1
|
Korbecki J, Bosiacki M, Pilarczyk M, Gąssowska-Dobrowolska M, Jarmużek P, Szućko-Kociuba I, Kulik-Sajewicz J, Chlubek D, Baranowska-Bosiacka I. Phospholipid Acyltransferases: Characterization and Involvement of the Enzymes in Metabolic and Cancer Diseases. Cancers (Basel) 2024; 16:2115. [PMID: 38893234 PMCID: PMC11171337 DOI: 10.3390/cancers16112115] [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: 04/15/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
This review delves into the enzymatic processes governing the initial stages of glycerophospholipid (phosphatidylcholine, phosphatidylethanolamine, and phosphatidylserine) and triacylglycerol synthesis. The key enzymes under scrutiny include GPAT and AGPAT. Additionally, as most AGPATs exhibit LPLAT activity, enzymes participating in the Lands cycle with similar functions are also covered. The review begins by discussing the properties of these enzymes, emphasizing their specificity in enzymatic reactions, notably the incorporation of polyunsaturated fatty acids (PUFAs) such as arachidonic acid and docosahexaenoic acid (DHA) into phospholipids. The paper sheds light on the intricate involvement of these enzymes in various diseases, including obesity, insulin resistance, and cancer. To underscore the relevance of these enzymes in cancer processes, a bioinformatics analysis was conducted. The expression levels of the described enzymes were correlated with the overall survival of patients across 33 different types of cancer using the GEPIA portal. This review further explores the potential therapeutic implications of inhibiting these enzymes in the treatment of metabolic diseases and cancer. By elucidating the intricate enzymatic pathways involved in lipid synthesis and their impact on various pathological conditions, this paper contributes to a comprehensive understanding of these processes and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland;
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (D.C.)
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (D.C.)
| | - Maciej Pilarczyk
- Department of Nervous System Diseases, Neurosurgery Center University Hospital in Zielona Góra, Collegium Medicum, University of Zielona Gora, 65-417 Zielona Góra, Poland; (M.P.); (P.J.)
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland;
| | - Paweł Jarmużek
- Department of Nervous System Diseases, Neurosurgery Center University Hospital in Zielona Góra, Collegium Medicum, University of Zielona Gora, 65-417 Zielona Góra, Poland; (M.P.); (P.J.)
| | | | - Justyna Kulik-Sajewicz
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (D.C.)
| |
Collapse
|
2
|
Ding N, Wang W, Teng J, Zeng Y, Zhang Q, Dong L, Tang H. miR-26a-5p Regulates Adipocyte Differentiation via Directly Targeting ACSL3 in Adipocytes. Adipocyte 2023; 12:1-10. [PMID: 36710425 PMCID: PMC9891161 DOI: 10.1080/21623945.2023.2166345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Preadipocytes become mature adipocytes after proliferation and differentiation, and although many genes and microRNAs have been identified in intramuscular fat, their physiological function and regulatory mechanisms remain largely unexplored. miR-26a-5p has been reported to be related to fat deposition, but its effect on porcine preadipocyte differentiation has not been explored. In this study, bioinformatics analysis and luciferase reporter assay identified that miR-26a-5p binds to the 3'UTR of Acyl-CoA synthetase long-chain family member 3 (ACSL3) mRNA. The model for porcine intramuscular preadipocyte differentiation was established to explore the function of miR-6a-5p-ACSL3 on adipocyte differentiation. ACSL3 knockdown markedly reduced the triglycerides (TG) content of cells, as well as the mRNA levels of adipogenic marker genes (PPAR-γ and SREBP-1c). The number of lipid droplets in cells transfected with a miR-26a-5p mimic is significantly reduced, consistent with ACSL3 knockdown results, while the miR-26a-5p inhibitor resulted in opposite results. Taken together, miR-26a-5p is a repressor of porcine preadipocyte differentiation and plays a vital role in ACSL3-mediated adipogenesis.
Collapse
Affiliation(s)
- Ning Ding
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Wenwen Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Jun Teng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Yongqing Zeng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Qin Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Licai Dong
- Shandong Futong Agriculture & Animal Husbandry Development Co. LTD, Linyi, China
| | - Hui Tang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China,CONTACT Hui Tang No. 61, Daizong Street, Tai’an City, Shandong Province, 271018, China
| |
Collapse
|
3
|
Uehara K, Santoleri D, Whitlock AEG, Titchenell PM. Insulin Regulation of Hepatic Lipid Homeostasis. Compr Physiol 2023; 13:4785-4809. [PMID: 37358513 PMCID: PMC10760932 DOI: 10.1002/cphy.c220015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
The incidence of obesity, insulin resistance, and type II diabetes (T2DM) continues to rise worldwide. The liver is a central insulin-responsive metabolic organ that governs whole-body metabolic homeostasis. Therefore, defining the mechanisms underlying insulin action in the liver is essential to our understanding of the pathogenesis of insulin resistance. During periods of fasting, the liver catabolizes fatty acids and stored glycogen to meet the metabolic demands of the body. In postprandial conditions, insulin signals to the liver to store excess nutrients into triglycerides, cholesterol, and glycogen. In insulin-resistant states, such as T2DM, hepatic insulin signaling continues to promote lipid synthesis but fails to suppress glucose production, leading to hypertriglyceridemia and hyperglycemia. Insulin resistance is associated with the development of metabolic disorders such as cardiovascular and kidney disease, atherosclerosis, stroke, and cancer. Of note, nonalcoholic fatty liver disease (NAFLD), a spectrum of diseases encompassing fatty liver, inflammation, fibrosis, and cirrhosis, is linked to abnormalities in insulin-mediated lipid metabolism. Therefore, understanding the role of insulin signaling under normal and pathologic states may provide insights into preventative and therapeutic opportunities for the treatment of metabolic diseases. Here, we provide a review of the field of hepatic insulin signaling and lipid regulation, including providing historical context, detailed molecular mechanisms, and address gaps in our understanding of hepatic lipid regulation and the derangements under insulin-resistant conditions. © 2023 American Physiological Society. Compr Physiol 13:4785-4809, 2023.
Collapse
Affiliation(s)
- Kahealani Uehara
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dominic Santoleri
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna E. Garcia Whitlock
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul M. Titchenell
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Farese RV, Walther TC. Glycerolipid Synthesis and Lipid Droplet Formation in the Endoplasmic Reticulum. Cold Spring Harb Perspect Biol 2023; 15:a041246. [PMID: 36096640 PMCID: PMC10153804 DOI: 10.1101/cshperspect.a041246] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
More than 60 years ago, Eugene Kennedy and coworkers elucidated the endoplasmic reticulum (ER)-based pathways of glycerolipid synthesis, including the synthesis of phospholipids and triacylglycerols (TGs). The reactions of the Kennedy pathway were identified by studying the conversion of lipid intermediates and the isolation of biochemical enzymatic activities, but the molecular basis for most of these reactions was unknown. With recent progress in the cell biology, biochemistry, and structural biology in this area, we have a much more mechanistic understanding of this pathway and its reactions. In this review, we provide an overview of molecular aspects of glycerolipid synthesis, focusing on recent insights into the synthesis of TGs. Further, we go beyond the Kennedy pathway to describe the mechanisms for storage of TG in cytosolic lipid droplets and discuss how overwhelming these pathways leads to ER stress and cellular toxicity, as seen in diseases linked to lipid overload and obesity.
Collapse
Affiliation(s)
- Robert V Farese
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Center for Causes and Prevention of Cardiovascular Disease (CAP-CVD), Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Center for Causes and Prevention of Cardiovascular Disease (CAP-CVD), Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute Boston, Boston, Massachusetts 02115, USA
| |
Collapse
|
5
|
Fujii J, Yamada KI. Defense systems to avoid ferroptosis caused by lipid peroxidation-mediated membrane damage. Free Radic Res 2023; 57:353-372. [PMID: 37551716 DOI: 10.1080/10715762.2023.2244155] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
The presence of hydrogen peroxide along with ferrous iron produces hydroxyl radicals that preferably oxidize polyunsaturated fatty acids (PUFA) to alkyl radicals (L•). The reaction of L• with an oxygen molecule produces lipid peroxyl radical (LOO•) that collectively trigger chain reactions, which results in the accumulation of lipid peroxidation products (LOOH). Oxygenase enzymes, such as lipoxygenase, also stimulate the peroxidation of PUFA. The production of phospholipid hydroperoxides (P-LOOH) can result in the destruction of the architecture of cell membranes and ultimate cell death. This iron-dependent regulated cell death is generally referred to as ferroptosis. Radical scavengers, which include tocopherol and nitric oxide (•NO), react with lipid radicals and terminate the chain reaction. When tocopherol reductively detoxifies lipid radicals, the resultant tocopherol radicals are recycled via reduction by coenzyme Q or ascorbate. CoQ radicals are reduced back by the anti-ferroptotic enzyme FSP1. •NO reacts with lipid radicals and produces less reactive nitroso compounds. The resulting P-LOOH is reductively detoxified by the action of glutathione peroxidase 4 (GPX4) or peroxiredoxin 6 (PRDX6). The hydrolytic removal of LOOH from P-LOOH by calcium-independent phospholipase A2 leads the preservation of membrane structure. While the expression of such protective genes or the presence of these anti-oxidant compounds serve to maintain a healthy condition, tumor cells employ them to make themselves resistant to anti-tumor treatments. Thus, these defense mechanisms against ferroptosis are protective in ordinary cells but are also potential targets for cancer treatment.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Ken-Ichi Yamada
- Faculty of Pharmaceutical Sciences, Physical Chemistry for Life Science Laboratory, Kyushu University, Fukuoka, Japan
| |
Collapse
|
6
|
Xu J, Zhou F, Wang X, Mo C. Role of ferroptosis in pregnancy related diseases and its therapeutic potential. Front Cell Dev Biol 2023; 11:1083838. [PMID: 36968201 PMCID: PMC10031498 DOI: 10.3389/fcell.2023.1083838] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Ferroptosis is a form of regulated cell death characterized by iron overload, overwhelming lipid peroxidation, and disruption of antioxidant systems. Emerging evidence suggests that ferroptosis is associated with pregnancy related diseases, such as spontaneous abortion, pre-eclampsia, gestational diabetes mellitus, intrahepatic cholestasis of pregnancy, and spontaneous preterm birth. According to these findings, inhibiting ferroptosis might be a potential option to treat pregnancy related diseases. This review summarizes the mechanisms and advances of ferroptosis, the pathogenic role of ferroptosis in pregnancy related diseases and the potential medicines for its treatment.
