1
|
Kejun D, Hao H, Shuangshuang C, Yaoqin M, Wei Z, Ting Z, Jiarui Z, Wan S, Xiaoyu S, Hongbo W, Xianjina X. Multifunctional DNA nano-sponge system for targeted sensitization of ovarian cancer chemotherapy via metabolic reprogramming and ferroptosis induction. J Control Release 2025:113663. [PMID: 40158809 DOI: 10.1016/j.jconrel.2025.113663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
The inefficacy of chemotherapy in ovarian cancer, hindered by poor targeting and multiple drug resistance, is intricately linked to the tumor micro-environment and abnormal metabolic patterns. Here, we present ZnOov@DS-DOX, an innovative nanomaterial that integrates small-sized ZnO nanoparticles with oxygen vacancies, embedded within 2D-ZnMOF and encapsulated in a DNA nanosponge, to enhance doxorubicin delivery. This system targets ovarian cancer cells specifically through the MUC1 aptamer and remodels metabolic signaling within the tumor microenvironment. ZnOov alleviates hypoxia and reduces chemoresistance by catalyzing H2O2, while Zn2+ release activates DNAzymes to target HIF-1a mRNA. The system reduces the efflux of chemotherapy drugs and inhibits cellular glycolysis. Disrupting the Warburg effect enhances mitochondrial respiration, increases ROS levels, disrupts the redox system, and induces ferroptosis, thereby significantly improving therapeutic efficacy. The uptake rate of ovarian cancer cells for the system was nearly 15 times higher than that of normal cells. Following treatment, the IC50 value of ovarian cancer cells decreased by approximately 72.7 %. Our in vitro and in vivo studies demonstrate that ZnOov@DS-DOX effectively modulates tumor metabolic pathways, mitigates drug resistance, and enhances treatment outcomes, representing a significant advancement in nanomedicine and cancer therapy.
Collapse
Affiliation(s)
- Dong Kejun
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hu Hao
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Cheng Shuangshuang
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mu Yaoqin
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhang Wei
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhou Ting
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhang Jiarui
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shu Wan
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shen Xiaoyu
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wang Hongbo
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Clinical Research Center of Cancer Immunotherapy of Hubei, Wuhan 430022, China.
| | - Xiao Xianjina
- Department of Obstetrics and Gynecology, Union Hospital and Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
2
|
Zhang X, Peng L, Kuang S, Wang T, Wu W, Zuo S, Chen C, Ye J, Zheng G, Guo Y, He Y. Lactate accumulation from HIF-1α-mediated PMN-MDSC glycolysis restricts brain injury after acute hypoxia in neonates. J Neuroinflammation 2025; 22:59. [PMID: 40025545 PMCID: PMC11871681 DOI: 10.1186/s12974-025-03385-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
Fetal intrauterine distress (FD) during delivery can cause fetal intrauterine hypoxia, posing significant risks to the fetus, mother, and newborns. While studies highlight the role of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in neonatal diseases and tumor hypoxia, their specific involvement in newborns experiencing fetal distress during delivery (FDNB) is not well understood. Here, we found elevated PMN-MDSC activation, increased glycolysis, enhanced lactate production, and upregulated HIF-1α expression in the blood of FDNB neonates compared to healthy newborns (NNB). Importantly, PMN-MDSC levels were inversely correlated with neuron-specific enolase (NSE), a marker for neurological injury. In neonatal mice subjected to acute hypoxia, a 48-h exposure led to a shift from exacerbation to amelioration of brain damage when compared with a 24-h period. This change was associated with a reduction in microglial activation, a decrease in the expression of inflammatory factors within the microglia, alongside increased peripheral PMN-MDSC activation. Depleting PMN-MDSCs led to heightened microglial activation and aggravated brain injury. Mechanistically, enhanced activation of PMN-MDSCs promotes HIF-1α accumulation while enhancing glycolysis and lactate release, thereby mitigating neonatal brain injury. Notably, lactate supplementation in hypoxic mice rescued brain damage caused by insufficient PMN-MDSC activation due to HIF-1α deficiency. Our study clarifies the role of lactate in peripheral PMN-MDSCs after acute hypoxia and its effects on microglial activation and subsequent brain injury.
