1
|
Gupta S, Afzal M, Agrawal N, Almalki WH, Rana M, Gangola S, Chinni SV, Kumar K B, Ali H, Singh SK, Jha SK, Gupta G. Harnessing the FOXO-SIRT1 axis: insights into cellular stress, metabolism, and aging. Biogerontology 2025; 26:65. [PMID: 40011269 DOI: 10.1007/s10522-025-10207-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
Aging and metabolic disorders share intricate molecular pathways, with the Forkhead box O (FOXO)- Sirtuin 1 (SIRT1) axis emerging as a pivotal regulator of cellular stress adaptation, metabolic homeostasis, and longevity. This axis integrates nutrient signaling with oxidative stress defence, modulating glucose and lipid metabolism, mitochondrial function, and autophagy to maintain cellular stability. FOXO transcription factors, regulated by SIRT1 deacetylation, enhance antioxidant defence mechanisms, activating genes such as superoxide dismutase (SOD) and catalase, thereby counteracting oxidative stress and metabolic dysregulation. Recent evidence highlights the dynamic role of reactive oxygen species (ROS) as secondary messengers in redox signaling, influencing FOXO-SIRT1 activity in metabolic adaptation. Additionally, key redox-sensitive regulators such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) interact with this pathway, orchestrating mitochondrial biogenesis and adaptive stress responses. Pharmacological interventions, including alpha-lipoic acid (ALA), resveratrol, curcumin and NAD+ precursors, exhibit therapeutic potential by enhancing insulin sensitivity, reducing oxidative burden, and restoring metabolic balance. This review synthesizes current advancements in FOXO-SIRT1 regulation, its emerging role in redox homeostasis, and its therapeutic relevance, offering insights into future strategies for combating metabolic dysfunction and aging-related diseases.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Pharmacology, Chameli Devi Institute of Pharmacy, Khandwa Road, Village Umrikheda, Near Tollbooth, Indore, Madhya Pradesh, 452020, India
| | - Muhammad Afzal
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gangola
- Department of Microbiology, Graphic Era Deemed to be University, Dehradun, 248002, India
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, 42610, Jenjarom, Selangor, Malaysia
| | - Benod Kumar K
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, New Delhi, India
- Centre for Himalayan Studies, University of Delhi, Delhi, 110007, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
2
|
Rodríguez-Rodríguez R, Baena M, Zagmutt S, Paraiso WK, Reguera AC, Fadó R, Casals N. International Union of Basic and Clinical Pharmacology: Fundamental insights and clinical relevance regarding the carnitine palmitoyltransferase family of enzymes. Pharmacol Rev 2025; 77:100051. [PMID: 40106976 DOI: 10.1016/j.pharmr.2025.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
The carnitine palmitoyltransferases (CPTs) play a key role in controlling the oxidation of long-chain fatty acids and are potential therapeutic targets for diseases with a strong metabolic component, such as obesity, diabetes, and cancer. Four distinct proteins are CPT1A, CPT1B, CPT1C, and CPT2, differing in tissue expression and catalytic activity. CPT1s are finely regulated by malonyl-CoA, a metabolite whose intracellular levels reflect the cell's nutritional state. Although CPT1C does not exhibit significant catalytic activity, it is capable of modulating the functioning of other neuronal proteins. Structurally, all CPTs share a Y-shaped catalytic tunnel that allows the entry of 2 substrates and accommodation of the acyl group in a hydrophobic pocket. Several molecules targeting these enzymes have been described, some showing potential in normalizing blood glucose levels in diabetic patients, and others that, through a central mechanism, are anorexigenic and enhance energy expenditure. However, given the critical roles that CPTs play in certain tissues, such as the heart, liver, and brain, it is essential to fully understand the differences between the various isoforms. We analyze in detail the structure of these proteins, their cellular and physiological functions, and their potential as therapeutic targets in diseases such as obesity, diabetes, and cancer. We also describe drugs identified to date as having inhibitory or activating capabilities for these proteins. This knowledge will support the design of new drugs specific to each isoform, and the development of nanomedicines that can selectively target particular tissues or cells. SIGNIFICANCE STATEMENT: Carnitine palmitoyltransferase (CPT) proteins, as gatekeepers of fatty acid oxidation, have great potential as pharmacological targets to treat metabolic diseases like obesity, diabetes, and cancer. In recent years, significant progress has been made in understanding the 3-dimensional structure of CPTs and their pathophysiological functions. A deeper understanding of the differences between the various CPT family members will enable the design of selective drugs and therapeutic approaches with fewer side effects.
Collapse
Affiliation(s)
- Rosalía Rodríguez-Rodríguez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel Baena
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Sebastián Zagmutt
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - West Kristian Paraiso
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Ana Cristina Reguera
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Rut Fadó
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Núria Casals
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Bakhtiari M, Jordan SC, Mumme HL, Sharma R, Shanmugam M, Bhasin SS, Bhasin M. ARMH1 is a novel marker associated with poor pediatric AML outcomes that affect the fatty acid synthesis and cell cycle pathways. Front Oncol 2024; 14:1445173. [PMID: 39703843 PMCID: PMC11655347 DOI: 10.3389/fonc.2024.1445173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Despite remarkable progress in Pediatric Acute Myeloid Leukemia (pAML) treatments, the relapsed disease remains difficult to treat, making it pertinent to identify novel biomarkers of prognostic/therapeutic significance. Material and methods Bone marrow samples from 21 pAML patients were analyzed using single cell RNA sequencing, functional assays with ARMH1 knockdown and overexpression were performed in leukemia cell lines to evaluate impact on proliferation and migration, and chemotherapy sensitivity. Mitochondrial function was assessed via Seahorse assay, ARMH1 interacting proteins were studied using co-immunoprecipitation. Bulk RNA-seq was performed on ARMH1knockdown and over expressing cell lines to evaluate the pathways and networks impacted by ARMH1. Results Our data shows that ARMH1, a novel cancer-associated gene, is highly expressed in the malignant blast cells of multiple pediatric hematologic malignancies, including AML, T/B-ALL, and T/B-MPAL. Notably, ARMH1 expression is significantly elevated in blast cells of patients who relapsed or have a high-risk cytogenetic profile (MLL) compared to standard-risk (RUNX1, inv (16)). ARMH1 expression is also significantly correlated with the pediatric leukemia stem cell score of 6 genes (LSC6) associated with poor outcomes. Perturbation of ARMH1 (knockdown and overexpression) in leukemia cell lines significantly impacted cell proliferation and migration. The RNA-sequencing analysis on multiple ARMH1 knockdown and overexpressing cell lines established an association with mitochondrial fatty acid synthesis and cell cycle pathways.The investigation of the mitochondrial matrix shows that pharmacological inhibition of a key enzyme in fatty acid synthesis regulation, CPT1A, resulted in ARMH1 downregulation. ARMH1 knockdown also led to a significant reduction in CPT1A and ATP production as well as Oxygen Consumption Rate. Our data indicates that downregulating ARMH1 impacts cell proliferation by reducing key cell cycle regulators such as CDCA7 and EZH2. Further, we also established that ARMH1 is a key physical interactant of EZH2, associated with multiple cancers. Conclusion Our findings underscore further evaluation of ARMH1 as a potential candidate for targeted therapies and stratification of aggressive pAML to improve outcomes.
Collapse
Affiliation(s)
- Mojtaba Bakhtiari
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
| | - Sean C. Jordan
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
| | - Hope L. Mumme
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
| | - Richa Sharma
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Swati S. Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Manoj Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| |
Collapse
|
4
|
Feng T, Zhang H, Zhou Y, Zhu Y, Shi S, Li K, Lin P, Chen J. Roles of posttranslational modifications in lipid metabolism and cancer progression. Biomark Res 2024; 12:141. [PMID: 39551780 PMCID: PMC11571667 DOI: 10.1186/s40364-024-00681-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Lipid metabolism reprogramming has emerged as a hallmark of malignant tumors. Lipids represent a complex group of biomolecules that not only compose the essential components of biological membranes and act as an energy source, but also function as messengers to integrate various signaling pathways. In tumor cells, de novo lipogenesis plays a crucial role in acquiring lipids to meet the demands of rapid growth. Increasing evidence has suggested that dysregulated lipid metabolism serves as a driver of cancer progression. Posttranslational modifications (PTMs), which occurs in most eukaryotic proteins throughout their lifetimes, affect the activity, abundance, function, localization, and interactions of target proteins. PTMs of crucial molecules are potential intervention sites and are emerging as promising strategies for the cancer treatment. However, there is limited information available regarding the PTMs that occur in cancer lipid metabolism and the potential treatment strategies associated with these PTMs. Herein, we summarize current knowledge of the roles and regulatory mechanisms of PTMs in lipid metabolism. Understanding the roles of PTMs in lipid metabolism in cancer could provide valuable insights into tumorigenesis and progression. Moreover, targeting PTMs in cancer lipid metabolism might represent a promising novel therapeutic strategy.
Collapse
Affiliation(s)
- Tianyu Feng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - He Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - Yanjie Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - Yalan Zhu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - Shiya Shi
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - Kai Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China.
| | - Ping Lin
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China.
| | - Jie Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China.
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China.
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
5
|
Wang X, Yang C, Huang C, Wang W. Dysfunction of the carnitine cycle in tumor progression. Heliyon 2024; 10:e35961. [PMID: 39211923 PMCID: PMC11357771 DOI: 10.1016/j.heliyon.2024.e35961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
The carnitine cycle is responsible for the transport of cytoplasmic fatty acids to the mitochondria for subsequent β-oxidation to maintain intracellular energy homeostasis. Recent studies have identified abnormalities in the carnitine cycle in various types of tumors; these abnormalities include the altered expression levels of carnitine cycle-related metabolic enzymes and transport proteins. Dysfunction of the carnitine cycle has been shown to influence tumorigenesis and progression by altering intracellular oxidative and inflammatory status or regulating tumor metabolic flexibility. Many therapeutic strategies targeting the carnitine cycle are actively being explored to modify the dysfunction of the carnitine cycle in patients with malignant tumors; such approaches include carnitine cycle-related enzyme inhibitors and exogenous carnitine supplementation. Therefore, here, we review the studies of carnitine in tumors, aiming to scientifically illustrate the dysfunction of the carnitine cycle in tumor progression and provide new ideas for further research.
Collapse
Affiliation(s)
- Xiangjun Wang
- Department of Hepatobiliary and Pancreatic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chuanxin Yang
- Department of Hepatobiliary and Pancreatic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chao Huang
- Department of Cell Biology, Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Wei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
6
|
Li B, Mi J, Yuan Q. Fatty acid metabolism-related enzymes in colorectal cancer metastasis: from biological function to molecular mechanism. Cell Death Discov 2024; 10:350. [PMID: 39103344 DOI: 10.1038/s41420-024-02126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024] Open
Abstract
Colorectal cancer (CRC) is a highly aggressive and life-threatening malignancy that metastasizes in ~50% of patients, posing significant challenges to patient survival and treatment. Fatty acid (FA) metabolism regulates proliferation, immune escape, metastasis, angiogenesis, and drug resistance in CRC. FA metabolism consists of three pathways: de novo synthesis, uptake, and FA oxidation (FAO). FA metabolism-related enzymes promote CRC metastasis by regulating reactive oxygen species (ROS), matrix metalloproteinases (MMPs), angiogenesis and epithelial-mesenchymal transformation (EMT). Mechanistically, the PI3K/AKT/mTOR pathway, wnt/β-catenin pathway, and non-coding RNA signaling pathway are regulated by crosstalk of enzymes related to FA metabolism. Given the important role of FA metabolism in CRC metastasis, targeting FA metabolism-related enzymes and their signaling pathways is a potential strategy to treat CRC metastasis.
Collapse
Affiliation(s)
- Biao Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Jing Mi
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Qi Yuan
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China.
| |
Collapse
|
7
|
Wang D, Mahmud I, Thakur VS, Kiat Tan S, Isom DG, Lombard DB, Gonzalgo ML, Kryvenko ON, Lorenzi PL, Tcheuyap VT, Brugarolas J, Welford SM. GPR1 and CMKLR1 Control Lipid Metabolism to Support the Development of Clear Cell Renal Cell Carcinoma. Cancer Res 2024; 84:2141-2154. [PMID: 38640229 PMCID: PMC11290988 DOI: 10.1158/0008-5472.can-23-2926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 04/21/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC), the most common type of kidney cancer, is largely incurable in the metastatic setting. ccRCC is characterized by excessive lipid accumulation that protects cells from stress and promotes tumor growth, suggesting that the underlying regulators of lipid storage could represent potential therapeutic targets. Here, we evaluated the regulatory roles of GPR1 and CMKLR1, two G protein-coupled receptors of the protumorigenic adipokine chemerin that is involved in ccRCC lipid metabolism. Both genetic and pharmacologic suppression of either receptor suppressed lipid formation and induced multiple forms of cell death, including apoptosis, ferroptosis, and autophagy, thereby significantly impeding ccRCC growth in cell lines and patient-derived xenograft models. Comprehensive lipidomic and transcriptomic profiling of receptor competent and depleted cells revealed overlapping and unique signaling of the receptors granting control over triglyceride synthesis, ceramide production, and fatty acid saturation and class production. Mechanistically, both receptors enforced suppression of adipose triglyceride lipase, but each receptor also demonstrated distinct functions, such as the unique ability of CMKLR1 to control lipid uptake through regulation of sterol regulatory element-binding protein 1c and the CD36 scavenger receptor. Treating patient-derived xenograft models with the CMKLR1-targeting small molecule 2-(α-naphthoyl) ethyltrimethylammonium iodide (α-NETA) led to a dramatic reduction in tumor growth, lipid storage, and clear-cell morphology. Together, these findings provide mechanistic insights into lipid regulation in ccRCC and identify a targetable axis at the core of the histologic definition of this tumor that could be exploited therapeutically. Significance: Extracellular control of lipid accumulation via G protein receptor-mediated cell signaling is a metabolic vulnerability in clear cell renal cell carcinoma, which depends on lipid storage to avoid oxidative toxicity.