Collapse
Affiliation(s)
- Jinfeng Xu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Fan Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaodong Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Chunheng Mo, ; Xiaodong Wang,
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Chunheng Mo, ; Xiaodong Wang,
| |
Collapse
|
7
|
Wu Z, Sun J, Liao Z, Qiao J, Chen C, Ling C, Wang H. An update on the therapeutic implications of long-chain acyl-coenzyme A synthetases in nervous system diseases. Front Neurosci 2022; 16:1030512. [PMID: 36507355 PMCID: PMC9731139 DOI: 10.3389/fnins.2022.1030512] [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: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Long-chain acyl-coenzyme A synthetases (ACSLs) are a family of CoA synthetases that activate fatty acid (FA) with chain lengths of 12-20 carbon atoms by forming the acyl-AMP derivative in an isozyme-specific manner. This family mainly includes five members (ACSL1, ACSL3, ACSL4, ACSL5, and ACSL6), which are thought to have specific and different functions in FA metabolism and oxidative stress of mammals. Accumulating evidence shows that the dysfunction of ACSLs is likely to affect cell proliferation and lead to metabolic diseases in multiple organs and systems through different signaling pathways and molecular mechanisms. Hence, a central theme of this review is to emphasize the therapeutic implications of ACSLs in nervous system disorders.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi Liao
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia Qiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China,*Correspondence: Hui Wang,
| |
Collapse
|
8
|
Marteau R, Ravez S, Mazhari Dorooee D, Bouchaoui H, Porte K, Devedjian JC, Melnyk P, Devos D, Frédérick R, El Bakali J. Repositioning of FDA-Approved antifungal agents to interrogate Acyl-CoA synthetase long chain family member 4 (ACSL4) in ferroptosis. Biochem Pharmacol 2022; 204:115239. [PMID: 36075462 DOI: 10.1016/j.bcp.2022.115239] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 01/06/2023]
Abstract
Ferroptosis, first coined in 2012, is an iron-dependent regulated cell death (RCD) characterized by the accumulation of lipid peroxides to toxic levels. This mechanism is currently being evaluated as a target for a variety of diseases offering new opportunities for drug design and development. Recent reports uncovered acyl-CoA synthetase long-chain 4 (ACSL4) as a critical contributor to ferroptosis execution. Therefore, ACSL4 inhibitors are emerging as attractive anti-ferroptotic agents. Herein, we developed a robust screening cascade with orthogonal biophysical and biochemical techniques to identify original human ACSL4 inhibitors. By screening an FDA-approved drug library, we were able to identify and validate new inhibitors with micromolar-range activities against ACSL4. With an IC50 of 280 nM against hACSL4, antifungal agent sertaconazole is to our knowledge, the most potent ACSL4 inhibitor identified so far. In addition, sertaconazole significantly reduced lipid peroxidation and ferroptosis in human differentiated dopaminergic neurons (Lund human mesencephalic LUHMES cells), demonstrating that it is a valuable chemical tool for further investigating the role of ACSL4 in ferroptosis. This study highlights the phenethyl-imidazole scaffold as a novel and promising starting point for the development of anti-ferroptotic agents targeting ACSL4.
Collapse
Affiliation(s)
- Romain Marteau
- Medicinal Chemistry Research Group (CMFA), Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCLouvain), 73 Avenue Mounier, B1.73.10, 1200 Bruxelles, Belgium
| | - Séverine Ravez
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Darius Mazhari Dorooee
- Medicinal Chemistry Research Group (CMFA), Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCLouvain), 73 Avenue Mounier, B1.73.10, 1200 Bruxelles, Belgium
| | - Hind Bouchaoui
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Karine Porte
- Medicinal Chemistry Research Group (CMFA), Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCLouvain), 73 Avenue Mounier, B1.73.10, 1200 Bruxelles, Belgium
| | - Jean-Christophe Devedjian
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France; Université du Littoral Côte d'Opale-1, place de l'Yser, BP 72033, 59375 Dunkerque Cedex, France
| | - Patricia Melnyk
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - David Devos
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Raphaël Frédérick
- Medicinal Chemistry Research Group (CMFA), Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCLouvain), 73 Avenue Mounier, B1.73.10, 1200 Bruxelles, Belgium.
| | - Jamal El Bakali
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France.
| |
Collapse
|
9
|
Targeting the Sphingolipid Rheostat in Gliomas. Int J Mol Sci 2022; 23:ijms23169255. [PMID: 36012521 PMCID: PMC9408832 DOI: 10.3390/ijms23169255] [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: 07/30/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/26/2022] Open
Abstract
Gliomas are highly aggressive cancer types that are in urgent need of novel drugs and targeted therapies. Treatment protocols have not improved in over a decade, and glioma patient survival remains among the worst of all cancer types. As a result, cancer metabolism research has served as an innovative approach to identifying novel glioma targets and improving our understanding of brain tumors. Recent research has uncovered a unique metabolic vulnerability in the sphingolipid pathways of gliomas that possess the IDH1 mutation. Sphingolipids are a family of lipid signaling molecules that play a variety of second messenger functions in cellular regulation. The two primary metabolites, sphingosine-1-phosphate (S1P) and ceramide, maintain a rheostat balance and play opposing roles in cell survival and proliferation. Altering the rheostat such that the pro-apoptotic signaling of the ceramides outweighs the pro-survival S1P signaling in glioma cells diminishes the hallmarks of cancer and enhances tumor cell death. Throughout this review, we discuss the sphingolipid pathway and identify the enzymes that can be most effectively targeted to alter the sphingolipid rheostat and enhance apoptosis in gliomas. We discuss each pathway’s steps based on their site of occurrence in the organelles and postulate novel targets that can effectively exploit this vulnerability.
Collapse
|
10
|
Molecular Characterization, Tissue Distribution Profile, and Nutritional Regulation of acsl Gene Family in Golden Pompano ( Trachinotus ovatus). Int J Mol Sci 2022; 23:ijms23126437. [PMID: 35742881 PMCID: PMC9224283 DOI: 10.3390/ijms23126437] [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: 05/08/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Long chain acyl-coA synthase (acsl) family genes activate the conversion of long chain fatty acids into acyl-coA to regulate fatty acid metabolism. However, the evolutionary characteristics, tissue expression and nutritional regulation of the acsl gene family are poorly understood in fish. The present study investigated the molecular characterization, tissue expression and nutritional regulation of the acsl gene family in golden pompano (Trachinotus ovatus). The results showed that the coding regions of acsl1, acsl3, acsl4, acsl5 and acsl6 cDNA were 2091 bp, 2142 bp, 2136 bp, 1977 bp and 2007 bp, encoding 697, 714, 712, 659 and 669 amino acids, respectively. Five acsl isoforms divided into two branches, namely, acsl1, acsl5 and acsl6, as well as acsl3 and acsl4. The tissue expression distribution of acsl genes showed that acsl1 and acsl3 are widely expressed in the detected tissues, while acsl4, acsl5 and acsl6 are mainly expressed in the brain. Compared to the fish fed with lard oil diets, the fish fed with soybean oil exhibited high muscular C18 PUFA contents and acsl1 and acsl3 mRNA levels, as well as low muscular SFA contents and acsl4 mRNA levels. High muscular n-3 LC-PUFA contents, and acsl3, acsl4 and acsl6 mRNA levels were observed in the fish fed with fish oil diets compared with those of fish fed with lard oil or soybean oil diets. High n-3 LC-PUFA levels and DHA contents, as well as the acsl3, acsl4 and acsl6 mRNA levels were exhibited in the muscle of fish fed diets with high dietary n-3 LC-PUFA levels. Additionally, the muscular acsl3, acsl4 and acsl6 mRNA expression levels, n-3 LC-PUFA and DHA levels were significantly up-regulated by the increase of dietary DHA proportions. Collectively, the positive relationship among dietary fatty acids, muscular fatty acids and acsl mRNA, indicated that T. ovatus Acsl1 and Acsl3 are beneficial for the C18 PUFA enrichment, and Acsl3, Acsl4 and Acsl6 are for n-3 LC-PUFA and DHA enrichment. The acquisition of fish Acsl potential function in the present study will play the foundation for ameliorating the fatty acids nutrition in farmed fish products.
Collapse
|
11
|
Ma Y, Nenkov M, Chen Y, Press AT, Kaemmerer E, Gassler N. Fatty acid metabolism and acyl-CoA synthetases in the liver-gut axis. World J Hepatol 2021; 13:1512-1533. [PMID: 34904027 PMCID: PMC8637682 DOI: 10.4254/wjh.v13.i11.1512] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/28/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
Fatty acids are energy substrates and cell components which participate in regulating signal transduction, transcription factor activity and secretion of bioactive lipid mediators. The acyl-CoA synthetases (ACSs) family containing 26 family members exhibits tissue-specific distribution, distinct fatty acid substrate preferences and diverse biological functions. Increasing evidence indicates that dysregulation of fatty acid metabolism in the liver-gut axis, designated as the bidirectional relationship between the gut, microbiome and liver, is closely associated with a range of human diseases including metabolic disorders, inflammatory disease and carcinoma in the gastrointestinal tract and liver. In this review, we depict the role of ACSs in fatty acid metabolism, possible molecular mechanisms through which they exert functions, and their involvement in hepatocellular and colorectal carcinoma, with particular attention paid to long-chain fatty acids and small-chain fatty acids. Additionally, the liver-gut communication and the liver and gut intersection with the microbiome as well as diseases related to microbiota imbalance in the liver-gut axis are addressed. Moreover, the development of potentially therapeutic small molecules, proteins and compounds targeting ACSs in cancer treatment is summarized.
