused on the novel lncRNA TENM3-AS1. RT-qPCR and FISH analyses revealed an increased expression of TENM3-AS1 in GC cell lines and patients. In vitro and in vivo functional experiments validated the effects of TENM3-AS1 to GC metastasis and the reprogramming of FA metabolism. ChIP, Biotinylated RNA pull-down, RIP, CHX-chase assay, ubiquitination assay, and RNA stabilization assay were employed to perceive the mechanisms underlying the effects of TENM3-AS1 in GC cells. RESULTS TENM3-AS1 expression was significantly elevated in metastatic tumors and advanced primary tumors of GC patients. This increased expression was also associated with a worsened overall survival and progression-free survival. Functionally, TENM3-AS1 enhanced the migration and invasiveness of GC cells in vitro, promoted tumorigenesis and liver metastasis in vivo, and increased FA biosynthesis in GC cells. Mechanistically, our studies showed that the transcription factor EGR1 activated TENM3-AS1, which in turn upregulated the expression of FASN and hnRNPK. Furthermore, TENM3-AS1 interacted with and stabilized hnRNPK by increasing its deubiquitination. This interaction reprogrammed FA metabolism and promoted GC progression by increasing FASN mRNA stability through hnRNPK. CONCLUSIONS In this study, by comparing lncRNA sequencing data from paired primary and peritoneal metastatic tumors and public transcriptome data from non-metastatic and metastatic samples, we clarified a novel lncRNA, TENM3-AS1. It was found that TENM3-AS1 was aberrantly activated in metastatic and advanced primary tumors, and was strongly correlated with a shorter survival in GC patients. Our study reveals the EGR1/TENM3-AS1/ hnRNPK/FASN axis as a novel curative target in metastatic GC.
Collapse
Affiliation(s)
- Yuhui Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Baiwei Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Wanchuan Wang
- The Sixth Affiliated Hospital, South China University of Technology, Foshan, 528000, P. R. China
| | - Haoming Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Junsheng Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yi Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yongming Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Feizhi Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yuanfang Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Xiaohui Zhai
- Department of Medical Oncology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, P. R. China.
| | - Wen Zhou
- The Sixth Affiliated Hospital, South China University of Technology, Foshan, 528000, P. R. China.
| |
Collapse
|
6
|
Yuan C, Hu C, Zhou H, Liu W, Lai W, Liu Y, Yin Y, Li G, Zhang R. L-methionine promotes CD8 + T cells killing hepatocellular carcinoma by inhibiting NR1I2/PCSK9 signaling. Neoplasia 2025; 64:101160. [PMID: 40158232 PMCID: PMC11997342 DOI: 10.1016/j.neo.2025.101160] [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: 10/23/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Liver cancer has consistently high incidence and mortality rates among malignant tumors. PCSK9, a target for hypercholesterolemia therapy, has recently been identified as an inhibitor of anti-tumor immunity, and targeting PCSK9 effectively inhibits tumor progression. However, small molecule inhibitors are lacking due to its flat protein structure. METHODS PCSK9 transcription inhibitor screening was conducted using a PCSK9 promoter-driven td-Tomato plasmid. Quantitative real-time PCR and immunoblotting were employed to assess the effect of L-methionine on PCSK9 expression in HCC cell lines. Co-culture experiments were performed to evaluate the impact of L-methionine on CD8+ T cell-mediated killing of liver cancer cells. RNA sequencing, CUT&Tag, gene editing, and luciferase reporter assays were utilized to identify the transcription factor regulating PCSK9. Additionally, liver cancer xenograft and spontaneous liver cancer mouse models were used to evaluate the anti-cancer efficacy of L-methionine. RESULTS Our study identified L-methionine, an essential amino acid, as a transcriptional inhibitor of PCSK9. The optimal dose of L-methionine to inhibit PCSK9 expression and enhance CD8+ T cell-mediated killing of liver cancer cells in vitro is 50 μM. Furthermore, intraperitoneal injection of 5 mg/kg/day of L-methionine significantly inhibited tumor growth in both liver cancer xenograft and spontaneous liver cancer mouse models. Mechanistically, we identified NR1I2 as a key transcription factor for PCSK9 and their crucial binding site was TGCACCCTGACAC. L-methionine inhibits PCSK9 transcription by downregulating NR1I2. CONCLUSIONS This work demonstrates that L-methionine promotes CD8+ T cell-mediated killing of hepatocellular carcinoma by inhibiting NR1I2/PCSK9 signaling. Our study introduces a novel and convenient approach to inhibit PCSK9 and provides a theoretical basis for the rational supplementation of L-methionine in liver cancer patients.
Collapse
Affiliation(s)
- Chengsha Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Huyue Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yafeng Liu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yue Yin
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China.
| |
Collapse
|
7
|
Zhang J, Kong X, Zhou B, Li R, Yu Z, Zhu J, Xi Q, Li Y, Zhao Z, Zhang R. Metabolic reprogramming of drug resistance in pancreatic cancer: mechanisms and effects. Mol Aspects Med 2025; 103:101368. [PMID: 40398192 DOI: 10.1016/j.mam.2025.101368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 05/10/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
Pancreatic cancer is a highly aggressive gastrointestinal malignancy, often termed the "king of cancers" due to its notoriously high mortality rate. Its clinical characteristics, including late diagnosis, low surgical resectability, high recurrence rates, significant chemoresistance, and poor prognosis have collectively driven the persistent rise in incidence and mortality. Despite ongoing advancements in therapeutic strategies, the management of pancreatic cancer, particularly at advanced stages, remains challenging. Chemotherapy remains the mainstay of current treatment. However, the prevalent problem of chemotherapy resistance poses a significant obstacle to effective treatment. Metabolic reprogramming, characterized by alterations in glucose metabolism, lipid biosynthesis, and amino acid utilization, supports the high energy demands and rapid proliferation of cancer cells. Emerging evidence suggests that these metabolic changes, possibly mediated by epigenetic mechanisms, also contribute to tumorigenesis and metastasis. These findings highlight the critical role of metabolic alterations in pancreatic cancer pathogenesis. This review explores the relationship between metabolic reprogramming and chemotherapy resistance, discussing underlying mechanisms and summarizing preclinical studies and drug development targeting metabolism. The aim is to provide a comprehensive perspective on potential therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Jinyi Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xueqing Kong
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Boyan Zhou
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Rui Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhaoan Yu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jinrong Zhu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Xi
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zichao Zhao
- Department of Emergency Medicine, Shaodong People's Hospital, Shaodong City, Hunan Province, China.
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
8
|
Huang J, Zhang L, Duan W, Li L, Liu X, Wang X. Lipidomics reveals effect of EHHADH in lung squamous cell. Cell Biol Toxicol 2025; 41:94. [PMID: 40450155 DOI: 10.1007/s10565-025-10044-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2025] [Accepted: 05/17/2025] [Indexed: 06/03/2025]
Abstract
Lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD) are two major pathological types of non-small cell lung cancer (NSCLC), characterized by distinct patterns of lipid metabolism. However, the molecular mechanisms underlying lipid metabolism reprogramming specific to LUSC remain poorly understood. This study aims to fill this gap by identifying and characterizing EHHADH (enoyl-CoA, hydratase/3-hydroxyacyl CoA dehydrogenase) as a key regulator of medium-chain fatty acid metabolism in LUSC. The peroxisomal L-bifunctional enzyme is one of the important elements to control the peroxisomal fatty acid beta-oxidation pathway. Through high-expression genes related to lipid metabolism were identified by data mining, the expression and regulatory effects of EHHADH in different cell lines were investigated. EHHADH was highly expressed in LUSC cells and exhibited different expression patterns from those in LUAD cells. Knockdown of EHHADH in LUSC cell lines led to a marked reduction in cell proliferation. RNA sequencing following EHHADH silencing demonstrated significant changes in the expression of lipid metabolism-related genes in different cell lines, such as AZGP1, CAV1, CYP3A4, NR2F2, NR3C2, and RARG. Lipidomics analysis further demonstrated that EHHADH plays a crucial role in regulating intracellular and extracellular lipid profiles. EHHADH knockdown resulted in increased levels of long-chain fatty acids and storage lipids, while decreased levels of medium-chain fatty acids. Conversely, overexpression of EHHADH reduced long-chain fatty acids and storage lipids, while increasing specific medium-chain fatty acids. These metabolic alterations were consistent with changes in lipid metabolism-related protein expression, supporting the molecular mechanistic role of EHHADH in lipid regulation. In conclusion, EHHADH functions as an important regulator of lipid metabolism in LUSC and plays a key role in the occurrence, progression, and treatment of lung cancer. The important impact of EHHADH in lipid metabolism disorders suggests potential utility as a biomarker for diagnosis and a target for personalized treatment strategies in lung cancer.
Collapse
Affiliation(s)
- Jianan Huang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linlin Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wanxin Duan
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liyang Li
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoxia Liu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Sajic T, Vizovišek M, Arni S, Ciuffa R, Mehnert M, Lenglet S, Weder W, Gallart-Ayala H, Ivanisevic J, Buljan M, Thomas A, Hillinger S, Aebersold R. Depletion-dependent activity-based protein profiling using SWATH/DIA-MS detects serine hydrolase lipid remodeling in lung adenocarcinoma progression. Nat Commun 2025; 16:4889. [PMID: 40425563 PMCID: PMC12117057 DOI: 10.1038/s41467-025-59564-x] [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: 11/16/2023] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Systematic inference of enzyme activity in human tumors is key to understanding cancer progression and resistance to therapy. However, standard protein or transcript abundances are blind to the activity status of the measured enzymes, regulated, for example, by active-site amino acid mutations or post-translational protein modifications. Current methods for activity-based proteome profiling (ABPP), which combine mass spectrometry (MS) with chemical probes, quantify the fraction of enzymes that are catalytically active. Here, we describe depletion-dependent ABPP (dd-ABPP) combined with automated SWATH/DIA-MS, which simultaneously determines three molecular layers of studied enzymes: i) catalytically active enzyme fractions, ii) enzyme and background protein abundances, and iii) context-dependent enzyme-protein interactions. We demonstrate the utility of the method in advanced lung adenocarcinoma (LUAD) by monitoring nearly 4000 protein groups and 200 serine hydrolases (SHs) in tumor and adjacent tissue sections routinely collected for patient histopathology. The activity profiles of 23 SHs and the abundance of 59 proteins associated with these enzymes retrospectively classified aggressive LUAD. The molecular signature revealed accelerated lipoprotein depalmitoylation via palmitoyl(protein)hydrolase activities, further confirmed by excess palmitate and its metabolites. The approach is universal and applicable to other enzyme families with available chemical probes, providing clinicians with a biochemical rationale for tumor sample classification.
Collapse
Affiliation(s)
- Tatjana Sajic
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland.
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| | - Matej Vizovišek
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland
| | - Stephan Arni
- Division of Thoracic Surgery, University Hospital Zurich (UHZ), Zürich, Switzerland
| | - Rodolfo Ciuffa
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland
| | - Martin Mehnert
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland
| | - Sébastien Lenglet
- Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne, Geneva, Switzerland
| | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich (UHZ), Zürich, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics and Lipidomics Platform, Faculty of Biology and Medicine, University of Lausanne, Quartier UNIL-CHUV, Rue du Bugnon 19, CH-1005, Lausanne, Switzerland
| | - Julijana Ivanisevic
- Metabolomics and Lipidomics Platform, Faculty of Biology and Medicine, University of Lausanne, Quartier UNIL-CHUV, Rue du Bugnon 19, CH-1005, Lausanne, Switzerland
| | - Marija Buljan
- Empa, Swiss Federal Laboratories for Materials Science and Technology, 9014 St Gallen, Dübendorf, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Aurelien Thomas
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne, Geneva, Switzerland
| | - Sven Hillinger
- Division of Thoracic Surgery, University Hospital Zurich (UHZ), Zürich, Switzerland.
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland.
