1
|
He Y, Chen Y, Gao C. A mendelian randomization study on the association between type 2 diabetes and the risk of bladder cancer. Discov Oncol 2025; 16:446. [PMID: 40172743 PMCID: PMC11965069 DOI: 10.1007/s12672-025-02218-7] [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: 09/23/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
PURPOSE We used Mendelian randomization (MR) analyses to examine the potential causal effects of type 2 diabetes and glycemic traits on bladder cancer risk. METHODS Two-sample MR analyses were conducted using summary data from genome-wide association studies (GWAS). Exposures included type 2 diabetes, fasting glucose, glycosylated hemoglobin (HbA1c), fasting insulin, and proinsulin levels, with bladder cancer as the outcome. Four methods-inverse variance weighted, MR-Egger, weighted median, and weighted mode-were used to assess the causal effects. Sensitivity analyses were conducted to ensure that the results were robust. RESULTS In the inverse variance weighted model, a weak positive effect was detected between genetically predicted HbA1c and bladder cancer (OR = 1.003, 95% CI = 1.0001 to 1.0052, P = 0.043). Other MR methods produced results with the same trend, although not all were statistically significant. However, there was no evidence to support the effect of type 2 diabetes, fasting insulin, or proinsulin levels on bladder cancer. No significant heterogeneity or pleiotropy was detected. CONCLUSION Mendelian randomization analysis indicated a mild promoting effect of increased HbA1c levels on bladder cancer risk. Further studies with larger sample sizes are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Yuan He
- Department of Endocrinology, Second Hospital of Tianjin Medical University, No.23 of Pingjiang Road, Hexi District, Tianjin, 300211, China.
| | - Yu Chen
- Department of Endocrinology, Second Hospital of Tianjin Medical University, No.23 of Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Chang Gao
- Department of Endocrinology, Second Hospital of Tianjin Medical University, No.23 of Pingjiang Road, Hexi District, Tianjin, 300211, China
| |
Collapse
|
2
|
Nishikiori N, Ohguro H, Watanabe M, Higashide M, Ogawa T, Furuhashi M, Sato T. High-Glucose-Induced Metabolic and Redox Alterations Are Distinctly Modulated by Various Antidiabetic Agents and Interventions Against FABP5/7, MITF and ANGPTL4 in Melanoma A375 Cells. Int J Mol Sci 2025; 26:1014. [PMID: 39940783 PMCID: PMC11817646 DOI: 10.3390/ijms26031014] [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: 12/26/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Hyperglycemia-induced effects on cellular metabolic properties and reactive oxygen species (ROS) generation play pivotal roles in the pathogenesis of malignant melanoma (MM). This study assessed how metabolic states, ROS production, and related gene expression are modulated by antidiabetic agents. The anti-diabetic agents metformin (Met) and imeglimin (Ime), inhibitors of fatty acid-binding proteins 5/7 (MF6) and microphthalmia-associated transcription factor (MITF) (ML329), and siRNA-mediated knockdown of angiopoietin-like protein 4 (ANGPTL4), which affect mitochondrial respiration, ROS production, and related gene expression, were tested in A375 (MM cell line) cells cultured in low (5.5 mM) and high glucose (50 mM) conditions. Cellular metabolic functions were significantly and differently modulated by Met, Ime, MF6, or ML329 and knockdown of ANGPTL4. High glucose significantly enhanced ROS production, which was alleviated by Ime but not by Met. Both MF6 and ML329 reduced ROS levels under both low and high glucose conditions. Knockdown of ANGPTL4 enhanced the change in glucose-dependent ROS production. Gene expression related to mitochondrial respiration and the pathogenesis of MM was significantly modulated by different glucose conditions, antidiabetic agents, MF6, and ML329. These findings suggest that glucose-dependent changes in cellular metabolism and redox status are differently modulated by antidiabetic agents, inhibition of fatty acid-binding proteins or MITF, and ANGPTL4 knockdown in A375 cells.
Collapse
Affiliation(s)
- Nami Nishikiori
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.)
| | - Megumi Watanabe
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.)
| | - Megumi Higashide
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.)
| | - Toshifumi Ogawa
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.O.); (M.F.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| |
Collapse
|
3
|
Chen A, Huang H, Fang S, Hang Q. ROS: A "booster" for chronic inflammation and tumor metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189175. [PMID: 39218404 DOI: 10.1016/j.bbcan.2024.189175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Reactive oxygen species (ROS) are a group of highly active molecules produced by normal cellular metabolism and play a crucial role in the human body. In recent years, researchers have increasingly discovered that ROS plays a vital role in the progression of chronic inflammation and tumor metastasis. The inflammatory tumor microenvironment established by chronic inflammation can induce ROS production through inflammatory cells. ROS can then directly damage DNA or indirectly activate cellular signaling pathways to promote tumor metastasis and development, including breast cancer, lung cancer, liver cancer, colorectal cancer, and so on. This review aims to elucidate the relationship between ROS, chronic inflammation, and tumor metastasis, explaining how chronic inflammation can induce tumor metastasis and how ROS can contribute to the evolution of chronic inflammation toward tumor metastasis. Interestingly, ROS can have a "double-edged sword" effect, promoting tumor metastasis in some cases and inhibiting it in others. This article also highlights the potential applications of ROS in inhibiting tumor metastasis and enhancing the precision of tumor-targeted therapy. Combining ROS with nanomaterials strategies may be a promising approach to enhance the efficacy of tumor treatment.
Collapse
Affiliation(s)
- Anqi Chen
- Medical College, Yangzhou University, Yangzhou 225009, China
| | - Haifeng Huang
- Department of Laboratory Medicine, The First People's Hospital of Yancheng, Yancheng 224006, China; Department of Laboratory Medicine, Yancheng Clinical Medical College of Jiangsu University, Yancheng 224006, China
| | - Sumeng Fang
- School of Mathematics, Tianjin University, Tianjin 300350, China
| | - Qinglei Hang
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining 221200, China; Key Laboratory of Jiangsu Province University for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou 225009, China; Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
4
|
Genard GC, Tirinato L, Pagliari F, Da Silva J, Giammona A, Alquraish F, Reyes MP, Bordas M, Marafioti MG, Franco SD, Janssen J, Garcia‐Calderón D, Hanley R, Nistico C, Fukasawa Y, Müller T, Krijgsveld J, Todaro M, Costanzo FS, Stassi G, Nessling M, Richter K, Maass KK, Liberale C, Seco J. Lipid droplets and small extracellular vesicles: More than two independent entities. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e162. [PMID: 39257626 PMCID: PMC11386333 DOI: 10.1002/jex2.162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 09/12/2024]
Abstract
Despite increasing knowledge about small extracellular vesicle (sEV) composition and functions in cell-cell communication, the mechanism behind their biogenesis remains unclear. Here, we reveal for the first time that sEV biogenesis and release into the microenvironment are tightly connected with another important organelle, Lipid Droplets (LDs). The correlation was observed in several human cancer cell lines as well as patient-derived colorectal cancer stem cells (CR-CSCs). Our results demonstrated that external stimuli such as radiation, pH, hypoxia or lipid-interfering drugs, known to affect the number of LDs/cell, similarly influenced sEV secretion. Importantly, through multiple omics data, at both mRNA and protein levels, we revealed RAB5C as a potential important molecular player behind this organelle connection. Altogether, the potential to fine-tune sEV biogenesis by targeting LDs could significantly impact the amount, cargos and properties of these sEVs, opening new clinical perspectives.