Collapse
Affiliation(s)
- Xiaogang Zhang
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Laiqin Peng
- Department of Gynecology and Obstetrics, Huizhou Central People's Hospital, Huizhou, China
| | - Shuyi Kuang
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tianci Wang
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Weibin Wu
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaowen Zuo
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chunling Chen
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences,, Guangzhou, China
| | - Jiaxiu Ye
- Department of Gynecology and Obstetrics, Huizhou Central People's Hospital, Huizhou, China
| | - Guilang Zheng
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences,, Guangzhou, China.
| | - Yuxiong Guo
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences,, Guangzhou, China.
| | - Yumei He
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Peng Z, Song J, Zhu W, Bao H, Hu Y, Shi Y, Cheng X, Jiang M, Fang F, Chen J, Shu X. Impact of sleep deprivation on colon cancer: Unraveling the KynA-P4HA2-HIF-1α axis in tumor lipid metabolism and metastasis. Mol Metab 2025; 93:102109. [PMID: 39920992 PMCID: PMC11869867 DOI: 10.1016/j.molmet.2025.102109] [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: 10/14/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVE There is growing evidence that sleep deprivation promotes cancer progression. In addition, colon cancer patients often experience sleep deprivation due to factors such as cancer pain and side effects of treatment. The occurrence of liver metastases is an important factor in the mortality of colon cancer patients. However, the relationship between sleep deprivation and liver metastases from colon cancer has not been elucidated. METHODS A sleep deprivation liver metastasis model was constructed to evaluate the effect of sleep deprivation on liver metastasis of colon cancer. Subsequently, mice feces were collected for untargeted metabolomics to screen and identify the key mediator, Kynurenic acid (KynA). Furthermore, HILPDA was screened by transcriptomics, and its potential mechanism was explored through ChIP, co-IP, ubiquitination experiments, phenotyping experiments, etc. RESULTS: Sleep deprivation promotes liver metastases in colon cancer. Functionally, sleep deprivation aggravates lipid accumulation and decreases the production of the microbiota metabolite KynA. In contrast, KynA inhibited colon cancer progression in vitro. In vivo, KynA supplementation reversed the promoting effects of sleep deprivation on liver metastases from colon cancer. Mechanistically, KynA downregulates the expression of P4HA2 to promote the ubiquitination and degradation of HIF-1α, which leads to a decrease in the transcription of HILPDA, and ultimately leads to an increase in lipolysis of colon cancer cells. CONCLUSIONS Our findings reveal that sleep deprivation impairs intracellular lipolysis by KynA, leading to lipid droplets accumulation in colon cancer cells. This process ultimately promotes colon cancer liver metastasis. This suggests a promising strategy for colon cancer treatment.
Collapse
Affiliation(s)
- Zuojie Peng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Jia Song
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Wenzhong Zhu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Haijun Bao
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Yuan Hu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Yongping Shi
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Xukai Cheng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Mi Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Feifei Fang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China
| | - Jinhuang Chen
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China.
| | - Xiaogang Shu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No.1277, Wuhan 430022, Hubei, China.
| |
Collapse
|
4
|
Zhang J, Ruan K, Chu Z, Wang X, Gu Y, Jin H, Zhang X, Liu Q, Yang J. Reprogramming of fatty acid metabolism: a hidden force regulating the occurrence and progression of cholangiocarcinoma. Cell Death Discov 2025; 11:72. [PMID: 39984452 PMCID: PMC11845788 DOI: 10.1038/s41420-025-02351-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/22/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor that originates from the bile duct epithelium and with a poor outcome due to lack of effective early diagnostic methods. Surgical resection is the preferred method for cure, but treatment options are limited for advanced diseases, such as distant metastatic or locally progressive tumors. Therefore, it is urgent to explore other new treatment methods. As modern living standards rise, the acceptance of high-fat, high-protein, and high-carbohydrate diets is growing among the public, and the resulting metabolic abnormalities are intimately linked to the initiation and spread of tumors. Metabolic reprogramming is a key mechanism in the process of tumor development and progression and is closely related to cancer cell proliferation, metastasis and drug resistance. Fatty acid (FA) metabolism, an integral component of cancer cell metabolism, can provide an energy source for cancer cells and participate in cell signaling, the regulation of the immune response and the maintenance of homeostasis of the internal environment, which are closely linked to the development and progression of CCA. Therefore, a better understanding of FA metabolism may provide promising strategies for early diagnosis, prognostic assessment and targeted therapy for CCA patients. In this paper, we review the effects of FA metabolism on CCA development and progression, summarize related mechanisms and the existing clinical applications of targeted lipid metabolism in CCA, and explore new targets for CCA metabolic therapy.