- Faculty of Science, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
10
|
Liu M, Zhu Y, McIlwain SJ, Deng H, Brasier AR, Ge Y, Kimple ME, Baschnagel AM. Characterizing Plasma-Based Metabolomic Signatures for Metastasis in Non-Small Cell Lung Cancer. Metabolites 2025; 15:340. [PMID: 40422916 PMCID: PMC12113581 DOI: 10.3390/metabo15050340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Background/Objectives: The current staging of non-small cell lung cancer (NSCLC) relies on conventional imaging, which lacks the sensitivity to detect micrometastatic disease. The functional assessment of NSCLC progression may provide independent information to enhance the prediction of metastatic risk. The objective of this study was to determine if we could identify a metabolomic signature predictive of metastasis in patients with NSCLC treated with definitive radiation. Methods: Plasma samples were collected prospectively from patients enrolled in a clinical trial with non-metastatic NSCLC treated with definitive radiation. Metabolites were extracted, and mass spectrometry-based analysis was performed using a flow injection electrospray (FIE)-Fourier transform ion cyclotron resonance (FTICR) mass spectrometry (MS) method. Early metastasis was defined as metastasis within 1 year of radiation treatment. Results: The study cohort included 28 patients. FIE-FITCR produced highly reproducible profiles in technical replicates. A total of 51 metabolic features were identified to be different in patients with early metastasis compared to patients without early metastasis (all adjusted p-values < 0.05, Welch's t-test), including glycerophospholipids, sphingolipids, and fatty acyls. In the follow-up samples collected after the initiation of chemotherapy and radiation treatment, a total of 174 metabolic features were significantly altered in patients who developed early metastasis compared to those who did not. Conclusions: We identified several distinct changes in the metabolic profiles of patients with NSCLC who developed metastatic disease within 1 year of definitive radiation. These findings highlight the potential of metabolomic profiling as a predictive tool for assessing metastatic risk in NSCLC.
Collapse
Affiliation(s)
- Manlu Liu
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA;
| | - Yanlong Zhu
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA; (Y.Z.); (H.D.); (Y.G.)
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA;
| | - Sean J. McIlwain
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA;
| | - Haotian Deng
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA; (Y.Z.); (H.D.); (Y.G.)
| | - Allan R. Brasier
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA;
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA; (Y.Z.); (H.D.); (Y.G.)
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA;
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michelle E. Kimple
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA; (Y.Z.); (H.D.); (Y.G.)
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Andrew M. Baschnagel
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA;
- University of Wisconsin Carbone Cancer Center, Madison, WI 53792, USA
| |
Collapse
|
11
|
Liu X, Qiu R, Gui P, Wei L, Lu Y, Deng Y, Xue Y, Su Y, Huang Q, Du Y. Osteoclast-derived arachidonic acid triggers dormant lung adenocarcinoma cell activation. iScience 2025; 28:112167. [PMID: 40271019 PMCID: PMC12018030 DOI: 10.1016/j.isci.2025.112167] [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: 10/15/2024] [Revised: 01/05/2025] [Accepted: 03/03/2025] [Indexed: 04/25/2025] Open
Abstract
Dormant lung adenocarcinoma (LUAD) cells in the bone microenvironment can re-emerge as metastatic disease through osteoclast interactions. Using a 3D dormancy model and a mouse bone metastasis model, this study reveals that arachidonic acid (AA) is the initiating molecule transferred from osteoclasts to dormant LUAD cells, triggering their activation. Dormant LUAD cells uptake AA through CD36, which activates the PPARγ-ANGPTL4 pathway and activates tumor cells. There is a dose-response relationship in the activation effect of AA, and inhibiting AA metabolism prevents this reactivation. The study also finds that the serum levels of AA and ANGPTL4 are significantly elevated in patients with clinical bone metastases compared to those without. This research confirms that osteoclasts transmit AA via the CD36-PPARγ-ANGPTL4 axis to activate dormant LUAD cells, suggesting that AA and ANGPTL4 may serve as valuable biomarkers and potential clinical applications in treatment and prediction of LUAD bone metastasis.
Collapse
Affiliation(s)
- Xingyu Liu
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Qiu
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengcheng Gui
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lirong Wei
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Lu
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Yan Deng
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Yang Xue
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingyang Su
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Qin Huang
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuzhen Du
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Severin M, Hansen RK, Rolver MG, Hels T, Maeda K, Pardo LA, Pedersen SF. Tumor acidosis supports cancer cell lipid uptake via a rapid transporter-independent mechanism. J Cell Sci 2025; 138:jcs263688. [PMID: 40190115 DOI: 10.1242/jcs.263688] [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: 09/06/2024] [Accepted: 03/25/2025] [Indexed: 05/17/2025] Open
Abstract
Tumor acidosis alters cancer cell metabolism and favors aggressive disease progression. Cancer cells in acidic environments increase lipid droplet accumulation and oxidative phosphorylation, which are characteristics of aggressive cancers. Here, we used live imaging, shotgun lipidomics and immunofluorescence analyses of mammary and pancreatic cancer cells to demonstrate that both acute acidosis and adaptation to acidic growth drive rapid uptake of fatty acids (FAs), which are converted to triacylglycerols and stored in lipid droplets. Consistent with being independent of de novo synthesis, triacylglycerol and lipid droplet accumulation in acid-adapted cells was unaffected by FA synthetase (FAS, encoded by FASN) inhibitors. Macropinocytosis, which is upregulated in acid-adapted cells, partially contributed to FA uptake, which was independent of other protein-facilitated lipid uptake mechanisms, including uptake via CD36 and FATP2, and caveolin- and clathrin-dependent endocytosis. We propose that a major mechanism by which tumor acidosis drives FA uptake is through neutralizing protonation of negatively charged FAs allowing their diffusive, transporter-independent uptake. We suggest that this could be a major factor triggering acidosis-driven metabolic rewiring.
Collapse
Affiliation(s)
- Marc Severin
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Rikke K Hansen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Michala G Rolver
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, 2200 Copenhagen, Denmark
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tove Hels
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Kenji Maeda
- Section for Cell Death and Metabolism, Danish Cancer Institute, 2100 Copenhagen, Denmark
| | - Luis A Pardo
- Max Planck Institute for Multidisciplinary Sciences, Oncophysiology Group, 37077 Göttingen, Germany
| | - Stine F Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
13
|
Liu F, Li K, Zhu Q. Targeting Metabolic Reprogramming in Bladder Cancer Immunotherapy: A Precision Medicine Approach. Biomedicines 2025; 13:1145. [PMID: 40426972 PMCID: PMC12108893 DOI: 10.3390/biomedicines13051145] [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: 02/27/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 05/29/2025] Open
Abstract
Bladder cancer, as a highly heterogeneous malignant tumor of the urinary system, is significantly affected by tumor metabolic reprogramming in its response to immunotherapy. This review systematically elaborates on the molecular mechanisms of abnormal glucose and lipid metabolism in the bladder cancer microenvironment and immune escape, and discusses precision treatment strategies based on metabolic regulation. In the future, it will be necessary to combine spatiotemporal omics and artificial intelligence technologies to construct a multi-target intervention system for the metabolic-immune interaction network, promoting a paradigm shift in precision treatment for bladder cancer.
Collapse
Affiliation(s)
| | | | - Qingyi Zhu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| |
Collapse
|
14
|
Fan S, Zhao K, Lei J, Ge Y. Preoperative total bile acid can be used as a prognostic biomarker in patients with operable biliary tract cancers. Discov Oncol 2025; 16:696. [PMID: 40338467 PMCID: PMC12061807 DOI: 10.1007/s12672-025-02527-x] [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: 12/08/2024] [Accepted: 04/28/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Biliary tract cancers (BTCs) are highly invasive malignancies with poor prognoses. However, reliable biomarkers for survival prediction remain lacking. Notably, abnormal lipid metabolism has elicited increasing interest in digestive tract tumors, with the liver playing an important role in lipid metabolism. OBJECTIVE To explore the relationship between hepatic lipid metabolism-related indicators, assessed through routine clinical biochemical testing and survival prognosis in patients with BTCs. METHODS Overall, 109 patients with a pathological diagnosis of BTC from 2017 to 2023 were included in this study. Univariate and multivariate Cox regression analyses were performed using R Studio software, and survival curves were plotted. RESULTS Univariate analysis revealed that tumor location and preoperative total bile acid (TBA), carcinoembryonic antigen, cancer antigen (CA)125, and CA19-9 levels were correlated with patient survival (P < 0.05). Multivariate Cox regression analysis identified increased TBA level [hazard ratio (HR) = 0.445, P = 0.004] as an independent prognostic factor for longer survival. Conversely, tumor location [intrahepatic cholangiocarcinoma (iCCA) and/or extrahepatic cholangiocarcinoma (eCCA)] (HR = 2.463, P = 0.036) and increased CA125 and CA19-9 levels (HR = 2.549, P = 0.008 and HR = 2.100, P = 0.019) were independent prognostic factors for shorter survival. Additionally, Kaplan‒Meier survival curves revealed significantly longer survival in patients with increased TBA levels than those in the normal group (P = 0.012). Conversely, patients with iCCA and/or eCCA tumor location and increased CA125 and CA19-9 levels had significantly shorter median survival (P = 0.044, P = 0.013, and P = 0.012, respectively). CONCLUSION TBA may be a biomarker for predicting survival in patients with operable BTC, highlighting its clinical significance and application potential.
Collapse
Affiliation(s)
- Shanshan Fan
- Department of Oncology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Kexin Zhao
- The Third Clinical School of Medicine, Capital Medical University, Beijing, China
| | - Jiabao Lei
- The Third Clinical School of Medicine, Capital Medical University, Beijing, China
| | - Yang Ge
- Department of Oncology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| |
Collapse
|
15
|
Edwards DN, Wang S, Kane K, Song W, Kim LC, Ngwa VM, Hwang Y, Ess K, Boothby MR, Chen J. Increased fatty acid delivery by tumor endothelium promotes metastatic outgrowth. JCI Insight 2025; 10:e187531. [PMID: 40198126 DOI: 10.1172/jci.insight.187531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Metastatic outgrowth in distant microscopic niches requires sufficient nutrients, including fatty acids (FAs), to support tumor growth and to generate an immunosuppressive tumor microenvironment (TME). However, despite the important role of FAs in metastasis, the regulation of FA supply in metastatic niches has not been defined. In this report, we show that tumor endothelium actively promotes outgrowth and restricts antitumor cytolysis by transferring FAs into developing metastatic tumors. We describe a process of transendothelial FA delivery via endosomes that requires mTORC1 activity. Thus, endothelial cell-specific targeted deletion of Raptor (RptorECKO), a unique component of the mTORC1 complex, significantly reduced metastatic tumor burden that was associated with improved markers of T cell cytotoxicity. Low-dose everolimus that selectively inhibited endothelial mTORC1 improves immune checkpoint responses in metastatic disease models. This work reveals the importance of transendothelial nutrient delivery to the TME, highlighting a future target for therapeutic development.
Collapse
Affiliation(s)
- Deanna N Edwards
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Shan Wang
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kelby Kane
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Wenqiang Song
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Division of Epidemiology, and
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Laura C Kim
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Verra M Ngwa
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yoonha Hwang
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kevin Ess
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mark R Boothby
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
| | - Jin Chen
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
16
|
Mehnert SA, Lee KJ, McLuckey SA. Enthalpies and Entropies of Activation for sn-1/ sn-2 Acyl Chain Loss in Glycerophospholipid Anions via Dipolar DC Kinetics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2025; 36:1041-1051. [PMID: 40173190 DOI: 10.1021/jasms.4c00477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Glycerophospholipids (GPs) have been observed to prefer losing a particular fatty acyl chain over the other, with the preference for the chain in either the sn-1 or sn-2 position being different between various GP classes. It has been assumed that the sn preference results from the entropic favorability of the transition state conformation; however, this has not been measured previously. Here, we demonstrate the application of our previously established collision-based activation method to GP fragmentation. The method utilizes a dipolar direct current (DDC) potential across a pair of opposing rods to force ions out of the center of the ion trap, causing them to undergo radio frequency (RF) heating by absorbing power from the trapping RF field. We confirmed that the previous assumption holds for some species studied here, wherein the ΔH‡ values were nearly identical and the ΔS‡ values showed greater differences between the sn positions. However, some species and ion types seem to be more driven by ΔH‡. Additionally, the loss of the fatty acyl chains as neutrals rather than ions should also be considered if one is to thoroughly weigh which chain is indeed the preferred loss, as including all forms of acyl chain loss results in an overall favorability for the acyl chain in the sn-2 position to be lost. The driving force behind these different losses seems to be a mixture of entropic and enthalpic reasons, with the identity and presence of the headgroup playing an important role in the observed fragmentation and the measured activation parameters.