Collapse
Affiliation(s)
- Yunxia Ma
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Miljana Nenkov
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Yuan Chen
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Adrian T Press
- Department of Anesthesiology and Intensive Care Medicine and Center for Sepsis Control and Care, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Elke Kaemmerer
- Department of Pediatrics, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Nikolaus Gassler
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany.
| |
Collapse
|
12
|
Kurotaki A, Kuwata H, Hara S. Substrate Specificity of Human Long-Chain Acyl-CoA Synthetase ACSL6 Variants. Biol Pharm Bull 2021; 44:1571-1575. [PMID: 34602568 DOI: 10.1248/bpb.b21-00551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Long-chain acyl-CoA synthetases (ACSLs) are a family of enzymes that convert long-chain free fatty acids into their active form, acyl-CoAs. Recent knock-out mouse studies revealed that among ACSL isoenzymes, ACSL6 plays an important role in the maintenance of docosahexaenoic acid (DHA)-containing glycerophospholipids. Several transcript variants of the human ACSL6 gene have been found; the two major ACSL6 variants, ACSL6V1 and V2, encode slightly different short motifs that both contain a conserved structural domain, the fatty acid Gate domain. In the present study, we expressed recombinant human ACSL6V1 and V2 in Spodoptera frugiperda 9 (Sf9) cells using the baculovirus expression system, and then, using our novel ACSL assay system with liquid chromatography-tandem mass spectrometry (LC-MS/MS), we examined the substrate specificities of the recombinant human ACSL6V1 and V2 proteins. The results showed that both ACSL6V1 and V2 could convert various kinds of long-chain fatty acids into their acyl-CoAs. Oleic acid was a good common substrate and eicosapolyenoic acids were poor common substrates for both variants. However, ACSL6V1 and V2 differed considerably in their preferences for octadecapolyenoic acids, such as linoleic acid, and docosapolyenoic acids, such as DHA and docosapentaenoic acid (DPA): ACSL6V1 preferred octadecapolyenoic acids, whereas V2 strongly preferred docosapolyenoic acids. Moreover, our kinetic studies revealed that ACSL6V2 had a much higher affinity for DHA than ACSL6V1. Our results suggested that ACSL6V1 and V2 might exert different physiological functions and indicated that ACSL6V2 might be critical for the maintenance of membrane phospholipids bearing docosapolyenoic acids such as DHA.
Collapse
Affiliation(s)
- Anri Kurotaki
- Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University
| | - Hiroshi Kuwata
- Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University
| | - Shuntaro Hara
- Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University
| |
Collapse
|
13
|
Fernandez RF, Pereyra AS, Diaz V, Wilson ES, Litwa KA, Martínez-Gardeazabal J, Jackson SN, Brenna JT, Hermann BP, Eells JB, Ellis JM. Acyl-CoA synthetase 6 is required for brain docosahexaenoic acid retention and neuroprotection during aging. JCI Insight 2021; 6:e144351. [PMID: 34100386 PMCID: PMC8262339 DOI: 10.1172/jci.insight.144351] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 04/23/2021] [Indexed: 12/27/2022] Open
Abstract
The omega-3 fatty acid docosahexaenoic acid (DHA) inversely relates to neurological impairments with aging; however, limited nondietary models manipulating brain DHA have hindered a direct linkage. We discovered that loss of long-chain acyl-CoA synthetase 6 in mice (Acsl6–/–) depletes brain membrane phospholipid DHA levels, independent of diet. Here, Acsl6–/– brains contained lower DHA compared with controls across the life span. The loss of DHA- and increased arachidonate-enriched phospholipids were visualized by MALDI imaging predominantly in neuron-rich regions where single-molecule RNA in situ hybridization localized Acsl6 to neurons. ACSL6 is also astrocytic; however, we found that astrocyte-specific ACSL6 depletion did not alter membrane DHA because astrocytes express a non–DHA-preferring ACSL6 variant. Across the life span, Acsl6–/– mice exhibited hyperlocomotion, impairments in working spatial memory, and increased cholesterol biosynthesis genes. Aging caused Acsl6–/– brains to decrease the expression of membrane, bioenergetic, ribosomal, and synaptic genes and increase the expression of immune response genes. With age, the Acsl6–/– cerebellum became inflamed and gliotic. Together, our findings suggest that ACSL6 promotes membrane DHA enrichment in neurons, but not in astrocytes, and is important for neuronal DHA levels across the life span. The loss of ACSL6 impacts motor function, memory, and age-related neuroinflammation, reflecting the importance of neuronal ACSL6-mediated lipid metabolism across the life span.
Collapse
Affiliation(s)
- Regina F Fernandez
- Department of Physiology, Brody School of Medicine, and East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Andrea S Pereyra
- Department of Physiology, Brody School of Medicine, and East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Victoria Diaz
- Department of Biology, University of Texas San Antonio, San Antonio, Texas, USA
| | - Emily S Wilson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Karen A Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | | | - Shelley N Jackson
- Structural Biology Core, Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, Maryland, USA
| | - J Thomas Brenna
- Departments of Pediatrics, Chemistry, and Nutrition and.,Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Brian P Hermann
- Department of Biology, University of Texas San Antonio, San Antonio, Texas, USA
| | - Jeffrey B Eells
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Jessica M Ellis
- Department of Physiology, Brody School of Medicine, and East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
14
|
Dattilo MA, Benzo Y, Herrera LM, Prada JG, Lopez PF, Caruso CM, Lasaga M, García CI, Paz C, Maloberti PM. Regulation and role of Acyl-CoA synthetase 4 in glial cells. J Steroid Biochem Mol Biol 2021; 208:105792. [PMID: 33246155 DOI: 10.1016/j.jsbmb.2020.105792] [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: 08/05/2020] [Revised: 10/23/2020] [Accepted: 11/14/2020] [Indexed: 10/22/2022]
Abstract
Acyl-CoA synthetase 4 (Acsl4), an enzyme involved in arachidonic acid (AA) metabolism, participates in physiological and pathological processes such as steroidogenesis and cancer. The role of Acsl4 in neurons and in nervous system development has also been documented but little is known regarding its functionality in glial cells. In turn, several processes in glial cells, including neurosteroidogenesis, stellation and AA uptake, are regulated by cyclic adenosine monophosphate (cAMP) signal. In this context, the aim of this work was to analyze the expression and functional role of Acsl4 in primary rat astrocyte cultures and in the C6 glioma cell line by chemical inhibition and stable silencing, respectively. Results show that Acsl4 expression was regulated by cAMP in both models and that cAMP stimulation of steroidogenic acute regulatory protein mRNA levels was reduced by Acsl4 inhibition or silencing. Also, Acsl4 inhibition reduced progesterone synthesis stimulated by cAMP and also affected cAMP-induced astrocyte stellation, decreasing process elongation and increasing branching complexity. Similar effects were observed for Acsl4 silencing on cAMP-induced C6 cell morphological shift. Moreover, Acsl4 inhibition and silencing reduced proliferation and migration of both cell types. Acsl4 silencing in C6 cells reduced the capacity for colony proliferation and neurosphere formation, the latter ability also being abolished by Acsl4 inhibition. In sum, this work presents novel evidence of Acsl4 involvement in neurosteroidogenesis and the morphological changes of glial cells promoted by cAMP. Furthermore, Acsl4 participates in migration and proliferation, also affecting cell self-renewal. Altogether, these findings provide insights into Acsl4 functions in glial cells.
Collapse
Affiliation(s)
- Melina A Dattilo
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Yanina Benzo
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Lucia M Herrera
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Jesica G Prada
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Paula F Lopez
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Carla M Caruso
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología, Buenos Aires, Argentina
| | - Mercedes Lasaga
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología, Buenos Aires, Argentina
| | - Corina I García
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina; Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Cristina Paz
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Paula M Maloberti
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina.
| |
Collapse
|
15
|
Bi Y, Yuan X, Chen Y, Chang G, Chen G. Expression analysis of genes related to lipid metabolism in peripheral blood lymphocytes of chickens challenged with reticuloendotheliosis virus. Poult Sci 2021; 100:101081. [PMID: 33813326 PMCID: PMC8047978 DOI: 10.1016/j.psj.2021.101081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 01/20/2021] [Accepted: 02/22/2021] [Indexed: 01/05/2023] Open
Abstract
The underlying molecular mechanism of lipid metabolism in peripheral blood lymphocytes from chicken infected with reticuloendotheliosis virus (REV) remains poorly understood. Therefore, this scientific question was explored in vitro and in vivo. The results indicated that triglyceride content was significantly reduced, but the free fatty acid content and carnitine palmitoyltransferase-1 activity were significantly increased in blood lymphocytes after REV infection. By RNA sequencing, 97 known differentially expressed genes (DEG) related to lipid metabolism or glycometabolism were screened via Gene Ontology term analysis. On the basis of these 97 DEG, enriched pathways, including the peroxisome proliferators-activated receptor (PPAR) signaling pathway, were identified. Among these 97 DEG, some representative genes were related to lipolysis and fatty acid utilization (PPARG, LPL, PLIN2, ACOX1, ACSL1, FABP3, and FABP4). However, other genes related to lipid biosynthesis (ACSL3, ACSL6, DGAT2, LPIN1, and LPIN2) were downregulated. The quantitative polymerase chain reaction results confirmed the accuracy of the RNA sequencing data, and the in vivo outcome supports theses in vitro results. Our findings revealed that REV regulates fatty acid and lipid metabolism in peripheral blood lymphocytes from chicken. After the lymphocytes were infected with REV, the exogenous fatty acids were preferentially used; genes involved in fatty acid utilization were upregulated via the PPAR pathway, whereas genes involved in lipid and fatty acid biosynthesis were downregulated.
Collapse
Affiliation(s)
- Yulin Bi
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P.R. China; Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou, 225009, P.R. China; Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou, 225009, P.R. China
| | - Xiaoya Yuan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P.R. China
| | - Ying Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P.R. China
| | - Guobin Chang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P.R. China.
| | - Guohong Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P.R. China
| |
Collapse
|
16
|
Drijvers JM, Gillis JE, Muijlwijk T, Nguyen TH, Gaudiano EF, Harris IS, LaFleur MW, Ringel AE, Yao CH, Kurmi K, Juneja VR, Trombley JD, Haigis MC, Sharpe AH. Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8 + T Cells. Cancer Immunol Res 2021; 9:184-199. [PMID: 33277233 PMCID: PMC7864883 DOI: 10.1158/2326-6066.cir-20-0384] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/19/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022]
Abstract
Metabolic constraints in the tumor microenvironment constitute a barrier to effective antitumor immunity and similarities in the metabolic properties of T cells and cancer cells impede the specific therapeutic targeting of metabolism in either population. To identify distinct metabolic vulnerabilities of CD8+ T cells and cancer cells, we developed a high-throughput in vitro pharmacologic screening platform and used it to measure the cell type-specific sensitivities of activated CD8+ T cells and B16 melanoma cells to a wide array of metabolic perturbations during antigen-specific killing of cancer cells by CD8+ T cells. We illustrated the applicability of this screening platform by showing that CD8+ T cells were more sensitive to ferroptosis induction by inhibitors of glutathione peroxidase 4 (GPX4) than B16 and MC38 cancer cells. Overexpression of ferroptosis suppressor protein 1 (FSP1) or cytosolic GPX4 yielded ferroptosis-resistant CD8+ T cells without compromising their function, while genetic deletion of the ferroptosis sensitivity-promoting enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) protected CD8+ T cells from ferroptosis but impaired antitumor CD8+ T-cell responses. Our screen also revealed high T cell-specific vulnerabilities for compounds targeting NAD+ metabolism or autophagy and endoplasmic reticulum (ER) stress pathways. We focused the current screening effort on metabolic agents. However, this in vitro screening platform may also be valuable for rapid testing of other types of compounds to identify regulators of antitumor CD8+ T-cell function and potential therapeutic targets.