Collapse
Affiliation(s)
- Jinglei Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, Zhejiang Province, 310053, China
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, 310006, China
| | - Kaiyi Ruan
- Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China
| | - Zhuohuan Chu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, Zhejiang Province, 310053, China
| | - Xiang Wang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, 310006, China
| | - Ye Gu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, 310006, China
| | - Hangbin Jin
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, 310006, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province, 310006, China
- Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province, 310006, China
| | - Xiaofeng Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, Zhejiang Province, 310053, China
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, 310006, China
- Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province, 310006, China
- Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province, 310006, China
| | - Qiang Liu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, 310006, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province, 310006, China.
- Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province, 310006, China.
| | - Jianfeng Yang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, Zhejiang Province, 310053, China.
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, 310006, China.
- Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province, 310006, China.
- Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province, 310006, China.
| |
Collapse
|
5
|
Huang S, Wang H, Cao J, Pang Q. Hypoxia and lipid metabolism related genes drive proliferation migration and immune infiltration mechanisms in colorectal cancer subtyping. Sci Rep 2025; 15:2394. [PMID: 39827204 PMCID: PMC11742731 DOI: 10.1038/s41598-025-85809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
Hypoxia and lipid metabolism play crucial roles in the progression of colorectal cancer (CRC). However, the specific functions of hypoxia- and lipid metabolism-related genes (HLPG) in CRC and their relationships with patient prognosis remain unclear. Differential expression analysis using the TCGA-COAD and GEO databases identified 117 HLPGs through the intersection of the two gene sets. After univariate Cox regression analysis, 17 prognostically relevant HLPG were identified. Consensus clustering classified CRC samples into two subtypes, and the immune microenvironment differences between them were evaluated. A risk scoring model utilizing seven prognostically significant HLPGs was created and its predictive performance was assessed through survival analysis and ROC curves. Finally, the key genes ITLN1 and SFRP2 were functionally validated in CRC cell lines. HLPG was closely linked to CRC prognosis. Two molecular subtypes were identified: Cluster A, characterized by enriched immune pathways and higher immune infiltration, and Cluster B, associated with improved overall survival. The seven HLPG-based risk scoring model effectively stratified patients into high- and low-risk groups, with high-risk patients exhibiting significantly poorer survival outcomes. Functional studies confirmed that SFRP2 and ITLN1 play essential roles in CRC cell proliferation, migration, and epithelial-mesenchymal transition (EMT). Furthermore, ITLN1 upregulated PD-L1 expression, increasing sensitivity to immunotherapy. Hypoxia was found to promote lipid metabolic alterations by modulating SFRP2 and ITLN1 expression. This study highlights the prognostic significance of HLPGs in CRC and introduces a robust risk scoring model for patient outcome prediction. ITLN1 could be a target for enhancing immunotherapy response in CRC.
