1
|
Freitas-Cortez MA, Masrorpour F, Jiang H, Mahmud I, Lu Y, Huang A, Duong LK, Wang Q, Voss TA, Kettlun Leyton CS, Wei B, Chan WK, Lin K, Zhang J, Tsouko E, Ganjoo S, Barsoumian HB, Riad TS, Hu Y, Leuschner C, Puebla-Osorio N, Wang J, Hu J, Davies MA, Puduvalli VK, Billon C, Burris TP, Lorenzi PL, Gan B, Welsh JW. Cancer cells avoid ferroptosis induced by immune cells via fatty acid binding proteins. Mol Cancer 2025; 24:40. [PMID: 39901247 PMCID: PMC11789333 DOI: 10.1186/s12943-024-02198-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/11/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Cancer creates an immunosuppressive environment that hampers immune responses, allowing tumors to grow and resist therapy. One way the immune system fights back is by inducing ferroptosis, a type of cell death, in tumor cells through CD8 + T cells. This involves lipid peroxidation and enzymes like lysophosphatidylcholine acyltransferase 3 (Lpcat3), which makes cells more prone to ferroptosis. However, the mechanisms by which cancer cells avoid immunotherapy-mediated ferroptosis are unclear. Our study reveals how cancer cells evade ferroptosis and anti-tumor immunity through the upregulation of fatty acid-binding protein 7 (Fabp7). METHODS To explore how cancer cells resist immune cell-mediated ferroptosis, we used a comprehensive range of techniques. We worked with cell lines including PD1-sensitive, PD1-resistant, B16F10, and QPP7 glioblastoma cells, and conducted in vivo studies in syngeneic 129 Sv/Ev, C57BL/6, and conditional knockout mice with Rora deletion specifically in CD8+ T cells, Cd8 cre;Rorafl mice. Methods included mass spectrometry-based lipidomics, targeted lipidomics, Oil Red O staining, Seahorse analysis, quantitative PCR, immunohistochemistry, PPARγ transcription factor assays, ChIP-seq, untargeted lipidomic analysis, ROS assay, ex vivo co-culture of CD8+ T cells with cancer cells, ATAC-seq, RNA-seq, Western blotting, co-immunoprecipitation assay, flow cytometry and Imaging Mass Cytometry. RESULTS PD1-resistant tumors upregulate Fabp7, driving protective metabolic changes that shield cells from ferroptosis and evade anti-tumor immunity. Fabp7 decreases the transcription of ferroptosis-inducing genes like Lpcat3 and increases the transcription of ferroptosis-protective genes such as Bmal1 through epigenetic reprogramming. Lipidomic profiling revealed that Fabp7 increases triglycerides and monounsaturated fatty acids (MUFAs), which impede lipid peroxidation and ROS generation. Fabp7 also improves mitochondrial function and fatty acid oxidation (FAO), enhancing cancer cell survival. Furthermore, cancer cells increase Fabp7 expression in CD8+ T cells, disrupting circadian clock gene expression and triggering apoptosis through p53 stabilization. Clinical trial data revealed that higher FABP7 expression correlates with poorer overall survival and progression-free survival in patients undergoing immunotherapy. CONCLUSIONS Our study uncovers a novel mechanism by which cancer cells evade immune-mediated ferroptosis through Fabp7 upregulation. This protein reprograms lipid metabolism and disrupts circadian regulation in immune cells, promoting tumor survival and resistance to immunotherapy. Targeting Fabp7 could enhance immunotherapy effectiveness by re-sensitizing resistant tumors to ferroptosis.
Collapse
Affiliation(s)
- Maria Angelica Freitas-Cortez
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA.