Collapse
Affiliation(s)
- Dazhi Wang
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Iqbal Mahmud
- Department of Bioinformatics & Computational Biology, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vijay S. Thakur
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Sze Kiat Tan
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Daniel G. Isom
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - David B. Lombard
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Department of Pathology & Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Bruce W. Carter VAMC, Miami FL 33125, USA
| | - Mark L. Gonzalgo
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Oleksandr N. Kryvenko
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Department of Pathology & Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Philip L. Lorenzi
- Department of Bioinformatics & Computational Biology, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vanina T Tcheuyap
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine/Hematology-Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Scott M. Welford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| |
Collapse
|
8
|
Zhang H, Sun Q, Dong H, Jin Z, Li M, Jin S, Zeng X, Fan J, Kong Y. Long-chain acyl-CoA synthetase-4 regulates endometrial decidualization through a fatty acid β-oxidation pathway rather than lipid droplet accumulation. Mol Metab 2024; 84:101953. [PMID: 38710444 PMCID: PMC11099325 DOI: 10.1016/j.molmet.2024.101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024] Open
Abstract
OBJECTIVE Lipid metabolism plays an important role in early pregnancy, but its effects on decidualization are poorly understood. Fatty acids (FAs) must be esterified by fatty acyl-CoA synthetases to form biologically active acyl-CoA in order to enter the anabolic and/or catabolic pathway. Long-chain acyl-CoA synthetase 4 (ACSL4) is associated with female reproduction. However, whether it is involved in decidualization is unknown. METHODS The expression of ACSL4 in human and mouse endometrium was detected by immunohistochemistry. ACSL4 levels were regulated by the overexpression of ACSL4 plasmid or ACSL4 siRNA, and the effects of ACSL4 on decidualization markers and morphology of endometrial stromal cells (ESCs) were clarified. A pregnant mouse model was established to determine the effect of ACSL4 on the implantation efficiency of mouse embryos. Modulation of ACSL4 detects lipid anabolism and catabolism. RESULTS Through examining the expression level of ACSL4 in human endometrial tissues during proliferative and secretory phases, we found that ACSL4 was highly expressed during the secretory phase. Knockdown of ACSL4 suppressed decidualization and inhibited the mesenchymal-to-epithelial transition induced by MPA and db-cAMP in ESCs. Further, the knockdown of ACSL4 reduced the efficiency of embryo implantation in pregnant mice. Downregulation of ACSL4 inhibited FA β-oxidation and lipid droplet accumulation during decidualization. Interestingly, pharmacological and genetic inhibition of lipid droplet synthesis did not affect FA β-oxidation and decidualization, while the pharmacological and genetic inhibition of FA β-oxidation increased lipid droplet accumulation and inhibited decidualization. In addition, inhibition of β-oxidation was found to attenuate the promotion of decidualization by the upregulation of ACSL4. The decidualization damage caused by ACSL4 knockdown could be reversed by activating β-oxidation. CONCLUSIONS Our findings suggest that ACSL4 promotes endometrial decidualization by activating the β-oxidation pathway. This study provides interesting insights into our understanding of the mechanisms regulating lipid metabolism during decidualization.
Collapse
Affiliation(s)
- Hongshuo Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China; Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Qianyi Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Haojie Dong
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Zeen Jin
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Mengyue Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Shanyuan Jin
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaolan Zeng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jianhui Fan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| | - Ying Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| |
Collapse
|
9
|
Zhao H, Cheng X, Yan L, Mi F, Wang W, Hu Y, Liu X, Fan Y, Min Q, Wang Y, Zhang W, Wu Q, Zhan Q. APC/C-regulated CPT1C promotes tumor progression by upregulating the energy supply and accelerating the G1/S transition. Cell Commun Signal 2024; 22:283. [PMID: 38783346 PMCID: PMC11112774 DOI: 10.1186/s12964-024-01657-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND In addition to functioning as a precise monitoring mechanism in cell cycle, the anaphase-promoting complex/cyclosome (APC/C) is reported to be involved in regulating multiple metabolic processes by facilitating the ubiquitin-mediated degradation of key enzymes. Fatty acid oxidation is a metabolic pathway utilized by tumor cells that is crucial for malignant progression; however, its association with APC/C remains to be explored. METHODS Cell cycle synchronization, immunoblotting, and propidium iodide staining were performed to investigate the carnitine palmitoyltransferase 1 C (CPT1C) expression manner. Proximity ligation assay and co-immunoprecipitation were performed to detect interactions between CPT1C and APC/C. Flow cytometry, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2 H-tetrazolium, inner salt (MTS) assays, cell-scratch assays, and transwell assays and xenograft transplantation assays were performed to investigate the role of CPT1C in tumor progression in vitro and in vivo. Immunohistochemistry was performed on tumor tissue microarray to evaluate the expression levels of CPT1C and explore its potential clinical value. RESULTS We identified CPT1C as a novel APC/C substrate. CPT1C protein levels exhibited cell cycle-dependent fluctuations, peaking at the G1/S boundary. Elevated CPT1C accelerated the G1/S transition, facilitating tumor cell proliferation in vitro and in vivo. Furthermore, CPT1C enhanced fatty acid utilization, upregulated ATP levels, and decreased reactive oxygen species levels, thereby favoring cell survival in a harsh metabolic environment. Clinically, high CPT1C expression correlated with poor survival in patients with esophageal squamous cell carcinoma. CONCLUSIONS Overall, our results revealed a novel interplay between fatty acid utilization and cell cycle machinery in tumor cells. Additionally, CPT1C promoted tumor cell proliferation and survival by augmenting cellular ATP levels and preserving redox homeostasis, particularly under metabolic stress. Therefore, CPT1C could be an independent prognostic indicator in esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Huihui Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xinxin Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Liping Yan
- Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Fang Mi
- State Key Laboratory of Molecular Oncology, National Cancer center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenqing Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuying Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xingyang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuyan Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Weimin Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Peking University International Cancer Institute, Beijing, 100142, China.
- Soochow University Cancer Institute, Suzhou, 215000, China.
| |
Collapse
|
10
|
Li J, Wang C, Xu X, Chen J, Guo H. An extensive analysis of the prognostic and immune role of FOXO1 in various types of cancer. Braz J Med Biol Res 2024; 57:e13378. [PMID: 38716982 PMCID: PMC11085032 DOI: 10.1590/1414-431x2024e13378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/27/2024] [Indexed: 05/12/2024] Open
Abstract
Forkhead Box O1 (FOXO1) has been reported to play important roles in many tumors. However, FOXO1 has not been studied in pan-cancer. The purpose of this study was to reveal the roles of FOXO1 in pan-cancer (33 cancers in this study). Through multiple public platforms, a pan-cancer analysis of FOXO1 was conducted to obtained FOXO1 expression profiles in various tumors to explore the relationship between FOXO1 expression and prognosis of these tumors and to disclose the potential mechanism of FOXO1 in these tumors. FOXO1 was associated with the prognosis of multiple tumors, especially LGG (low grade glioma), OV (ovarian carcinoma), and KIRC (kidney renal clear cell carcinoma). FOXO1 might play the role of an oncogenic gene in LGG and OV, while playing the role of a cancer suppressor gene in KIRC. FOXO1 expression had a significant correlation with the infiltration of some immune cells in LGG, OV, and KIRC. By combining FOXO1 expression and immune cell infiltration, we found that FOXO1 might influence the overall survival of LGG through the infiltration of myeloid dendritic cells or CD4+ T cells. Functional enrichment analysis and gene set enrichment analysis showed that FOXO1 might play roles in tumors through immunoregulatory interactions between a lymphoid and a non-lymphoid cell, TGF-beta signaling pathway, and transcriptional misregulation in cancer. FOXO1 was associated with the prognosis of multiple tumors, especially LGG, OV, and KIRC. In these tumors, FOXO1 might play its role via the regulation of the immune microenvironment.
Collapse
Affiliation(s)
- Jie Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated
Hangzhou First People's Hospital, West Lake University School of Medicine,
Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine
of Zhejiang Province, Hangzhou, China
| | - Chao Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated
Hangzhou First People's Hospital, West Lake University School of Medicine,
Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine
of Zhejiang Province, Hangzhou, China
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine
of Zhejiang Province, Hangzhou, China
| | - Jun Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated
Hangzhou First People's Hospital, West Lake University School of Medicine,
Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine
of Zhejiang Province, Hangzhou, China
| | - Haijun Guo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated
Hangzhou First People's Hospital, West Lake University School of Medicine,
Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine
of Zhejiang Province, Hangzhou, China
| |
Collapse
|
11
|
Zhang FB, Gan L, Zhu TH, Ding HQ, Wu CH, Guan YT, Chen XQ. Pan-cancer analyses reveal genomics and clinical outcome association of the fatty acid oxidation regulators in cancer. Heliyon 2024; 10:e28441. [PMID: 38590909 PMCID: PMC10999922 DOI: 10.1016/j.heliyon.2024.e28441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/10/2024] Open
Abstract
Background Fatty acid oxidation (FAO) is considered to play a vital part in tumor metabolic reprogramming. But the comprehensive description of FAO dysregulation in tumors has not been unknown. Methods We obtained FAO genes, RNA-seq data and clinical information from the Msigdb, TCGA and GTEx databases. We assessed their prognosis value using univariate cox analysis, survival analysis and Kaplan-Meier curve. We determined the function of FAO genes using gene set variation analysis. The correlation analysis was calculated by corrplot R package. Immunotherapy response was assessed through TIDE scores. The protein expression levels of FAO genes were validated using immunohistochemistry (IHC). Results The FAO scores were highest in COAD but lowest in PCPG. FAO scores were significantly associated with the prognosis of some cancers in OS, DSS, DFI and PFI. Besides, gene set variation analysis identified that FAO scores were related to immune-related pathways, and immune infiltration analysis showed FAO scores were positively related to cancer-associated fibroblasts and various immune-related genes. TIDE scores were significantly decreased in ACC, CHOL, ESCA, GBM, LAML, SARC, SKCM and THCA compared with normal samples, while it was significantly increased in BLCA, LUAD, LUSC, PCPG, PRAD and STAD. Besides, most FAO genes were downregulated in pan-cancer compared with normal samples. Moreover, we found copy number variation (CNV) of FAO genes played a positive role in their mRNA expression, while methylation was negative. We determined FAO genes were closely related to some drugs in pan-cancer. Conclusions FAO score is a novel and promising factor for predicting outcomes.
Collapse
Affiliation(s)
- Fu-bin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Lei Gan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Tian-hong Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Hui-qing Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Cheng-hao Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No.389 Xincun Road, Shanghai, 200065, China
| | - Yu-tao Guan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Xue-qin Chen
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| |
Collapse
|
12
|
Saorin A, Saorin G, Duzagac F, Parisse P, Cao N, Corona G, Cavarzerani E, Rizzolio F. Microfluidic production of amiodarone loaded nanoparticles and application in drug repositioning in ovarian cancer. Sci Rep 2024; 14:6280. [PMID: 38491077 PMCID: PMC10943008 DOI: 10.1038/s41598-024-55801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
Amiodarone repositioning in cancer treatment is promising, however toxicity limits seem to arise, constraining its exploitability. Notably, amiodarone has been investigated for the treatment of ovarian cancer, a tumour known for metastasizing within the peritoneal cavity. This is associated with an increase of fatty acid oxidation, which strongly depends on CPT1A, a transport protein which has been found overexpressed in ovarian cancer. Amiodarone is an inhibitor of CPT1A but its role still has to be explored. Therefore, in the present study, amiodarone was tested on ovarian cancer cell lines with a focus on lipid alteration, confirming its activity. Moreover, considering that drug delivery systems could lower drug side effects, microfluidics was employed for the development of drug delivery systems of amiodarone obtaining simultaneously liposomes with a high payload and amiodarone particles. Prior to amiodarone loading, microfluidics production was optimized in term of temperature and flow rate ratio. Moreover, stability over time of particles was evaluated. In vitro tests confirmed the efficacy of the drug delivery systems.
Collapse
Affiliation(s)
- Asia Saorin
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy
| | - Gloria Saorin
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy
| | - Fahriye Duzagac
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pietro Parisse
- Elettra-Sincrotrone Trieste S.C.p.A., Area Science Park, Strada Statale 14 km 163.5, Basovizza, 34149, Trieste, Italy
- CNR-IOM - Istituto Officina dei Materiali, Area Science Park, s.s. 14 Km 163.5, Basovizza, 34149, Trieste, Italy
| | - Ni Cao
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy
| | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Enrico Cavarzerani
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy.