Collapse
Affiliation(s)
- Samantha A Mehnert
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, Indiana 47906-2084, United States
| | - Katherine J Lee
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, Indiana 47906-2084, United States
| | - Scott A McLuckey
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, Indiana 47906-2084, United States
| |
Collapse
|
17
|
Wen J, He JQ. Associations between lipids and lung cancer subtypes. Discov Oncol 2025; 16:656. [PMID: 40314895 PMCID: PMC12048372 DOI: 10.1007/s12672-025-02450-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
OBJECTIVE The causative relationship between lung cancer subtypes and lipids is yet unknown. This research aims to elucidate the potential causative link connecting lipid levels to lung cancer subtypes, particularly focusing on non-small cell lung cancer (NSCLC), using the Mendelian randomization (MR) method and meta-analysis. METHODS Summary statistics were obtained from genome-wide association study (GWAS) datasets. A comprehensive MR analysis was performed to explore the causal role of lipids in NSCLC subtypes. To ensure the reliability of the results, an external dataset was used for validation, and a meta-analysis was performed for further synthesis. RESULTS Two of the 179 lipids examined showed potential causal connection with lung adenocarcinoma (LUAD) and three with lung squamous cell carcinoma (LUSC). Specifically, phosphatidylcholine (PC) (16:0_20:4) and PC (18:0_20:4) might be connected to an elevated risk of LUSC, but PC (18:0_20:2) might be linked to a decreased risk. It was discovered that PC (16:1_20:4) and PC (18:0_20:4) might raise the risk for LUAD. CONCLUSION Complex lipid metabolic pathways, especially involving PC, are present in NSCLC, and distinct lipid isomers may influence various molecular subtypes in different ways.
Collapse
Affiliation(s)
- Jiayu Wen
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Meishan City, Renshou County, Meishan, China
| | - Jian-Qing He
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Xiong L, Cheng J. Rewiring lipid metabolism to enhance immunotherapy efficacy in melanoma: a frontier in cancer treatment. Front Oncol 2025; 15:1519592. [PMID: 40376583 PMCID: PMC12078133 DOI: 10.3389/fonc.2025.1519592] [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: 10/30/2024] [Accepted: 03/31/2025] [Indexed: 05/18/2025] Open
Abstract
Immunotherapy has transformed the landscape of melanoma treatment, offering significant extensions in survival for many patients. Despite these advancements, nearly 50% of melanoma cases remain resistant to such therapies, highlighting the need for novel approaches. Emerging research has identified lipid metabolism reprogramming as a key factor in promoting melanoma progression and resistance to immunotherapy. This reprogramming not only supports tumor growth and metastasis but also creates an immunosuppressive environment that impairs the effectiveness of treatments such as immune checkpoint inhibitors (ICIs). This review delves into the intricate relationship between lipid metabolism and immune system interactions in melanoma. We will explore how alterations in lipid metabolic pathways contribute to immune evasion and therapy resistance, emphasizing recent discoveries in this area. Additionally, we also highlights novel therapeutic strategies targeting lipid metabolism to enhance immune checkpoint inhibitor (ICI) efficacy.
Collapse
Affiliation(s)
- Lihua Xiong
- Department of Dermatology, Cheng Du Xinjin District Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Jian Cheng
- Department of Chinese Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
19
|
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
|
20
|
An Y, Song H, Qiu H, Jiang J, Shi J. Lipid Metabolism in Gastrointestinal Malignancies: Exploring Dysregulation, Biomarkers, and Treatment Strategies. Cancer Med 2025; 14:e70975. [PMID: 40391753 PMCID: PMC12090204 DOI: 10.1002/cam4.70975] [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: 01/14/2025] [Revised: 04/09/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Gastrointestinal malignancies are a major public health concern worldwide, characterized by high incidence and mortality rates. Despite continuous advancements in existing treatment methods, overall survival rates remain low. Lipid metabolism plays a crucial role in the occurrence, progression, and treatment of gastrointestinal malignancies. Its involvement in the metabolic reprogramming of tumor cells, regulation of the tumor microenvironment, and drug response has become a research hotspot. MATERIALS & METHODS This review summarizes current research related to lipid metabolism mechanisms, biomarkers, and therapies in GI cancers, with emphasis on its interaction with the tumor microenvironment.
Collapse
Affiliation(s)
- Yan An
- Department of AnesthesiologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Huihui Song
- Obstetrical Medicine Center, Weifang People's HospitalShandong Second Medical UniversityWeifangChina
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical UniversityShandong Second Medical UniversityWeifangChina
- Clinical Research CenterAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Jun Jiang
- Department of AnesthesiologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Junfeng Shi
- Department of Endocrinology and Metabolism, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical UniversityShandong Second Medical UniversityWeifangChina
- Clinical Research CenterAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| |
Collapse
|
21
|
Fries B, Sekera E, Holbrook J, Hummon A. Multi-Omic Evaluation of PLK1 Inhibitor-Onvansertib-In Colorectal Cancer Spheroids. JOURNAL OF MASS SPECTROMETRY : JMS 2025; 60:e5137. [PMID: 40197665 PMCID: PMC11976698 DOI: 10.1002/jms.5137] [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: 01/15/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Polo-like kinase 1 (Plk1) is a serine/threonine kinase involved in regulating the cell cycle. It is activated by aurora kinase B along with the cofactors Borealin, INCE, and survivin. Plk1 is involved in the development of resistances to chemotherapeutics such as doxorubicin, Taxol, and gemcitabine. It has been shown that patients with higher levels of Plk1 have lower survival rates. Onvansertib is a competitive ATP inhibitor for Plk1 in clinical trials for the treatment of tumors and has recently entered a trial for the treatment of KRAS mutant colorectal cancers (CRCs). In this study, we conducted an untargeted liquid chromatography-mass spectrometry (LC-MS) proteomics study as well as an untargeted lipidomics analysis of HCT 116 spheroids treated with onvansertib over a 72-h treatment time-course experiment. Mass spectrometry imaging (MSI) showed that onvansertib begins to accumulate most prominently after 12 h of treatment and continues to accumulate through 72 h. Proteomic results displayed alterations to cell cycle control proteins and an increasing abundance of aurora kinase B and Borealin. The proteomics data also showed alterations to many lipid metabolism enzymes. The MSI lipidomics data indicated alterations to phosphatidylcholine lipids, with many lipids increasing in abundance over time or increasing until 12 h of onvansertib treatment and decreasing after that time point. In summary, these results suggest that onvansertib is causing cells within the spheroid to halt at a certain phase of the cell cycle in accordance with previous literature. Our findings suggest the S phase is likely interrupted, with observed alterations in cell cycle control proteins and PC lipid abundance.
Collapse
Affiliation(s)
- Brian D. Fries
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOhioUSA
| | - Emily R. Sekera
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOhioUSA
| | | | - Amanda B. Hummon
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
22
|
Zhuo Y, Xu Y, Qu X, Li Q, Sun M, Gao X, Yuan F, Cao M, Pan B. Research on Peripheral Nerve Aging and Degeneration: Cellular Changes and Mechanism Exploration From the Perspective of Single-Cell Sequencing. Eur J Neurosci 2025; 61:e70129. [PMID: 40317786 DOI: 10.1111/ejn.70129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
As age increases, there are structural and functional alterations in the peripheral nervous system (PNS), significantly affecting movement, sensation and autonomic function. Understanding the characteristics and mechanisms of PNS aging is crucial for preventing and treating related diseases. This study employed single-cell sequencing technology to analyse the dorsal root ganglia (DRG) and sciatic nerve (SN) of aging rats, in comparison with adult rats. The research investigated the mechanisms underlying PNS aging and degeneration, revealing the transcriptional profiles of various cell types. Significant differences were observed in the proportion of Schwann cells between the DRG and SN of adult and aged rats. The Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) revealed that pathways related to neurodegeneration were upregulated in Schwann cells. Additionally, lipid metabolism pathways were upregulated in the SN of aged rats, suggesting that certain lipid signalling molecules may influence cell proliferation. Through further re-clustering of myelinating Schwann cells, six distinct subtypes were identified. The anti-aging protein protocadherin 9 (PCDH9) was preliminarily screened and found to be significantly downregulated with age. In vitro experiments confirmed that PCDH9 expression is associated with Schwann cell proliferation and differentiation. By using gene expression analysis and cell type across several age groups, this study offers important insights into the mechanisms of PNS aging and degeneration.
Collapse
Affiliation(s)
- Yuyang Zhuo
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yingcai Xu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xinzhe Qu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qupeng Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Maji Sun
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiao Gao
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Feng Yuan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Menghan Cao
- Center of Clinical Oncology, The Afffliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bin Pan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
23
|
Chang CC, Chang CB, Shen CH, Lee MY, Jou YC, Tung CL, Lai WH, Hung CF, Wang M, Lai YY, Chen PC, Wu SF. Immunosuppression of Tumor-Derived Factors Modulated Neutrophils in Upper Tract Urothelial Carcinoma Through Upregulation of Arginase-1 via ApoA1-STAT3 Axis. Cells 2025; 14:660. [PMID: 40358184 PMCID: PMC12072159 DOI: 10.3390/cells14090660] [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: 04/07/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Upper tract urothelial carcinoma (UTUC) presents aggressive features and a tumor microenvironment with T cell depletion. However, the role of tumor-associated neutrophils in UTUC remains unclear. This study aimed to investigate how UTUC tumor-derived factors modulate neutrophils and their impact on T cell immune responses. Our findings demonstrate that UTUC secreted tumor-derived factors, with apolipoprotein A1 (Apo-A1) being the predominant factor, which upregulated arginase-1 expression in neutrophils. STAT3 activation was responsible for the upregulation of arginase-1 in neutrophils. Blocking the interactions between Apo-A1 and its receptors reduced arginase-1 expression in neutrophils treated with tumor tissue culture supernatant (TTCS). Moreover, both CD4+ T and CD8+ T cell proliferation were inhibited by neutrophils treated with Apo-A1 or TTCS. Importantly, blocking Apo-A1 signaling in neutrophils reversed the inhibitory effects on T cells. In UTUC patients, the neutrophil-to-lymphocyte ratio was higher than that in healthy subjects. The expression of arginase-1 in neutrophils and the level of Apo-A1 within UTUC tumors were negatively correlated with tumor-infiltrating CD4+ T cells. Additionally, neutrophils from UTUC patients showed increased expression of arginase-1 and exhibited inhibitory effects of T cell functions. These findings suggest that UTUC orchestrates an immune-suppressive microenvironment through Apo-A1-mediated upregulation of arginase-1 in neutrophils, ultimately leading to the inhibition of T cell proliferation.