Collapse
Affiliation(s)
- Géraldine C. Genard
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Luca Tirinato
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Experimental and Clinical Medicine, Nanotechnology Research CenterUniversity of Magna GraeciaCatanzaroItaly
- Department of Medical and Surgical ScienceUniversity Magna GraeciaCatanzaroItaly
| | - Francesca Pagliari
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jessica Da Silva
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Alessandro Giammona
- Biological and Environmental Science and EngineeringKing Abdullah University of Science and Technology (KAUST)ThuwalSaudi Arabia
- Institute of Molecular Bioimaging and Physiology (IBFM)National Research Council (CNR)SegrateItaly
| | - Fatema Alquraish
- Biological and Environmental Science and EngineeringKing Abdullah University of Science and Technology (KAUST)ThuwalSaudi Arabia
| | - Maria Parra Reyes
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Marie Bordas
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Maria Grazia Marafioti
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Simone Di Franco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE)University of PalermoPalermoItaly
| | - Jeannette Janssen
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Daniel Garcia‐Calderón
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Physics and AstronomyHeidelberg UniversityHeidelbergGermany
| | - Rachel Hanley
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Clelia Nistico
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Experimental and Clinical MedicineUniversity of Magna GraeciaCatanzaroItaly
| | - Yoshinori Fukasawa
- Core Labs, King Abdullah University of Science and Technology(KAUST)ThuwalSaudi Arabia
| | - Torsten Müller
- German Cancer Research Center, DKFZHeidelbergGermany
- Medical FacultyHeidelberg UniversityHeidelbergGermany
| | - Jeroen Krijgsveld
- Medical FacultyHeidelberg UniversityHeidelbergGermany
- Proteomics of Stem Cells and CancerGerman Cancer Research CenterHeidelbergGermany
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE)University of PalermoPalermoItaly
| | | | - Giorgio Stassi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE)University of PalermoPalermoItaly
| | - Michelle Nessling
- Electron Microscopy FacilityGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Karsten Richter
- Electron Microscopy FacilityGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Kendra K. Maass
- Hopp‐Children's Cancer Center Heidelberg (KiTZ)HeidelbergGermany
| | - Carlo Liberale
- Biological and Environmental Science and EngineeringKing Abdullah University of Science and Technology (KAUST)ThuwalSaudi Arabia
| | - Joao Seco
- Division of Biomedical Physics in Radiation OncologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Physics and AstronomyHeidelberg UniversityHeidelbergGermany
| |
Collapse
|
5
|
Xie Y, Shao X, Zhang P, Zhang H, Yu J, Yao X, Fu Y, Wei J, Wu C. High Starch Induces Hematological Variations, Metabolic Changes, Oxidative Stress, Inflammatory Responses, and Histopathological Lesions in Largemouth Bass ( Micropterus salmoides). Metabolites 2024; 14:236. [PMID: 38668364 PMCID: PMC11051861 DOI: 10.3390/metabo14040236] [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: 03/25/2024] [Revised: 04/09/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
This study evaluated effects of high starch (20%) on hematological variations, glucose and lipid metabolism, antioxidant ability, inflammatory responses, and histopathological lesions in largemouth bass. Results showed hepatic crude lipid and triacylglycerol (TAG) contents were notably increased in fish fed high starch. High starch could increase counts of neutrophils, lymphocytes, monocytes, eosinophils, and basophils and serum contents of TAG, TBA, BUN, and LEP (p < 0.05). There were increasing trends in levels of GLUT2, glycolysis, gluconeogenesis, and LDH in fish fed high starch through the AKT/PI3K signal pathway. Meanwhile, high starch not only triggered TAG and cholesterol synthesis, but mediated cholesterol accumulation by reducing ABCG5, ABCG8, and NPC1L1. Significant increases in lipid droplets and vacuolization were also shown in hepatocytes of D3-D7 groups fed high starch. In addition, high starch could decrease levels of mitochondrial Trx2, TrxR2, and Prx3, while increasing ROS contents. Moreover, high starch could notably increase amounts of inflammatory factors (IL-1β, TNF-α, etc.) by activating NLRP3 inflammasome key molecules (GSDME, caspase 1, etc.). In conclusion, high starch could not only induce metabolic disorders via gluconeogenesis and accumulation of glycogen, TAG, and cholesterol, but could disturb redox homeostasis and cause inflammatory responses by activating the NLRP3 inflammasome in largemouth bass.
Collapse
Affiliation(s)
| | - Xianping Shao
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Department of Fisheries, School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (Y.X.); (P.Z.); (H.Z.); (J.Y.); (X.Y.); (Y.F.); (J.W.)
| | | | | | | | | | | | | | - Chenglong Wu
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Department of Fisheries, School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (Y.X.); (P.Z.); (H.Z.); (J.Y.); (X.Y.); (Y.F.); (J.W.)