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, 33081, Aviano, Italy.
| |
Collapse
|
13
|
Zhao S, Wang Q, Zhang X, Ma B, Shi Y, Yin Y, Kong W, Zhang W, Li J, Yang H. MARCH5-mediated downregulation of ACC2 promotes fatty acid oxidation and tumor progression in ovarian cancer. Free Radic Biol Med 2024; 212:464-476. [PMID: 38211832 DOI: 10.1016/j.freeradbiomed.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Lipid metabolic reprogramming has been recognized as a hallmark of human cancer. Acetyl-CoA Carboxylases (ACCs) are key rate-limiting enzymes involved in fatty acid metabolism regulation by catalyzing the carboxylation of acetyl-CoA to malonyl-CoA. Previously, most studies focused on the role of ACC1 in fatty acid metabolism in cancer, while the function of ACC2 remains largely uncharacterized in human cancers, especially in ovarian cancer (OC). Here, we show that ACC2 was significantly downregulated in cancerous tissue of OC, and the downregulation of ACC2 is closely associated with lager tumor size, metastases and worse prognosis in OC patients. Downregulation of ACC2 promoted proliferation and metastasis of OC both in vitro and in vivo by enhancing FAO. Notably, mitochondria-associated ubiquitin ligase (MARCH5) was identified to interact with and downregulate ACC2 by ubiquitination and degradation in OC. Moreover, ACC2 downregulation-enhanced FAO contributed to the progression of OC promoted by MARCH5. In conclusion, our findings demonstrate that MARCH5-mediated downregulation of ACC2 promotes FAO and tumorigenesis in OC, suggesting MARCH5-ACC2 axis as a potent candidate for the treatment and prevention of OC.
Collapse
Affiliation(s)
- Shuhua Zhao
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Qingqiang Wang
- General Department, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xiaohong Zhang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Boyi Ma
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yuan Shi
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yadong Yin
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Weina Kong
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wei Zhang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Jibin Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, China.
| | - Hong Yang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, China.
| |
Collapse
|
14
|
Wu K, Lin F. Lipid Metabolism as a Potential Target of Liver Cancer. J Hepatocell Carcinoma 2024; 11:327-346. [PMID: 38375401 PMCID: PMC10875169 DOI: 10.2147/jhc.s450423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/25/2024] [Indexed: 02/21/2024] Open
Abstract
Hepatocellular carcinoma (HCC) stands as a severe malignant tumor with a profound impact on overall health, often accompanied by an unfavorable prognosis. Despite some advancements in the diagnosis and treatment of this disease, improving the prognosis of HCC remains a formidable challenge. It is noteworthy that lipid metabolism plays a pivotal role in the onset, development, and progression of tumor cells. Existing research indicates the potential application of targeting lipid metabolism in the treatment of HCC. This review aims to thoroughly explore the alterations in lipid metabolism in HCC, offering a detailed account of the potential advantages associated with innovative therapeutic strategies targeting lipid metabolism. Targeting lipid metabolism holds promise for potentially enhancing the prognosis of HCC.
Collapse
Affiliation(s)
- Kangze Wu
- Department of Hepatobiliary Surgery, Shaoxing People’s Hospital, Shaoxing, People’s Republic of China
| | - Feizhuan Lin
- Department of Hepatobiliary Surgery, Shaoxing People’s Hospital, Shaoxing, People’s Republic of China
| |
Collapse
|
15
|
Yang C, Zhu L, Lin Q. Anoikis related genes may be novel markers associated with prognosis for ovarian cancer. Sci Rep 2024; 14:1564. [PMID: 38238592 PMCID: PMC10796408 DOI: 10.1038/s41598-024-52117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/14/2024] [Indexed: 01/22/2024] Open
Abstract
The aim of this study was to determine the prognostic significance of anoikis related genes (ARGs) in ovarian cancer (OC) and to develop a prognostic signature based on ARG expression. We analyzed cohorts of OC patients and used nonnegative matrix factorization (NMF) for clustering. Single-sample gene-set enrichment analysis (ssGSEA) was employed to quantify immune infiltration. Survival analyses were performed using the Kaplan-Meier method, and differences in survival were determined using the log-rank test. The extent of anoikis modification was quantified using a risk score generated from ARG expression. The analysis of single-cell sequencing data was performed by the Tumor Immune Single Cell Hub (TISCH). Our analyses revealed two distinct patterns of anoikis modification. The risk score was used to evaluate the anoikis modification patterns in individual tumors. Three hub-genes were screened using the LASSO (Least Absolute Shrinkage and Selection Operator) method and patients were classified into different risk groups based on their individual score and the median score. The low-risk subtype was characterized by decreased expression of hub-genes and better overall survival. The risk score, along with patient age and gender, were considered to identify the prognostic signature, which was visualized using a nomogram. Our findings suggest that ARGs may play a novel role in the prognosis of OC. Based on ARG expression, we have developed a prognostic signature for OC that can aid in patient stratification and treatment decision-making. Further studies are needed to validate these results and to explore the underlying mechanisms.
Collapse
Affiliation(s)
- Chen Yang
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
| | - LuChao Zhu
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
| | - Qin Lin
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China.
| |
Collapse
|
16
|
Yang H, Gu W, Ni J, Ma Y, Li S, Neumann D, Ding X, Zhu L. Carnitine palmitoyl-transferase 1A is potentially involved in bovine herpesvirus 1 productive infection. Vet Microbiol 2024; 288:109932. [PMID: 38043447 PMCID: PMC10919102 DOI: 10.1016/j.vetmic.2023.109932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/21/2023] [Accepted: 11/26/2023] [Indexed: 12/05/2023]
Abstract
Bovine herpesvirus 1(BoHV-1) is an important bovine pathogen that causes great economic loss to cattle farms worldwide. The virus-productive infection in bovine kidney (MDBK) cells results in ATP depletion. The mechanisms are not well understood. Mitochondrial fatty acid β-oxidation (FAO) is an important energy source in many tissues with high energy demand. Since carnitine palmitoyl-transferase 1 A (CPT1A) is the rate-limiting enzyme of FAO, we investigated the interactions between virus-productive infection and CPT1A signaling. Here, we found that virus-productive infection at the later stage significantly decreased CPT1A protein levels in all the detected cells, including MDBK, A549, and Neuro-2A cells, differentially altered the accumulation of CPT1A proteins in the nucleus and cytosol, and re-localized the protein in the nucleus. Etomoxir (ETO), an irreversible inhibitor of CPT1A, inhibited viral replication and partially interfered with the ability of BoHV-1 to alter CPT1A accumulation in the nucleus but not in the cytosol. Furthermore, ETO consistently reduced RNA levels of two viral regulatory proteins (bICP0 and bICP22) and protein expression of virion-associated proteins during productive infection, further supporting the important roles of CPT1A signaling in BoHV-1 productive infection. These data, for the first time, suggest that CPT1A is potentially involved in BoHV-1 productive infection.
Collapse
Affiliation(s)
- Hao Yang
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Wenyuan Gu
- Center for Animal Diseases Control and Prevention of Hebei Province, Shijiazhuang 050035, China
| | - Junqing Ni
- Hebei Province Animal Husbandry and Improved Breeds Work Station, Shijiazhuang 050061, China
| | - Yabin Ma
- Hebei Province Animal Husbandry and Improved Breeds Work Station, Shijiazhuang 050061, China
| | - Shitao Li
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA 70118, USA
| | - Donna Neumann
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 537006, USA
| | - Xiuyan Ding
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Liqian Zhu
- College of Life Sciences, Hebei University, Baoding 071002, China
| |
Collapse
|
17
|
Alarcon-Zapata P, Perez AJ, Toledo-Oñate K, Contreras H, Ormazabal V, Nova-Lamperti E, Aguayo CA, Salomon C, Zuniga FA. Metabolomics profiling and chemoresistance mechanisms in ovarian cancer cell lines: Implications for targeting glutathione pathway. Life Sci 2023; 333:122166. [PMID: 37827232 DOI: 10.1016/j.lfs.2023.122166] [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: 08/15/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/14/2023]
Abstract
Ovarian cancer presents a significant challenge due to its high rate of chemoresistance, which complicates the effectiveness of drug-response therapy. This study provides a comprehensive metabolomic analysis of ovarian cancer cell lines OVCAR-3 and SK-OV-3, characterizing their distinct metabolic landscapes. Metabolomics coupled with chemometric analysis enabled us to discriminate between the metabolic profiles of these two cell lines. The OVCAR-3 cells, which are sensitive to doxorubicin (DOX), exhibited a preference for biosynthetic pathways associated with cell proliferation. Conversely, DOX-resistant SK-OV-3 cells favored fatty acid oxidation for energy maintenance. Notably, a marked difference in glutathione (GSH) metabolism was observed between these cell lines. Our investigations further revealed that GSH depletion led to a profound change in drug sensitivity, inducing a shift from a cytostatic to a cytotoxic response. The results derived from this comprehensive metabolomic analysis offer potential targets for novel therapeutic strategies to overcome drug resistance. Our study suggests that targeting the GSH pathway could potentially enhance chemotherapy's efficacy in treating ovarian cancer.
Collapse
Affiliation(s)
- Pedro Alarcon-Zapata
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile; Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomás, Concepción, Chile
| | - Andy J Perez
- Department of Instrumental Analysis, Faculty of Pharmacy, University of Concepcion, Chile
| | - Karin Toledo-Oñate
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile
| | - Hector Contreras
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile
| | - Valeska Ormazabal
- Department of Pharmacology, Faculty of Biological Sciences, University of Concepcion, Chile
| | - Estefania Nova-Lamperti
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile
| | - Claudio A Aguayo
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane QLD 4029, Australia
| | - Felipe A Zuniga
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile.
| |
Collapse
|
18
|
Kolb S, Hoffmann I, Monjé N, Dragomir MP, Jank P, Bischoff P, Keunecke C, Pohl J, Kunze CA, Marchenko S, Schmitt WD, Kulbe H, Sers C, Sehouli J, Braicu EI, Denkert C, Darb-Esfahani S, Horst D, Sinn BV, Taube ET. LRP1B-a prognostic marker in tubo-ovarian high-grade serous carcinoma. Hum Pathol 2023; 141:158-168. [PMID: 37742945 DOI: 10.1016/j.humpath.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
Low-density lipoprotein (LDL) receptor-related protein 1B (LRP1B) is a member of the LDL receptor family and has often been discussed as a tumor suppressor gene, as its down-regulation is correlated with a poor prognosis in multiple carcinoma entities. Due to the high metastasis rate into the fatty peritoneal cavity and current research findings showing a dysregulation of lipid metabolism in tubo-ovarian high-grade serous carcinoma (HGSC), we questioned the prognostic impact of the LRP1B protein expression. We examined a well-characterized large cohort of 571 patients with primary HGSC and analyzed the LRP1B protein expression via immunohistochemical staining (both in tumor and stroma cells separately), performed precise bioimage analysis with QuPath, and calculated the prognostic impact using SPSS. Our results demonstrate that LRP1B functions as a significant prognostic marker for overall survival (OS) and progression-free survival (PFS) in HGSC on the protein level. High cytoplasmic expression of LRP1B in tumor, stroma, and combined tumor and stroma cells has a significantly positive association with a mean prolongation of the OS by 42 months (P = .005), 29 months (P = .005), and 25 months (P = .001), respectively. Additionally, the mean PFS was 18 months longer in tumor (P = .002), 19 months in stroma (P = .004), and 19 months in both cell types combined (P = .01). Our results remained significant in multivariate analysis. We envision LRP1B as a potential prognostic tool that could help us understand the functional role of lipid metabolism in advanced HGSC, especially regarding liposomal medications.
Collapse
Affiliation(s)
- Svenja Kolb
- Department of Gynecology, Vivantes Netzwerk für Gesundheit GmbH Berlin, Vivantes Hospital Neukölln, 12351, Berlin, Germany
| | - Inga Hoffmann
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Nanna Monjé
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Mihnea P Dragomir
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Paul Jank
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg, 35043 Marburg, Germany
| | - Philip Bischoff
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Carlotta Keunecke
- Department of Gynecology, European Competence Center for Ovarian Cancer, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Jonathan Pohl
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Catarina Alisa Kunze
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Sofya Marchenko
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Wolfgang D Schmitt
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Hagen Kulbe
- Department of Gynecology, European Competence Center for Ovarian Cancer, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; Tumorbank Ovarian Cancer Network, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Christine Sers
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, European Competence Center for Ovarian Cancer, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; Tumorbank Ovarian Cancer Network, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Elena Ioana Braicu
- Department of Gynecology, European Competence Center for Ovarian Cancer, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; Tumorbank Ovarian Cancer Network, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg, 35043 Marburg, Germany
| | - Silvia Darb-Esfahani
- MVZ Pathologie Spandau, 13589 Berlin, Spandau, Germany; MVZ Pathologie Berlin-Buch, 13125 Berlin, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Bruno V Sinn
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Eliane T Taube
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany.
| |
Collapse
|
19
|
Szrok-Jurga S, Czumaj A, Turyn J, Hebanowska A, Swierczynski J, Sledzinski T, Stelmanska E. The Physiological and Pathological Role of Acyl-CoA Oxidation. Int J Mol Sci 2023; 24:14857. [PMID: 37834305 PMCID: PMC10573383 DOI: 10.3390/ijms241914857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Fatty acid metabolism, including β-oxidation (βOX), plays an important role in human physiology and pathology. βOX is an essential process in the energy metabolism of most human cells. Moreover, βOX is also the source of acetyl-CoA, the substrate for (a) ketone bodies synthesis, (b) cholesterol synthesis, (c) phase II detoxication, (d) protein acetylation, and (d) the synthesis of many other compounds, including N-acetylglutamate-an important regulator of urea synthesis. This review describes the current knowledge on the importance of the mitochondrial and peroxisomal βOX in various organs, including the liver, heart, kidney, lung, gastrointestinal tract, peripheral white blood cells, and other cells. In addition, the diseases associated with a disturbance of fatty acid oxidation (FAO) in the liver, heart, kidney, lung, alimentary tract, and other organs or cells are presented. Special attention was paid to abnormalities of FAO in cancer cells and the diseases caused by mutations in gene-encoding enzymes involved in FAO. Finally, issues related to α- and ω- fatty acid oxidation are discussed.