Collapse
Affiliation(s)
- Shansong Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Huiying Wang
- Department of Rheumatology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, 313000, China
| | - Jiaqing Cao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Qiang Pang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
6
|
Huang Y, Zhang R, Fan S, Shi M, Tang X, Wang X, Deng X. OSBPL10-CNBP axis mediates hypoxia-induced pancreatic cancer development. Biofactors 2025; 51:e2124. [PMID: 39329194 DOI: 10.1002/biof.2124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of malignancies with worst outcomes among digestive system tumors. Identification of novel biomarkers is of great significance for treatment researches and prognosis prediction of pancreatic cancer patients. Due to OSBPL10 known involvement in oncogenic activity in other tumors, we elucidated the mechanism underlying its contribution to pancreatic cancer progression. We employed data from the Gene Expression Omnibus database to detect the expression of OSBPL10 in normal and pancreatic cancer tissues. A series of assays were conducted to assess the impact of OSBPL10 on the proliferation and metastatic capacities of pancreatic cancer cells and the influence of OSBPL10 on macrophages were evaluated by Flow cytometry. In addition, Co-immunoprecipitation, mass spectrometry, and western blot assays were utilized to investigate the potential mechanisms of OSBPL10 activity. From our study, OSBPL10 is revealed to be upregulated in pancreatic cancer, with poor prognosis. The overexpression promotes malignant behaviors of pancreatic cancer cells and has an impact on tumor immune microenvironment by stimulating the transformation M1 macrophages into M2 macrophages. Mechanistically, hypoxia induces the expression of OSBPL10 through interaction between hypoxia-inducible factor 1-α and the promoter region of OSBPL10. Additionally, OSBPL10 directly bound to CNBP, mediating CNBP expression and ultimately regulating the proliferation and metastasis capacity of pancreatic cancer cells, as well as influencing macrophage polarization. The research emphasized the oncogenic role of OSBPL10 in pancreatic cancer, uncovering key mechanisms involving hypoxia, HIF-1α, and CNBP. The finding suggests that OSBPL10 is a novel biomarker in pancreatic cancer, making it a potential therapeutic target for intervention in this malignancy.
Collapse
Affiliation(s)
- Yishu Huang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Neoplasms Translational Medicine, Shanghai, China
| | - Ronghao Zhang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuyang Fan
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Minmin Shi
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Xiaomei Tang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xinjing Wang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Li J, Ping P, Li Y, Xu X. Fatty acid metabolism: A new target for nasopharyngeal carcinoma therapy. Chin J Cancer Res 2024; 36:652-668. [PMID: 39802901 PMCID: PMC11724175 DOI: 10.21147/j.issn.1000-9604.2024.06.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid metabolic reprogramming is considered one of the most prominent metabolic abnormalities in cancer, and fatty acid metabolism is a key aspect of lipid metabolism. Recent studies have shown that fatty acid metabolism and its related lipid metabolic pathways play important roles in the malignant progression of nasopharyngeal carcinoma (NPC). NPC cells adapt to harsh environments by enhancing biological processes such as fatty acid metabolism, uptake, production, and oxidation, thereby accelerating their growth. In addition, the reprogramming of fatty acid metabolism plays a central role in the tumor microenvironment (TME) of NPC, and the phenotypic transformation of immune cells is closely related to fatty acid metabolism. Moreover, the reprogramming of fatty acid metabolism in NPC contributes to immune escape, which significantly affects disease treatment, progression, recurrence, and metastasis. This review explores recent advances in fatty acid metabolism in NPC and focuses on the interconnections among metabolic reprogramming, tumor immunity, and corresponding therapies. In conclusion, fatty acid metabolism represents a potential target for NPC treatment, and further exploration is needed to develop strategies that target the interaction between fatty acid metabolic reprogramming and immunotherapy.
Collapse
Affiliation(s)
- Juan Li
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Pengbin Ping
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yanhua Li
- Department of International Medical, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Xiaoying Xu
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
8
|
Lin XT, Luo YD, Mao C, Gong Y, Hou Y, Zhang LD, Gu YP, Wu D, Zhang J, Zhang YJ, Tan DH, Xie CM. Integrated ubiquitomics characterization of hepatocellular carcinomas. Hepatology 2024:01515467-990000000-01044. [PMID: 39348425 DOI: 10.1097/hep.0000000000001096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/02/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND AND AIMS Patients with aggressive HCC have limited therapeutic options. Therefore, a better understanding of HCC pathogenesis is needed to improve treatment. Genomic studies of HCC have improved our understanding of cancer biology. However, the ubiquitomic characteristics of HCC remain poorly understood. We aimed to reveal the ubiquitomic characteristics of HCC and provide clinical feature biomarkers of the aggressive HCC that may be used for diagnosis or therapy in the clinic. APPROACH AND RESULTS The comprehensive proteomic, phosphoproteomic, and ubiquitomic analyses were performed on tumors and adjacent normal liver tissues from 85 patients with HCC. HCCs displayed overexpression of drugable targets CBR1-S151 and CPNE1-S55. COL4A1, LAMC1, and LAMA4 were highly expressed in the disease free survival-poor patients. Phosphoproteomic and ubiquitomic features of HCC revealed cross talk in metabolism and metastasis. Ubiquitomics predicted diverse prognosis and clarified HCC subtype-specific proteomic signatures. Expression of biomarkers TUBA1A, BHMT2, BHMT, and ACY1 exhibited differential ubiquitination levels and displayed high prognostic risk scores, suggesting that targeting these proteins or their modified forms may be beneficial for future clinical treatment. We validated that TUBA1A K370 deubiquitination drove severe HCC and labeled an aggressive subtype of HCCs. TUBA1A K370 deubiquitination was at least partly attributed to protein kinase B-mediated USP14 activation in HCC. Notably, targeting AKT-USP14-TUBA1A complex promoted TUBA1A degradation and blocked liver tumorigenesis in vivo. CONCLUSIONS This study expands our knowledge of ubiquitomic signatures, biomarkers, and potential therapeutic targets in HCC.