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Children's Research Institute at UT Southwestern, Pediatrics 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Hong Jiang
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Iqbal Mahmud
- Department of Bioinformatics and Computational Biology, Metabolomics Core Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ailing Huang
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Lisa K Duong
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Qi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Tiffany A Voss
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Claudia S Kettlun Leyton
- Department of Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bo Wei
- Department of Bioinformatics and Computational Biology, Metabolomics Core Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wai-Kin Chan
- Department of Bioinformatics and Computational Biology, Metabolomics Core Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Efrosini Tsouko
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shonik Ganjoo
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Thomas S Riad
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Yun Hu
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Carola Leuschner
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vinay K Puduvalli
- Department of Neuro-Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Cyrielle Billon
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy at University of Health and Sciences, St. Louis, MO, 63110, USA
| | - Thomas P Burris
- University of Florida Genetics Institute, Gainesville, FL, 32610, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, Metabolomics Core Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - James W Welsh
- Department of Radiation Oncology, UT MD Anderson Cancer Center, 6565 MD Anderson Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Warren WG, Osborn M, Duffy P, Yates A, O'Sullivan SE. Potential safety implications of fatty acid-binding protein inhibition. Toxicol Appl Pharmacol 2024; 491:117079. [PMID: 39218163 DOI: 10.1016/j.taap.2024.117079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Fatty acid-binding proteins (FABPs) are small intracellular proteins that regulate fatty acid metabolism, transport, and signalling. There are ten known human isoforms, many of which are upregulated and involved in clinical pathologies. As such, FABP inhibition may be beneficial in disease states such as cancer, and those involving the cardiovascular system, metabolism, immunity, and cognition. Recently, a potent, selective FABP5 inhibitor (ART26.12), with 90-fold selectivity to FABP3 and 20-fold selectivity to FABP7, was found to be remarkably benign, with a no-observed-adverse-effect level of 1000 mg/kg in rats and dogs, showing no genotoxicity, cardiovascular, central, or respiratory toxicity. To understand the potential implication of FABP inhibition more fully, this review systematically assessed literature investigating genetic knockout, knockdown, and pharmacological inhibition of FABP3, FABP4, FABP5, or FABP7. Analysis of the literature revealed that animals bred not to express FABPs showed the most biological effects, suggesting key roles of these proteins during development. FABP ablation sometimes exacerbated symptoms of disease models, particularly those linked to metabolism, inflammatory and immune responses, cardiac contractility, neurogenesis, and cognition. However, FABP inhibition (genetic silencing or pharmacological) had a positive effect in many more disease conditions. Several polymorphisms of each FABP gene have also been linked to pathological conditions, but it was unclear how several polymorphisms affected protein function. Overall, analysis of the literature to date suggests that pharmacological inhibition of FABPs in adults is of low risk.
Collapse
Affiliation(s)
- William G Warren
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom.
| | - Myles Osborn
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Paul Duffy
- Apconix Ltd., Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Andrew Yates
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | | |
Collapse
|
3
|
George Warren W, Osborn M, Yates A, O'Sullivan SE. The emerging role of fatty acid binding protein 7 (FABP7) in cancers. Drug Discov Today 2024; 29:103980. [PMID: 38614160 DOI: 10.1016/j.drudis.2024.103980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/27/2024] [Accepted: 04/05/2024] [Indexed: 04/15/2024]
Abstract
Fatty acid binding protein 7 (FABP7) is an intracellular protein involved in the uptake, transportation, metabolism, and storage of fatty acids (FAs). FABP7 is upregulated up to 20-fold in multiple cancers, usually correlated with poor prognosis. FABP7 silencing or pharmacological inhibition suggest FABP7 promotes cell growth, migration, invasion, colony and spheroid formation/increased size, lipid uptake, and lipid droplet formation. Xenograft studies show that suppression of FABP7 inhibits tumour formation and tumour growth, and improves host survival. The molecular mechanisms involve promotion of FA uptake, lipid droplets, signalling [focal adhesion kinase (FAK), proto-oncogene tyrosine-protein kinase Src (Src), mitogen-activated protein kinase kinase/p-extracellular signal-regulated kinase (MEK/ERK), and Wnt/β-catenin], hypoxia-inducible factor 1-alpha (Hif1α), vascular endothelial growth factor A/prolyl 4-hydroxylase subunit alpha-1 (VEGFA/P4HA1), snail family zinc finger 1 (Snail1), and twist-related protein 1 (Twist1). The oncogenic capacity of FABP7 makes it a promising pharmacological target for future cancer treatments.