Collapse
Affiliation(s)
- Sylwia Szrok-Jurga
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Jacek Turyn
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Areta Hebanowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Julian Swierczynski
- Institue of Nursing and Medical Rescue, State University of Applied Sciences in Koszalin, 75-582 Koszalin, Poland;
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Ewa Stelmanska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| |
Collapse
|
20
|
Liu Z, Liu W, Wang W, Ma Y, Wang Y, Drum DL, Cai J, Blevins H, Lee E, Shah S, Fisher PB, Wang X, Fang X, Guo C, Wang XY. CPT1A-mediated fatty acid oxidation confers cancer cell resistance to immune-mediated cytolytic killing. Proc Natl Acad Sci U S A 2023; 120:e2302878120. [PMID: 37722058 PMCID: PMC10523454 DOI: 10.1073/pnas.2302878120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/14/2023] [Indexed: 09/20/2023] Open
Abstract
Although tumor-intrinsic fatty acid β-oxidation (FAO) is implicated in multiple aspects of tumorigenesis and progression, the impact of this metabolic pathway on cancer cell susceptibility to immunotherapy remains unknown. Here, we report that cytotoxicity of killer T cells induces activation of FAO and upregulation of carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme of FAO in cancer cells. The repression of CPT1A activity or expression renders cancer cells more susceptible to destruction by cytotoxic T lymphocytes. Our mechanistic studies reveal that FAO deficiency abrogates the prosurvival signaling in cancer cells under immune cytolytic stress. Furthermore, we identify T cell-derived IFN-γ as a major factor responsible for induction of CPT1A and FAO in an AMPK-dependent manner, indicating a dynamic interplay between immune effector cells and tumor targets. While cancer growth in the absence of CPT1A remains largely unaffected, established tumors upon FAO inhibition become significantly more responsive to cellular immunotherapies including chimeric antigen receptor-engineered human T cells. Together, these findings uncover a mode of cancer resistance and immune editing that can facilitate immune escape and limit the benefits of immunotherapies.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Wenjie Liu
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Wei Wang
- Department of Biochemistry, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Yibao Ma
- Department of Biochemistry, Virginia Commonwealth University School of Medicine, Richmond, VA23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Yufeng Wang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
| | - David L. Drum
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
| | - Jinyang Cai
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Hallie Blevins
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Eun Lee
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Syed Shah
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298
- Hunter Holmes McGuire VA Medical Center, Richmond, VA23249
| | - Paul B. Fisher
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Xinhui Wang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
| | - Xianjun Fang
- Department of Biochemistry, Virginia Commonwealth University School of Medicine, Richmond, VA23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Chunqing Guo
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| | - Xiang-Yang Wang
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298
- Hunter Holmes McGuire VA Medical Center, Richmond, VA23249
- Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA23298
| |
Collapse
|
21
|
Wang Y, Chen Q, Wu S, Sun X, Yin R, Ouyang Z, Yin H, Wei Y. Amelioration of ethanol-induced oxidative stress and alcoholic liver disease by in vivo RNAi targeting Cyp2e1. Acta Pharm Sin B 2023; 13:3906-3918. [PMID: 37719371 PMCID: PMC10502278 DOI: 10.1016/j.apsb.2023.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/16/2022] [Accepted: 12/10/2022] [Indexed: 09/19/2023] Open
Abstract
Alcoholic liver disease (ALD) results from continuous and heavy alcohol consumption. The current treatment strategy for ALD is based on alcohol withdrawal coupled with antioxidant drug intervention, which is a long process with poor efficacy and low patient compliance. Alcohol-induced CYP2E1 upregulation has been demonstrated as a key regulator of ALD, but CYP2E1 knockdown in humans was impractical, and pharmacological inhibition of CYP2E1 by a clinically relevant approach for treating ALD was not shown. In this study, we developed a RNAi therapeutics delivered by lipid nanoparticle, and treated mice fed on Lieber-DeCarli ethanol liquid diet weekly for up to 12 weeks. This RNAi-based inhibition of Cyp2e1 expression reduced reactive oxygen species and oxidative stress in mouse livers, and contributed to improved ALD symptoms in mice. The liver fat accumulation, hepatocyte inflammation, and fibrosis were reduced in ALD models. Therefore, this study suggested the feasibility of RNAi targeting to CYP2E1 as a potential therapeutic tool to the development of ALD.
Collapse
Affiliation(s)
- Yalan Wang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Qiubing Chen
- Department of Urology, Frontier Science Centre for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Shuang Wu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Xinyu Sun
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Runting Yin
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Zhen Ouyang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Hao Yin
- Department of Urology, Frontier Science Centre for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- RNA Institute, Wuhan University, Wuhan 430072, China
- Wuhan Research Centre for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430010, China
| | - Yuan Wei
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
22
|
Tang Y, Zhang W, Wang Y, Li H, Zhang C, Wang Y, Lin Y, Shi H, Xiang H, Huang L, Zhu J. Expression Variation of CPT1A Induces Lipid Reconstruction in Goat Intramuscular Precursor Adipocytes. Int J Mol Sci 2023; 24:13415. [PMID: 37686221 PMCID: PMC10488119 DOI: 10.3390/ijms241713415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Intramuscular fat (IMF) deposition is one of the most important factors affecting meat quality and is closely associated with the expression of carnitine palmitoyl transferase 1A (CPT1A) which facilitates the transfer of long-chain fatty acids (LCFAs) into the mitochondria. However, the role of how CPT1A regulates the IMF formation remains unclear. Herein, we established the temporal expression profile of CPT1A during the differentiation of goat intramuscular precursor adipocytes. Functionally, the knockdown of CPT1A by siRNA treatment significantly increased the mRNA expression of adipogenic genes and promoted lipid deposition in goat intramuscular precursor adipocytes. Meanwhile, a CPT1A deficiency inhibited cell proliferation and promoted cell apoptosis significantly. CPT1A was then supported by the overexpression of CPT1A which significantly suppressed the cellular triglyceride deposition and promoted cell proliferation although the cell apoptosis also was increased. For RNA sequencing, a total of 167 differential expression genes (DEGs), including 125 upregulated DEGs and 42 downregulated DEGs, were observed after the RNA silencing of CPT1A compared to the control, and were predicted to enrich in the focal adhesion pathway, cell cycle, apoptosis and the MAPK signaling pathway by KEGG analysis. Specifically, blocking the MAPK signaling pathway by a specific inhibitor (PD169316) rescued the promotion of cell proliferation in CPT1A overexpression adipocytes. In conclusion, the expression variation of CPT1A may reconstruct the lipid distribution between cellular triglyceride deposition and cell proliferation in goat intramuscular precursor adipocyte. Furthermore, we demonstrate that CPT1A promotes the proliferation of goat adipocytes through the MAPK signaling pathway. This work widened the genetic regulator networks of IMF formation and delivered theoretical support for improving meat quality from the aspect of IMF deposition.
Collapse
Affiliation(s)
- Yinmei Tang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Wenyang Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| | - Yinggui Wang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Haiyang Li
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Changhui Zhang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Yong Wang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| | - Yaqiu Lin
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| | - Hengbo Shi
- College of Animal Science, Zhejiang University, Hangzhou 310058, China;
| | - Hua Xiang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| | - Lian Huang
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
| | - Jiangjiang Zhu
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu 610225, China; (Y.T.); (Y.W.); (H.L.); (C.Z.); (Y.W.); (Y.L.); (H.X.); (L.H.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Ministry of Education, Chengdu 610041, China;
| |
Collapse
|
23
|
Liu B, Meng Q, Gao X, Sun H, Xu Z, Wang Y, Zhou H. Lipid and glucose metabolism in senescence. Front Nutr 2023; 10:1157352. [PMID: 37680899 PMCID: PMC10481967 DOI: 10.3389/fnut.2023.1157352] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Senescence is an inevitable biological process. Disturbances in glucose and lipid metabolism are essential features of cellular senescence. Given the important roles of these types of metabolism, we review the evidence for how key metabolic enzymes influence senescence and how senescence-related secretory phenotypes, autophagy, apoptosis, insulin signaling pathways, and environmental factors modulate glucose and lipid homeostasis. We also discuss the metabolic alterations in abnormal senescence diseases and anti-cancer therapies that target senescence through metabolic interventions. Our work offers insights for developing pharmacological strategies to combat senescence and cancer.
Collapse
Affiliation(s)
- Bin Liu
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Xin Gao
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huihui Sun
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Zhixiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
24
|
Mahé M, Rios-Fuller TJ, Karolin A, Schneider RJ. Genetics of enzymatic dysfunctions in metabolic disorders and cancer. Front Oncol 2023; 13:1230934. [PMID: 37601653 PMCID: PMC10433910 DOI: 10.3389/fonc.2023.1230934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Inherited metabolic disorders arise from mutations in genes involved in the biogenesis, assembly, or activity of metabolic enzymes, leading to enzymatic deficiency and severe metabolic impairments. Metabolic enzymes are essential for the normal functioning of cells and are involved in the production of amino acids, fatty acids and nucleotides, which are essential for cell growth, division and survival. When the activity of metabolic enzymes is disrupted due to mutations or changes in expression levels, it can result in various metabolic disorders that have also been linked to cancer development. However, there remains much to learn regarding the relationship between the dysregulation of metabolic enzymes and metabolic adaptations in cancer cells. In this review, we explore how dysregulated metabolism due to the alteration or change of metabolic enzymes in cancer cells plays a crucial role in tumor development, progression, metastasis and drug resistance. In addition, these changes in metabolism provide cancer cells with a number of advantages, including increased proliferation, resistance to apoptosis and the ability to evade the immune system. The tumor microenvironment, genetic context, and different signaling pathways further influence this interplay between cancer and metabolism. This review aims to explore how the dysregulation of metabolic enzymes in specific pathways, including the urea cycle, glycogen storage, lysosome storage, fatty acid oxidation, and mitochondrial respiration, contributes to the development of metabolic disorders and cancer. Additionally, the review seeks to shed light on why these enzymes represent crucial potential therapeutic targets and biomarkers in various cancer types.
Collapse
Affiliation(s)
| | | | | | - Robert J. Schneider
- Department of Microbiology, Grossman NYU School of Medicine, New York, NY, United States
| |
Collapse
|
25
|
Kalathil AA, Guin S, Ashokan A, Basu U, Surnar B, Delma KS, Lima LM, Kryvenko ON, Dhar S. New Pathway for Cisplatin Prodrug to Utilize Metabolic Substrate Preference to Overcome Cancer Intrinsic Resistance. ACS CENTRAL SCIENCE 2023; 9:1297-1312. [PMID: 37521786 PMCID: PMC10375877 DOI: 10.1021/acscentsci.3c00286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Indexed: 08/01/2023]
Abstract
Tumor cells adapt to diverse survival strategies defying our pursuit of multimodal cancer therapy. Prostate cancer (PCa) is an example that is resistant to one of the most potent chemotherapeutics, cisplatin. PCa cells survive and proliferate using fatty acid oxidation (FAO), and the dependence on fat utilization increases as the disease progresses toward a resistant form. Using a pool of patient biopsies, we validated the expression of a key enzyme carnitine palmitoyltransferase 1 A (CPT1A) needed for fat metabolism. We then discovered that a cisplatin prodrug, Platin-L, can inhibit the FAO of PCa cells by interacting with CPT1A. Synthesizing additional cisplatin-based prodrugs, we documented that the presence of an available carboxylic acid group near the long chain fatty acid linker on the Pt(IV) center is crucial for CPT1A binding. As a result of fat metabolism disruption by Platin-L, PCa cells transition to an adaptive glucose-dependent chemosensitive state. Potential clinical translation of Platin-L will require a delivery vehicle to direct it to the prostate tumor microenvironment. Thus, we incorporated Platin-L in a biodegradable prostate tumor-targeted orally administrable nanoformulation and demonstrated its safety and efficacy. The distinctive FAO inhibitory property of Platin-L can be of potential clinical relevance as it offers the use of cisplatin for otherwise resistant cancer.