Collapse
Affiliation(s)
- Jefte M Drijvers
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Jacob E Gillis
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Tara Muijlwijk
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Thao H Nguyen
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Emily F Gaudiano
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Isaac S Harris
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Martin W LaFleur
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Alison E Ringel
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Cong-Hui Yao
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Kiran Kurmi
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Vikram R Juneja
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Justin D Trombley
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts.
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts.
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
17
|
Zheng M, Wang W, Liu J, Zhang X, Zhang R. Lipid Metabolism in Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:49-69. [PMID: 33740243 DOI: 10.1007/978-981-33-6785-2_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Metabolic reprogramming is one of the most critical hallmarks in cancer cells. In the past decades, mounting evidence has demonstrated that, besides the Warburg Effect, lipid metabolism dysregulation is also one of the essential characteristics of cancer cell metabolism. Lipids are water-insoluble molecules with diverse categories of phosphoglycerides, triacylglycerides, sphingolipids, sterols, etc. As the major utilization for energy storage, fatty acids are the primary building blocks for synthesizing triacylglycerides. And phosphoglycerides, sphingolipids, and sterols are the main components constructing biological membranes. More importantly, lipids play essential roles in signal transduction by functioning as second messengers or hormones. Much evidence has shown specific alterations of lipid metabolism in cancer cells. Consequently, the structural configuration of biological membranes, the energy homeostasis under nutrient stress, and the abundance of lipids in the intracellular signal transduction are affected by these alterations. Furthermore, lipid droplets accumulate in cancer cells and function adaptively to different types of harmful stress. This chapter reviews the regulation, functions, and therapeutic benefits of targeting lipid metabolism in cancer cells. Overall, this chapter highlights the significance of exploring more potential therapeutic strategies for malignant diseases by unscrambling lipid metabolism regulation in cancer cells.
Collapse
Affiliation(s)
- Minhua Zheng
- Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Wei Wang
- Department of Immunology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xiao Zhang
- Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Rui Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, People's Republic of China.
- Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
18
|
Xu Y, Li X, Cheng Y, Yang M, Wang R. Inhibition of ACSL4 attenuates ferroptotic damage after pulmonary ischemia-reperfusion. FASEB J 2020; 34:16262-16275. [PMID: 33070393 DOI: 10.1096/fj.202001758r] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 02/05/2023]
Abstract
Lung ischemia-reperfusion (IR) injury is a common clinical pathology associated with high mortality. Ferroptosis, a novel mode of cell death elicited by iron-dependent phospholipid peroxidation, has been implicated in ischemic events. Acyl-CoA synthetase long-chain family member 4 (ACSL4) is one of the main enzymes in pro-ferroptotic lipid metabolism. In this study, the involvement of ferroptotic death in different durations of reperfusion was evaluated by assessing the iron content, malondialdehyde, and glutathione levels, ferroptosis-related protein expression, and mitochondria morphology. The roles of ferroptosis-specific inhibitor, liproxastin-1 (Lip-1), and ACSL4 modulation in a preventive regimen were assessed in vivo and in vitro. The hallmarks of pulmonary function, such as histological lung injury score, wet/dry ratio, and oxygenation index, were evaluated as well. Results showed that lung IR increased the tissue iron content and lipid peroxidation accumulation, along with key protein (GPX4 and ACSL4) expression alteration during reperfusion. Pretreatment with Lip-1 inhibited ferroptosis and ameliorated lung IR-induced injury in animal and cell models. In addition, administering ACSL4 inhibitor rosiglitazone before ischemia diminished the ferroptotic damage in IR-injured lung tissue, consistent with the protective effect of ACSL4 knockdown on lung epithelial cells subjected to hypoxia/reoxygenation. Thus, this study delineated that IR-induced ferroptotic cell death in lung tissue and ACSL4 were correlated with this process. Inhibition of ferroptosis and ACSL4 mitigated the ferroptotic damage in IR-induced lung injury by reducing lipid peroxidation and increasing the glutathione and GPX4 levels.
Collapse
Affiliation(s)
- Yixin Xu
- Department of Anesthesiology, West China Hospital, Sichuan University, The Research Units of West China (2018RU12), Chinese Academy of Medical Sciences, Chengdu, China.,Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuehan Li
- Department of Anesthesiology, West China Hospital, Sichuan University, The Research Units of West China (2018RU12), Chinese Academy of Medical Sciences, Chengdu, China.,Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Cheng
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Mingan Yang
- Division of Biostatistics & Epidemiology, School of Public Health, San Diego State University, San Diego, CA, USA
| | - Rurong Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, The Research Units of West China (2018RU12), Chinese Academy of Medical Sciences, Chengdu, China
| |
Collapse
|
19
|
Fernandez RF, Ellis JM. Acyl-CoA synthetases as regulators of brain phospholipid acyl-chain diversity. Prostaglandins Leukot Essent Fatty Acids 2020; 161:102175. [PMID: 33031993 PMCID: PMC8693597 DOI: 10.1016/j.plefa.2020.102175] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/22/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022]
Abstract
Each individual cell-type is defined by its distinct morphology, phenotype, molecular and lipidomic profile. The importance of maintaining cell-specific lipidomic profiles is exemplified by the numerous diseases, disorders, and dysfunctional outcomes that occur as a direct result of altered lipidome. Therefore, the mechanisms regulating cellular lipidome diversity play a role in maintaining essential biological functions. The brain is an organ particularly rich in phospholipids, the main constituents of cellular membranes. The phospholipid acyl-chain profile of membranes in the brain is rather diverse due in part to the high degree of cellular heterogeneity. These membranes and the acyl-chain composition of their phospholipids are highly regulated, but the mechanisms that confer this tight regulation are incompletely understood. A family of enzymes called acyl-CoA synthetases (ACSs) stands at a pinnacle step allowing influence over cellular acyl-chain selection and subsequent metabolic flux. ACSs perform the initial reaction for cellular fatty acid metabolism by ligating a Coenzyme A to a fatty acid which both traps a fatty acid within a cell and activates it for metabolism. The ACS family of enzymes is large and diverse consisting of 25-26 family members that are nonredundant, each with unique distribution across and within cell types, and differential fatty acid substrate preferences. Thus, ACSs confer a critical intracellular fatty acid selecting step in a cell-type dependent manner providing acyl-CoA moieties that serve as essential precursors for phospholipid synthesis and remodeling, and therefore serve as a key regulator of cellular membrane acyl-chain compositional diversity. Here we will discuss how the contribution of individual ACSs towards brain lipid metabolism has only just begun to be elucidated and discuss the possibilities for how ACSs may differentially regulate brain lipidomic diversity.
Collapse
Affiliation(s)
- Regina F Fernandez
- Department of Physiology and East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, NC, United States
| | - Jessica M Ellis
- Department of Physiology and East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, NC, United States.
| |
Collapse
|
20
|
RNA-Seq Study of Hepatic Response of Yellow-Feather Chickens to Acute Heat Stress. ANNALS OF ANIMAL SCIENCE 2020. [DOI: 10.2478/aoas-2019-0060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
The yellow-feather broiler is a popular poultry breed in Asia, particularly in China. In this study, we performed RNA-seq analysis to identify differentially expressed genes (deGs) in the liver of yellow-feather broilers that had been subjected to acute heat stress treatment (38±1°C for 4 h, recovery 2 h) and determine the response of the liver to high temperature and its effects on yellow-feather broiler physiology. We found that the cloacal temperature and respiratory rate of yellow-feather chickens were significantly increased immediately after the initiation of acute heat stress (38°c) treatment. And after recovery for 2 h, there was no difference in the cloacal temperature and respiratory rate between the acute heat stress and control groups. A total of 834 DEGs were observed in response to heat stress by RNA-seq. Almost half of the DEGs were involved in the lipid and energy metabolism, including fatty acid metabolism (ACOX1, ACACA, ACSL1, ACSL6, ACAA1, ACAA2, HADHB, and FASN) and propanoate metabolism (ACSS2, ALDH2, ACACA, DLAT, ALDH7A1, MDH1, ME1, ABAT, SUCLG2, and ACSS3). Our findings provide the context for RNA-seq studies in the liver of yellow-feather chickens and suggest that the liver of yellow-feather broilers has the lipid and energy metabolism physiological mechanisms activated in response to heat stress.
Collapse
|
21
|
Huang M, Wu J, Ling R, Li N. Quadruple negative breast cancer. Breast Cancer 2020; 27:527-533. [PMID: 31939077 DOI: 10.1007/s12282-020-01047-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 01/05/2020] [Indexed: 12/15/2022]
Abstract
Quadruple negative breast cancer (QNBC), lacking the expression of ER (estrogen receptor), PR (progesterone receptor), HER2 (human epidermal growth factor receptor-2) and AR (androgen receptor), was regarded as one breast cancer subtype with the worst prognosis. Recently, the molecular features of QNBC are not well understood. Different from AR-positive triple-negative breast cancer, QNBC is insensitive to conventional chemotherapeutic agents and has no efficient treatment targets. However, QNBC has been shown to express unique proteins that may be amenable to use in the development of targeted therapies. Here we reviewed the features of QNBC and proteins that may serve as effective targets for QNBC treatment, such as ACSL4, SKP2, immune checkpoint inhibitors, EGFR, MicroRNA signatures and Engrailed 1.