Collapse
Affiliation(s)
- Chih-Chia Chang
- Department of Radiation Therapy and Oncology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan;
| | - Chia-Bin Chang
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Cheng-Huang Shen
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Ming-Yang Lee
- Department of Hematology and Oncology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan;
| | - Yeong-Chin Jou
- Department of Urology, St. Martin De Porres Hospital, Chiayi 60069, Taiwan;
| | - Chun-Liang Tung
- Department of Pathology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan;
| | - Wei-Hong Lai
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Chi-Feng Hung
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Meilin Wang
- Department of Microbiology and Immunology, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
| | - Ya-Yan Lai
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 60004, Taiwan;
| | - Pi-Che Chen
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Shu-Fen Wu
- Department of Biomedical Sciences, and Epigenomics Human Disease Research Center, National Chung Cheng University, Chiayi 62102, Taiwan
| |
Collapse
|
24
|
Uti DE, Atangwho IJ, Alum EU, Ntaobeten E, Obeten UN, Bawa I, Agada SA, Ukam CIO, Egbung GE. Antioxidants in cancer therapy mitigating lipid peroxidation without compromising treatment through nanotechnology. DISCOVER NANO 2025; 20:70. [PMID: 40272665 PMCID: PMC12021792 DOI: 10.1186/s11671-025-04248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/27/2025]
Abstract
BACKGROUND Cancer treatments often exploit oxidative stress to selectively kill tumour cells by disrupting their lipid peroxidation membranes and inhibiting antioxidant enzymes. However, lipid peroxidation plays a dual role in cancer progression, acting as both a tumour promoter and a suppressor. Balancing oxidative stress through antioxidant therapy remains a challenge, as excessive antioxidant activity may compromise the efficacy of chemotherapy and radiotherapy. AIM This review explores the role of antioxidants in mitigating lipid peroxidation in cancer therapy while maintaining treatment efficacy. It highlights recent advancements in nanotechnology-based targeted antioxidant delivery to optimize therapeutic outcomes. METHODS A comprehensive literature review was conducted using reputable databases, including PubMed, Scopus, Web of Science, and ScienceDirect. The search focused on publications from the past five years (2020-2025), supplemented by relevant studies from earlier years. Keywords such as "antioxidants," "lipid peroxidation," "nanotechnology in cancer therapy," and "oxidative stress" were utilized. Relevant articles were critically analysed, and graphical illustrations were created. RESULTS Emerging evidence suggests that nanoparticles, including liposomes, polymeric nanoparticles, metal-organic frameworks, and others, can effectively encapsulate and control the release of antioxidants in tumour cells while minimizing systemic toxicity. Stimuli-responsive carriers with tumour-specific targeting mechanisms further enhance antioxidant delivery. Studies indicate that these strategies help preserve normal cells, mitigate oxidative stress-related damage, and improve treatment efficacy. However, challenges such as bioavailability, stability, and potential interactions with standard therapies remain. CONCLUSION Integrating nanotechnology with antioxidant-based interventions presents a promising approach for optimizing cancer therapy. Future research should focus on refining lipid peroxidation modulation strategies, assessing oxidative stress profiles during treatment, and employing biomarkers to determine optimal antioxidant dosing. A balanced approach to antioxidant use may enhance therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
- Daniel Ejim Uti
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria.
| | - Item Justin Atangwho
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Esther Ugo Alum
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda
| | - Emmanuella Ntaobeten
- Department of Cancer and Haematology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Uket Nta Obeten
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| | - Inalegwu Bawa
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | - Samuel A Agada
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | | | - Godwin Eneji Egbung
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
25
|
Wang C, Ou Z, Deng H, Zhang Y, Li X, Wang X, Zhao D. GDF15 drives de novo lipogenesis and contributes to ovarian cancer metastasis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167868. [PMID: 40280199 DOI: 10.1016/j.bbadis.2025.167868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/10/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Ovarian cancer is frequently diagnosed at an advanced stage, characterized by extensive metastasis. Recent studies indicate that metastatic and primary tumors exhibit similar mutational landscape, suggesting that non-mutational factors significantly contribute to the metastatic process. Enhanced lipid metabolism has been implicated across various stages of cancer progression, making the targeting of metabolic vulnerabilities a promising therapeutic strategy. In this study, we demonstrate that growth differentiation factor 15 (GDF15), a member of the TGF-β superfamily, which has been Indicated to be associated with several metabolic diseases, is significantly elevated in the serum of ovarian cancer patients, particularly in metastatic lesions compared to primary tumors. Elevated GDF15 levels correlate with reduced overall survival and progression-free survival. Furthermore, we found that GDF15 facilitates tumor metastasis by regulating de novo lipogenesis through the PI3K/AKT signaling pathway. These findings suggest that targeting GDF15-mediated lipid metabolism could provide a novel therapeutic approach to inhibit ovarian cancer metastasis.
Collapse
Affiliation(s)
- Chenxi Wang
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhengjie Ou
- Department of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongming Deng
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ying Zhang
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoyang Li
- School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Xiaobing Wang
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Dan Zhao
- Department of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
26
|
Chai R, Zheng K, Xu T, Wang H, Cheng X, Lu C, Kang Y. SNX10 Is Involved in Ovarian Cancer Cell Metastasis by Repolarizing Tumor-Associated Macrophages Through mTOR1/Lysosomes Pathway. Biomedicines 2025; 13:1021. [PMID: 40426851 PMCID: PMC12109050 DOI: 10.3390/biomedicines13051021] [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: 03/11/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Tumor-associated macrophages (TAMs) are prevalent in advanced ovarian cancer tissues and ascites, significantly influencing disease prognosis. However, the mechanisms driving TAM polarization and their tumor-promoting effects remain poorly understood. Methods: The subcellular distribution of SNX10 in ovarian cancer tissues was analyzed using single-cell datasets (GSE147082, GSE58937). The Kaplan-Meier Plotter and GEPIA2 databases were used to evaluate SNX10's prognostic relevance. Lentivirus-mediated SNX10 overexpression in THP-1 cells was employed in tumor cell-macrophage co-culture experiments. Transwell assays and flow cytometry assessed SNX10's effects on ovarian cancer cell metastasis and cisplatin-induced apoptosis. RNA sequencing, Western blotting, lysosomal pH detection, lipid droplet staining, and RT-qPCR were performed to explore SNX10's molecular mechanisms in TAM polarization and immune modulation. Results: SNX10 was specifically expressed in TAMs, promoting their polarization into the M2 phenotype. This enhanced the migration and invasion of ovarian cancer cell lines A2780 and A2780/CP70 while reducing cisplatin-induced apoptosis. SNX10 decreased lipid droplet content, downregulated p-mTOR1, and impaired lysosomal function in TAMs. Additionally, SNX10 differentially modulated PD-L1 mRNA expression in platinum-sensitive and platinum-resistant ovarian cancer cells. Conclusions: SNX10 regulates the mTOR1/lysosome pathway in TAMs, influencing lipid metabolism and indirectly modulating ovarian cancer cell metastasis. It also alters PD-L1 mRNA expression, suggesting a role in shaping the tumor immune microenvironment.
Collapse
Affiliation(s)
- Ranran Chai
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (R.C.); (K.Z.); (T.X.); (H.W.); (X.C.)
| | - Kewei Zheng
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (R.C.); (K.Z.); (T.X.); (H.W.); (X.C.)
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Ting Xu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (R.C.); (K.Z.); (T.X.); (H.W.); (X.C.)
| | - Hui Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (R.C.); (K.Z.); (T.X.); (H.W.); (X.C.)
| | - Xiaobo Cheng
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (R.C.); (K.Z.); (T.X.); (H.W.); (X.C.)
| | - Chong Lu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (R.C.); (K.Z.); (T.X.); (H.W.); (X.C.)
| | - Yu Kang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (R.C.); (K.Z.); (T.X.); (H.W.); (X.C.)
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| |
Collapse
|
27
|
Poswal J, Mandal CC. Lipid metabolism dysregulation for bone metastasis and its prevention. Expert Rev Anticancer Ther 2025:1-17. [PMID: 40219980 DOI: 10.1080/14737140.2025.2492784] [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: 02/16/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION Bone metastasis often develops in advanced malignancies. Lipid metabolic dysregulation might play pivotal role in cancer progression and subsequent deterioration of bone health at metastatic condition. In-depth understanding of lipid reprogramming in metastasized cancer cells and other stromal cells including bone marrow adipocyte (BMA) is an urgent need to develop effective therapy. AREA COVERED This paper emphasizes providing an overview of multifaceted role of dysregulated lipids and BMA in cancer cells in association with bone metastasis by utilizing search terms lipid metabolism, lipid and metastasis in PubMed. This study extends to address mechanism linked with lipid metabolism and various crucial genes (e.g. CSF-1, RANKL, NFkB and NFATc1) involved in bone metastasis. This review examines therapeutic strategies targeting lipid metabolism to offer potential avenues to disrupt lipid-driven metastasis. EXPERT OPINION On metastatic condition, dysregulated lipid molecules especially in BMA and other stromal cells not only favors cancer progression but also potentiate lipid reprogramming within cancer cells. Distinct dysregulated lipid-metabolism associated genes may act as biomarker, and targeting these is challenging task for specific treatment. Curbing function of bone resorption associated genes by lipid controlling drugs (e.g. statins, omega-3 FA and metformin) may provide additional support to curtail lipid-associated bone metastasis.
Collapse
Affiliation(s)
- Jyoti Poswal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
28
|
Wang Y, Chen J, Wang Z, Luo X, Wu N, Wang J. HKDC1 promotes ovarian cancer progression through boosting lipid metabolism and immune escape by stabilizing G6PC/G6PC2. Commun Biol 2025; 8:615. [PMID: 40234623 PMCID: PMC12000390 DOI: 10.1038/s42003-025-08031-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
Ovarian cancer (OC) is a significant health challenge, yet the mechanisms driving its progression remain unclear. This study explored the role of hexokinase domain-containing protein 1 (HKDC1) in OC, focusing on tumor growth, lipid metabolism, and immune evasion. Human OC cell lines (SKOV3 and HEY) and the murine OC cell line (ID8) were used to knock down and overexpress HKDC1. An ID8-based epithelial OC mouse model was established to validate the in vitro findings. Our results demonstrated that HKDC1 was upregulated in OC and promoted cell proliferation, migration, and invasion. HKDC1 enhanced lipid accumulation by elevating levels of free fatty acids (FFA), triglycerides, phospholipids, cholesterol, and neutral lipid, while upregulating key enzymes (ACC1, FASN, SCD1, HMGCS1, and HMGCR). It promoted immune escape through PD-L1 upregulation, inhibiting T cell proliferation and reducing IFN-γ, granzyme B, and perforin levels while increasing PD-1 levels. HKDC1 knockdown reversed these effects, which were restored by adding FFA. Mechanistically, HKDC1 interacted with and stabilized glucose-6-phosphatase catalytic subunits (G6PC/G6PC2), supporting its tumor-promoting functions. These findings were confirmed in an OC mouse model, highlighting HKDC1 as a key driver of OC progression through lipid biosynthesis and immune suppression, offering potential therapeutic targets.