Collapse
Affiliation(s)
- Akil A. Kalathil
- NanoTherapeutics
Research Laboratory, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Subham Guin
- NanoTherapeutics
Research Laboratory, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Akash Ashokan
- NanoTherapeutics
Research Laboratory, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Sylvester
Comprehensive Cancer Centre, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Uttara Basu
- NanoTherapeutics
Research Laboratory, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Sylvester
Comprehensive Cancer Centre, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Bapurao Surnar
- NanoTherapeutics
Research Laboratory, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Sylvester
Comprehensive Cancer Centre, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Katiana S. Delma
- Department
of Pathology and Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Leonor M. Lima
- Department
of Pathology and Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Oleksandr N. Kryvenko
- Sylvester
Comprehensive Cancer Centre, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Department
of Pathology and Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Department
of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Desai Sethi
Urology Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Shanta Dhar
- NanoTherapeutics
Research Laboratory, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Sylvester
Comprehensive Cancer Centre, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Department
of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| |
Collapse
|
26
|
Cacciola NA, Sepe F, Fioriniello S, Petillo O, Margarucci S, Scivicco M, Peluso G, Balestrieri A, Bifulco G, Restucci B, Severino L. The Carnitine Palmitoyltransferase 1A Inhibitor Teglicar Shows Promising Antitumour Activity against Canine Mammary Cancer Cells by Inducing Apoptosis. Pharmaceuticals (Basel) 2023; 16:987. [PMID: 37513899 PMCID: PMC10383333 DOI: 10.3390/ph16070987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Canine mammary tumours (CMTs) are the most common cancer in intact female dogs. In addition to surgery, additional targeted and non-targeted therapies may offer survival benefits to these patients. Therefore, exploring new treatments for CMT is a promising area in veterinary oncology. CMT cells have an altered lipid metabolism and use the oxidation of fatty acids for their energy needs. Here we investigated the tumoricidal effects of teglicar, a reversible inhibitor of carnitine palmitoyl transferase 1A (CPT1A), the rate-limiting enzyme for fatty acid import into mitochondria, on two CMT cells, P114 and CMT-U229. Viability and apoptosis were examined in CMT cells using the crystal violet assay, trypan blue assay, and flow cytometry analysis. The expression of mediators of apoptosis signalling (e.g., caspase-9, caspase-8, and caspase-3) was assessed by quantitative real-time polymerase chain reaction and western blot analyses. Teglicar was able to decrease cell viability and induce apoptosis in P114 and CMT-U229 cells. At the molecular level, the effect of teglicar was associated with an upregulation of the mRNA expression levels of caspase-9, caspase-8, and caspase-3 and an increase in their protein levels. In summary, our results show that teglicar has a potential effect against CMTs through the induction of apoptotic cell death, making it a promising therapeutic agent against CMTs.
Collapse
Affiliation(s)
- Nunzio Antonio Cacciola
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Fabrizia Sepe
- Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Salvatore Fioriniello
- Institute of Genetics and Biophysics "A. Buzzati-Traverso" (IGB), National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Orsolina Petillo
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Sabrina Margarucci
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Marcello Scivicco
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
| | - Gianfranco Peluso
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
- Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, Via di Sant'Alessandro 8, 00131 Rome, Italy
| | - Anna Balestrieri
- Food Safety Department, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Via Salute 2, 80055 Portici, Italy
| | - Giovanna Bifulco
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
| | - Brunella Restucci
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
| | - Lorella Severino
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
| |
Collapse
|
27
|
Zhu Y, Wang Y, Li Y, Li Z, Kong W, Zhao X, Chen S, Yan L, Wang L, Tong Y, Shao H. Carnitine palmitoyltransferase 1A promotes mitochondrial fission by enhancing MFF succinylation in ovarian cancer. Commun Biol 2023; 6:618. [PMID: 37291333 PMCID: PMC10250469 DOI: 10.1038/s42003-023-04993-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Mitochondria are dynamic organelles that are important for cell growth and proliferation. Dysregulated mitochondrial dynamics are highly associated with the initiation and progression of various cancers, including ovarian cancer. However, the regulatory mechanism underlying mitochondrial dynamics is still not fully understood. Previously, our study showed that carnitine palmitoyltransferase 1A (CPT1A) is highly expressed in ovarian cancer cells and promotes the development of ovarian cancer. Here, we find that CPT1A regulates mitochondrial dynamics and promotes mitochondrial fission in ovarian cancer cells. Our study futher shows that CPT1A regulates mitochondrial fission and function through mitochondrial fission factor (MFF) to promote the growth and proliferation of ovarian cancer cells. Mechanistically, we show that CPT1A promotes succinylation of MFF at lysine 302 (K302), which protects against Parkin-mediated ubiquitin-proteasomal degradation of MFF. Finally, the study shows that MFF is highly expressed in ovarian cancer cells and that high MFF expression is associated with poor prognosis in patients with ovarian cancer. MFF inhibition significantly inhibits the progression of ovarian cancer in vivo. Overall, CPT1A regulates mitochondrial dynamics through MFF succinylation to promote the development of ovarian cancer. Moreover, our findings suggest that MFF is a potential therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Yaqin Zhu
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Yue Wang
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Ying Li
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Zhongqi Li
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Wenhui Kong
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Xiaoxuan Zhao
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Shuting Chen
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Liting Yan
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Lenan Wang
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Yunli Tong
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Huanjie Shao
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China.
| |
Collapse
|
28
|
Wang Y, Huang X, Fan H, Xu Y, Qi Z, Zhang Y, Huang Y. Identification of fatty acid-related subtypes, the establishment of a prognostic signature, and immune infiltration characteristics in lung adenocarcinoma. Aging (Albany NY) 2023; 15:204725. [PMID: 37199651 DOI: 10.18632/aging.204725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/03/2023] [Indexed: 05/19/2023]
Abstract
Abnormal fatty acid (FA) metabolism can change the inflammatory microenvironment and promote tumor progression and metastasis, however, the potential association between FA-related genes (FARGs) and lung adenocarcinoma (LUAD) is still unclear. In this study, we described the genetic and transcriptomic changes of FARGs in LUAD patients and identified two different FA subtypes, which were significantly correlated with overall survival and tumor microenvironment infiltrating cells in LUAD patients. In addition, the FA score was also constructed through the LASSO Cox to evaluate the FA dysfunction of each patient. Multivariate Cox analysis proved that the FA score was an independent predictor and created the FA score integrated nomogram, which offered a quantitative tool for clinical practice. The performance of the FA score has been substantiated in numerous datasets for its commendable accuracy in estimating overall survival in LUAD patients. The groups with high and low FA scores exhibited different mutation spectrums, copy number variations, enrichment pathways, and immune status. Noteworthy differences between the two groups in terms of immunophenoscore and Tumor Immune Dysfunction and Exclusion were observed, suggesting that the group with a low FA score was more responsive to immunotherapy, and similar results were also confirmed in the immunotherapy cohort. In addition, seven potential chemotherapeutic drugs related to FA score targeting were predicted. Ultimately, we ascertained that the attenuation of KRT6A expression impeded the proliferation, migration, and invasion of LUAD cell lines. In summary, this research offers novel biomarkers to facilitate prognostic forecasting and clinical supervision for individuals afflicted with LUAD.
Collapse
Affiliation(s)
- Yuzhi Wang
- Department of Laboratory Medicine, Deyang People’s Hospital, Deyang 618000, Sichuan, People’s Republic of China
| | - Xiaoxiao Huang
- Department of Laboratory Medicine, Liuzhou Hospital of Guangzhou Women and Children’s Medical Center, Liuzhou 545000, Guangxi, People’s Republic of China
- Guangxi Clinical Research Center for Obstetrics and Gynecology, Liuzhou 545000, Guangxi, People’s Republic of China
| | - Hong Fan
- Department of Pathology, Shanghai Jianding District Anting Hospital, Shanghai 200000, People’s Republic of China
| | - Yunfei Xu
- Department of Laboratory Medicine, Chengdu Women’s and Children’s Central Hospital, Chengdu 610031, Sichuan, People’s Republic of China
| | - Zelin Qi
- Department of Laboratory Medicine, Deyang People’s Hospital, Deyang 618000, Sichuan, People’s Republic of China
| | - Yi Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, Fujian, People’s Republic of China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou 350001, Fujian, People’s Republic of China
| | - Yi Huang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, Fujian, People’s Republic of China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou 350001, Fujian, People’s Republic of China
- Central Laboratory, Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou 350001, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fuzhou 350001, Fujian, People’s Republic of China
| |
Collapse
|
29
|
Jiang X, Du W, Shi C, Kang M, Song Q, Zhang L, Pei D. Identification of a lipid metabolism-related gene for cancer immunotherapy. Front Pharmacol 2023; 14:1186064. [PMID: 37251324 PMCID: PMC10213444 DOI: 10.3389/fphar.2023.1186064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Background: Tumors frequently evade immune surveillance through multiple pathways to escape T cell recognition and destruction. Previous studies indicated that lipid metabolism alteration could affect the anti-tumor immunity of cancer cells. Nonetheless, the studies that investigated lipid metabolism-related gene for cancer immunotherapy are still few. Materials and methods: By mining the TCGA database, we screened out carnitine palmitoyltransferase-2 (CPT2), a key enzyme in the fatty acid β-oxidation (FAO) process associated with anti-tumor immunity. We then analyzed the gene expression and clinicopathological features of CPT2 using open-source platforms and databases. Molecular proteins interacting with CPT2 were also identified using web interaction tools. Subsequently, the relationship between CPT2 and survival was analyzed in cancer patients. Results: Our study revealed that CPT2 played a vital role in tumor microenvironment and immune response signaling pathways. We have also demonstrated that increased CPT2 gene expression could enhance the level of tumor immune cell infiltration. Furthermore, high CPT2 expression positively related with overall survival associated with immunotherapy. CPT2 expression was also associated with the prognosis of human cancers, suggesting that CPT2 may be a potential biomarker for predicting the efficacy of cancer immunotherapy. Conclusion: To the best of our knowledge, the relationship between CPT2 and tumor immune microenvironment was first proposed in this study. Therefore, further studies on CPT2 may provide new insights into the development of effective cancer immunotherapy.
Collapse
Affiliation(s)
- Xin Jiang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Wenqi Du
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Ce Shi
- Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Mengjie Kang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Qiuya Song
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Lansheng Zhang
- Department of Oncological Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongsheng Pei
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
30
|
Chen M, Chao B, Xu J, Liu Z, Tao Y, He J, Wang J, Yang H, Luo X, Qi H. CPT1A modulates PI3K/Akt/mTOR pathway to promote preeclampsia. Placenta 2023; 133:23-31. [PMID: 36702079 DOI: 10.1016/j.placenta.2023.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Preeclampsia (PE) refers to a syndrome of new-onset hypertension with multisystem involvement and damage after 20 weeks of gestation. Defective placentation due to dysregulated behaviors of trophoblast cells is considered a predominant cause of PE. METHODS Immunofluorescence (if) and Western blot were used to detect the expression and localization of Carnitine palmitoyltransferase 1A (CPT1A) in placenta. CPT1A protein was overexpressed/knocked down in HTR8/SVneo cells by lentiviral/siRNA interference method. CCK-8 Assay, Western blot, flow cytometry, Wound healing and Transwell assay were used to detect the functional impact of CPT1A on HTR8/SVneo cells. Transcriptomics and bioinformatics analysis were used to predict the possible pathway of CPT1A participating in PE. RESULTS CPT1A was upregulated in preeclamptic placentas when compared with normal controls. The abnormal expression of CPT1A in HTR8/SVneo cells is associated with the invasion and migration of HTR8/SVneo cells but is not related to the proliferation, cycle, and apoptosis of HTR8/SVneo cells. The results of Transcriptomic and Western blots suggest that phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway are activated in the si-CPT1A-1796 group. Compared with the si-NC group, the epithelial-mesenchymal transition (EMT) process of HTR8/SVneo cells in the si-CPT1A- 1796 group was significantly enhanced. DISCUSSION CPT1A may participate in the pathogenesis of PE by inhibiting the EMT process of HTR8/SVneo cells through the PI3K/AKT/mTOR signaling axis. Thus, the newly unveiled novel function of CPT1A in PE via the PI3K/Akt/mTOR pathway provides a novel insight into the pathogenesis of PE.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Affiliated Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Bingdi Chao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jiacheng Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zheng Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yuelan Tao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jie He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jie Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Huan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Xin Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Hongbo Qi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China; China-Canada-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China; Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, China.
| |
Collapse
|
31
|
Nwosu ZC, Song MG, di Magliano MP, Lyssiotis CA, Kim SE. Nutrient transporters: connecting cancer metabolism to therapeutic opportunities. Oncogene 2023; 42:711-724. [PMID: 36739364 PMCID: PMC10266237 DOI: 10.1038/s41388-023-02593-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 02/05/2023]
Abstract
Cancer cells rely on certain extracellular nutrients to sustain their metabolism and growth. Solute carrier (SLC) transporters enable cells to acquire extracellular nutrients or shuttle intracellular nutrients across organelles. However, the function of many SLC transporters in cancer is unknown. Determining the key SLC transporters promoting cancer growth could reveal important therapeutic opportunities. Here we summarize recent findings and knowledge gaps on SLC transporters in cancer. We highlight existing inhibitors for studying these transporters, clinical trials on treating cancer by blocking transporters, and compensatory transporters used by cancer cells to evade treatment. We propose targeting transporters simultaneously or in combination with targeted therapy or immunotherapy as alternative strategies for effective cancer therapy.
Collapse
Affiliation(s)
- Zeribe Chike Nwosu
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Mun Gu Song
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea
| | | | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, USA
| | - Sung Eun Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea.