Collapse
Affiliation(s)
- Meiling Huang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jiang Wu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Ling
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Nanlin Li
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
22
|
Roelands J, Garand M, Hinchcliff E, Ma Y, Shah P, Toufiq M, Alfaki M, Hendrickx W, Boughorbel S, Rinchai D, Jazaeri A, Bedognetti D, Chaussabel D. Long-Chain Acyl-CoA Synthetase 1 Role in Sepsis and Immunity: Perspectives From a Parallel Review of Public Transcriptome Datasets and of the Literature. Front Immunol 2019; 10:2410. [PMID: 31681299 PMCID: PMC6813721 DOI: 10.3389/fimmu.2019.02410] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022] Open
Abstract
A potential role for the long-chain acyl-CoA synthetase family member 1 (ACSL1) in the immunobiology of sepsis was explored during a hands-on training workshop. Participants first assessed the robustness of the potential gap in biomedical knowledge identified via an initial screen of public transcriptome data and of the literature associated with ACSL1. Increase in ACSL1 transcript abundance during sepsis was confirmed in several independent datasets. Querying the ACSL1 literature also confirmed the absence of reports associating ACSL1 with sepsis. Inferences drawn from both the literature (via indirect associations) and public transcriptome data (via correlation) point to the likely participation of ACSL1 and ACSL4, another family member, in inflammasome activation in neutrophils during sepsis. Furthermore, available clinical data indicate that levels of ACSL1 and ACSL4 induction was significantly higher in fatal cases of sepsis. This denotes potential translational relevance and is consistent with involvement in pathways driving potentially deleterious systemic inflammation. Finally, while ACSL1 expression was induced in blood in vitro by a wide range of pathogen-derived factors as well as TNF, induction of ACSL4 appeared restricted to flagellated bacteria and pathogen-derived TLR5 agonists and IFNG. Taken together, this joint review of public literature and omics data records points to two members of the acyl-CoA synthetase family potentially playing a role in inflammasome activation in neutrophils. Translational relevance of these observations in the context of sepsis and other inflammatory conditions remain to be investigated.
Collapse
Affiliation(s)
- Jessica Roelands
- Sidra Medicine, Doha, Qatar.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | | | - Emily Hinchcliff
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ying Ma
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Parin Shah
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | | | | | | | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | |
Collapse
|
23
|
Role of acyl-CoA synthetase ACSL4 in arachidonic acid metabolism. Prostaglandins Other Lipid Mediat 2019; 144:106363. [DOI: 10.1016/j.prostaglandins.2019.106363] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/15/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022]
|
24
|
Hale BJ, Fernandez RF, Kim SQ, Diaz VD, Jackson SN, Liu L, Brenna JT, Hermann BP, Geyer CB, Ellis JM. Acyl-CoA synthetase 6 enriches seminiferous tubules with the ω-3 fatty acid docosahexaenoic acid and is required for male fertility in the mouse. J Biol Chem 2019; 294:14394-14405. [PMID: 31399511 PMCID: PMC6768642 DOI: 10.1074/jbc.ra119.009972] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/01/2019] [Indexed: 12/31/2022] Open
Abstract
Docosahexaenoic acid (DHA) is an ω-3 dietary-derived polyunsaturated fatty acid of marine origin enriched in testes and necessary for normal fertility, yet the mechanisms regulating the enrichment of DHA in the testes remain unclear. Long-chain ACSL6 (acyl-CoA synthetase isoform 6) activates fatty acids for cellular anabolic and catabolic metabolism by ligating a CoA to a fatty acid, is highly expressed in testes, and has high preference for DHA. Here, we investigated the role of ACSL6 for DHA enrichment in the testes and its requirement for male fertility. Acsl6-/- males were severely subfertile with smaller testes, reduced cauda epididymal sperm counts, germ cell loss, and disorganization of the seminiferous epithelium. Total fatty acid profiling of Acsl6-/- testes revealed reduced DHA and increased ω-6 arachidonic acid, a fatty acid profile also reflected in phospholipid composition. Strikingly, lipid imaging demonstrated spatial redistribution of phospholipids in Acsl6-/- testes. Arachidonic acid-containing phospholipids were predominantly interstitial in control testes but diffusely localized across Acsl6-/- testes. In control testes, DHA-containing phospholipids were predominantly within seminiferous tubules, which contain Sertoli cells and spermatogenic cells but relocalized to the interstitium in Acsl6-/- testes. Taken together, these data demonstrate that ACSL6 is an initial driving force for germ cell DHA enrichment and is required for normal spermatogenesis and male fertility.
Collapse
Affiliation(s)
- Benjamin J Hale
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina 27834
| | - Regina F Fernandez
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina 27834
| | - Sora Q Kim
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana 47907
| | - Victoria D Diaz
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas 78249
| | - Shelley N Jackson
- National Institute on Drug Abuse, Intramural Research Program, Structural Biology Core, Baltimore, Maryland 21224
| | - Lei Liu
- Departments of Pediatrics, Chemistry, and Nutrition, Dell Pediatric Research Institute, University of Texas at Austin, Austin, Texas 78723
- Department of Veterinary Sciences, Hunan Agricultural University, Changsha 410128, China
| | - J Thomas Brenna
- Departments of Pediatrics, Chemistry, and Nutrition, Dell Pediatric Research Institute, University of Texas at Austin, Austin, Texas 78723
| | - Brian P Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas 78249
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina 27834
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina 27834
| | - Jessica M Ellis
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina 27834
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina 27834
| |
Collapse
|
25
|
Lv Y, Cao Y, Gao Y, Yun J, Yu Y, Zhang L, Hu Z, Liu L, Xue J, Zhang G. Effect of ACSL3 Expression Levels on Preadipocyte Differentiation in Chinese Red Steppe Cattle. DNA Cell Biol 2019; 38:945-954. [DOI: 10.1089/dna.2018.4443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yang Lv
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yang Cao
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- Key Laboratory of Beef Cattle Genetics and Breeding in Ministry of Agriculture and Rural Agriculture, Changchun, China
| | - Yi Gao
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Jinyan Yun
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Yongsheng Yu
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Lichun Zhang
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Zhongchang Hu
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Lixiang Liu
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jiajia Xue
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Guoliang Zhang
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Beef Cattle Genetics and Breeding in Ministry of Agriculture and Rural Agriculture, Changchun, China
| |
Collapse
|
26
|
Angius A, Uva P, Pira G, Muroni MR, Sotgiu G, Saderi L, Uleri E, Caocci M, Ibba G, Cesaraccio MR, Serra C, Carru C, Manca A, Sanges F, Porcu A, Dolei A, Scanu AM, Rocca PC, De Miglio MR. Integrated Analysis of miRNA and mRNA Endorses a Twenty miRNAs Signature for Colorectal Carcinoma. Int J Mol Sci 2019; 20:E4067. [PMID: 31434359 PMCID: PMC6720928 DOI: 10.3390/ijms20164067] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) ranks as the most frequent carcinoma worldwide. CRC patients show strong prognostic differences and responses to treatment, and 20% have incurable metastatic disease at diagnosis. We considered it essential to investigate mechanisms that control cellular regulatory networks, such as the miRNA-mRNA interaction, known to be involved in cancer pathogenesis. We conducted a human miRNome analysis by TaqMan low density array, comparing CRC to normal colon tissue (NCT, and experimentally identified gene targets of miRNAs deregulated, by anti-correlation analysis, with the CRC whole-transcriptome profile obtained from RNASeq experiments. We identified an integrated signature of 20 deregulated miRNAs in CRC. Enrichment analyses of the gene targets controlled by these miRNAs brought to light 25 genes, members of pathways known to lead to cell growth and death (CCND1, NKD1, FZD3, MAD2L1, etc.), such as cell metabolism (ACSL6, PRPS1-2). A screening of prognosis-mediated miRNAs underlined that the overexpression of miR-224 promotes CRC metastasis, and is associated with high stage and poor survival. These findings suggest that the biology and progression of CRC depend on deregulation of multiple miRNAs that cause a complex dysfunction of cellular molecular networks. Our results have further established miRNA-mRNA interactions and defined multiple pathways involved in CRC pathogenesis.
Collapse
Affiliation(s)
- Andrea Angius
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Cittadella Universitaria di Cagliari, 09042 Monserrato (CA), Italy
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Piscina Manna, 09050 Pula (CA), Italy
| | - Giovanna Pira
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Maria Rosaria Muroni
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Giovanni Sotgiu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Laura Saderi
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Elena Uleri
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Maurizio Caocci
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Gabriele Ibba
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Maria Rosaria Cesaraccio
- Department of Prevention, Registro Tumori Provincia di Sassari, ASSL Sassari-ATS Sardegna, Via Rizzeddu 21, Sassari, Italy
| | - Caterina Serra
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Alessandra Manca
- Department of Pathology, AOU Sassari, Via Matteotti 60, 07100 Sassari, Italy
| | - Francesca Sanges
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Alberto Porcu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Antonia Dolei
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Antonio Mario Scanu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
| | - Paolo Cossu Rocca
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
- Department of Diagnostic Services, "Giovanni Paolo II" Hospital, ASSL Olbia-ATS Sardegna, Via Bazzoni-Sircana, 07026 Olbia, Italy.
| | - Maria Rosaria De Miglio
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| |
Collapse
|
27
|
Valente RS, de Almeida TG, Alves MF, de Camargo J, Basso AC, Belaz KRA, Eberlin MN, Landim-Alvarenga FDC, Fontes PK, Nogueira MFG, Sudano MJ. Modulation of long-chain Acyl-CoA synthetase on the development, lipid deposit and cryosurvival of in vitro produced bovine embryos. PLoS One 2019; 14:e0220731. [PMID: 31381602 PMCID: PMC6681945 DOI: 10.1371/journal.pone.0220731] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/22/2019] [Indexed: 11/19/2022] Open
Abstract
In this study, we evaluated the modulation effect of long-chain Acyl-CoA synthetase during early embryo development. Bovine embryos were cultured in four groups: positive modulation (ACS+) with GW3965 hydrochloride, negative modulation (ACS-) with Triacsin C, association of both modulators (ACS±), and control. Embryo development rates were not altered (P>0.05) by treatments. Embryonic cytoplasmic lipid content increased in ACS+ but reduced in ACS- compared to the control (P < 0.05), whereas the membrane phospholipids profile was not altered by treatments. The total number of blastomeres did not differ (P > 0.05) between groups; however, an increased apoptotic cells percentage was found in ACS- compared to control. Twenty-four hours after warming, ACS+ and control grade I embryos presented the best hatching rates, whereas the ACS+ group equaled the hatching rates between their embryos of grades I, II and III 48 hours after warming. The relative abundance of transcripts for genes associated with lipid metabolism (ACSL3, ACSL6, ACAT1, SCD, and AUH), heatshock (HSP90AA1 and HSF1), oxidative stress (GPX4), and angiogenesis (VEGF), among other important genes for embryo development were affected by at least one of the treatments. The treatments were effective in modulating the level of transcripts for ACSL3 and the cytoplasmic lipid content. The ACS- was not effective in increasing embryonic cryosurvival, whereas ACS+ restored survival rates after vitrification of embryos with low quality, making them equivalent to embryos of excellent quality.