Collapse
Affiliation(s)
- Ying Wang
- Department of the Central Laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, P. R. China.
| | - Juan Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Zhan Wang
- Lung Cancer and Gastrointestinal Unit, Department of Medical Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, P. R. China
| | - Xia Luo
- Department of the Central Laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, P. R. China
| | - Nayiyuan Wu
- Department of the Central Laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, P. R. China
| | - Jing Wang
- Department of the Central Laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, P. R. China.
| |
Collapse
|
29
|
Prajumwongs P, Titapun A, Thanasukarn V, Jareanrat A, Khuntikeo N, Namwat N, Klanrit P, Wangwiwatsin A, Chindaprasirt J, Koonmee S, Sa-Ngiamwibool P, Muangritdech N, Roytrakul S, Loilome W. Identification of serum metabolite biomarkers and metabolic reprogramming mechanisms to predict recurrence in cholangiocarcinoma. Sci Rep 2025; 15:12782. [PMID: 40229491 PMCID: PMC11997029 DOI: 10.1038/s41598-025-97641-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
Cholangiocarcinoma (CCA) has high recurrence rates that severely limit long-term survival. Effective tools for accurate recurrence monitoring and diagnosis remain lacking. Metabolic reprogramming, a key driver of CCA growth and recurrence, is underutilized in cancer screening and management. This study aimed to identify metabolite-based biomarkers to evaluate recurrence severity, enhance disease management, and elucidate the molecular mechanisms underlying CCA recurrence. A comprehensive, non-targeted serum metabolomics analysis using ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry was conducted. Support Vector Machine (SVM) modeling was employed to develop a predictive framework based on metabolite biomarkers. The analysis revealed significant alterations in metabolomics and lipidomics across CCA recurrence subtypes. Notably, changes in metabolites such as amino acids, lipid-derived carnitines, and glycerophospholipids were associated with cancer progression through enhanced energy production and lipid remodeling. The SVM-constructed metabolite-based predictive model demonstrated predictive accuracy comparable to current clinical diagnostic standards. These findings provide novel insights into the metabolic mechanisms underlying CCA recurrence, addressing critical clinical challenges. By advancing early diagnostic approaches, particularly for preoperative detection, this study offers a reliable method for predicting recurrence in CCA patients. This enables effective treatment planning and supports the development of personalized therapeutic strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Piya Prajumwongs
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Attapol Titapun
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Vasin Thanasukarn
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Apiwat Jareanrat
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Natcha Khuntikeo
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Poramate Klanrit
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Arporn Wangwiwatsin
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jarin Chindaprasirt
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Medical Oncology Unit, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Supinda Koonmee
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Prakasit Sa-Ngiamwibool
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nattha Muangritdech
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Sittiruk Roytrakul
- Proteomics Research Laboratory, Genome Institute, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Watcharin Loilome
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
30
|
Tian X, Bai X, Han Y, Ye Y, Peng M, Cui H, Li K. PPAR γ changing ALDH1A3 content to regulate lipid metabolism and inhibit lung cancer cell growth. Mol Genet Genomics 2025; 300:41. [PMID: 40198404 PMCID: PMC11978687 DOI: 10.1007/s00438-025-02243-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/08/2025] [Indexed: 04/10/2025]
Abstract
PPAR γ, as a widely present receptor in tissues, plays a key role in lipid metabolism, energy balance, inflammatory response, and cell differentiation. It plays an important role in the occurrence and development of various tumors, including prostate cancer, gastric cancer, lung cancer, etc., by regulating lipid metabolism. However, the specific mechanism by which it affects lung cancer growth is not yet clear. To investigate how PPAR γ affects lung cancer cell growth by altering ALDH1A3 levels through its impact on lipid metabolism. Bioinformatics analysis was used to predict the correlation between PPAR γ, ALDH1A3 and lung cancer. Based on the results of bioinformatics analysis, PPAR γ activator (Pioglitazone, Pio) and ALDH1A3 inhibitor (diethylaminobenzaldehyde, DEAB) were used to act on lung cancer cells and observe their growth. After measuring the IC50 value of the drug in vitro experiments, lipid metabolomics analysis was conducted to identify the significant changes in differential metabolites and metabolic pathways under the combined influence of Pio and DEAB. Through bioinformatics analysis, it was found that there were significant differences in the levels of PPAR γ and ALDH1A3 between lung cancer and normal lung tissues, and ALDH1A3 was positively correlated with PPAR γ. AUC analysis found that PPAR γ and ALDH1A3 have good predictive value in the diagnosis and prognosis of lung cancer. GSEA enrichment analysis showed that PPAR γ and ALDH1A3 were significantly correlated with lipid oxidation. Combining relevant literature to demonstrate the inhibitory effect of PPAR γ receptors on lung cancer cells and the ability of PPAR γ activation to inhibit ALDH1A3 levels. Further in vitro CCK-8 and IC50 measurements of lung cancer cells A549 and H1299 were conducted, followed by non targeted lipidomics analysis. It was found that the metabolic pathways upregulated by activation of PPAR γ and inhibition of ALDH1A3 included glycerophospholipid metabolism, cholesterol metabolism, arachidonic acid metabolism, and fat digestion and absorption, with glycerophospholipid metabolism pathway accounting for the highest percentage. Conclusion: PPAR γ activation can inhibit the production of ALDH1A3, alter the glycerophospholipid metabolism pathway, and thus inhibit the proliferation of lung cancer cells. This study confirms that PPAR γ affects lung cancer proliferation by influencing the glycerophospholipid metabolism pathway.
Collapse
Affiliation(s)
- Xinyuan Tian
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, 010107, Inner Mongolia Autonomous Region, China
| | - Xiaoping Bai
- Department of Radiation Oncology, Baotou Cancer Hospital, Baotou, 014030, Inner Mongolia Autonomous Region, China
| | - Yunqi Han
- The Fourth Hospital of Inner Mongolia Autonomous Region, Hohhot, 010107, Inner Mongolia Autonomous Region, China
| | - Yu Ye
- Peking University Cancer Hospital (Inner Mongolia Campus) and Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, 010107, Inner Mongolia Autonomous Region, China
| | - Meiling Peng
- Hohhot First Hospital, Hohhot, 010107, Inner Mongolia Autonomous Region, China
| | - Hongwei Cui
- Peking University Cancer Hospital (Inner Mongolia Campus) and Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, 010107, Inner Mongolia Autonomous Region, China.
| | - Kai Li
- Department of Oncology, The Second Afliated Hospital of Baotou Medical College, Baotou, 014030, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
31
|
Guo Z, Li K, Ren X, Wang X, Yang D, Ma S, Zeng X, Zhang P. The role of the tumor microenvironment in HNSCC resistance and targeted therapy. Front Immunol 2025; 16:1554835. [PMID: 40236700 PMCID: PMC11996806 DOI: 10.3389/fimmu.2025.1554835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
The prognosis for head and neck squamous cell carcinoma (HNSCC) remains unfavorable, primarily due to significant therapeutic resistance and the absence effective interventions. A major obstacle in cancer treatment is the persistent resistance of cancer cells to a variety of therapeutic modalities. The tumor microenvironment (TME) which includes encompasses all non-malignant components and their metabolites within the tumor tissue, plays a crucial role in this context. The distinct characteristics of the HNSCC TME facilitate tumor growth, invasion, metastasis, and resistance to treatment. This review provides a comprehensive overview of the HNSCC TME components, with a particular focus on tumor-associated macrophages (TAMs), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), the extracellular matrix, reprogrammed metabolic processes, and metabolic products. It elucidates their contributions to modulating resistance to chemotherapy, radiotherapy, targeted therapy, and immunotherapy in HNSCC, and explores novel therapeutic strategies targeting the TME for HNSCC management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Peng Zhang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| |
Collapse
|
32
|
Yang L, Wang X, Wang S, Shen J, Li Y, Wan S, Xiao Z, Wu Z. Targeting lipid metabolism in regulatory T cells for enhancing cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189259. [PMID: 39798823 DOI: 10.1016/j.bbcan.2025.189259] [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: 06/11/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
As immunosuppressive cells, Regulatory T cells (Tregs) exert their influence on tumor immune escape within the tumor microenvironment (TME) by effectively suppressing the activity of other immune cells, thereby significantly impeding the anti-tumor immune response. In recent years, the metabolic characteristics of Tregs have become a focus of research, especially the important role of lipid metabolism in maintaining the function of Tregs. Consequently, targeted interventions aimed at modulating lipid metabolism in Tregs have been recognized as an innovative and promising approach to enhance the effectiveness of tumor immunotherapy. This review presents a comprehensive overview of the pivotal role of lipid metabolism in regulating the function of Tregs, with a specific focus on targeting Tregs lipid metabolism as an innovative approach to augment anti-tumor immune responses. Furthermore, we discuss potential opportunities and challenges associated with this strategy, aiming to provide novel insights for enhancing the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Liu Yang
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shurong Wang
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yaling Li
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shengli Wan
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Zhigui Wu
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
33
|
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
|
34
|
Yuan X, Yu S, Lin L, Chen Y, Wu Z, Fang X, Zhang W. Brusatol inhibits malignant phenotypes and lipid metabolism of osteosarcoma cells by regulating PI3K/AKT and MAPK pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156464. [PMID: 39970856 DOI: 10.1016/j.phymed.2025.156464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/06/2025] [Accepted: 02/02/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Osteosarcoma (OS), the most frequent type of primary bone cancer, has a poor prognosis in metastatic cases, with overall 5-year survival rates stagnating at 20 %-30 %. This highlights the critical need for innovative therapies to address the significant survival gap between metastatic and non-metastatic cases. Brusatol (BRU), a compound extracted from Brucea javanica, has shown promising anti-tumor properties in various cancers; however, its effects on OS have yet to be investigated. PURPOSE To investigate the anti-tumor mechanisms of BRU in OS and evaluate its potential therapeutic efficacy, with a particular focus on its impact on lipid metabolism and related signaling pathways. METHODS In vitro experiments to assess the anti-tumor effects of BRU involved colony formation, CCK-8, Transwell analysis, as well as flow cytometry. RNA sequencing was conducted to identify transcriptional changes in BRU-treated cells. The mechanism of action was investigated through analysis of lipid metabolism and key signaling pathways. Therapeutic efficacy and safety were evaluated in vivo using xenograft models. RESULTS BRU significantly inhibited OS cell proliferation, migration, and invasion, while also inducing G2/M phase cell cycle arrest as well as promoting apoptosis. Transcriptome analysis revealed that BRU affected lipid metabolism-related genes and suppressed the PI3K/AKT and MAPK pathways. BRU treatment reduced fatty acid synthase expression and free fatty acid content in OS cells. In vivo experiments demonstrated that BRU effectively restricted xenograft growth. CONCLUSION This study revealed that BRU exhibits potent anti-tumor effects in OS by modulating lipid metabolism through the PI3K/AKT and MAPK pathways.
Collapse
Affiliation(s)
- Xuhui Yuan
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Shaolin Yu
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Lan Lin
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Yang Chen
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Zhaoyang Wu
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Xinyu Fang
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China.
| | - Wenming Zhang
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China.
| |
Collapse
|
35
|
Liu X, Lu J, Ni X, He Y, Wang J, Deng Z, Zhang G, Shi T, Chen W. FASN promotes lipid metabolism and progression in colorectal cancer via the SP1/PLA2G4B axis. Cell Death Discov 2025; 11:122. [PMID: 40148316 PMCID: PMC11950308 DOI: 10.1038/s41420-025-02409-9] [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: 11/09/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Abnormal metabolic reprogramming is essential for tumorigenesis, metastasis, and the regulation of immune responses. Fatty acid synthase (FASN), a key enzyme in lipid metabolism, plays a crucial role in these processes. However, the relationship between FASN-mediated lipid reprogramming and the immune response in colorectal cancer (CRC) remains unclear. The present study demonstrated that FASN expression is elevated in CRC tissues and is significantly associated with poor prognosis. Functional experiments revealed that FASN promotes proliferation, migration, invasion, and phosphatidylcholine (PC) production in CRC cells. Additionally, in vivo experiments revealed that FASN knockdown significantly inhibits tumor growth and the spread of CRC cells to the lungs. Mechanistically, FASN, which is upregulated in CRC tissues, drives cancer cell proliferation, metastasis, and PC metabolism through the SP1/PLA2G4B axis, subsequently suppressing the antitumor response of natural killer (NK) cells in a PC-dependent manner. These findings provide new insights into lipid metabolism and the immunobiology of CRC, suggesting potential targets for the treatment and prevention of CRC. Schematic diagram showing the mechanism by which FASN promotes cancer cell proliferation, metastasis, and PC metabolism in CRC via the SP1/PLA2G4B axis, subsequently suppressing the antitumor response of NK cells in a PC-dependent manner. FFA free fatty acid, LPA lysophosphatidic acid, PA phosphatidate, DAG diglyceride, PC phosphatidylcholine, LPC lysophosphatidylcholine, CE cholesterol ester, TAG triacylglycerol.