| |
Collapse
|
6
|
Wang Y, Xu C, Yang X, Liu X, Guo Z, Lin X, Li L, Huang Z. Glycerol-3-phosphate acyltransferase 3-mediated lipid droplets accumulation confers chemoresistance of colorectal cancer. MedComm (Beijing) 2024; 5:e486. [PMID: 38344398 PMCID: PMC10857777 DOI: 10.1002/mco2.486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/18/2023] [Accepted: 12/25/2023] [Indexed: 10/23/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy worldwide. It is well known that lipid metabolism reprogramming contributes to the tumor progression. However, the lipid metabolic alterations and potential remodeling mechanism underlying the chemoresistance of CRC remain largely unclear. In this study, we compared the gene expression profiles of chemoresistant versus control CRC cells from the GEO database and identified a key factor, Glycerol-3-phosphate acyltransferase 3 (GPAT3), that promotes lipid droplet (LD) production and confers chemoresistance of CRC. With applying of HPLC-MS and molecular dynamics simulation, we also demonstrated that the activity of lysophosphatidic acid synthesis by GPAT3 was dependent on its acetylation at K316 site. In particular, GPAT3-mediated LD accumulation inhibited immunogenic cell death of tumor, and thus facilitated CD8+ T-cell exhaustion and malignant progression in mouse xenografts and hepatic-metastasis tumors in CRC patients. High GPAT3 expression turned CRC cells into nonimmunogenic cells after (Oxaliplatin) Oxa treatment, which was supported by a decrease in cytotoxic IFN-γ release and CD8+ T-cell exhaustion. In conclusion, these findings revealed the role of GPAT3-associated LD accumulation, which conferred a malignant phenotype (chemoresistance) and regulated the tumor microenvironment of CRC. These results suggest that GPAT3 is a potential target to enhance CRC chemosensitivity and develop novel therapeutic interventions.
Collapse
Affiliation(s)
- Ying Wang
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
| | - Caihua Xu
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Xianfeng Yang
- Department of RadiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Xiaofei Liu
- Department of thyroid breast surgery, First Clinical CollegeShandong University of Traditional Chinese MedicineJinanShandongChina
| | - Zijian Guo
- Department of Oncological SurgeryAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
| | - Xinyu Lin
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
| | - Lihua Li
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
| | - Zhaohui Huang
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
| |
Collapse
|
7
|
Deng J, Wang X, Zhao Y, Zhao X, Yang L, Qi Z. A dual donor-acceptor fluorescent probe with viscosity response and lipid droplets targeting to initiate oxidative stress for tumor elimination. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 305:123503. [PMID: 37857075 DOI: 10.1016/j.saa.2023.123503] [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: 07/03/2023] [Revised: 09/09/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023]
Abstract
A dual donor-acceptor photosensitizer TCN-2 prepared based on single donor-acceptor could fulfil lipid droplets targeting to trigger apoptosis and tumor growth arrest. Meanwhile, all of experiments both in phosphate buffer solution and intracellular surroundings have demonstrated that TCN-2 catalyzed the production of type I as well as type II reactive oxygen species, forming a hybrid reactive oxygen species pattern, indicating that TCN-2 could be applied to initiate a series of biological responses triggered by oxidative stress within most high-viscosity solid tumors. In addition, TCN-2 also has the capability of fluorescence imaging, which could perfectly combine therapeutic imaging to achieve therapeutic effects while identifying cancerous lesions. Due to the structural design of double electron-absorbing groups, TCN-2 retained excellent lipophilicity while enhancing solubility in the biological environment. Terrific biocompatibility, minimal phototoxic damage to normal cells and tissues, and specific driving to prescriptive organelles to maximize therapeutic effects were used to enhance the therapeutic effect of photodynamic therapy to cease disease progression.
Collapse
Affiliation(s)
- Jing Deng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xing Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yongfei Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinxin Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Li Yang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Zhengjian Qi
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
8
|
Li S, Ruan B, Wang Z, Xia J, Lin Q, Xu R, Zhu H, Yu Z. Glucose dysregulation promotes oncogenesis in human bladder cancer by regulating autophagy and YAP1/TAZ expression. J Cell Mol Med 2023; 27:3744-3759. [PMID: 37665055 PMCID: PMC10718143 DOI: 10.1111/jcmm.17943] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/08/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023] Open
Abstract
Glucose dysregulation is strongly correlated with cancer development, and cancer is prevalent in patients with Type 2 diabetes (T2D). We aimed to elucidate the mechanism underlying autophagy in response to glucose dysregulation in human bladder cancer (BC). 220 BC patients were included in this retrospective study. The expression of YAP1, TAZ and AMPK, EMT-associated markers, and autophagy marker proteins was analysed by immunohistochemistry, western blotting, and quantitative real-time PCR (qPCR). Further, T24 and UMUC-3 BC cells were cultured in media with different glucose concentrations, and the expression of YAP1, TAZ, AMPK and EMT-associated markers, and autophagy marker proteins was analysed by western blotting and qPCR. Autophagy was observed by immunofluorescence and electron microscopy. BC cell viability was tested using MTT assays. A xenograft nude mouse model of diabetes was used to evaluate tumour growth, metastasis and survival. A poorer pathologic grade and tumour-node-metastasis stage were observed in patients with BC with comorbid T2D than in others with BC. YAP1 and TAZ were upregulated in BC samples from patients with T2D. Mechanistically, high glucose (HG) promoted BC progression both in vitro and in vivo and inhibited autophagy. Specifically, various autophagy marker proteins and AMPK were negatively regulated under HG conditions and correlated with YAP1 and TAZ expression. These results demonstrate that HG inhibits autophagy and promotes cancer development in BC. YAP1/TAZ/AMPK signalling plays a crucial role in regulating glucose dysregulation during autophagy. Targeting these effectors exhibits therapeutic significance and can serve as prognostic markers in BC patients with T2D.
Collapse
Affiliation(s)
- Shi Li
- Department of Urology, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang ProvinceThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Banzhan Ruan
- Department of Oncology of The First Affiliated Hospital and Tumor InstituteHainan Medical UniversityHaikouHainanChina
| | - Zhi Wang
- Department of Urology and Chest SurgeryThe People Hospital of TongjiangBazhongSichuanChina
| | - Jianling Xia
- Department of Oncology and HematologyThe People Hospital of TongjiangBazhongSichuanChina
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalHospital of the University of Electronic Science and Technology of ChinaChengduChina
| | - Qi Lin
- Department of UrologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ruoting Xu
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Hua Zhu
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Zhixian Yu
- Department of UrologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
9
|
Bhowmick S, Biswas T, Ahmed M, Roy D, Mondal S. Caveolin-1 and lipids: Association and their dualism in oncogenic regulation. Biochim Biophys Acta Rev Cancer 2023; 1878:189002. [PMID: 37848094 DOI: 10.1016/j.bbcan.2023.189002] [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: 08/14/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023]
Abstract
Caveolin-1 (Cav-1) is a structural protein of caveolae that functions as a molecular organizer for different cellular functions including endocytosis and cellular signaling. Cancer cells take advantage of the physical position of Cav-1, as it can communicate with extracellular matrix, help to organize growth factor receptors, redistribute cholesterol and glycosphingolipids, and finally transduce signals within the cells for oncogenesis. Recent studies emphasize the exceeding involvement of Cav-1 with different lipid bodies and in altering the metabolism, especially lipid metabolism. However, the association of Cav-1 with different lipid bodies like lipid rafts, lipid droplets, cholesterols, sphingolipids, and fatty acids is remarkably dynamic. The lipid-Cav-1 alliance plays a dual role in carcinogenesis. Both cancer progression and regression are modified and affected by the type of lipid molecule's association with Cav-1. Accordingly, this Cav-1-lipid cooperation exemplifies a cancer-type-specific treatment strategy for a better prognosis of the disease. In this review, we first present Cav-1 as an oncogenic molecule and its communication via lipid raft. We discussed the involvement of Cav-1 with lipid droplets, Cholesterol, sphingolipids, gangliosides, and ceramides. Further, we describe the Cav-1-mediated altered Fatty acid metabolism in cancer and the strategic therapeutic approaches toward Cav-1 targeting.