Collapse
Affiliation(s)
- Xiao-Tong Lin
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuan-Deng Luo
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Hepatobiliary Surgery, General Hospital of Xinjiang Military Region, Urumchi, Xinjiang, China
| | - Cui Mao
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Gong
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu Hou
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Lei-Da Zhang
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yong-Peng Gu
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Di Wu
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Zhang
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu-Jun Zhang
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - De-Hong Tan
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuan-Ming Xie
- Department of Hepatobiliary Surgery, Key Laboratory of Hepatobiliary and Pancreatic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
9
|
Huang T, Cao H, Liu C, Sun X, Dai S, Liu L, Wang Y, Guo C, Wang X, Gao Y, Tang W, Xia Y. MAL2 reprograms lipid metabolism in intrahepatic cholangiocarcinoma via EGFR/SREBP-1 pathway based on single-cell RNA sequencing. Cell Death Dis 2024; 15:411. [PMID: 38866777 PMCID: PMC11169275 DOI: 10.1038/s41419-024-06775-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 06/14/2024]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a highly aggressive cancer characterized by a poor prognosis and resistance to chemotherapy. In this study, utilizing scRNA-seq, we discovered that the tetra-transmembrane protein mal, T cell differentiation protein 2 (MAL2), exhibited specific enrichment in ICC cancer cells and was strongly associated with a poor prognosis. The inhibition of MAL2 effectively suppressed cell proliferation, invasion, and migration. Transcriptomics and metabolomics analyses suggested that MAL2 promoted lipid accumulation in ICC by stabilizing EGFR membrane localization and activated the PI3K/AKT/SREBP-1 axis. Molecular docking and Co-IP proved that MAL2 interacted directly with EGFR. Based on constructed ICC organoids, the downregulation of MAL2 enhanced apoptosis and sensitized ICC cells to cisplatin. Lastly, we conducted a virtual screen to identify sarizotan, a small molecule inhibitor of MAL2, and successfully validated its ability to inhibit MAL2 function. Our findings highlight the tumorigenic role of MAL2 and its involvement in cisplatin sensitivity, suggesting the potential for novel combination therapeutic strategies in ICC.
Collapse
Affiliation(s)
- Tian Huang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China
| | - Hengsong Cao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China
| | - Chuan Liu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China
| | - Xiaohu Sun
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China
| | - Shipeng Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China
| | - Li Liu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuliang Wang
- School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Guo
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou First People's Hospital, Guangzhou, Guangdong, China
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China.
| | - Yun Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China.
| | - Weiwei Tang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China.
| | - Yongxiang Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key laboratory of Hepatobiliary cancers,Nanjing, China, Nanjing, Jiangsu, China.