Collapse
Affiliation(s)
- Xin Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Jiachun Lu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Xiangyu Ni
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Yuxin He
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Jiayu Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Zilin Deng
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
| |
Collapse
|
36
|
Chua AE, Go EP, Desaire H. Exploring Sample Storage Conditions for the Mass Spectrometric Analysis of Extracted Lipids from Latent Fingerprints. Biomolecules 2025; 15:477. [PMID: 40305181 PMCID: PMC12024923 DOI: 10.3390/biom15040477] [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: 02/26/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
In large-scale studies, uncontrolled systematic variability introduced during sample preparation, processing, and storage can interfere with the detection of subtle biological signals. This study evaluates storage conditions, including two sample preparation methods and storage durations, to minimize systematic variability in the analysis of extracted lipids from latent fingerprints. In the traditional approach, samples are prepared immediately, stored as lipid extracts, and processed in multiple batches. In an alternative method, samples are stored directly on the deposition foil, and preparation is delayed until all can be processed in a single batch. Storage duration is evaluated to determine if shorter storage with analysis in multiple batches is more effective than longer storage with analysis in a single batch. Our findings demonstrate that storage of latent fingerprint samples on the deposition foil is a viable option, with minimal degradation of key features even after eight months of storage. While some differences in lipid profiles were observed across storage conditions, these differences were minor and would likely have little impact in larger studies where biological variability is greater. These insights offer practical guidance for implementing latent fingerprint sampling in large-scale studies by identifying optimal conditions that preserve sample quality and streamline workflows.
Collapse
Affiliation(s)
| | | | - Heather Desaire
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA; (A.E.C.); (E.P.G.)
| |
Collapse
|
37
|
Liu J, Ma S, Deng D, Yang Y, Li J, Zhang Y, Yin P, Shang D. Multi-Omics Profiling Reveals Glycerolipid Metabolism-Associated Molecular Subtypes and Identifies ALDH2 as a Prognostic Biomarker in Pancreatic Cancer. Metabolites 2025; 15:207. [PMID: 40137171 PMCID: PMC11943634 DOI: 10.3390/metabo15030207] [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: 02/26/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Background: The reprogramming of lipid metabolism, especially glycerolipid metabolism (GLM), plays a key role in cancer progression and response to therapy. However, the role and molecular characterization of GLM in pancreatic cancer (PC) remain unclear. Methods: A pan-cancer analysis of glycerolipid metabolism-related genes (GMRGs) was first conducted to assess copy-number variants, single-nucleotide variations, methylation, and mRNA expression. Subsequently, GLM in PC was characterized using lipidomics, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomic analysis. A cluster analysis based on bulk RNA sequencing data from 930 PC samples identified GLM-associated subtypes, which were then analyzed for differences in prognosis, biological function, immune microenvironment, and drug sensitivity. To prioritize prognostically relevant GMRGs in PC, we employed a random forest (RF) algorithm to rank their importance across 930 PC samples. Finally, the key biomarker of PC was validated using PCR and immunohistochemistry. Results: Pan-cancer analysis identified molecular features of GMRGs in cancers, while scRNA-seq, spatial transcriptomics, and lipidomics highlighted GLM heterogeneity in PC. Two GLM-associated subtypes with significant prognostic, biofunctional, immune microenvironmental, and drug sensitivity differences were identified in 930 PC samples. Finally, ALDH2 was identified as a novel prognostic biomarker in PC and validated in a large number of datasets and clinical samples. Conclusions: This study highlights the crucial role of GLM in PC and defines a new PC subtype and prognostic biomarker. These findings establish a novel avenue for studying prognostic prediction and precision medicine in PC patients.
Collapse
Affiliation(s)
- Jifeng Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
| | - Shurong Ma
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
| | - Dawei Deng
- Department of Hepato-Biliary-Pancreas, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China;
| | - Yao Yang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Junchen Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Yunshu Zhang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Peiyuan Yin
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Dong Shang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
38
|
Wang H, Tu T, Yin L, Liu Z, Lu H. Single nucleotide polymorphisms in ovarian cancer impacting lipid metabolism and prognosis: an integrated TCGA database analysis. BMC Cancer 2025; 25:462. [PMID: 40082829 PMCID: PMC11907782 DOI: 10.1186/s12885-025-13841-6] [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: 11/11/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Ovarian cancer (OC) stands as a formidable adversary among women, remaining a leading cause of cancer-related mortality owing to its aggressive and invasive nature. Investigating prognostic markers intricately linked to OC's molecular pathogenesis represents a critical avenue for enhancing patient outcomes and survival prospects. In this comprehensive study, we embarked on a bioinformatics journey, leveraging the vast repository of single nucleotide polymorphism (SNP) data from OC patients available within the TCGA database. Our overarching goal was to unearth the genetic underpinnings of OC, shedding light on potential prognostic markers that could significantly impact clinical decision-making and patient care. Our meticulous analysis led to the discovery of five mutated genes-APOB, BRCA1, COL6A3, LRP1, and LRP1B-engaged in the intricate world of lipid metabolism. These genes, previously unexplored in the context of OC, emerged as prominent figures in our investigation, showcasing their potential roles in OC progression. The intricate interplay between lipid metabolism and cancer development has garnered considerable attention in recent years, and our findings underscore the relevance of these genes in the context of OC. To fortify our discoveries, we delved into the realm of survival analysis, a pivotal component of our investigation. The results yielded compelling evidence of significant correlations between patient survival and the expression levels of the aforementioned genes. This critical insight underscores the potential utility of these genes as prognostic markers, illuminating a path toward more personalized and effective approaches to patient care. Our study represents a multifaceted approach to unraveling the complex molecular pathogenesis of OC. By harnessing the power of high-throughput data mining, we uncovered genetic insights that may reshape our understanding of this formidable disease. We complemented these findings with advanced techniques such as RT-qPCR and Western blot, further dissecting the intricacies of OC's molecular landscape. This holistic approach not only deepens our understanding but also provides essential bioinformatics information that holds promise in assessing patient prognosis. In summary, our study represents a significant stride in the quest to decode the molecular intricacies of ovarian cancer. Our findings spotlight the potential prognostic significance of APOB, BRCA1, COL6A3, LRP1, and LRP1B, inviting further exploration into their roles in OC progression. Ultimately, our research carries the potential to shape the future of OC management, offering a glimpse into a more personalized and effective approach to patient care.
Collapse
Affiliation(s)
- Haoyu Wang
- Zhejiang University School of Medicine, #866 Yuhangtang RoadZhejiang Province, Hangzhou, 3100058, People's Republic of China
- Plastic & Cosmetic Center, College of Medicine, The First Affiliated Hospital, Zhejiang University, #79 Qingchun RoadZhejiang Province, Hangzhou, 310003, People's Republic of China
| | - Tian Tu
- Plastic & Cosmetic Center, College of Medicine, The First Affiliated Hospital, Zhejiang University, #79 Qingchun RoadZhejiang Province, Hangzhou, 310003, People's Republic of China
| | - Lijun Yin
- Department of Gynaecology and Obstetrics, College of Medicine, The First Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, 310003, People's Republic of China
| | - Zhenfeng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China
| | - Hui Lu
- Department of Orthopedics, College of Medicine, The First Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, 310003, People's Republic of China.
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, #866 Yuhangtang RoadZhejiang Province, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
39
|
Wan Y, Li G, Cui G, Duan S, Chang S. Reprogramming of Thyroid Cancer Metabolism: from Mechanism to Therapeutic Strategy. Mol Cancer 2025; 24:74. [PMID: 40069775 PMCID: PMC11895238 DOI: 10.1186/s12943-025-02263-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/06/2025] [Indexed: 03/15/2025] Open
Abstract
Thyroid cancer as one of the most prevalent malignancies of endocrine system, has raised public concern and more research on its mechanism and treatment. And metabolism-based therapies have advanced rapidly, for the exclusive metabolic profiling of thyroid cancer. In thyroid cancer cells, plenty of metabolic pathways are reprogrammed to accommodate tumor microenvironment. In this review, we initiatively summarize recent progress in the full-scale thyroid cancer metabolic rewiring and the interconnection of various metabolites. We also discuss the efficacy and prospect of metabolic targeted detection as well as therapy. Comprehending metabolic mechanism and characteristics of thyroid cancer roundly will be highly beneficial to managing individual patients.
Collapse
Affiliation(s)
- Yuxuan Wan
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China
- Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Guoqing Li
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China
- Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Gaoyuan Cui
- Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Saili Duan
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Department of Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Thyroid Disease in Hunan Province, Changsha, 410008, Hunan, People's Republic of China.
- Hunan Provincial Engineering Research Center for Thyroid and Related Diseases Treatment Technology, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
40
|
Xie X, Liu W, Yuan Z, Chen H, Mao W. Bridging epigenomics and tumor immunometabolism: molecular mechanisms and therapeutic implications. Mol Cancer 2025; 24:71. [PMID: 40057791 PMCID: PMC11889836 DOI: 10.1186/s12943-025-02269-y] [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: 11/12/2024] [Accepted: 02/11/2025] [Indexed: 04/02/2025] Open
Abstract
Epigenomic modifications-such as DNA methylation, histone acetylation, and histone methylation-and their implications in tumorigenesis, progression, and treatment have emerged as a pivotal field in cancer research. Tumors undergo metabolic reprogramming to sustain proliferation and metastasis in nutrient-deficient conditions, while suppressing anti-tumor immunity in the tumor microenvironment (TME). Concurrently, immune cells within the immunosuppressive TME undergo metabolic adaptations, leading to alterations in their immune function. The complicated interplay between metabolites and epigenomic modulation has spotlighted the significance of epigenomic regulation in tumor immunometabolism. In this review, characteristics of the epigenomic modification associated with tumors are systematically summarized alongside with their regulatory roles in tumor metabolic reprogramming and immunometabolism. Classical and emerging approaches are delineated to broaden the boundaries of research on the crosstalk research on the crosstalk between tumor immunometabolism and epigenomics. Furthermore, we discuss potential therapeutic strategies that target tumor immunometabolism to modulate epigenomic modifications, highlighting the burgeoning synergy between metabolic therapies and immunotherapy as a promising avenue for cancer treatment.
Collapse
Affiliation(s)
- Xiaowen Xie
- Department of Thoracic Surgery, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, 214023, China
| | - Weici Liu
- Department of Thoracic Surgery, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, 214023, China
- Center of Clinical Research, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China.
| | - Hanqing Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Wenjun Mao
- Department of Thoracic Surgery, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, 214023, China.
| |
Collapse
|
41
|
Yin X, Shen H, Wang H, Wang Q, Zhang S, Zhang C, Jia Q, Guo S, Xu X, Zhang W, Li B, Shi X, Gao S, Shi M, Zhao X, Wang S, Han J, Zhang G, Li Y, Li P, Jing W, Song B, Zheng K, Li G, Zhang Y, Jiang H, Wu C, Song Z, Niu G, Zhang Q, Guo J, Sun Z, Han F, Li Y, Gao D, Jin H, Yang H, Li J, Jin G. Pathogenic germline variants in Chinese pancreatic adenocarcinoma patients. Nat Commun 2025; 16:2214. [PMID: 40044664 PMCID: PMC11882848 DOI: 10.1038/s41467-025-57520-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
Putting pancreatic adenocarcinoma (PAAD) screening into perspective for high-risk individuals could significantly reduce cancer morbidity and mortality. Previous studies have profiled somatic mutations in PAAD. In contrast, the prevalence of mutations in PAAD predisposition genes has not been defined, especially in the Asian population. Using a multi-tier cohort design and whole genome/exome sequencing, we create a comprehensive germline mutation map of PAAD in 1,123 Chinese cancer patients in comparison with 11 pan-ethnic studies. For well-known pathogenic/likely pathogenic germline variants, Chinese patients exhibit overlapping but distinct germline mutation patterns comparing with Western cohorts, highlighted by lower mutation rates in known PAAD genes including BRCA1, BRCA2, ATM, CDKN2A, and CHEK2, and distinct mutations in CFTR, RAD51D, FANCA, ERCC2, and GNAS exclusive to Chinese patients. CFTR emerges as a top candidate gene following loss of heterozygosity analysis. Using an integrative multi-omics and functional validation paradigm, we discover that deleterious variants of uncertain significance may compromise CFTR's tumor suppressor function, and demonstrate the clinical relevance by using patient derived organoids for drug screen. Our multifaceted approach not only deepens the knowledge of population differences in PAAD germline mutations but also unveils potential avenues for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Xiaoyi Yin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
- Department of Pathology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Hui Shen
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, 264000, China
- Center for Translational Medicine, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Qingchen Wang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Shan Zhang
- Center for Translational Medicine, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Chunming Zhang
- Western Institute of Advanced Technology, Chinese Academy of Science, Chongqing, China
| | - Qi Jia
- Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Xiongfei Xu
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Wenhui Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Bo Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Meilong Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Xuenan Zhao
- Center for Translational Medicine, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Sheng Wang
- Center for Translational Medicine, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Jiawei Han
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
- Department of General Surgery, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
| | - Guoxiao Zhang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
- Department of General Surgery, The 72nd Group Army Hospital of Chinese People's Liberation Army, Huzhou, China
| | - Yikai Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Penghao Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Bin Song
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Kailian Zheng
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Gang Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Yijie Zhang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | - Cong Wu
- Clinical Research Unit, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
| | | | - Gang Niu
- Western Institute of Advanced Technology, Chinese Academy of Science, Chongqing, China
| | - Qiangzu Zhang
- Western Institute of Advanced Technology, Chinese Academy of Science, Chongqing, China
| | - Jianglong Guo
- Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Zhen Sun
- Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Fengxian Han
- Center for Translational Medicine, Second Military Medical University (Naval Medical University), Shanghai, 200433, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yunguang Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Haojie Jin
- Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China.
| | - Hongbo Yang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
| | - Jing Li
- Center for Translational Medicine, Second Military Medical University (Naval Medical University), Shanghai, 200433, China.