Collapse
Affiliation(s)
- Sramana Bhowmick
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Tannishtha Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Mehnaz Ahmed
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Debarshi Roy
- Department of Biological Sciences, Alcorn State University, Lorman, MS 39096, USA
| | - Susmita Mondal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India.
| |
Collapse
|
10
|
Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K, Yang JL. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics. J Hematol Oncol 2023; 16:103. [PMID: 37700339 PMCID: PMC10498649 DOI: 10.1186/s13045-023-01498-2] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Lipid metabolic reprogramming is an emerging hallmark of cancer. In order to sustain uncontrolled proliferation and survive in unfavorable environments that lack oxygen and nutrients, tumor cells undergo metabolic transformations to exploit various ways of acquiring lipid and increasing lipid oxidation. In addition, stromal cells and immune cells in the tumor microenvironment also undergo lipid metabolic reprogramming, which further affects tumor functional phenotypes and immune responses. Given that lipid metabolism plays a critical role in supporting cancer progression and remodeling the tumor microenvironment, targeting the lipid metabolism pathway could provide a novel approach to cancer treatment. This review seeks to: (1) clarify the overall landscape and mechanisms of lipid metabolic reprogramming in cancer, (2) summarize the lipid metabolic landscapes within stromal cells and immune cells in the tumor microenvironment, and clarify their roles in tumor progression, and (3) summarize potential therapeutic targets for lipid metabolism, and highlight the potential for combining such approaches with other anti-tumor therapies to provide new therapeutic opportunities for cancer patients.
Collapse
Affiliation(s)
- Hao-Ran Jin
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zi-Jing Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ming-Jia Xi
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bi-Han Xia
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Deng
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Jin-Lin Yang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Singh L, Nair L, Kumar D, Arora MK, Bajaj S, Gadewar M, Mishra SS, Rath SK, Dubey AK, Kaithwas G, Choudhary M, Singh M. Hypoxia induced lactate acidosis modulates tumor microenvironment and lipid reprogramming to sustain the cancer cell survival. Front Oncol 2023; 13:1034205. [PMID: 36761981 PMCID: PMC9906992 DOI: 10.3389/fonc.2023.1034205] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023] Open
Abstract
It is well known that solid hypoxic tumour cells oxidise glucose through glycolysis, and the end product of this pathway is fermented into lactate which accumulates in the tumour microenvironment (TME). Initially, it was proclaimed that cancer cells cannot use lactate; therefore, they dump it into the TME and subsequently augment the acidity of the tumour milieu. Furthermore, the TME acts as a lactate sink with stope variable amount of lactate in different pathophysiological condition. Regardless of the amount of lactate pumped out within TME, it disappears immediately which still remains an unresolved puzzle. Recent findings have paved pathway in exploring the main role of lactate acidosis in TME. Cancer cells utilise lactate in the de novo fatty acid synthesis pathway to initiate angiogenesis and invasiveness, and lactate also plays a crucial role in the suppression of immunity. Furthermore, lactate re-programme the lipid biosynthetic pathway to develop a metabolic symbiosis in normoxic, moderately hypoxic and severely hypoxic cancer cells. For instance: severely hypoxic cancer cells enable to synthesizing poly unsaturated fatty acids (PUFA) in oxygen scarcity secretes excess of lactate in TME. Lactate from TME is taken up by the normoxic cancer cells whereas it is converted back to PUFAs after a sequence of reactions and then liberated in the TME to be utilized in the severely hypoxic cancer cells. Although much is known about the role of lactate in these biological processes, the exact molecular pathways that are involved remain unclear. This review attempts to understand the molecular pathways exploited by lactate to initiate angiogenesis, invasiveness, suppression of immunity and cause re-programming of lipid synthesis. This review will help the researchers to develop proper understanding of lactate associated bimodal regulations of TME.
Collapse
Affiliation(s)
- Lakhveer Singh
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Lakshmi Nair
- Department of Pharmaceutical Science, Assam University (A Central University), Silchar, Assam, India
| | - Dinesh Kumar
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Mandeep Kumar Arora
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Sakshi Bajaj
- Chaudhary Devi Lal College of Pharmacy, Yamuna Nagar, India
| | - Manoj Gadewar
- School of Medical and Allied Sciences, KR Mangalam University, Gurgaon, India
| | | | - Santosh Kumar Rath
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Amit Kumar Dubey
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Gaurav Kaithwas
- Department of Pharmaceutical Science, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Manjusha Choudhary
- University Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, India,*Correspondence: Manjusha Choudhary, ; Manjari Singh,
| | - Manjari Singh
- Department of Pharmaceutical Science, Assam University (A Central University), Silchar, Assam, India,*Correspondence: Manjusha Choudhary, ; Manjari Singh,
| |
Collapse
|
12
|
Moreira-Barbosa C, Matos A, Fernandes R, Mendes-Ferreira M, Rodrigues R, Cruz T, Costa ÂM, Cardoso AP, Ghilardi C, Oliveira MJ, Ribeiro R. The role of fatty acids metabolism on cancer progression and therapeutics development. BIOACTIVE LIPIDS 2023:101-132. [DOI: 10.1016/b978-0-12-824043-4.00007-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Pagliari F, Sogne E, Panella D, Perozziello G, Liberale C, Das G, Turdo A, Di Franco S, Seco J, Falqui A, Gratteri S, Pujia A, Di Fabrizio E, Candeloro P, Tirinato L. Correlative Raman-Electron-Light (CREL) Microscopy Analysis of Lipid Droplets in Melanoma Cancer Stem Cells. BIOSENSORS 2022; 12:1102. [PMID: 36551069 PMCID: PMC9776032 DOI: 10.3390/bios12121102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
Among all neoplasms, melanoma is characterized by a very high percentage of cancer stem cells (CSCs). Several markers have been proposed for their identification, and lipid droplets (LDs) are among them. Different techniques are used for their characterization such as mass spectrometry, imaging techniques, and vibrational spectroscopies. Some emerging experimental approaches for the study of LDs are represented by correlative light-electron microscopy and by correlative Raman imaging-scanning electron microscopy (SEM). Based on these scientific approaches, we developed a novel methodology (CREL) by combining Raman micro-spectroscopy, confocal fluorescence microscopy, and SEM coupled with an energy-dispersive X-ray spectroscopy module. This procedure correlated cellular morphology, chemical properties, and spatial distribution from the same region of interest, and in this work, we presented the application of CREL for the analysis of LDs within patient-derived melanoma CSCs (MCSCs).