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
32
|
Liang K. Mitochondrial CPT1A: Insights into structure, function, and basis for drug development. Front Pharmacol 2023; 14:1160440. [PMID: 37033619 PMCID: PMC10076611 DOI: 10.3389/fphar.2023.1160440] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Carnitine Palmitoyl-Transferase1A (CPT1A) is the rate-limiting enzyme in the fatty acid β-oxidation, and its deficiency or abnormal regulation can result in diseases like metabolic disorders and various cancers. Therefore, CPT1A is a desirable drug target for clinical therapy. The deep comprehension of human CPT1A is crucial for developing the therapeutic inhibitors like Etomoxir. CPT1A is an appealing druggable target for cancer therapies since it is essential for the survival, proliferation, and drug resistance of cancer cells. It will help to lower the risk of cancer recurrence and metastasis, reduce mortality, and offer prospective therapy options for clinical treatment if the effects of CPT1A on the lipid metabolism of cancer cells are inhibited. Targeted inhibition of CPT1A can be developed as an effective treatment strategy for cancers from a metabolic perspective. However, the pathogenic mechanism and recent progress of CPT1A in diseases have not been systematically summarized. Here we discuss the functions of CPT1A in health and diseases, and prospective therapies targeting CPT1A. This review summarizes the current knowledge of CPT1A, hoping to prompt further understanding of it, and provide foundation for CPT1A-targeting drug development.
Collapse
|
33
|
Hu T, Chen X, Lu S, Zeng H, Guo L, Han Y. Biological Role and Mechanism of Lipid Metabolism Reprogramming Related Gene ECHS1 in Cancer. Technol Cancer Res Treat 2022; 21:15330338221140655. [PMID: 36567598 PMCID: PMC9806408 DOI: 10.1177/15330338221140655] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cancer is a major threat to human health today. Although the existing anticancer treatments have effectively improved the prognosis of some patients, there are still other patients who cannot benefit from these well-established strategies. Reprogramming of lipid metabolism is one of the typical features of cancers. Recent studies have revealed that key enzymes involved in lipid metabolism may be effective anticancer therapeutic targets, but the development of therapeutic lipid metabolism targets is still insufficient. ECHS1 (enoyl-CoA hydratase, short chain 1) is a key enzyme mediating the hydration process of mitochondrial fatty acid β-oxidation and has been observed to be abnormally expressed in a variety of cancers. Therefore, with ECHS1 and cancer as the main keywords, we searched the relevant studies of ECHS1 in the field of cancer in Pubmed, summarized the research status and functions of ECHS1 in different cancer contexts, and explored its potential regulatory mechanisms, with a view to finding new therapeutic targets for anti-metabolic therapy. By reviewing and summarizing the retrieved literatures, we found that ECHS1 regulates malignant biological behaviors such as cell proliferation, metastasis, apoptosis, autophagy, and drug resistance by remodeling lipid metabolism and regulating intercellular oncogenic signaling pathways. Not only that, ECHS1 exhibits early diagnostic and prognostic value in clear cell renal cell carcinoma, and small-molecule inhibitors that regulate ECHS1 also show therapeutic significance in preclinical studies. Taken together, we propose that ECHS1 has the potential to serve as a therapeutic target of lipid metabolism.
Collapse
Affiliation(s)
- Teng Hu
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Xiaojing Chen
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Simin Lu
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Hao Zeng
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Lu Guo
- Department of Ophthalmology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China,Yunwei Han, Department of Oncology, The
Affiliated Hospital of Southwest Medical University, Taiping Street, No. 25,
Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
34
|
Yang Q, Bae G, Nadiradze G, Castagna A, Berezhnoy G, Zizmare L, Kulkarni A, Singh Y, Weinreich FJ, Kommoss S, Reymond MA, Trautwein C. Acidic ascites inhibits ovarian cancer cell proliferation and correlates with the metabolomic, lipidomic and inflammatory phenotype of human patients. J Transl Med 2022; 20:581. [PMID: 36503580 PMCID: PMC9743551 DOI: 10.1186/s12967-022-03763-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/05/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The poor prognosis of ovarian cancer patients is strongly related to peritoneal metastasis with the production of malignant ascites. However, it remains largely unclear how ascites in the peritoneal cavity influences tumor metabolism and recurrence. This study is an explorative approach aimed at for a deeper molecular and physical-chemical characterization of malignant ascites and to investigate their effect on in vitro ovarian cancer cell proliferation. METHODS This study included 10 malignant ascites specimens from patients undergoing ovarian cancer resection. Ascites samples were deeply phenotyped by 1H-NMR based metabolomics, blood-gas analyzer based gas flow analysis and flow cytomertry based a 13-plex cytokine panel. Characteristics of tumor cells were investigated in a 3D spheroid model by SEM and metabolic activity, adhesion, anti-apoptosis, migratory ability evaluated by MTT assay, adhesion assay, flowcytometry and scratch assay. The effect of different pH values was assessed by adding 10% malignant ascites to the test samples. RESULTS The overall extracellular (peritoneal) environment was alkaline, with pH of ascites at stage II-III = 7.51 ± 0.16, and stage IV = 7.78 ± 0.16. Ovarian cancer spheroids grew rapidly in a slightly alkaline environment. Decreasing pH of the cell culture medium suppressed tumor features, metabolic activity, adhesion, anti-apoptosis, and migratory ability. However, 10% ascites could prevent tumor cells from being affected by acidic pH. Metabolomics analysis identified stage IV patients had significantly higher concentrations of alanine, isoleucine, phenylalanine, and glutamine than stage II-III patients, while stage II-III patients had significantly higher concentrations of 3-hydroxybutyrate. pH was positively correlated with acetate, and acetate positively correlated with lipid compounds. IL-8 was positively correlated with lipid metabolites and acetate. Glutathione and carnitine were negatively correlated with cytokines IL-6 and chemokines (IL-8 & MCP-1). CONCLUSION Alkaline malignant ascites facilitated ovarian cancer progression. Additionally, deep ascites phenotyping by metabolomics and cytokine investigations allows for a refined stratification of ovarian cancer patients. These findings contribute to the understanding of ascites pathology in ovarian cancer.
Collapse
Affiliation(s)
- Qianlu Yang
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany
| | - Gyuntae Bae
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Giorgi Nadiradze
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany ,grid.411544.10000 0001 0196 8249Department of General and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Arianna Castagna
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany ,grid.411544.10000 0001 0196 8249Department of General and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Laimdota Zizmare
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Aditi Kulkarni
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Yogesh Singh
- grid.411544.10000 0001 0196 8249Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany ,grid.411544.10000 0001 0196 8249Research Institute of Women’s Health, Women’s Hospital, University Hospital Tübingen, Tübingen, Germany
| | - Frank J. Weinreich
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany
| | - Stefan Kommoss
- grid.411544.10000 0001 0196 8249Research Institute of Women’s Health, Women’s Hospital, University Hospital Tübingen, Tübingen, Germany
| | - Marc A. Reymond
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany ,grid.411544.10000 0001 0196 8249Department of General and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Trautwein
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
35
|
Park S, Mossmann D, Chen Q, Wang X, Dazert E, Colombi M, Schmidt A, Ryback B, Ng CKY, Terracciano LM, Heim MH, Hall MN. Transcription factors TEAD2 and E2A globally repress acetyl-CoA synthesis to promote tumorigenesis. Mol Cell 2022; 82:4246-4261.e11. [PMID: 36400009 DOI: 10.1016/j.molcel.2022.10.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/22/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022]
Abstract
Acetyl-coenzyme A (acetyl-CoA) plays an important role in metabolism, gene expression, signaling, and other cellular processes via transfer of its acetyl group to proteins and metabolites. However, the synthesis and usage of acetyl-CoA in disease states such as cancer are poorly characterized. Here, we investigated global acetyl-CoA synthesis and protein acetylation in a mouse model and patient samples of hepatocellular carcinoma (HCC). Unexpectedly, we found that acetyl-CoA levels are decreased in HCC due to transcriptional downregulation of all six acetyl-CoA biosynthesis pathways. This led to hypo-acetylation specifically of non-histone proteins, including many enzymes in metabolic pathways. Importantly, repression of acetyl-CoA synthesis promoted oncogenic dedifferentiation and proliferation. Mechanistically, acetyl-CoA synthesis was repressed by the transcription factors TEAD2 and E2A, previously unknown to control acetyl-CoA synthesis. Knockdown of TEAD2 and E2A restored acetyl-CoA levels and inhibited tumor growth. Our findings causally link transcriptional reprogramming of acetyl-CoA metabolism, dedifferentiation, and cancer.
Collapse
Affiliation(s)
- Sujin Park
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Dirk Mossmann
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Qian Chen
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland; Division of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, 4031 Basel, Switzerland
| | - Xueya Wang
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland; Division of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, 4031 Basel, Switzerland
| | - Eva Dazert
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Marco Colombi
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | | | - Brendan Ryback
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Charlotte K Y Ng
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Markus H Heim
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland; Division of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, 4031 Basel, Switzerland
| | - Michael N Hall
- Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
36
|
Gu L, He Q, Xia W, Rong G, Wang D, Li M, Ji F, Sun W, Cao T, Zhou H, Xu T. Integrated analysis of lncRNA and gene expression in longissimus dorsi muscle at two developmental stages of Hainan black goats. PLoS One 2022; 17:e0276004. [PMID: 36315512 PMCID: PMC9621442 DOI: 10.1371/journal.pone.0276004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022] Open
Abstract
It is deemed that meat quality of kids’ is better than that of adults’ for Hainan black goat. Generally, meat quality is affected by many indicators, such as intramuscular fat (IMF) content, muscle fiber diameter and shear force. It is indicated that long non-coding RNAs (lncRNAs) play essential roles in meat quality of goats. However, it is unclear whether and how lncRNAs and genes play their roles in meat quality of Hainan Black goats. Here, we firstly compared the meat quality between two-month-old kids (kids) and adult goats (adults). Then, the lncRNA-seq and RNA-seq data were integrated and analyzed to explore the potential functions of lncRNAs and genes. The results showed that adults’ IMF content and muscle fiber diameter were extremely significantly higher than that of kids (P<0.01). For the sequenced data, average 84,970,398, and 83,691,250 clean reads were obtained respectively for Kids and adults, among which ~96% were mapped to the reference genome of goats. Through analyzing, 18,242 goat annotated genes, 1,429 goat annotated lncRNAs and 2,967 novel lncRNAs were obtained. Analysis of differential expression genes (DEGs) and lncRNAs (DELs) showed that 328 DEGs and 98 DELs existed between kids and adults. Furthermore, functional enrichment analysis revealed that a number of DEGs and DELs were mainly associated with IMF. Primarily, DGAT2 expressed higher in adults than that in kids and CPT1A expressed higher in kids than that in adults. Both of them were overlapped by DEGs and targets of DELs, suggesting the two DEGs and the DELs targeted by the two DEGs might be the potential regulators of goat IMF deposition. Taken together, our results provide basic support for further understanding the function and mechanism of lncRNAs and genes in meat quality of Hainan black goats.
Collapse
Affiliation(s)
- Lihong Gu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Science, Haikou, China
| | - Qijie He
- Shengzhou Animal Husbandry Development Center, Shaoxing City, Zhejiang Province, China
| | - Wanliang Xia
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Guang Rong
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Dingfa Wang
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Mao Li
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Fengjie Ji
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Weiping Sun
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Ting Cao
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Hanlin Zhou
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- * E-mail: (TX); (HZ)
| | - Tieshan Xu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- * E-mail: (TX); (HZ)
| |
Collapse
|
37
|
Guryleva MV, Penzar DD, Chistyakov DV, Mironov AA, Favorov AV, Sergeeva MG. Investigation of the Role of PUFA Metabolism in Breast Cancer Using a Rank-Based Random Forest Algorithm. Cancers (Basel) 2022; 14:cancers14194663. [PMID: 36230586 PMCID: PMC9562210 DOI: 10.3390/cancers14194663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Polyunsaturated fatty acids (PUFAs) and their derivatives, oxylipins, are a constant focus of cancer research due to the relationship between cancer and processes of energy metabolism and inflammation, where a PUFA system is an active player. Only recently have methods been developed that allow for studying such complex systems. Using the Rank-based Random Forest (RF) model, we show that PUFA metabolism genes are critical for the pathogenesis of breast cancer (BC); BC subtypes differ in PUFA metabolism gene expression. The enrichment of BC subtypes with various genes associated with oxylipin signaling pathways indicates a different contribution of these compounds to the biology of subtypes. Abstract Polyunsaturated fatty acid (PUFA) metabolism is currently a focus in cancer research due to PUFAs functioning as structural components of the membrane matrix, as fuel sources for energy production, and as sources of secondary messengers, so called oxylipins, important players of inflammatory processes. Although breast cancer (BC) is the leading cause of cancer death among women worldwide, no systematic study of PUFA metabolism as a system of interrelated processes in this disease has been carried out. Here, we implemented a Boruta-based feature selection algorithm to determine the list of most important PUFA metabolism genes altered in breast cancer tissues compared with in normal tissues. A rank-based Random Forest (RF) model was built on the selected gene list (33 genes) and applied to predict the cancer phenotype to ascertain the PUFA genes involved in cancerogenesis. It showed high-performance of dichotomic classification (balanced accuracy of 0.94, ROC AUC 0.99) We also retrieved a list of the important PUFA genes (46 genes) that differed between molecular subtypes at the level of breast cancer molecular subtypes. The balanced accuracy of the classification model built on the specified genes was 0.82, while the ROC AUC for the sensitivity analysis was 0.85. Specific patterns of PUFA metabolic changes were obtained for each molecular subtype of breast cancer. These results show evidence that (1) PUFA metabolism genes are critical for the pathogenesis of breast cancer; (2) BC subtypes differ in PUFA metabolism genes expression; and (3) the lists of genes selected in the models are enriched with genes involved in the metabolism of signaling lipids.