- Department of Precision Medicine, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200433, China.
| |
Collapse
|
42
|
Wan M, Pan S, Shan B, Diao H, Jin H, Wang Z, Wang W, Han S, Liu W, He J, Zheng Z, Pan Y, Han X, Zhang J. Lipid metabolic reprograming: the unsung hero in breast cancer progression and tumor microenvironment. Mol Cancer 2025; 24:61. [PMID: 40025508 PMCID: PMC11874147 DOI: 10.1186/s12943-025-02258-1] [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: 12/03/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Aberrant lipid metabolism is a well-recognized hallmark of cancer. Notably, breast cancer (BC) arises from a lipid-rich microenvironment and depends significantly on lipid metabolic reprogramming to fulfill its developmental requirements. In this review, we revisit the pivotal role of lipid metabolism in BC, underscoring its impact on the progression and tumor microenvironment. Firstly, we delineate the overall landscape of lipid metabolism in BC, highlighting its roles in tumor progression and patient prognosis. Given that lipids can also act as signaling molecules, we next describe the lipid signaling exchanges between BC cells and other cellular components in the tumor microenvironment. Additionally, we summarize the therapeutic potential of targeting lipid metabolism from the aspects of lipid metabolism processes, lipid-related transcription factors and immunotherapy in BC. Finally, we discuss the possibilities and problems associated with clinical applications of lipid‑targeted therapy in BC, and propose new research directions with advances in spatiotemporal multi-omics.
Collapse
Affiliation(s)
- Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haizhou Diao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Ziqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Shuya Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wan Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaying He
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Graduate School of Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zihan Zheng
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
43
|
Winkelkotte AM, Al-Shami K, Chaves-Filho AB, Vogel FCE, Schulze A. Interactions of Fatty Acid and Cholesterol Metabolism with Cellular Stress Response Pathways in Cancer. Cold Spring Harb Perspect Med 2025; 15:a041548. [PMID: 38951029 PMCID: PMC11875093 DOI: 10.1101/cshperspect.a041548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Lipids have essential functions as structural components of cellular membranes, as efficient energy storage molecules, and as precursors of signaling mediators. While deregulated glucose and amino acid metabolism in cancer have received substantial attention, the roles of lipids in the metabolic reprogramming of cancer cells are less well understood. However, since the first description of de novo fatty acid biosynthesis in cancer tissues almost 70 years ago, numerous studies have investigated the complex functions of altered lipid metabolism in cancer. Here, we will summarize the mechanisms by which oncogenic signaling pathways regulate fatty acid and cholesterol metabolism to drive rapid proliferation and protect cancer cells from environmental stress. The review also discusses the role of fatty acid metabolism in metabolic plasticity required for the adaptation to changing microenvironments during cancer progression and the connections between fatty acid and cholesterol metabolism and ferroptosis.
Collapse
Affiliation(s)
- Alina M Winkelkotte
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kamal Al-Shami
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Adriano B Chaves-Filho
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute of Chemistry, University of São Paulo, 05508000 São Paulo, Brazil
| | - Felix C E Vogel
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
44
|
Jiang S, Lin X, Chen B, Chen G, Kwan KJ, Liu J, Sun Q, Wang J, Lu Y, Tong J, Deng Y, Yu B, Tang J. ACSL1 Aggravates Thromboinflammation by LPC/LPA Metabolic Axis in Hyperlipidemia Associated Myocardial Ischemia-Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406359. [PMID: 39853712 PMCID: PMC11923997 DOI: 10.1002/advs.202406359] [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: 06/09/2024] [Revised: 12/24/2024] [Indexed: 01/26/2025]
Abstract
Acute myocardial infarction (AMI) is associated with well-established metabolic risk factors, especially hyperlipidemia and obesity. Myocardial ischemia-reperfusion injury (mIRI) significantly offsets the therapeutic efficacy of revascularization. Previous studies indicated that disrupted lipid homeostasis can lead to lipid peroxidation damage and inflammation, yet the underlying mechanisms remain unclear. Here, the study demonstrates that hyperlipidemia is a key driver of mIRI. Long-chain fatty acyl-CoA synthetase 1 (ACSL1) is upregulated in both hyperlipidemia and AMI patients. ACSL1 expression is induced by a high-fat microenvironment (oxLDL and palmitic acid) in a concentration-dependent manner. Interestingly, the protein level is positively correlated with total cholesterol level and thromboinflammatory biomarkers. Furthermore, ACSL1 reprogrammed lipid metabolism in monocytes, leading to the accumulation of lysophosphatidylcholine (LPC)/lysophosphatidic acid (LPA). The monocytic LPC/LPA axis accelerated lipid peroxidation and neutrophil extracellular traps (NETs)-induced thromboinflammation via the paracrine effect. The main LPA producer Autotaxinis is also induced under high-fat conditions and then exerts thromboinflammation response through converted LPC to LPA. Finally, ACSL1 knockdown or NETs release inhibitor (DNase I or GSK484) significantly alleviated mIRI in mice. These findings highlight ACSL1 and NETosis as potential key targets for preventing mIRI and underscore the lipid peroxidation in the mechanisms of ACSL1-mediated thromboinflammation.
Collapse
Affiliation(s)
- Shuai Jiang
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Xueguang Lin
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Bo Chen
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Gang Chen
- Department of CardiologyShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Kristine J.S. Kwan
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Jing Liu
- State Key Laboratory of Genetic EngineeringCollaborative Innovation Center for Genetics and DevelopmentSchool of Life Sciencesand Human Phenome InstituteFudan UniversityShanghai200438China
| | - Qi Sun
- Department of Endocrinology and MetabolismAffiliated Hospital of Nantong UniversityNantong226006China
| | - Jie Wang
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Yijie Lu
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Jindong Tong
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Ying Deng
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Bo Yu
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
- Department of Vascular SurgeryHuashan HospitalFudan UniversityShanghai200040China
| | - Jingdong Tang
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| |
Collapse
|
45
|
Chen Y, Zhong Z, Ruan X, Zhan X, Ding Y, Wei F, Qin X, Yu H, Lu Y. Novel biomarker in hepatocellular carcinoma: Stearoyl-CoA desaturase 1. Dig Liver Dis 2025; 57:770-781. [PMID: 39638727 DOI: 10.1016/j.dld.2024.11.008] [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: 07/19/2024] [Revised: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND In recent years, more and more studies have shown that reprogramming lipid metabolism plays an important role in the occurrence and development of hepatocellular carcinoma (HCC). However, there is a lack of systematic exploration of fatty acid (FA) profiles in HCC. AIMS This study aims to systematically investigate the FA profile in HCC and assess the diagnostic potential of stearoyl-CoA desaturase 1 (SCD1) as a biomarker for HCC. METHODS The FA profile in HCC tissues was detected by gas chromatography mass spectrometry. Abnormal FA metabolism was analyzed by qRT-PCR, Western blot. Immunohistochemical and bioinformatics analysis were used to analyze SCD1 expression and function. Receiver operating characteristic curves were used to analyze the diagnostic efficacy of SCD1, and the relationship between SCD1 and immune infiltration in HCC was analyzed by the biological information method. RESULTS FAs were found to accumulate in the HCC samples, and abnormal FA metabolism in HCC related to the upregulation of the expression and activity of SCD1. The combination of SCD1 and alpha-fetoprotein produced a greater area under the receiver operating characteristic curve (0.925, P < 0.001) than SCD1 or alpha-fetoprotein alone. It also showed better sensitivity (77.5 %). Besides, high SCD1 expression was found to be related to immune infiltration in HCC. CONCLUSION SCD1 can serve as a reliable biomarker for HCC diagnosis.
Collapse
Affiliation(s)
- Yongling Chen
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, No.6 Shuangyong Road, Nanning, Guangxi 530021, China
| | - Ziqing Zhong
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, No.6 Shuangyong Road, Nanning, Guangxi 530021, China
| | - Xuelian Ruan
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, No.6 Shuangyong Road, Nanning, Guangxi 530021, China
| | - Xiuyu Zhan
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital affiliated to Guangxi Medical University, No.8 Wenchang Road, Liuzhou, Guangxi 545006, China
| | - Yanting Ding
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital affiliated to Guangxi Medical University, No.8 Wenchang Road, Liuzhou, Guangxi 545006, China
| | - Fangyi Wei
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, No.6 Shuangyong Road, Nanning, Guangxi 530021, China
| | - Xue Qin
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, No.6 Shuangyong Road, Nanning, Guangxi 530021, China
| | - Hongli Yu
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital affiliated to Guangxi Medical University, No.8 Wenchang Road, Liuzhou, Guangxi 545006, China.
| | - Yu Lu
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital affiliated to Guangxi Medical University, No.8 Wenchang Road, Liuzhou, Guangxi 545006, China.
| |
Collapse
|
46
|
Jin N, Qian YY, Jiao XF, Wang Z, Li X, Pan W, Jiang JK, Huang P, Wang SY, Jin P, Gao QL, Liu D, Xia Y. Niraparib restricts intraperitoneal metastases of ovarian cancer by eliciting CD36-dependent ferroptosis. Redox Biol 2025; 80:103528. [PMID: 39922130 PMCID: PMC11851289 DOI: 10.1016/j.redox.2025.103528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/01/2025] [Indexed: 02/10/2025] Open
Abstract
Ovarian cancer (OC) is prone to peritoneum or omentum dissemination, thus giving rise to the formidable challenge of unresectable surgery and a dismal survival rate. Although niraparib holds a pivotal role in the maintenance treatment of OC, its effect on suppressing metastases during primary intervention remains enigmatic. Recently, we initiated a prospective clinical study (NCT04507841) in order to evaluate the therapeutic efficacy of neoadjuvant niraparib monotherapy for advanced OC with homologous recombination deficiency. An analysis of patient tumor burden before and after the niraparib challenge showed a remarkable vulnerability of OC intraperitoneal metastases to niraparib exposure. This killing capacity of niraparib was closely associated with the accumulation of fatty acids within the abdomen, which was confirmed by the increased susceptibility of tumor cells to niraparib treatment in the presence of fatty acids. In the context of abundant fatty acids, niraparib elevated intracellular levels of fatty acids and lipid peroxidation, leading to subsequent tumor cell ferroptosis in a p53 and BRCA-independent manner. Notably, under niraparib exposure, a critical fatty acid transporter CD36 was dramatically upregulated in tumors, facilitating excessive uptake of fatty acids. Pharmacological inhibition of either ferroptosis or CD36 impaired the anti-tumor activity of niraparib both in vitro and in murine intraperitoneal ID8 tumor models. Our findings demonstrate ferroptosis as a novel mechanism underlying the regression of OC metastases induced by niraparib, thereby offering tantalizing prospects for the frontline application of this agent in the management of OC.