Collapse
Affiliation(s)
- Francesca Pagliari
- Division of Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Biological and Environmental Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Elisa Sogne
- Biological and Environmental Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- PoliFAB, Polytechnic of Milan, Via Giuseppe Colombo, 81, 20133 Milan, Italy
| | - Davide Panella
- Nanotechnology Research Center, Department of Experimental and Clinical Medicine, University of Magna Graecia, 88100 Catanzaro, Italy
| | - Gerardo Perozziello
- Nanotechnology Research Center, Department of Experimental and Clinical Medicine, University of Magna Graecia, 88100 Catanzaro, Italy
| | - Carlo Liberale
- Biological and Environmental Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Gobind Das
- Biological and Environmental Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Department of Physics, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Simone Di Franco
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Joao Seco
- Division of Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, 69120 Heidelberg, Germany
| | - Andrea Falqui
- Department of Physics “Aldo Pontremoli”, University of Milan, Via Celoria 16, 20133 Milan, Italy
| | - Santo Gratteri
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
- Department of Medical and Surgical Science, University Magna Graecia, 88100 Catanzaro, Italy
| | - Arturo Pujia
- Department of Medical and Surgical Science, University Magna Graecia, 88100 Catanzaro, Italy
| | - Enzo Di Fabrizio
- Department of Applied Science and Technology, Polytechnic of Turin, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Patrizio Candeloro
- Nanotechnology Research Center, Department of Experimental and Clinical Medicine, University of Magna Graecia, 88100 Catanzaro, Italy
| | - Luca Tirinato
- Division of Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Biological and Environmental Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Department of Medical and Surgical Science, University Magna Graecia, 88100 Catanzaro, Italy
| |
Collapse
|
14
|
Hong X, Hu Y, Yuan Z, Fang Z, Zhang X, Yuan Y, Guo C. Oxidatively Damaged Nucleic Acid: Linking Diabetes and Cancer. Antioxid Redox Signal 2022; 37:1153-1167. [PMID: 35946074 DOI: 10.1089/ars.2022.0096] [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] [Indexed: 11/13/2022]
Abstract
Significance: Our current knowledge of the mechanism between diabetes and cancer is limited. Oxidatively damaged nucleic acid is considered a critical factor to explore the connections between these two diseases. Recent Advances: The link between diabetes mellitus and cancer has attracted increasing attention in recent years. Emerging evidence supports that oxidatively damaged nucleic acid caused by an imbalance between reactive oxygen species generation and elimination is a bridge connecting diabetes and cancer. 8-Oxo-7,8-dihydro-2'-deoxyguanosine and 8-oxo-7,8-dihydroguanosine assume important roles as biomarkers in assessing the relationship between oxidatively damaged nucleic acid and cancer. Critical Issues: The consequences of diabetes are extensive and may lead to the occurrence of cancer by influencing a combination of factors. At present, there is no direct evidence that diabetes causes cancer by affecting a single factor. Furthermore, the difficulty in controlling variables and differences in detection methods lead to poor reliability and repeatability of results, and there are no clear cutoff values for biomarkers to indicate cancer risk. Future Directions: A better understanding of connections as well as mechanisms between diabetes and cancer is still needed. Both diabetes and cancer are currently intractable diseases. Further exploration of the specific mechanism of oxidatively damaged nucleic acid in the connection between diabetes and cancer is urgently needed. In the future, it is necessary to further take oxidatively damaged nucleic acid as an entry point to provide new ideas for the diagnosis and treatment of diabetes and cancer. Experimental drugs targeting the repair process of oxidatively generated damage require an extensive preclinical evaluation and could ultimately provide new treatment strategies for these diseases. Antioxid. Redox Signal. 37, 1153-1167.
Collapse
Affiliation(s)
- Xiujuan Hong
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqiu Hu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijun Yuan
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihao Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxiao Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Cheng Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Wen GM, Xu XY, Xia P. Metabolism in Cancer Stem Cells: Targets for Clinical Treatment. Cells 2022; 11:3790. [PMID: 36497050 PMCID: PMC9736883 DOI: 10.3390/cells11233790] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer stem cells (CSCs) have high tumorigenicity, high metastasis and high resistance to treatment. They are the key factors for the growth, metastasis and drug resistance of malignant tumors, and are also the important reason for the occurrence and recurrence of tumors. Metabolic reprogramming refers to the metabolic changes that occur when tumor cells provide sufficient energy and nutrients for themselves. Metabolic reprogramming plays an important role in regulating the growth and activity of cancer cells and cancer stem cells. In addition, the immune cells or stromal cells in the tumor microenvironment (TME) will change due to the metabolic reprogramming of cancer cells. Summarizing the characteristics and molecular mechanisms of metabolic reprogramming of cancer stem cells will provide new ideas for the comprehensive treatment of malignant tumors. In this review, we summarized the changes of the main metabolic pathways in cancer cells and cancer stem cells.
Collapse
Affiliation(s)
- Gui-Min Wen
- Department of Basic Nursing, College of Nursing, Jinzhou Medical University, Jinzhou 121001, China
| | - Xiao-Yan Xu
- College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Pu Xia
- Biological Anthropology Institute, College of Basic Medical Science, Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|
16
|
Zhou Y, Li X, Guan A, Zhou H, Zhu Y, Wang R, Li R. EPHX2 Inhibits Colon Cancer Progression by Promoting Fatty Acid Degradation. Front Oncol 2022; 12:870721. [PMID: 35433439 PMCID: PMC9005964 DOI: 10.3389/fonc.2022.870721] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor cells use metabolic reprogramming to keep up with the need for bioenergy, biosynthesis, and oxidation balance needed for rapid tumor division. This phenomenon is considered a marker of tumors, including colon cancer (CRC). As an important pathway of cellular energy metabolism, fatty acid metabolism plays an important role in cellular energy supply and oxidation balance, but presently, our understanding of the exact role of fatty acid metabolism in CRC is limited. Currently, no lipid metabolism therapy is available for the treatment of CRC. The establishment of a lipidmetabolism model regulated by oncogenes/tumor suppressor genes and associated with the clinical characteristics of CRC is necessary to further understand the mechanism of fatty acid metabolism in CRC. In this study, through multi-data combined with bioinformatic analysis and basic experiments, we introduced a tumor suppressor gene, EPHX2, which is rarely reported in CRC, and confirmed that its inhibitory effect on CRC is related to fatty acid degradation.