Collapse
Affiliation(s)
- Roniele Santana Valente
- School of Veterinary Medicine, Federal University of Pampa, Uruguaiana, RS, Brazil
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, SP, Brazil
| | | | | | - Janine de Camargo
- School of Veterinary Medicine, Federal University of Pampa, Uruguaiana, RS, Brazil
| | | | | | | | | | - Patricia Kubo Fontes
- Department of Biological Sciences, São Paulo State University, Assis, SP, Brazil
| | | | - Mateus José Sudano
- School of Veterinary Medicine, Federal University of Pampa, Uruguaiana, RS, Brazil
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, SP, Brazil
- * E-mail:
| |
Collapse
|
28
|
Park S, Lee M, Chun CH, Jin EJ. The lncRNA, Nespas, Is Associated with Osteoarthritis Progression and Serves as a Potential New Prognostic Biomarker. Cartilage 2019; 10:148-156. [PMID: 28805067 PMCID: PMC6425538 DOI: 10.1177/1947603517725566] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION In this article, we explored the hypothesis that the long noncoding RNA, Nespas, promotes osteoarthritis (OA) by supporting abnormal lipid metabolism in human chondrocytes. MATERIALS AND METHODS Human articular chondrocytes from osteoarthritis patients were used and expression level of Nespas were determined by real-time polymerase chain reaction. Introduction of Nespas and Nespas-associated genes/miRNAs were performed by using a lentiviral system. The effect of Nespas on mitochondrial function was determined by staining mitochondria and analyzing mitopotential and mitochondrial genes. Moreover, to identify the responsible molecules in Nespas-induced pathogenesis, profiling of peroxisomal genes and miRNAs were applied and interactome analysis was performed. RESULTS Highly elevated levels of Nespas and Acyl-CoA synthetase 6 (ACSL6) were observed in OA patients. Both Nespas overexpression and ACSL6 upregulation into human chondrocytes induced typical OA characteristics, such as downregulation of type II collagen; upregulation of type I collagen, metalloproteinase 13, and caspase-1 and -3; and dysfunction of mitochondria and peroxisome. Co-expression of Nespas and ACSL6 siRNA reduced caspase-1 and -3 levels. Moreover, Nespas overexpression significantly suppressed levels of miR-291a-3p, -196a-5p, -23a-3p, -24-3p, and let-7a-5p, and these miRs are known to potentially target ACSL6 according to ingenuity pathway analysis. We also confirmed that these miRs were significantly suppressed in human OA chondrocytes. Overexpression of miR-291a-3p, -196a-5p, -23a-3p, -24-3p, or let-7a-5p in the presence of Nespas suppressed levels of ACSL6, caspase-1 and -3. DISCUSSION Overall, we suggest that elevated Nespas level in OA are associated with OA pathogenesis by suppressing miRs targeting ACSL6 and subsequent ACSL6 upregulation.
Collapse
Affiliation(s)
- Sujung Park
- Department of Biological Sciences,
College of Natural Sciences, Wonkwang University, Iksan, Chunbuk, Korea,These authors contributed equally to
this work
| | - Myeungsoo Lee
- Department of Internal Medicine,
Division of Rheumatology, Wonkwang University School of Medicine, Iksan, Chunbuk,
Korea,These authors contributed equally to
this work
| | - Churl-Hong Chun
- Department of Orthopedic Surgery,
Wonkwang University School of Medicine, Iksan, Chunbuk, Korea
| | - Eun-Jung Jin
- Department of Biological Sciences,
College of Natural Sciences, Wonkwang University, Iksan, Chunbuk, Korea,Eun-Jung Jin, Department of Biological
Sciences, College of Natural Sciences, Wonkwang University, Iksan, Chunbuk
570-749, Korea.
| |
Collapse
|
29
|
Adaptive evolution of the ACSL gene family in Carnivora. Genetica 2019; 147:141-148. [DOI: 10.1007/s10709-019-00057-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
|
30
|
Zhu XG, Nicholson Puthenveedu S, Shen Y, La K, Ozlu C, Wang T, Klompstra D, Gultekin Y, Chi J, Fidelin J, Peng T, Molina H, Hang HC, Min W, Birsoy K. CHP1 Regulates Compartmentalized Glycerolipid Synthesis by Activating GPAT4. Mol Cell 2019; 74:45-58.e7. [PMID: 30846317 DOI: 10.1016/j.molcel.2019.01.037] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 11/26/2018] [Accepted: 01/25/2019] [Indexed: 01/10/2023]
Abstract
Cells require a constant supply of fatty acids to survive and proliferate. Fatty acids incorporate into membrane and storage glycerolipids through a series of endoplasmic reticulum (ER) enzymes, but how these enzymes are regulated is not well understood. Here, using a combination of CRISPR-based genetic screens and unbiased lipidomics, we identified calcineurin B homologous protein 1 (CHP1) as a major regulator of ER glycerolipid synthesis. Loss of CHP1 severely reduces fatty acid incorporation and storage in mammalian cells and invertebrates. Mechanistically, CHP1 binds and activates GPAT4, which catalyzes the initial rate-limiting step in glycerolipid synthesis. GPAT4 activity requires CHP1 to be N-myristoylated, forming a key molecular interface between the two proteins. Interestingly, upon CHP1 loss, the peroxisomal enzyme, GNPAT, partially compensates for the loss of ER lipid synthesis, enabling cell proliferation. Thus, our work identifies a conserved regulator of glycerolipid metabolism and reveals plasticity in lipid synthesis of proliferating cells.
Collapse
Affiliation(s)
- Xiphias Ge Zhu
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Shirony Nicholson Puthenveedu
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA; Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, Graz 8036, Austria
| | - Yihui Shen
- Department of Chemistry and Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Konnor La
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Can Ozlu
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Tim Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Diana Klompstra
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Yetis Gultekin
- Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Jingyi Chi
- Laboratory of Molecular Metabolism, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Justine Fidelin
- The Proteomics Resource Center, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Tao Peng
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Henrik Molina
- The Proteomics Resource Center, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Howard C Hang
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Wei Min
- Department of Chemistry and Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
31
|
Abstract
Ferroptosis is a form of cell death that requires phospholipid peroxidation and has attracted increased attention, both as a means to eradicate tumors resistant to standard chemotherapy and for its potential contribution to tissue damage such as in ischemia/reperfusion. The center stage taken by phospholipid peroxidation in ferroptosis is highlighted by recent discoveries that demonstrate an intricate regulation of both the metabolism of polyunsaturated fatty acids as well as mechanisms leading to their oxidation. These metabolic steps converge at the point of ferroptosis execution through mechanisms that are now only starting to be understood. In this short review, we provide an appraisal of some of the recent advances in the understanding of the ferroptosis process and also provide some perspectives of where this knowledge could take us.
Collapse
Affiliation(s)
| | - Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química , Universidade de São Paulo , São Paulo 05508-000 , Brazil
| | - Almut Schulze
- Theodor-Boveri-Institute, Biocenter , University of Würzburg , 97074 Würzburg , Germany
| |
Collapse
|
32
|
Zamzow DR, Elias V, Acosta VA, Escobedo E, Magnusson KR. Higher Levels of Protein Palmitoylation in the Frontal Cortex across Aging Were Associated with Reference Memory and Executive Function Declines. eNeuro 2019; 6:ENEURO.0310-18.2019. [PMID: 30740518 PMCID: PMC6366935 DOI: 10.1523/eneuro.0310-18.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/17/2022] Open
Abstract
Cognitive decline with aging is often due to altered levels of protein expression. The NMDA receptor (NMDAR) and the complex of proteins surrounding the receptor are susceptible to age-related changes in expression. In the frontal cortex of aged mice, there is a significant loss of expression of the GluN2B subunit of the NMDAR, an increase in Fyn expression, and no change in PSD-95. Studies have also found that, in the frontal cortex, phosphorylation of GluN2B subunits and palmitoylation of GluN2 subunits and NMDAR complex proteins are affected by age. In this study, we examined some of the factors that may lead to the differences in the palmitoylation levels of NMDAR complex proteins in the frontal cortex of aged animals. The Morris water maze was used to test spatial learning in 3- and 24-month-old mice. The acyl-biotinyl exchange method was used to precipitate palmitoylated proteins from the frontal cortices and hippocampi of the mice. Additionally, brain lysates from old and young mice were probed for the expression of fatty acid transporter proteins. An age-related increase of palmitoylated GluN2A, GluN2B, Fyn, PSD-95, and APT1 (acyl protein thioesterase 1) in the frontal cortex was associated with poorer reference memory and/or executive functions. These data suggest that there may be a perturbation in the palmitoylation cycle in the frontal cortex of aged mice that contributes to age-related cognitive declines.