Collapse
Affiliation(s)
- Ning Jin
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi-yu Qian
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-fei Jiao
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhen Wang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Medicine Research Centre of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, China
| | - Xin Li
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen Pan
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jin-kai Jiang
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pu Huang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Si-yuan Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ping Jin
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qing-lei Gao
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Liu
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Xia
- Cancer Biology Research Center (Key Laboratory of Chinese Ministry of Education), Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Gynecology and Obstetrics, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
47
|
Tu Y, Chen Y, Mo L, Yan G, Xie J, Ji X, Chen S, Niu C, Liao P. Multi-Omic Analysis Reveals a Lipid Metabolism Gene Signature and Predicts Prognosis and Chemotherapy Response in Thyroid Carcinoma. Cancer Med 2025; 14:e70819. [PMID: 40119647 PMCID: PMC11928767 DOI: 10.1002/cam4.70819] [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: 03/23/2024] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
OBJECTIVE Lipid metabolic reprogramming is closely intertwined with the development and progression of thyroid carcinoma (TC); however, its specific mechanism remains elusive. This study aimed to investigate the association between lipid metabolism and TC progression. METHODS We employed liquid chromatography-mass spectrometry (LC/MS) for an untargeted metabolomics analysis, comparing 12 TC patients and 12 healthy controls (HC). Additionally, we conducted the screening of differentially expressed genes (DEGs) and identified differentially expressed lipid metabolism genes (LMGs). Multi-omic findings related to lipid metabolism were integrated to establish a prognostic risk model. The resulting risk score stratified TC patients into high- and low-risk groups. Overall survival (O.S.) was assessed using Kaplan-Meier (K-M) analysis. The immune landscape was evaluated using the CIBERSORT algorithm, and chemotherapeutic response was predicted utilizing the "pRRophetic" R package. RESULTS Our metabolomic analysis revealed heightened lipid metabolic activity in TC, corroborated by similar findings in transcriptomic analysis. Multi-omic analysis identified key LMGs (FABP4, PPARGC1A, AGPAT4, ALDH1A1, TGFA, and GPAT3) associated with fatty acids and glycerophospholipids metabolism. A novel risk model, incorporating these LMGs, confirmed significantly worse O.S. (p = 0.0045) in the high-risk group based on TCGA_THCA. Furthermore, high-risk TC patients exhibited lower immune cell infiltration, and predictive outcomes indicated the efficacy of potential therapeutic drugs across risk groups. CONCLUSION This multi-omic analysis underscores the potential utility of the lipid metabolism risk model in guiding clinical treatment and improving outcomes for TC patients.
Collapse
Affiliation(s)
- Yuqin Tu
- Chongqing Medical UniversityChongqingChina
- Department of Clinical LaboratoryChongqing General HospitalChongqingChina
| | | | - Linlong Mo
- Department of Breast and Thyroid SurgeryChongqing General HospitalChongqingChina
| | - Guiling Yan
- Chongqing Medical UniversityChongqingChina
- Department of Clinical LaboratoryChongqing General HospitalChongqingChina
| | - Jingling Xie
- Chongqing Medical UniversityChongqingChina
- Department of Clinical LaboratoryChongqing General HospitalChongqingChina
| | - Xinyao Ji
- Chongqing Medical UniversityChongqingChina
- Department of Clinical LaboratoryChongqing General HospitalChongqingChina
| | - Shu Chen
- Chongqing Medical UniversityChongqingChina
- Department of Clinical LaboratoryChongqing General HospitalChongqingChina
| | - Changchun Niu
- Chongqing Medical UniversityChongqingChina
- Department of Clinical LaboratoryChongqing General HospitalChongqingChina
| | - Pu Liao
- Chongqing Medical UniversityChongqingChina
- Department of Clinical LaboratoryChongqing General HospitalChongqingChina
| |
Collapse
|
48
|
Puente-Cobacho B, Esteo C, Altea-Manzano P, Garcia-Perez JL, Quiles JL, Sanchez-Rovira P, Martín-Salvago MD, Molina-Jiménez L, Luque RJ, Fendt SM, Vera-Ramirez L. De novo lipogenesis protects dormant breast cancer cells from ferroptosis and promotes metastasis. Redox Biol 2025; 80:103480. [PMID: 39787900 PMCID: PMC11764609 DOI: 10.1016/j.redox.2024.103480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
Dormant disseminated tumor cells (DTCs) remain viable for years to decades before establishing a clinically overt metastatic lesion. DTCs are known to be highly resilient and able to overcome the multiple biological hurdles imposed along the metastatic cascade. However, the specific metabolic adaptations of dormant DTCs remain to be elucidated. Here, we reveal that dormant DTCs upregulate de novo lipogenesis and favor the activation and incorporation of monounsaturated fatty acids (MUFAs) to their cellular membranes through the activation of acyl-coenzyme A synthetase long-chain family member 3 (ACSL3). Pharmacologic inhibition of de novo lipogenesis or genetic knockdown of ACSL3 results in lipid peroxidation and non-apoptotic cell death through ferroptosis. Clinically, ACSL3 was found to be overexpressed in quiescent DTCs in the lymph nodes of breast cancer patients and to significantly correlate with shorter disease-free and overall survival. Our work provides new insights into the molecular mechanisms enabling the survival of dormant DTCs and supports the use of de novo lipogenesis inhibitors to prevent breast cancer metastasis.
Collapse
Affiliation(s)
- Beatriz Puente-Cobacho
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Cintia Esteo
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Patricia Altea-Manzano
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jose Luis Garcia-Perez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | | | | | | | - Rafael J Luque
- Pathological Anatomy Unit, University Hospital of Jaén, Jaén, Spain
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Laura Vera-Ramirez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain.
| |
Collapse
|
49
|
Chen Z, Gong Y, Chen F, Lee HJ, Qian J, Zhao J, Zhang W, Li Y, Zhou Y, Xu Q, Xia Y, Zhou L, Cheng J. Orchestrated desaturation reprogramming from stearoyl-CoA desaturase to fatty acid desaturase 2 in cancer epithelial-mesenchymal transition and metastasis. Cancer Commun (Lond) 2025; 45:245-280. [PMID: 39722173 PMCID: PMC11947613 DOI: 10.1002/cac2.12644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Adaptative desaturation in fatty acid (FA) is an emerging hallmark of cancer metabolic plasticity. Desaturases such as stearoyl-CoA desaturase (SCD) and fatty acid desaturase 2 (FADS2) have been implicated in multiple cancers, and their dominant and compensatory effects have recently been highlighted. However, how tumors initiate and sustain their self-sufficient FA desaturation to maintain phenotypic transition remains elusive. This study aimed to explore the molecular orchestration of SCD and FADS2 and their specific reprogramming mechanisms in response to cancer progression. METHODS The potential interactions between SCD and FADS2 were explored by bioinformatics analyses across multiple cancer cohorts, which guided subsequent functional and mechanistic investigations. The expression levels of desaturases were investigated with online datasets and validated in both cancer tissues and cell lines. Specific desaturation activities were characterized through various isomer-resolved lipidomics methods and sensitivity assays using desaturase inhibitors. In-situ lipid profiling was conducted using multiplex stimulated Raman scattering imaging. Functional assays were performed both in vitro and in vivo, with RNA-sequencing employed for the mechanism verification. RESULTS After integration of the RNA-protein-metabolite levels, the data revealed that a reprogramming from SCD-dependent to FADS2-dependent desaturation was linked to cancer epithelial-mesenchymal transition (EMT) and progression in both patients and cell lines. FADS2 overexpression and SCD suppression concurrently maintained EMT plasticity. A FADS2/β-catenin self-reinforcing feedback loop facilitated the degree of lipid unsaturation, membrane fluidity, metastatic potential and EMT signaling. Moreover, SCD inhibition triggered a lethal apoptosis but boosted survival plasticity by inducing EMT and enhancing FA uptake via adenosine monophosphate-activated protein kinase activation. Notably, this desaturation reprogramming increased transforming growth factor-β2, effectively sustaining aggressive phenotypes and metabolic plasticity during EMT. CONCLUSIONS These findings revealed a metabolic reprogramming from SCD-dependent to FADS2-dependent desaturation during cancer EMT and progression, which concurrently supports EMT plasticity. Targeting desaturation reprogramming represents a potential vulnerability for cancer metabolic therapy.
Collapse
Affiliation(s)
- Zhicong Chen
- Department of Obstetrics and GynecologyCenter for Reproductive MedicineGuangdong Provincial Key Laboratory of Major Obstetric DiseasesGuangdong Provincial Clinical Research Center for Obstetrics and GynecologyGuangdong‐Hong Kong‐Macao Greater Bay Area Higher Education Joint Laboratory of Maternal‐Fetal MedicineThe Third Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongP. R. China
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Yanqing Gong
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Fukai Chen
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
| | - Hyeon Jeong Lee
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
- College of Biomedical Engineering & Instrument ScienceKey Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jinqin Qian
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Jing Zhao
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua UniversityBeijingP. R. China
| | - Wenpeng Zhang
- State Key Laboratory of Precision Measurement Technology and InstrumentsDepartment of Precision InstrumentTsinghua UniversityBeijingP. R. China
| | - Yamin Li
- Department of Biomedical EngineeringTufts UniversityMedfordMassachusettsUSA
| | - Yihui Zhou
- College of Biomedical Engineering & Instrument ScienceKey Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qiaobing Xu
- Department of Biomedical EngineeringTufts UniversityMedfordMassachusettsUSA
| | - Yu Xia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua UniversityBeijingP. R. China
| | - Liqun Zhou
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Ji‐Xin Cheng
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
50
|
Wang L, Duan W, Ruan C, Liu J, Miyagishi M, Kasim V, Wu S. YY2-CYP51A1 signaling suppresses hepatocellular carcinoma progression by restraining de novo cholesterol biosynthesis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167658. [PMID: 39761760 DOI: 10.1016/j.bbadis.2025.167658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Lipid accumulation is a frequently observed characteristic of cancer. Lipid accumulation is closely related to tumor progression, metastasis, and drug resistance; however, the mechanism underlying lipid metabolic reprogramming in tumor cells is not fully understood. Yin yang 2 (YY2) is a C2H2‑zinc finger transcription factor that exerts tumor-suppressive effects. However, its involvement in tumor cell lipid metabolic reprogramming remains unclear. In the present study, we identified YY2 as a novel regulator of cholesterol metabolism. We showed that YY2 suppressed cholesterol accumulation in hepatocellular carcinoma (HCC) cells by downregulating the transcriptional activity of cytochrome P450 family 51 subfamily A member 1 (CYP51A1), a key enzyme in de novo cholesterol biosynthesis. Subsequently, through in vitro and in vivo experiments, we demonstrated that this downregulation is crucial for the YY2 tumor suppressive effect. Together, our findings unraveled a previously unprecedented regulation of HCC cells cholesterol metabolism, and eventually, their tumorigenic potential, through YY2 negative regulation on CYP51A1 expression. This study revealed a novel regulatory mechanism of lipid metabolic reprogramming in tumor cells and provided insights into the molecular mechanism underlying the YY2 the suppressive effect. Furthermore, our findings suggest a potential antitumor therapeutic strategy targeting cholesterol metabolic reprogramming using YY2.
Collapse
Affiliation(s)
- Lingxian Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wei Duan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Cao Ruan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jingyi Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Makoto Miyagishi
- Life Science Innovation, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China.
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China.
| |
Collapse
|