Collapse
Affiliation(s)
- Yiran Zhou
- Department of General Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Yan'an Hospital of Kunming City, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Xiao Li
- Department of General Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Yan'an Hospital of Kunming City, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Aoran Guan
- Department of General Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Yan'an Hospital of Kunming City, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Haodong Zhou
- Department of General Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Yan'an Hospital of Kunming City, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Yankun Zhu
- Department of General Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Yan'an Hospital of Kunming City, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Ruotian Wang
- Department of General Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Yan'an Hospital of Kunming City, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Ruhong Li
- Department of General Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Yan'an Hospital of Kunming City, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| |
Collapse
|
17
|
Loughran R, Emerling BM. Mechanistic roles of mutant p53 governing lipid metabolism. Adv Biol Regul 2022; 83:100839. [PMID: 34840111 PMCID: PMC8858851 DOI: 10.1016/j.jbior.2021.100839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 01/03/2023]
Abstract
Metabolic reprogramming of cancer cells by various acquired mutations provides support for rapid proliferation and growth in the tumor microenvironment. Mutations in the TP53 gene are the most common mutation found across all human cancers. Commonly referred to as "the guardian of the genome", p53 has a well-established role as a tumor suppressor by mediating checkpoint integrity and protecting cells from DNA damage. To date, the many functional roles of p53 extending beyond its classical function and exerting control over metabolic processes continues to confound the field. Recently, emerging roles for p53 in mediating lipid metabolism have come to light with intriguing metabolic roles in regulating cholesterol homeostasis and lipid droplet formation. Herein, we will seek to unify the mechanisms by which absence of functional p53, as well as stable mutant forms of p53, exert control over these lipid metabolism programs. Of equal importance, synthetic lethal phenotypes in the context of mutant p53 and aberrant lipid homeostasis offer new possible targets in the therapeutic landscape. This review aims to characterize the mechanisms by which p53 exerts control over these pathways and examine how precision medicine may benefit from tumor subtyping of p53 mutations.
Collapse
Affiliation(s)
- Ryan Loughran
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys, La Jolla, CA 92037, USA
| | - Brooke M. Emerling
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys, La Jolla, CA 92037, USA,Correspondence:
| |
Collapse
|
18
|
Dadgar T, Ebrahimi N, Gholipour AR, Akbari M, Khani L, Ahmadi A, Hamblin MR. Targeting the metabolism of cancer stem cells by energy disruptor molecules. Crit Rev Oncol Hematol 2021; 169:103545. [PMID: 34838705 DOI: 10.1016/j.critrevonc.2021.103545] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/17/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) have been identified in various tumor types. CSCs are believed to contribute to tumor metastasis and resistance to conventional therapy. So targeting these cells could be an effective strategy to eliminate tumors and a promising new type of cancer treatment. Alterations in metabolism play an essential role in CSC biology and their resistance to treatment. The metabolic properties pathways in CSCs are different from normal cells, and to some extent, are different from regular tumor cells. Interestingly, CSCs can use other nutrients for their metabolism and growth. The different metabolism causes increased sensitivity of CSCs to agents that disrupt cellular homeostasis. Compounds that interfere with the central metabolic pathways are known as energy disruptors and can reduce CSC survival. This review highlights the differences between regular cancer cells and CSC metabolism and discusses the action mechanisms of energy disruptors at the cellular and molecular levels.
Collapse
Affiliation(s)
- Tahere Dadgar
- Department of Biology, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Nasim Ebrahimi
- Division of Genetics, Department of Cell and Molecular & Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Gholipour
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Maryam Akbari
- Department of Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Khani
- Department of Immunology, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Amirhossein Ahmadi
- Department of Biological Science and Technology, Faculty of Nano and Bio Science and Technology, Persian Gulf University, Bushehr, 75169, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| |
Collapse
|
19
|
Castelli S, De Falco P, Ciccarone F, Desideri E, Ciriolo MR. Lipid Catabolism and ROS in Cancer: A Bidirectional Liaison. Cancers (Basel) 2021; 13:cancers13215484. [PMID: 34771647 PMCID: PMC8583096 DOI: 10.3390/cancers13215484] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022] Open
Abstract
Although cancer cell metabolism was mainly considered to rely on glycolysis, with the concomitant impairment of mitochondrial metabolism, it has recently been demonstrated that several tumor types are sustained by oxidative phosphorylation (OXPHOS). In this context, endogenous fatty acids (FAs) deriving from lipolysis or lipophagy are oxidised into the mitochondrion, and are used as a source of energy through OXPHOS. Because the electron transport chain is the main source of ROS, cancer cells relying on fatty acid oxidation (FAO) need to be equipped with antioxidant systems that maintain the ROS levels under the death threshold. In those conditions, ROS can act as second messengers, favouring proliferation and survival. Herein, we highlight the different responses that tumor cells adopt when lipid catabolism is augmented, taking into account the different ROS fates. Many papers have demonstrated that the pro- or anti-tumoral roles of endogenous FA usage are hugely dependent on the tumor type, and on the capacity of cancer cells to maintain redox homeostasis. In light of this, clinical studies have taken advantage of the boosting of lipid catabolism to increase the efficacy of tumor therapy, whereas, in other contexts, antioxidant compounds are useful to reduce the pro-survival effects of ROS deriving from FAO.
Collapse
Affiliation(s)
- Serena Castelli
- Department of Biology, University of Rome “Tor Vergata”, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (S.C.); (P.D.F.); (E.D.)
| | - Pamela De Falco
- Department of Biology, University of Rome “Tor Vergata”, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (S.C.); (P.D.F.); (E.D.)
| | - Fabio Ciccarone
- IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Enrico Desideri
- Department of Biology, University of Rome “Tor Vergata”, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (S.C.); (P.D.F.); (E.D.)
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome “Tor Vergata”, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (S.C.); (P.D.F.); (E.D.)