Collapse
Affiliation(s)
| | - Valerie Elias
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon 97331
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon 97331
| | - Varinia A. Acosta
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon 97331
| | - Emily Escobedo
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon 97331
| | - Kathy R. Magnusson
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon 97331
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon 97331
| |
Collapse
|
33
|
|
34
|
Zhou Y, Wang Y, Zhang X, Bhar S, Jones Lipinski RA, Han J, Feng L, Butcher RA. Biosynthetic tailoring of existing ascaroside pheromones alters their biological function in C. elegans. eLife 2018; 7:33286. [PMID: 29863473 PMCID: PMC5986272 DOI: 10.7554/elife.33286] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 04/26/2018] [Indexed: 11/13/2022] Open
Abstract
Caenorhabditis elegans produces ascaroside pheromones to control its development and behavior. Even minor structural differences in the ascarosides have dramatic consequences for their biological activities. Here, we identify a mechanism that enables C. elegans to dynamically tailor the fatty-acid side chains of the indole-3-carbonyl (IC)-modified ascarosides it has produced. In response to starvation, C. elegans uses the peroxisomal acyl-CoA synthetase ACS-7 to activate the side chains of medium-chain IC-ascarosides for β-oxidation involving the acyl-CoA oxidases ACOX-1.1 and ACOX-3. This pathway rapidly converts a favorable ascaroside pheromone that induces aggregation to an unfavorable one that induces the stress-resistant dauer larval stage. Thus, the pathway allows the worm to respond to changing environmental conditions and alter its chemical message without having to synthesize new ascarosides de novo. We establish a new model for biosynthesis of the IC-ascarosides in which side-chain β-oxidation is critical for controlling the type of IC-ascarosides produced. Small roundworms such as Caenorhabditis elegans release chemical signals called ascarosides in order to communicate with other worms of the same species. Using the ascarosides, the worm can tell its friends, for example, how crowded the neighborhood is and whether there is enough food. The ascarosides thus help the worms in the population decide whether the neighborhood is good – meaning they should hang around, eat, and make babies – or whether the neighborhood is bad. If so, the worms should develop into a larval stage specialized for dispersal that will allow them to find a better neighborhood. Roundworms make the ascarosides by attaching a long chemical ‘side chain’ to an ascarylose sugar. Further chemical modifications allow the worms to produce different signals. In general, to signal a good neighborhood, worms attach a structure called an indole group to the ascarosides. To signal a bad neighborhood, worms make the side chain very short. But how does a worm control which ascarosides it makes? Zhou, Wang et al. now show that C. elegans can change the meaning of its chemical message by modifying the ascarosides that it has already produced instead of making new ones from scratch. Specifically, as their neighborhood runs out of food, C. elegans can use an enzyme called ACS-7 to initiate the shortening of the side chains of indole-ascarosides. The worm can thus change a favorable ascaroside signal that causes the worms to group together into an unfavorable ascaroside signal that causes the worms to enter their dispersal stage. Although Zhou, Wang et al. have focused on chemical communication in C. elegans, the findings could easily apply to the many other species of roundworm that produce ascarosides. Knowing how worms communicate will help us to understand how worms respond to their environment. This knowledge could potentially be used to interfere with the lifecycles and survival of parasitic worm species that harm health and crops.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Chemistry, University of Florida, Gainesville, United States
| | - Yuting Wang
- Department of Chemistry, University of Florida, Gainesville, United States
| | - Xinxing Zhang
- Department of Chemistry, University of Florida, Gainesville, United States
| | - Subhradeep Bhar
- Department of Chemistry, University of Florida, Gainesville, United States
| | | | - Jungsoo Han
- Department of Chemistry, University of Florida, Gainesville, United States
| | - Likui Feng
- Department of Chemistry, University of Florida, Gainesville, United States
| | - Rebecca A Butcher
- Department of Chemistry, University of Florida, Gainesville, United States
| |
Collapse
|
35
|
Poppelreuther M, Sander S, Minden F, Dietz MS, Exner T, Du C, Zhang I, Ehehalt F, Knüppel L, Domschke S, Badenhop A, Staudacher S, Ehehalt R, Stremmel W, Thiele C, Heilemann M, Füllekrug J. The metabolic capacity of lipid droplet localized acyl-CoA synthetase 3 is not sufficient to support local triglyceride synthesis independent of the endoplasmic reticulum in A431 cells. Biochim Biophys Acta Mol Cell Biol Lipids 2018. [PMID: 29526665 DOI: 10.1016/j.bbalip.2018.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
ACSL3 is the only long chain fatty acyl-CoA synthetase consistently found on growing and mature lipid droplets (LDs), suggesting that this specific localization has biological relevance. Current models for LD growth propose that triglycerides are synthesized by enzymes at the LD surface, with activated fatty acids provided by LD localized ACSL3, thus allowing growth independent of the ER. Here, we tested this hypothesis by quantifying ACSL3 on LDs from human A431 cells. RNAi of ACSL3 reduced the oleoyl-CoA synthetase activity by 83%, suggesting that ACSL3 is by far the dominant enzyme of A431 cells. Molar quantification revealed that there are 1.4 million ACSL3 molecules within a single cell. Metabolic labeling indicated that each ACSL3 molecule contributed a net gain of 3.1 oleoyl-CoA/s. 3D reconstruction of confocal images demonstrated that 530 individual lipid droplets were present in an average oleate fed A431 cell. A representative single lipid droplet with a diameter of 0.66 μm contained 680 ACSL3 molecules on the surface. Subcellular fractionation showed that at least 68% of ACSL3 remain at the ER even during extensive fatty acid supplementation. High resolution single molecule microscopy confirmed the abundance of cytoplasmic ACSL3 outside of LDs. Model calculations for triglyceride synthesis using only LD localized ACSL3 gave significant slower growth of LDs as observed experimentally. In conclusion, although ACSL3 is an abundant enzyme on A431 LDs, the metabolic capacity is not sufficient to account for LD growth solely by the local synthesis of triglycerides.
Collapse
Affiliation(s)
| | - Simone Sander
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Fadil Minden
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Marina S Dietz
- Institute for Physical and Theoretical Chemistry, Goethe-University Frankfurt, Germany
| | - Tarik Exner
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Chen Du
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Ingrid Zhang
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Friedrich Ehehalt
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Laura Knüppel
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Susanne Domschke
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Anna Badenhop
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Sarah Staudacher
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Robert Ehehalt
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Wolfgang Stremmel
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany
| | - Christoph Thiele
- Life and Medical Sciences Institute, University of Bonn, Germany
| | - Mike Heilemann
- Institute for Physical and Theoretical Chemistry, Goethe-University Frankfurt, Germany
| | - Joachim Füllekrug
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, Germany.
| |
Collapse
|
36
|
Brain docosahexaenoic acid uptake and metabolism. Mol Aspects Med 2018; 64:109-134. [PMID: 29305120 DOI: 10.1016/j.mam.2017.12.004] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/21/2017] [Accepted: 12/28/2017] [Indexed: 12/22/2022]
Abstract
Docosahexaenoic acid (DHA) is the most abundant n-3 polyunsaturated fatty acid in the brain where it serves to regulate several important processes and, in addition, serves as a precursor to bioactive mediators. Given that the capacity of the brain to synthesize DHA locally is appreciably low, the uptake of DHA from circulating lipid pools is essential to maintaining homeostatic levels. Although, several plasma pools have been proposed to supply the brain with DHA, recent evidence suggests non-esterified-DHA and lysophosphatidylcholine-DHA are the primary sources. The uptake of DHA into the brain appears to be regulated by a number of complementary pathways associated with the activation and metabolism of DHA, and may provide mechanisms for enrichment of DHA within the brain. Following entry into the brain, DHA is esterified into and recycled amongst membrane phospholipids contributing the distribution of DHA in brain phospholipids. During neurotransmission and following brain injury, DHA is released from membrane phospholipids and converted to bioactive mediators which regulate signaling pathways important to synaptogenesis, cell survival, and neuroinflammation, and may be relevant to treating neurological diseases. In the present review, we provide a comprehensive overview of brain DHA metabolism, encompassing many of the pathways and key enzymatic regulators governing brain DHA uptake and metabolism. In addition, we focus on the release of non-esterified DHA and subsequent production of bioactive mediators and the evidence of their proposed activity within the brain. We also provide a brief review of the evidence from post-mortem brain analyses investigating DHA levels in the context of neurological disease and mood disorder, highlighting the current disparities within the field.
Collapse
|
37
|
Soupene E, Kuypers FA. Phosphatidylserine decarboxylase CT699, lysophospholipid acyltransferase CT775, and acyl-ACP synthase CT776 provide membrane lipid diversity to Chlamydia trachomatis. Sci Rep 2017; 7:15767. [PMID: 29150677 PMCID: PMC5693948 DOI: 10.1038/s41598-017-16116-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/07/2017] [Indexed: 02/08/2023] Open
Abstract
De novo lipid synthesis and scavenging of fatty acids (FA) are processes essential for the formation of the membrane of the human pathogen Chlamydia trachomatis (C.t.). Host FA are assimilated via esterification by the bacterial acyl-acyl carrier protein (ACP) synthase AasC but inhibitors of the host acyl-CoA synthetase enymes ACSL also impaired growth of C.t. in human cells. In E. coli, activity of AasC was sensitive to triacsin C and rosiglitazone G. The absence of a triacsin C-insensitive pathway and the increased inhibition by rosiglitazone G confirmed the sensitivity of the bacterial acyl-ACP synthase to these drugs in infected human cells. We found no evidence that the human ACSL enzymes are required for lipid formation by C.t. The broad substrate specificity of acyltransferase CT775 provides C.t. with the capacity to incorporate straight-chain and bacterial specific branched-chain fatty acids. CT775 accepts both acyl-ACP and acyl-CoA as acyl donors and, 1- or 2-acyl isomers of lysophosphoplipids as acyl acceptors. The enzyme responsible for remodeling of human phosphatidylserine to bacterial phosphatidylethanolamine was identified as CT699. These findings provide evidence that the pathogen has the ability to extend the lipid diversity of its membrane.