Collapse
Affiliation(s)
| | - Myles Osborn
- Artelo Biosciences Limited, Alderley Park, Cheshire, UK
| | - Andrew Yates
- Artelo Biosciences Limited, Alderley Park, Cheshire, UK
| | | |
Collapse
|
4
|
Transcriptomics and Lipid Metabolomics Analysis of Subcutaneous, Visceral, and Abdominal Adipose Tissues of Beef Cattle. Genes (Basel) 2022; 14:genes14010037. [PMID: 36672778 PMCID: PMC9858949 DOI: 10.3390/genes14010037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Fat deposition traits are influenced by genetics and environment, which affect meat quality, growth rate, and energy metabolism of domestic animals. However, at present, the molecular mechanism of fat deposition is not entirely understood in beef cattle. Therefore, the current study conducted transcriptomics and lipid metabolomics analysis of subcutaneous, visceral, and abdominal adipose tissue (SAT, VAT, and AAT) of Huaxi cattle to investigate the differences among these adipose tissues and systematically explore how candidate genes interact with metabolites to affect fat deposition. These results demonstrated that compared with SAT, the gene expression patterns and metabolite contents of VAT and AAT were more consistent. Particularly, SCD expression, monounsaturated fatty acid (MUFA) and triglyceride (TG) content were higher in SAT, whereas PCK1 expression and the contents of saturated fatty acid (SFA), diacylglycerol (DG), and lysoglycerophosphocholine (LPC) were higher in VAT. Notably, in contrast to PCK1, 10 candidates including SCD, ELOVL6, ACACA, and FABP7 were identified to affect fat deposition through positively regulating MUFA and TG, and negatively regulating SFA, DG, and LPC. These findings uncovered novel gene resources and offered a theoretical basis for future investigation of fat deposition in beef cattle.
Collapse
|
5
|
Tao Q, Zhang D, Zhang Q, Liu C, Ye S, Feng Y, Liu R. Mitochondrial targeted ROS Scavenger based on nitroxide for Treatment and MRI imaging of Acute Kidney Injury. Free Radic Res 2022; 56:303-315. [PMID: 35746859 DOI: 10.1080/10715762.2022.2093724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Overproduction of reactive oxygen species (ROS) during oxidative stress is a hallmark of acute kidney injure (AKI), which induced the damage to the renal cells and mitochondrial injury. PURPOSE In this contribution, we prepared mitochondrial targeted nitroxide, which linked 3-carboxy-2,2,5,5-tetramethylpyrrolidine 1-oxyl (carboxy-PROXYL) with (2-aminoethyl)triphenylphosphonium bromide (TPP), named TPP-PROXYL to eliminate the ROS in situ and image the oxidative stress reaction by MRI. METHODS 2,7-Dichlorodihydrofluorescein diacetate (DCFH-DA) staining, mitochondrial membrane potential assay (JC-1) staining and transmission electron microscope (TEM) experiments were processed to verify that TPP-PROXYL could target mitochondria, scavenge the ROS, and prevent damage to mitochondria in live cells. Contrast enhanced MRI also been used to monitor these redox reaction in AKI model. RESULTS TPP-PROXYL demonstrated excellent ROS T1-weighted magnetic resonance imaging (MRI) enhancement in vitro and in vivo, with r1 value about 0.190 mM-1·s-1. In vivo AKI treatment experiments proved that TPP-PROXYL could improve the survival rate of mice and inhibit kidney damage. Moreover, the great ROS scavenging capability and the renal damage reduction during AKI treatment of TPP-PROXYL was verified via MR imaging technology. CONCLUSION Collectively, this research provides TPP-PROXYL would serve as a powerful platform to realize ROS scavenging, treatment and MR imaging of AKI.
Collapse
Affiliation(s)
- Quan Tao
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education & Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Di Zhang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education & Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Qianqian Zhang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education & Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Chuang Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education & Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Sheng Ye
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education & Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education & Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Ruiyuan Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education & Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|