- IRCCS San Raffaele Pisana, Via Della Pisana 235, 00163 Rome, Italy
- Correspondence:
| |
Collapse
|
20
|
Tan Y, Yu K, Liang L, Liu Y, Song F, Ge Q, Fang X, Yu T, Huang Z, Jiang L, Wang P. Sodium-Glucose Co-Transporter 2 Inhibition With Empagliflozin Improves Cardiac Function After Cardiac Arrest in Rats by Enhancing Mitochondrial Energy Metabolism. Front Pharmacol 2021; 12:758080. [PMID: 34712142 PMCID: PMC8546214 DOI: 10.3389/fphar.2021.758080] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022] Open
Abstract
Empagliflozin is a newly developed antidiabetic drug to reduce hyperglycaemia by highly selective inhibition of sodium–glucose co-transporter 2. Hyperglycaemia is commonly seen in patients after cardiac arrest (CA) and is associated with worse outcomes. In this study, we examined the effects of empagliflozin on cardiac function in rats with myocardial dysfunction after CA. Non-diabetic male Sprague–Dawley rats underwent ventricular fibrillation to induce CA, or sham surgery. Rats received 10 mg/kg of empagliflozin or vehicle at 10 min after return of spontaneous circulation by intraperitoneal injection. Cardiac function was assessed by echocardiography, histological analysis, molecular markers of myocardial injury, oxidative stress, mitochondrial ultrastructural integrity and metabolism. We found that empagliflozin did not influence heart rate and blood pressure, but left ventricular function and survival time were significantly higher in the empagliflozin treated group compared to the group treated with vehicle. Empagliflozin also reduced myocardial fibrosis, serum cardiac troponin I levels and myocardial oxidative stress after CA. Moreover, empagliflozin maintained the structural integrity of myocardial mitochondria and increased mitochondrial activity after CA. In addition, empagliflozin increased circulating and myocardial ketone levels as well as heart β-hydroxy butyrate dehydrogenase 1 protein expression. Together, these metabolic changes were associated with an increase in cardiac energy metabolism. Therefore, empagliflozin favorably affected cardiac function in non-diabetic rats with acute myocardial dysfunction after CA, associated with reducing glucose levels and increasing ketone body oxidized metabolism. Our data suggest that empagliflozin might benefit patients with myocardial dysfunction after CA.
Collapse
Affiliation(s)
- Yunke Tan
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Kai Yu
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Lian Liang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Yuanshan Liu
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Fengqing Song
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Qiulin Ge
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Xiangshao Fang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Tao Yu
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Zitong Huang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Longyuan Jiang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Peng Wang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
21
|
Xiao J, Tian M, Su L, Bao Y, Niu L, Zhang X. Detection of the effect of polydopamine (PDA)-coated polydimethylsiloxane (PDMS) substrates on the release of H 2O 2 from a single HeLa cell. Analyst 2021; 146:6445-6449. [PMID: 34585688 DOI: 10.1039/d1an01506k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Endogenous H2O2 generated by a single HeLa cell that was adhered on the PDA-coated PDMS substrates under 25 mM glucose culture conditions was detected using a home-built photoelectric dual detection platform. With PMA as the stimulus, the cell released a small amount of H2O2 and its mitochondrial membrane potential (MMP) decrease was smaller, compared with that on the PDMS substrates.
Collapse
Affiliation(s)
- Jingyu Xiao
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Meng Tian
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Lei Su
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518037, P.R. China, P. R. China.
| | - Yu Bao
- Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China.
| | - Li Niu
- Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China.
| | - Xueji Zhang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518037, P.R. China, P. R. China.
| |
Collapse
|
22
|
Royo-García A, Courtois S, Parejo-Alonso B, Espiau-Romera P, Sancho P. Lipid droplets as metabolic determinants for stemness and chemoresistance in cancer. World J Stem Cells 2021; 13:1307-1317. [PMID: 34630864 PMCID: PMC8474722 DOI: 10.4252/wjsc.v13.i9.1307] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/13/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Previously regarded as simple fat storage particles, new evidence suggests that lipid droplets (LDs) are dynamic and functional organelles involved in key cellular processes such as membrane biosynthesis, lipid metabolism, cell signalling and inflammation. Indeed, an increased LD content is one of the most apparent features resulting from lipid metabolism reprogramming necessary to support the basic functions of cancer cells. LDs have been associated to different cellular processes involved in cancer progression and aggressiveness, such as tumorigenicity, invasion and metastasis, as well as chemoresistance. Interestingly, all of these processes are controlled by a subpopulation of highly aggressive tumoral cells named cancer stem cells (CSCs), suggesting that LDs may be fundamental elements for stemness in cancer. Considering the key role of CSCs on chemoresistance and disease relapse, main factors of therapy failure, the design of novel therapeutic approaches targeting these cells may be the only chance for long-term survival in cancer patients. In this sense, their biology and functional properties render LDs excellent candidates for target discovery and design of combined therapeutic strategies. In this review, we summarise the current knowledge identifying LDs and CSCs as main contributors to cancer aggressiveness, metastasis and chemoresistance.
Collapse
Affiliation(s)
- Alba Royo-García
- Hospital Universitario Miguel Servet, IIS Aragón, Zaragoza 50009, Spain
| | - Sarah Courtois
- Hospital Universitario Miguel Servet, IIS Aragón, Zaragoza 50009, Spain
| | | | | | - Patricia Sancho
- Hospital Universitario Miguel Servet, IIS Aragón, Zaragoza 50009, Spain
| |
Collapse
|
23
|
Nisticò C, Pagliari F, Chiarella E, Fernandes Guerreiro J, Marafioti MG, Aversa I, Genard G, Hanley R, Garcia-Calderón D, Bond HM, Mesuraca M, Tirinato L, Spadea MF, Seco JC. Lipid Droplet Biosynthesis Impairment through DGAT2 Inhibition Sensitizes MCF7 Breast Cancer Cells to Radiation. Int J Mol Sci 2021; 22:10102. [PMID: 34576263 PMCID: PMC8466244 DOI: 10.3390/ijms221810102] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most frequent cancer in women worldwide and late diagnosis often adversely affects the prognosis of the disease. Radiotherapy is commonly used to treat breast cancer, reducing the risk of recurrence after surgery. However, the eradication of radioresistant cancer cells, including cancer stem cells, remains the main challenge of radiotherapy. Recently, lipid droplets (LDs) have been proposed as functional markers of cancer stem cells, also being involved in increased cell tumorigenicity. LD biogenesis is a multistep process requiring various enzymes, including Diacylglycerol acyltransferase 2 (DGAT2). In this context, we evaluated the effect of PF-06424439, a selective DGAT2 inhibitor, on MCF7 breast cancer cells exposed to X-rays. Our results demonstrated that 72 h of PF-06424439 treatment reduced LD content and inhibited cell migration, without affecting cell proliferation. Interestingly, PF-06424439 pre-treatment followed by radiation was able to enhance radiosensitivity of MCF7 cells. In addition, the combined treatment negatively interfered with lipid metabolism-related genes, as well as with EMT gene expression, and modulated the expression of typical markers associated with the CSC-like phenotype. These findings suggest that PF-06424439 pre-treatment coupled to X-ray exposure might potentiate breast cancer cell radiosensitivity and potentially improve the radiotherapy effectiveness.