Collapse
Affiliation(s)
- Mariia V. Guryleva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Dmitry D. Penzar
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Dmitry V. Chistyakov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Correspondence: ; Tel.: +7-495-939-4332
| | - Andrey A. Mironov
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Kharkevich Institute of Information Transmission Problems, Russian Academy of Sciences, 127051 Moscow, Russia
| | - Alexander V. Favorov
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- School of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Marina G. Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
38
|
Raeisi M, Zehtabi M, Velaei K, Fayyazpour P, Aghaei N, Mehdizadeh A. Anoikis in cancer: The role of lipid signaling. Cell Biol Int 2022; 46:1717-1728. [DOI: 10.1002/cbin.11896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/20/2022]
Affiliation(s)
- Mortaza Raeisi
- Hematology and Oncology Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Mojtaba Zehtabi
- Hematology and Oncology Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Kobra Velaei
- Department of Anatomical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Parisa Fayyazpour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| | - Negar Aghaei
- Department of Psycology, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
- Imam Sajjad Hospital Tabriz Azad University Tabriz Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
39
|
Decreased Levels of GSH Are Associated with Platinum Resistance in High-Grade Serous Ovarian Cancer. Antioxidants (Basel) 2022; 11:antiox11081544. [PMID: 36009263 PMCID: PMC9404763 DOI: 10.3390/antiox11081544] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 12/12/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is the most common and aggressive OC histotype. Although initially sensitive to standard platinum-based chemotherapy, most HGSOC patients relapse and become chemoresistant. We have previously demonstrated that platinum resistance is driven by a metabolic shift toward oxidative phosphorylation via activation of an inflammatory response, accompanied by reduced cholesterol biosynthesis and increased uptake of exogenous cholesterol. To better understand metabolic remodeling in OC, herein we performed an untargeted metabolomic analysis, which surprisingly showed decreased reduced glutathione (GSH) levels in resistant cells. Accordingly, we found reduced levels of enzymes involved in GSH synthesis and recycling, and compensatory increased expression of thioredoxin reductase. Cisplatin treatment caused an increase of reduced GSH, possibly due to direct binding hindering its oxidation, and consequent accumulation of reactive oxygen species. Notably, expression of the cysteine-glutamate antiporter xCT, which is crucial for GSH synthesis, directly correlates with post-progression survival of HGSOC patients, and is significantly reduced in patients not responding to platinum-based therapy. Overall, our data suggest that cisplatin treatment could positively select cancer cells which are independent from GSH for the maintenance of redox balance, and thus less sensitive to cisplatin-induced oxidative stress, opening new scenarios for the GSH pathway as a therapeutic target in HGSOC.
Collapse
|
40
|
Dong S, Li S, Wang X, Liang S, Zhang W, Li L, Xu Q, Shi B, Cheng Z, Zhang X, Zhong M, Zhang G, Hu S. CD147 Mediates 5-Fluorouracil Resistance in Colorectal Cancer by Reprogramming Glycolipid Metabolism. Front Oncol 2022; 12:813852. [PMID: 35898887 PMCID: PMC9309564 DOI: 10.3389/fonc.2022.813852] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Chemoresistance against 5-fluorouracil (5-FU) is a major issue for colorectal cancer (CRC) patients. Increasing evidence for the roles of CD147 in glycolipid metabolic reprogramming and chemoresistance of tumor cells has emerged in recent years. However, whether CD147 contributes to 5-FU resistance in CRC and the role of abnormal glycolipid metabolism in this process remain poorly understood. We analyzed CD147 expression in primary tumor samples of CRC patients and found that upregulated CD147 correlated with decreased 5-FU chemosensitivity and an unfavorable prognosis of CRC patients. Moreover, in vivo and in vitro experiments confirmed that CD147 regulates glycolipid metabolism through two separate pathways. Mechanistically, CD147 upregulates HIF-1α-mediated glycolysis by activating the PI3K/AKT/mTOR pathway and CD147 also attenuates PPARα-mediated fatty acid oxidation by activation of the MAPK pathway. Most importantly, we found that CD147 confers 5-FU resistance in CRC via these glycolipid metabolic signatures. Our results demonstrated that CD147 is a potential 5-FU resistance biomarker for CRC patients and a candidate therapeutic target to restore 5-FU sensitivity of 5-FU-resistant CRC by remodeling glycolipid metabolism.
Collapse
Affiliation(s)
- Shuohui Dong
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Songhan Li
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyan Wang
- Department of Neonatology, Weifang Yidu Central Hospital, Weifang, China
| | - Shuo Liang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Ear-nose-throat (ENT) Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjie Zhang
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Linchuan Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Qian Xu
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bowen Shi
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhiqiang Cheng
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mingwei Zhong
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Sanyuan Hu
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: SanYuan Hu,
| |
Collapse
|
41
|
Targeting lipid metabolism in the treatment of ovarian cancer. Oncotarget 2022; 13:768-783. [PMID: 35634242 PMCID: PMC9132258 DOI: 10.18632/oncotarget.28241] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 05/07/2022] [Indexed: 11/25/2022] Open
Abstract
Cancer cells undergo alterations in lipid metabolism to support their high energy needs, tumorigenesis and evade an anti-tumor immune response. Alterations in fatty acid production are controlled by multiple enzymes, chiefly Acetyl CoA Carboxylase, ATP-Citrate Lyase, Fatty Acid Synthase, and Stearoyl CoA Desaturase 1. Ovarian cancer (OC) is a common gynecological malignancy with a high rate of aggressive carcinoma progression and drug resistance. The accumulation of unsaturated fatty acids in ovarian cancer supports cell growth, increased cancer cell migration, and worse patient outcomes. Ovarian cancer cells also expand their lipid stores via increased uptake of lipids using fatty acid translocases, fatty acid-binding proteins, and low-density lipoprotein receptors. Furthermore, increased lipogenesis and lipid uptake promote chemotherapy resistance and dampen the adaptive immune response needed to eliminate tumors. In this review, we discuss the role of lipid synthesis and metabolism in driving tumorigenesis and drug resistance in ovarian cancer conferring poor prognosis and outcomes in patients. We also cover some aspects of how lipids fuel ovarian cancer stem cells, and how these metabolic alterations in intracellular lipid content could potentially serve as biomarkers of ovarian cancer.
Collapse
|
42
|
Khiewkamrop P, Surangkul D, Srikummool M, Richert L, Pekthong D, Parhira S, Somran J, Srisawang P. Epigallocatechin gallate triggers apoptosis by suppressing de novo lipogenesis in colorectal carcinoma cells. FEBS Open Bio 2022; 12:937-958. [PMID: 35243817 PMCID: PMC9063442 DOI: 10.1002/2211-5463.13391] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 12/18/2021] [Accepted: 03/02/2022] [Indexed: 11/11/2022] Open
Abstract
The de novo lipogenesis (DNL) pathway has been identified as a regulator of cancer progression and aggressiveness. Downregulation of key lipogenesis enzymes has been shown to activate apoptosis in cancerous cells. Epigallocatechin gallate (EGCG) inhibits cancer cell proliferation without causing cytotoxicity in healthy cells. The present study aimed to investigate the effects of EGCG on the promotion of apoptosis associated with the DNL pathway inhibition in cancer cells, both in vitro and in vivo. We observed that two colorectal cancer cell lines (HCT116 and HT-29) had a higher cytotoxic response to EGCG treatment than hepatocellular carcinoma cells, including HepG2 and HuH-7. EGCG treatment decreased cell viability and increased mitochondrial damage-triggered apoptosis in both HCT116 and HT-29 cancer cells. Additionally, we treated mice transplanted with HCT116 cells with 30 or 50 mg·kg-1 EGCG for 7 days to evaluate the apoptotic effects of EGCG treatment in a xenograft mouse model of cancer. We observed a decrease in intracellular fatty acid levels, which suggested that EGCG-induced apoptosis was associated with a decrease in fatty acid levels in cancer. Suppression of ATP synthesis by EGCG indicated that cell death induction in cancer cells could be mediated by shared components of the DNL and energy metabolism pathways. In addition, EGCG-induced apoptosis suppressed the expression of the phosphorylation protein kinase B and extracellular signal-regulated kinase 1/2 signaling proteins in tumors from xenografted mice. Cytotoxic effects in unaffected organs and tissues of the mouse xenograft model were absent upon EGCG treatment.
Collapse
Affiliation(s)
- Phuriwat Khiewkamrop
- Department of PhysiologyFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| | - Damratsamon Surangkul
- Department of BiochemistryFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| | - Metawee Srikummool
- Department of BiochemistryFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| | - Lysiane Richert
- KaLy‐CellPlobsheimFrance
- EA 4267 PEPITEUniversité de Bourgogne Franche‐ComtéBesançonFrance
| | - Dumrongsak Pekthong
- Department of Pharmacy PracticeFaculty of Pharmaceutical SciencesNaresuan UniversityPhitsanulokThailand
| | - Supawadee Parhira
- Department of Pharmaceutical TechnologyFaculty of Pharmaceutical SciencesNaresuan UniversityPhitsanulokThailand
| | - Julintorn Somran
- Department of PathologyFaculty of MedicineNaresuan UniversityPhitsanulokThailand
| | - Piyarat Srisawang
- Department of PhysiologyFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| |
Collapse
|
43
|
S SK, Swamy SN, Devaraj VR, Premalatha CS, Pallavi VR, Chandrashekar Sagar BK, Shinde DD, Gawari R. Metabolic Reprogramming and Lipophagy Mediates Survival of Ascites Derived Metastatic Ovarian Cancer Cells. Asian Pac J Cancer Prev 2022; 23:1699-1709. [PMID: 35633555 PMCID: PMC9587889 DOI: 10.31557/apjcp.2022.23.5.1699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/09/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE The study was aimed at understanding the survival of metastatic ovarian cancer spheroids in the malignant ascites microenvironment. METHODS All the assays were performed using aseptically collected patient samples. The cells were characterized for the expression of ovarian and cancer stem cell markers using immunocytochemistry. The presence of lipid in the primary metastatic cancer spheroids were confirmed by neutral fat staining using Oil Red-O and transmission electron microscopy. The mRNA expression of autophagy and lipid metabolism genes was analyzed using RT-PCR. The lipid content was analyzed using lipidomics analysis. Etomoxir and chloroquine were used to study the effect of inhibition of autophagy in the metastatic cells. The data were analyzed using appropriate statistical tools and a p-value <0.05 was considered to be statistically significant. RESULTS Metastatic ovarian cancer spheroids exhibit cancer stem like properties and undergo a metabolic reprogramming when they disseminate from the primary tumor. We report here the accumulation of numerous cytoplasmic lipid droplets and lipophagic vesicles in the metastatic cells in contrast to their primary tumors. In addition we also report that these cells depend on lipophagy for the utilization of lipids rather than the conventional lipolytic pathway. The lipidomics analysis data reveals that the metastatic cells possess high levels of unsaturated fatty acids. We have also reported the occurrence of distinct accumulation of multiple nuclei in the patient derived metastatic cells. Inhibition of beta-oxidation and autophagic machinery using etomoxir and chloroquine resulted in cell death suggesting a potential mode to suppress metastatic cancer cells. CONCLUSION Metabolic reprogramming is a characteristic feature of the metastatic ovarian cancer cells that are persisting in the malignant ascites. Targeting of the metastatic by gaining an insight into the various metabolic and molecular changes that occur in the metastatic niche provides a promising therapeutic approach in management of the disease.
Collapse
Affiliation(s)
- Sandeep Kumar S
- Department of Biochemistry, Kidwai Memorial Institute of Oncology Dr.M.H.Marigowda Road Bangalore, India.
| | - Shalini N Swamy
- Department of Biochemistry, Kidwai Memorial Institute of Oncology Dr.M.H.Marigowda Road Bangalore, India.
| | | | - Chennagiri S Premalatha
- Department of Pathology, Kidwai Memorial Institute of Oncology Dr.M.H.Marigowda raod Bangalore India.
| | - V R Pallavi
- Department of Gynecologic Oncology, Kidwai Memorial Institute of Oncology, Bangalore, India.
| | - B K Chandrashekar Sagar
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bangalore, India.
| | - Dhananjay D Shinde
- Department of Pathology and Microbiology, University of Nebraska Medical Centre, Omaha, NE, USA.
| | - Ramesh Gawari
- Department of Biochemistry, Kidwai Memorial Institute of Oncology Dr.M.H.Marigowda Road Bangalore, India.
| |
Collapse
|
44
|
Liu HX, Liu QJ. Logistic role of carnitine shuttle system on radiation-induced L-carnitine and acylcarnitines alteration. Int J Radiat Biol 2022; 98:1595-1608. [PMID: 35384773 DOI: 10.1080/09553002.2022.2063430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE With the development of radiation metabolomics, a large number of radiation-related metabolic biomarkers have been identified and validated. The L-carnitine and acylcarnitines have the potential to be the new promising candidate indicators of radiation exposure. This review summarizes the effect of carnitine shuttle system on the profile of acylcarnitines and correlates the radiation effects on upstream regulators of carnitine shuttle system with the change characteristics of L-carnitine and acylcarnitines after irradiation across different animal models as well as a few humans. CONCLUSIONS Studies report that acylcarnitines were ubiquitously elevated after irradiation, especially the free L-carnitine and short-chain acylcarnitines (C2-C5). However, the molecular mechanism underlying acylcarnitine alterations after irradiation is not fully investigated, and further studies are needed to explore the biological effect and its mechanism. The activity of the carnitine shuttle system plays a key role in the alteration of L-carnitine and acylcarnitines, and the upstream regulators of the system are known to be affected by irradiation. These evidences indicate that that there is a logistic role of carnitine shuttle system on radiation-induced L-carnitine and acylcarnitines alteration.