| |
Collapse
|
10
|
Feng D, Wang J, Xiao Y, Wu R, Li D, Tuo Z, Yu Q, Ye L, MIYAMOTO A, Yoo KH, Wei W, Ye X, Zhang C, Han P. SKA3 targeted therapies in cancer precision surgery: bridging bench discoveries to clinical applications - review article. Int J Surg 2024; 110:2323-2337. [PMID: 38241327 PMCID: PMC11020031 DOI: 10.1097/js9.0000000000001123] [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: 09/11/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024]
Abstract
Spindle and kinetochore-associated complex subunit 3 (SKA3) is a microtubule-binding subcomplex of the outer kinetochore, which plays a vital role in proper chromosomal segregation and cell division. Recently, SKA3 have been demonstrated its oncogenic role of tumorigenesis and development in cancers. In this review, the authors comprehensively deciphered SKA3 in human cancer from various aspects, including bibliometrics, pan-cancer analysis, and narrative summary. The authors also provided the top 10 predicted drugs targeting SKA3. The authors proposed that SKA3 was a potential target and brought new therapeutic opportunities for cancer patients.
Collapse
Affiliation(s)
- Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Yuhan Xiao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Akira MIYAMOTO
- Department of Rehabilitation, West Kyushu University, Japan
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Xing Ye
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Chi Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou
| | - Ping Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| |
Collapse
|
11
|
Li S, Yuan J, Cheng Z, Li Y, Cheng S, Liu X, Huang S, Xu Z, Wu A, Liu L, Dong J. Hsa_circ_0021205 enhances lipolysis via regulating miR-195-5p/HSL axis and drives malignant progression of glioblastoma. Cell Death Discov 2024; 10:71. [PMID: 38341418 DOI: 10.1038/s41420-024-01841-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Abnormal lipid metabolism is an essential hallmark of glioblastoma. Hormone sensitive lipase (HSL), an important rate-limiting enzyme contributed to lipolysis, which was involved in aberrant lipolysis of glioblastoma, however, its definite roles and the relevant regulatory pathway have not been fully elucidated. Our investigations disclosed high expression of HSL in glioblastoma. Knock-down of HSL restrained proliferation, migration, and invasion of glioblastoma cells while adding to FAs could significantly rescue the inhibitory effect of si-HSL on tumor cells. Overexpression of HSL further promoted tumor cell proliferation and invasion. Bioinformatics analysis and dual-luciferase reporter assay were performed to predict and verify the regulatory role of ncRNAs on HSL. Mechanistically, hsa_circ_0021205 regulated HSL expression by sponging miR-195-5p, which further promoted lipolysis and drove the malignant progression of glioblastoma. Besides, hsa_circ_0021205/miR-195-5p/HSL axis activated the epithelial-mesenchymal transition (EMT) signaling pathway. These findings suggested that hsa_circ_0021205 promoted tumorigenesis of glioblastoma through regulation of HSL, and targeting hsa_circ_0021205/miR-195-5p/HSL axis can serve as a promising new strategy against glioblastoma.
Collapse
Affiliation(s)
- Suwen Li
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiaqi Yuan
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, the Zhangjiagang Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Zhe Cheng
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, the Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yongdong Li
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shan Cheng
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinglei Liu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shilu Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhipeng Xu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Anyi Wu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Dong
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
12
|
Liu YT, Che Y, Qiu HL, Xia HX, Feng YZ, Deng JY, Yuan Y, Tang QZ. ADP-ribosylation: An emerging direction for disease treatment. Ageing Res Rev 2024; 94:102176. [PMID: 38141734 DOI: 10.1016/j.arr.2023.102176] [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: 09/07/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 12/25/2023]
Abstract
ADP-ribosylation (ADPr) is a dynamically reversible post-translational modification (PTM) driven primarily by ADP-ribosyltransferases (ADPRTs or ARTs), which have ADP-ribosyl transfer activity. ADPr modification is involved in signaling pathways, DNA damage repair, metabolism, immunity, and inflammation. In recent years, several studies have revealed that new targets or treatments for tumors, cardiovascular diseases, neuromuscular diseases and infectious diseases can be explored by regulating ADPr. Here, we review the recent research progress on ART-mediated ADP-ribosylation and the latest findings in the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Yu-Ting Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, PR China
| | - Yan Che
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, PR China
| | - Hong-Liang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, PR China
| | - Hong-Xia Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, PR China
| | - Yi-Zhou Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, PR China
| | - Jiang-Yang Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, PR China
| | - Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, PR China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, PR China.
| |
Collapse
|