Collapse
Affiliation(s)
- Clelia Nisticò
- Department of Clinical and Experimental Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.N.); (E.C.); (M.G.M.); (I.A.); (H.M.B.); (M.M.)
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
| | - Francesca Pagliari
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
| | - Emanuela Chiarella
- Department of Clinical and Experimental Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.N.); (E.C.); (M.G.M.); (I.A.); (H.M.B.); (M.M.)
| | - Joana Fernandes Guerreiro
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 1397), 2695-066 Bobadela LRS, Portugal
| | - Maria Grazia Marafioti
- Department of Clinical and Experimental Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.N.); (E.C.); (M.G.M.); (I.A.); (H.M.B.); (M.M.)
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
| | - Ilenia Aversa
- Department of Clinical and Experimental Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.N.); (E.C.); (M.G.M.); (I.A.); (H.M.B.); (M.M.)
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
| | - Geraldine Genard
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
| | - Rachel Hanley
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
- Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| | - Daniel Garcia-Calderón
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
- Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| | - Heather Mandy Bond
- Department of Clinical and Experimental Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.N.); (E.C.); (M.G.M.); (I.A.); (H.M.B.); (M.M.)
| | - Maria Mesuraca
- Department of Clinical and Experimental Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.N.); (E.C.); (M.G.M.); (I.A.); (H.M.B.); (M.M.)
| | - Luca Tirinato
- Department of Clinical and Experimental Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.N.); (E.C.); (M.G.M.); (I.A.); (H.M.B.); (M.M.)
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
| | - Maria Francesca Spadea
- Department of Clinical and Experimental Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.N.); (E.C.); (M.G.M.); (I.A.); (H.M.B.); (M.M.)
| | - Joao Carlos Seco
- Division of BioMedical Physics in Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany; (F.P.); (J.F.G.); (G.G.); (R.H.); (D.G.-C.)
- Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Wang Y, Pan H, chen D, Guo D, Wang X. Targeting at cancer energy metabolism and lipid droplet formation as new treatment strategies for epigallocatechin-3-gallate (EGCG) in colorectal cancer cells. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
25
|
Broadfield LA, Pane AA, Talebi A, Swinnen JV, Fendt SM. Lipid metabolism in cancer: New perspectives and emerging mechanisms. Dev Cell 2021; 56:1363-1393. [PMID: 33945792 DOI: 10.1016/j.devcel.2021.04.013] [Citation(s) in RCA: 339] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/15/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
Tumors undergo metabolic transformations to sustain uncontrolled proliferation, avoid cell death, and seed in secondary organs. An increased focus on cancer lipid metabolism has unveiled a number of mechanisms that promote tumor growth and survival, many of which are independent of classical cellular bioenergetics. These mechanisms include modulation of ferroptotic-mediated cell death, support during tumor metastasis, and interactions with the cells of the tumor microenvironment. As such, targeting lipid metabolism for anti-cancer therapies is attractive, with recent work on small-molecule inhibitors identifying compounds to target lipid metabolism. Here, we discuss these topics and identify open questions.
Collapse
Affiliation(s)
- Lindsay A Broadfield
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Antonino Alejandro Pane
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ali Talebi
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute (LKI), KU Leuven, University of Leuven, Leuven, Belgium
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute (LKI), KU Leuven, University of Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
26
|
Neef SK, Janssen N, Winter S, Wallisch SK, Hofmann U, Dahlke MH, Schwab M, Mürdter TE, Haag M. Metabolic Drug Response Phenotyping in Colorectal Cancer Organoids by LC-QTOF-MS. Metabolites 2020; 10:metabo10120494. [PMID: 33271860 PMCID: PMC7760698 DOI: 10.3390/metabo10120494] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
As metabolic rewiring is crucial for cancer cell proliferation, metabolic phenotyping of patient-derived organoids is desirable to identify drug-induced changes and trace metabolic vulnerabilities of tumor subtypes. We established a novel protocol for metabolomic and lipidomic profiling of colorectal cancer organoids by liquid chromatography quadrupole time-of-flight mass spectrometry (LC-QTOF-MS) facing the challenge of capturing metabolic information from a minimal sample amount (<500 cells/injection) in the presence of an extracellular matrix (ECM). The best procedure of the tested protocols included ultrasonic metabolite extraction with acetonitrile/methanol/water (2:2:1, v/v/v) without ECM removal. To eliminate ECM-derived background signals, we implemented a data filtering procedure based on the p-value and fold change cut-offs, which retained features with signal intensities >120% compared to matrix-derived signals present in blank samples. As a proof-of-concept, the method was applied to examine the early metabolic response of colorectal cancer organoids to 5-fluorouracil treatment. Statistical analysis revealed dose-dependent changes in the metabolic profiles of treated organoids including elevated levels of 2′-deoxyuridine, 2′-O-methylcytidine, inosine and 1-methyladenosine and depletion of 2′-deoxyadenosine and specific phospholipids. In accordance with the mechanism of action of 5-fluorouracil, changed metabolites are mainly involved in purine and pyrimidine metabolism. The novel protocol provides a first basis for the assessment of metabolic drug response phenotypes in 3D organoid models.
Collapse
Affiliation(s)
- Sylvia K. Neef
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
| | - Nicole Janssen
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
| | - Svenja K. Wallisch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
| | - Marc H. Dahlke
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
- Department of Surgery, Robert-Bosch Hospital, 70376 Stuttgart, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
- Departments of Clinical Pharmacology, and of Pharmacy and Biochemistry, University of Tuebingen, 72074 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180), Image-Guided and Functionally Instructed Tumor Therapies, University of Tuebingen, 72074 Tuebingen, Germany
| | - Thomas E. Mürdter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, 70376 Tuebingen, Germany; (S.K.N.); (N.J.); (S.W.); (S.K.W.); (U.H.); (M.H.D.); (M.S.); (T.E.M.)
- Correspondence: ; Tel.: +49-711-8101-5429
| |
Collapse
|