Collapse
Affiliation(s)
- Hai-Xiang Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Qing-Jie Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| |
Collapse
|
45
|
Mozihim AK, Chung I, Said NABM, Jamil AHA. Reprogramming of Fatty Acid Metabolism in Gynaecological Cancers: Is There a Role for Oestradiol? Metabolites 2022; 12:metabo12040350. [PMID: 35448537 PMCID: PMC9031151 DOI: 10.3390/metabo12040350] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Gynaecological cancers are among the leading causes of cancer-related death among women worldwide. Cancer cells undergo metabolic reprogramming to sustain the production of energy and macromolecules required for cell growth, division and survival. Emerging evidence has provided significant insights into the integral role of fatty acids on tumourigenesis, but the metabolic role of high endogenous oestrogen levels and increased gynaecological cancer risks, notably in obesity, is less understood. This is becoming a renewed research interest, given the recently established association between obesity and incidence of many gynaecological cancers, including breast, ovarian, cervical and endometrial cancers. This review article, hence, comprehensively discusses how FA metabolism is altered in these gynaecological cancers, highlighting the emerging role of oestradiol on the actions of key regulatory enzymes of lipid metabolism, either directly through its classical ER pathways, or indirectly via the IGIFR pathway. Given the dramatic rise in obesity and parallel increase in the prevalence of gynaecological cancers among premenopausal women, further clarifications of the complex mechanisms underpinning gynaecological cancers are needed to inform future prevention efforts. Hence, in our review, we also highlight opportunities where metabolic dependencies can be exploited as viable therapeutic targets for these hormone-responsive cancers.
Collapse
Affiliation(s)
- Azilleo Kristo Mozihim
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Nur Akmarina B. M. Said
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
| | - Amira Hajirah Abd Jamil
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
- Correspondence: ; Tel.: +60-3-7967-4909
| |
Collapse
|
46
|
Lu J, Zhang Y, Sun M, Ding C, Zhang L, Kong Y, Cai M, Miccoli P, Ma C, Yue X. Multi-Omics Analysis of Fatty Acid Metabolism in Thyroid Carcinoma. Front Oncol 2022; 11:737127. [PMID: 34976793 PMCID: PMC8717782 DOI: 10.3389/fonc.2021.737127] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/24/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Papillary thyroid carcinoma (PTC) accounts for the majority of thyroid cancer and affects a large number of individuals. The pathogenesis of PTC has not been completely elucidated thus far. Metabolic reprogramming is a common feature in tumours. Our previous research revealed the reprogramming of lipid metabolism in PTC. Further studies on lipid metabolism reprogramming may help elucidate the pathogenesis of PTC. Methods Clinical samples of PTC and para-tumour tissue were analysed using lipidomic, proteomic, and metabolomic approaches. A multi-omics integrative strategy was adopted to identify the important pathways in PTC. The findings were further confirmed using western blotting, tissue microarray, bioinformatics, and cell migration assays. Results Multi-omics data and the results of integrated analysis revealed that the three steps of fatty acid metabolism (hydrolysis, transportation, and oxidation) were significantly enhanced in PTC. Especially, the expression levels of LPL, FATP2, and CPT1A, three key enzymes in the respective steps, were elevated in PTC. Moreover, LPL, FATP2 and CPT1A expression was associated with the TNM stage, lymph node metastasis of PTC. Moreover, high levels of FATP2 and CPT1A contributed to poor prognosis of PTC. In addition, ectopic overexpression of LPL, FATP2 and CPT1A can each promote the migration of thyroid cancer cells. Conclusions Our data suggested that enhanced fatty acid metabolism supplied additional energy and substrates for PTC progression. This may help elucidating the underlying mechanism of PTC pathogenesis and identifying the potential therapeutic targets for PTC.
Collapse
Affiliation(s)
- Jinghui Lu
- Department of Hernia and Abdominal Wall Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yankun Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Sun
- Department of Hernia and Abdominal Wall Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Laboratory of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changyuan Ding
- Department of Thyroid Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Zhang
- Department of Obstetrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, China
| | - Youzi Kong
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Cell Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Cai
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Paolo Miccoli
- Department of Surgery, University of Pisa, Pisa, Italy
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuetian Yue
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Cell Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
47
|
Wang W, Ma Y, He T, Mooney E, Guo C, Wang XY, Fang X. Histopathological Diagnosis of Nonalcoholic Steatohepatitis (NASH). Methods Mol Biol 2022; 2455:49-62. [PMID: 35212985 DOI: 10.1007/978-1-0716-2128-8_5] [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] [Indexed: 06/14/2023]
Abstract
Fatty acid beta oxidation (FAO) is a predominant bioenergetic pathway in mammals. Substantial investigations have demonstrated that FAO activity is dysregulated in many pathophysiological conditions including nonalcoholic steatohepatitis (NASH). Convenient and quantitative assays of FAO activities are important for studies of cell metabolism and the biological relevance of FAO to health and diseases. However, most current FAO assays are based on non-physiological culture conditions, measure FAO activity indirectly or lack adequate quantification. We herein describe details of practical protocols for measurement of basal and genetically or pharmacologically regulated FAO activities in the mammalian system. We also discuss the advantages and disadvantages of these assays in the context of experimental purposes.
Collapse
Affiliation(s)
- Wei Wang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Yibao Ma
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Alliance Pharma Inc, Malvern, PA, USA
| | - Tianhai He
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Erin Mooney
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Xianjun Fang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
48
|
Downregulation of CPT2 promotes proliferation and inhibits apoptosis through p53 pathway in colorectal cancer. Cell Signal 2022; 92:110267. [DOI: 10.1016/j.cellsig.2022.110267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023]
|
49
|
Huang D, Chowdhury S, Wang H, Savage SR, Ivey RG, Kennedy JJ, Whiteaker JR, Lin C, Hou X, Oberg AL, Larson MC, Eskandari N, Delisi DA, Gentile S, Huntoon CJ, Voytovich UJ, Shire ZJ, Yu Q, Gygi SP, Hoofnagle AN, Herbert ZT, Lorentzen TD, Calinawan A, Karnitz LM, Weroha SJ, Kaufmann SH, Zhang B, Wang P, Birrer MJ, Paulovich AG. Multiomic analysis identifies CPT1A as a potential therapeutic target in platinum-refractory, high-grade serous ovarian cancer. Cell Rep Med 2021; 2:100471. [PMID: 35028612 PMCID: PMC8714940 DOI: 10.1016/j.xcrm.2021.100471] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 09/24/2021] [Accepted: 11/19/2021] [Indexed: 12/14/2022]
Abstract
Resistance to platinum compounds is a major determinant of patient survival in high-grade serous ovarian cancer (HGSOC). To understand mechanisms of platinum resistance and identify potential therapeutic targets in resistant HGSOC, we generated a data resource composed of dynamic (±carboplatin) protein, post-translational modification, and RNA sequencing (RNA-seq) profiles from intra-patient cell line pairs derived from 3 HGSOC patients before and after acquiring platinum resistance. These profiles reveal extensive responses to carboplatin that differ between sensitive and resistant cells. Higher fatty acid oxidation (FAO) pathway expression is associated with platinum resistance, and both pharmacologic inhibition and CRISPR knockout of carnitine palmitoyltransferase 1A (CPT1A), which represents a rate limiting step of FAO, sensitize HGSOC cells to platinum. The results are further validated in patient-derived xenograft models, indicating that CPT1A is a candidate therapeutic target to overcome platinum resistance. All multiomic data can be queried via an intuitive gene-query user interface (https://sites.google.com/view/ptrc-cell-line).
Collapse
Affiliation(s)
- Dongqing Huang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Shrabanti Chowdhury
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hong Wang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Sara R Savage
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Richard G Ivey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jacob J Kennedy
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jeffrey R Whiteaker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Chenwei Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Xiaonan Hou
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Melissa C Larson
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN 55905, USA
| | - Najmeh Eskandari
- Division of Hematology and Oncology, Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Davide A Delisi
- Division of Hematology and Oncology, Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Saverio Gentile
- Division of Hematology and Oncology, Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | | | - Uliana J Voytovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Zahra J Shire
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Qing Yu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew N Hoofnagle
- Department of Lab Medicine, University of Washington, Seattle, WA 98195, USA
| | - Zachary T Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Travis D Lorentzen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Anna Calinawan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Larry M Karnitz
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - S John Weroha
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael J Birrer
- University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Amanda G Paulovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
50
|
Wu T, Luo G, Lian Q, Sui C, Tang J, Zhu Y, Zheng B, Li Z, Zhang Y, Zhang Y, Bao J, Hu J, Shen S, Yang Z, Wu J, Wang K, Zhao Y, Yang S, Wang S, Qiu X, Wang W, Wu X, Wang H, Gu J, Chen L. Discovery of a Carbamoyl Phosphate Synthetase 1-Deficient HCC Subtype With Therapeutic Potential Through Integrative Genomic and Experimental Analysis. Hepatology 2021; 74:3249-3268. [PMID: 34343359 DOI: 10.1002/hep.32088] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/06/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Metabolic reprogramming plays an important role in tumorigenesis. However, the metabolic types of different tumors are diverse and lack in-depth study. Here, through analysis of big databases and clinical samples, we identified a carbamoyl phosphate synthetase 1 (CPS1)-deficient hepatocellular carcinoma (HCC) subtype, explored tumorigenesis mechanism of this HCC subtype, and aimed to investigate metabolic reprogramming as a target for HCC prevention. APPROACH AND RESULTS A pan-cancer study involving differentially expressed metabolic genes of 7,764 tumor samples in 16 cancer types provided by The Cancer Genome Atlas (TCGA) demonstrated that urea cycle (UC) was liver-specific and was down-regulated in HCC. A large-scale gene expression data analysis including 2,596 HCC cases in 7 HCC cohorts from Database of HCC Expression Atlas and 17,444 HCC cases from in-house hepatectomy cohort identified a specific CPS1-deficent HCC subtype with poor clinical prognosis. In vitro and in vivo validation confirmed the crucial role of CPS1 in HCC. Liquid chromatography-mass spectrometry assay and Seahorse analysis revealed that UC disorder (UCD) led to the deceleration of the tricarboxylic acid cycle, whereas excess ammonia caused by CPS1 deficiency activated fatty acid oxidation (FAO) through phosphorylated adenosine monophosphate-activated protein kinase. Mechanistically, FAO provided sufficient ATP for cell proliferation and enhanced chemoresistance of HCC cells by activating forkhead box protein M1. Subcutaneous xenograft tumor models and patient-derived organoids were employed to identify that blocking FAO by etomoxir may provide therapeutic benefit to HCC patients with CPS1 deficiency. CONCLUSIONS In conclusion, our results prove a direct link between UCD and cancer stemness in HCC, define a CPS1-deficient HCC subtype through big-data mining, and provide insights for therapeutics for this type of HCC through targeting FAO.
Collapse
Affiliation(s)
- Tong Wu
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Guijuan Luo
- Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Qiuyu Lian
- UM-SJTU Joint Institute, Shanghai Jiao Tong University, Shanghai, China.,MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, China
| | - Chengjun Sui
- Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Jing Tang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanjing Zhu
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Bo Zheng
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Zhixuan Li
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yani Zhang
- Institute of Metabolism and Integrative Biology and School of Life Sciences, Fudan University, Shanghai, China
| | - Yangqianwen Zhang
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jinxia Bao
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Ji Hu
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Siyun Shen
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Zhao Yang
- Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Jianmin Wu
- Institute of Metabolism and Integrative Biology and School of Life Sciences, Fudan University, Shanghai, China
| | - Kaiting Wang
- Institute of Metabolism and Integrative Biology and School of Life Sciences, Fudan University, Shanghai, China
| | - Yan Zhao
- Institute of Metabolism and Integrative Biology and School of Life Sciences, Fudan University, Shanghai, China
| | - Shuai Yang
- Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shan Wang
- Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyao Qiu
- Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenwen Wang
- Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuan Wu
- Department of Laboratory Medicine, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Hongyang Wang
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China.,Eastern Hepatobiliary Surgery Hospital, Shanghai, China.,National Center for Liver Cancer, Shanghai, China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, China
| | - Lei Chen
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China.,Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Liver Cancer, Shanghai, China
| |
Collapse
|