Collapse
Affiliation(s)
- Eric Soupene
- Children's Hospital Oakland Research Institute, Oakland, CA, USA.
| | - Frans A Kuypers
- Children's Hospital Oakland Research Institute, Oakland, CA, USA
| |
Collapse
|
38
|
Triacsin C reduces lipid droplet formation and induces mitochondrial biogenesis in primary rat hepatocytes. J Bioenerg Biomembr 2017; 49:399-411. [PMID: 28918598 DOI: 10.1007/s10863-017-9725-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/04/2017] [Indexed: 12/21/2022]
Abstract
Intracellular long-chain acyl-CoA synthetases (ACSL) activate fatty acids to produce acyl-CoA, which undergoes β-oxidation and participates in the synthesis of esterified lipids such as triacylglycerol (TAG). Imbalances in these metabolic routes are closely associated with the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Triacsin C is one of the few compounds that inhibit TAG accumulation into lipid droplets (LD) by suppressing ACSL activity. Here we report that treatment of primary rat hepatocytes with triacsin C at concentrations lower than the IC50 (4.1 μM) for LD formation: (i) diminished LD number in a concentration-dependent manner; (ii) increased mitochondrial amount; (iii) markedly improved mitochondrial metabolism by enhancing the β-oxidation efficiency, electron transport chain capacity, and degree of coupling - treatment of isolated rat liver mitochondria with the same triacsin C concentrations did not affect the last two parameters; (iv) decreased the GSH/GSSG ratio and elevated the protein carbonyl level, which suggested an increased reactive oxygen species production, as observed in isolated mitochondria. The hepatocyte mitochondrial improvements were not related to either the transcriptional levels of PGC-1α or the content of mTOR and phosphorylated AMPK. Triacsin C at 10 μM induced hepatocyte death by necrosis and/or apoptosis through mechanisms associated with mitochondrial permeability transition pore opening, as demonstrated by experiments using isolated mitochondria. Therefore, triacsin C at sub-IC50 concentrations modulates the lipid imbalance by shifting hepatocytes to a more oxidative state and enhancing the fatty acid consumption, which can in turn accelerate lipid oxidation and reverse NAFLD in long-term therapies.
Collapse
|
39
|
Geillon F, Gondcaille C, Raas Q, Dias AMM, Pecqueur D, Truntzer C, Lucchi G, Ducoroy P, Falson P, Savary S, Trompier D. Peroxisomal ATP-binding cassette transporters form mainly tetramers. J Biol Chem 2017; 292:6965-6977. [PMID: 28258215 DOI: 10.1074/jbc.m116.772806] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Indexed: 12/22/2022] Open
Abstract
ABCD1 and its homolog ABCD2 are peroxisomal ATP-binding cassette (ABC) half-transporters of fatty acyl-CoAs with both distinct and overlapping substrate specificities. Although it is established that ABC half-transporters have at least to dimerize to generate a functional unit, functional equivalents of tetramers (i.e. dimers of full-length transporters) have also been reported. However, oligomerization of peroxisomal ABCD transporters is incompletely understood but is of potential significance because more complex oligomerization might lead to differences in substrate specificity. In this work, we have characterized the quaternary structure of the ABCD1 and ABCD2 proteins in the peroxisomal membrane. Using various biochemical approaches, we clearly demonstrate that both transporters exist as both homo- and heterotetramers, with a predominance of homotetramers. In addition to tetramers, some larger molecular ABCD assemblies were also found but represented only a minor fraction. By using quantitative co-immunoprecipitation assays coupled with tandem mass spectrometry, we identified potential binding partners of ABCD2 involved in polyunsaturated fatty-acid metabolism. Interestingly, we identified calcium ATPases as ABCD2-binding partners, suggesting a role of ABCD2 in calcium signaling. In conclusion, we have shown here that ABCD1 and its homolog ABCD2 exist mainly as homotetramers in the peroxisomal membrane.
Collapse
Affiliation(s)
| | | | | | | | - Delphine Pecqueur
- CLIPP-ICMUB, Université Bourgogne-Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France and
| | - Caroline Truntzer
- CLIPP-ICMUB, Université Bourgogne-Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France and
| | - Géraldine Lucchi
- CLIPP-ICMUB, Université Bourgogne-Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France and
| | - Patrick Ducoroy
- CLIPP-ICMUB, Université Bourgogne-Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France and
| | - Pierre Falson
- the Drug Resistance and Membrane Proteins Team, Molecular Microbiology and Structural Biochemistry Laboratory, Institut de Biologie et Chimie des Protéines (IBCP), UMR5086 CNRS/Université Lyon 1, 7 Passage du Vercors, 69367 Lyon, France
| | | | | |
Collapse
|
40
|
Arlia-Ciommo A, Svistkova V, Mohtashami S, Titorenko VI. A novel approach to the discovery of anti-tumor pharmaceuticals: searching for activators of liponecrosis. Oncotarget 2017; 7:5204-25. [PMID: 26636650 PMCID: PMC4868681 DOI: 10.18632/oncotarget.6440] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/21/2015] [Indexed: 02/04/2023] Open
Abstract
A recently conducted chemical genetic screen for pharmaceuticals that can extend longevity of the yeast Saccharomyces cerevisiae has identified lithocholic acid as a potent anti-aging molecule. It was found that this hydrophobic bile acid is also a selective anti-tumor chemical compound; it kills different types of cultured cancer cells if used at concentrations that do not compromise the viability of non-cancerous cells. These studies have revealed that yeast can be successfully used as a model organism for high-throughput screens aimed at the discovery of selectively acting anti-tumor small molecules. Two metabolic traits of rapidly proliferating fermenting yeast, namely aerobic glycolysis and lipogenesis, are known to be similar to those of cancer cells. The mechanisms underlying these key metabolic features of cancer cells and fermenting yeast have been established; such mechanisms are discussed in this review. We also suggest how a yeast-based chemical genetic screen can be used for the high-throughput development of selective anti-tumor pharmaceuticals that kill only cancer cells. This screen consists of searching for chemical compounds capable of increasing the abundance of membrane lipids enriched in unsaturated fatty acids that would therefore be toxic only to rapidly proliferating cells, such as cancer cells and fermenting yeast.
Collapse
Affiliation(s)
| | | | - Sadaf Mohtashami
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | |
Collapse
|
41
|
Liver-specific ATP-citrate lyase inhibition by bempedoic acid decreases LDL-C and attenuates atherosclerosis. Nat Commun 2016; 7:13457. [PMID: 27892461 PMCID: PMC5133702 DOI: 10.1038/ncomms13457] [Citation(s) in RCA: 289] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/06/2016] [Indexed: 12/25/2022] Open
Abstract
Despite widespread use of statins to reduce low-density lipoprotein cholesterol (LDL-C) and associated atherosclerotic cardiovascular risk, many patients do not achieve sufficient LDL-C lowering due to muscle-related side effects, indicating novel treatment strategies are required. Bempedoic acid (ETC-1002) is a small molecule intended to lower LDL-C in hypercholesterolemic patients, and has been previously shown to modulate both ATP-citrate lyase (ACL) and AMP-activated protein kinase (AMPK) activity in rodents. However, its mechanism for LDL-C lowering, efficacy in models of atherosclerosis and relevance in humans are unknown. Here we show that ETC-1002 is a prodrug that requires activation by very long-chain acyl-CoA synthetase-1 (ACSVL1) to modulate both targets, and that inhibition of ACL leads to LDL receptor upregulation, decreased LDL-C and attenuation of atherosclerosis, independently of AMPK. Furthermore, we demonstrate that the absence of ACSVL1 in skeletal muscle provides a mechanistic basis for ETC-1002 to potentially avoid the myotoxicity associated with statin therapy.
Collapse
|
42
|
Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, Prokisch H, Trümbach D, Mao G, Qu F, Bayir H, Füllekrug J, Scheel CH, Wurst W, Schick JA, Kagan VE, Angeli JPF, Conrad M. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol 2016; 13:91-98. [PMID: 27842070 DOI: 10.1038/nchembio.2239] [Citation(s) in RCA: 2191] [Impact Index Per Article: 273.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 09/13/2016] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a form of regulated necrotic cell death controlled by glutathione peroxidase 4 (GPX4). At present, mechanisms that could predict sensitivity and/or resistance and that may be exploited to modulate ferroptosis are needed. We applied two independent approaches-a genome-wide CRISPR-based genetic screen and microarray analysis of ferroptosis-resistant cell lines-to uncover acyl-CoA synthetase long-chain family member 4 (ACSL4) as an essential component for ferroptosis execution. Specifically, Gpx4-Acsl4 double-knockout cells showed marked resistance to ferroptosis. Mechanistically, ACSL4 enriched cellular membranes with long polyunsaturated ω6 fatty acids. Moreover, ACSL4 was preferentially expressed in a panel of basal-like breast cancer cell lines and predicted their sensitivity to ferroptosis. Pharmacological targeting of ACSL4 with thiazolidinediones, a class of antidiabetic compound, ameliorated tissue demise in a mouse model of ferroptosis, suggesting that ACSL4 inhibition is a viable therapeutic approach to preventing ferroptosis-related diseases.
Collapse
Affiliation(s)
- Sebastian Doll
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Yulia Y Tyurina
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elena Panzilius
- Institute of Stem Cell Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Sho Kobayashi
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Irina Ingold
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Michaela Aichler
- Institute of Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Axel Walch
- Institute of Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Technische Universität München, Institute of Human Genetics, München, Germany
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gaowei Mao
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Feng Qu
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hulya Bayir
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joachim Füllekrug
- Department of Gastroenterology, University of Heidelberg, Heidelberg, Germany
| | - Christina H Scheel
- Institute of Stem Cell Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Joel A Schick
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
43
|
Teodoro BG, Sampaio IH, Bomfim LHM, Queiroz AL, Silveira LR, Souza AO, Fernandes AMAP, Eberlin MN, Huang TY, Zheng D, Neufer PD, Cortright RN, Alberici LC. Long-chain acyl-CoA synthetase 6 regulates lipid synthesis and mitochondrial oxidative capacity in human and rat skeletal muscle. J Physiol 2016; 595:677-693. [PMID: 27647415 DOI: 10.1113/jp272962] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/11/2016] [Indexed: 12/29/2022] Open
Abstract
KEY POINTS Long-chain acyl-CoA synthetase 6 (ACSL6) mRNA is present in human and rat skeletal muscle, and is modulated by nutritional status: exercise and fasting decrease ACSL6 mRNA, whereas acute lipid ingestion increase its expression. ACSL6 genic inhibition in rat primary myotubes decreased lipid accumulation, as well as activated the higher mitochondrial oxidative capacity programme and fatty acid oxidation through the AMPK/PGC1-α pathway. ACSL6 overexpression in human primary myotubes increased phospholipid species and decreased oxidative metabolism. ABSTRACT Long-chain acyl-CoA synthetases (ACSL 1 to 6) are key enzymes regulating the partitioning of acyl-CoA species toward different metabolic fates such as lipid synthesis or β-oxidation. Despite our understanding of ecotopic lipid accumulation in skeletal muscle being associated with metabolic diseases such as obesity and type II diabetes, the role of specific ACSL isoforms in lipid synthesis remains unclear. In the present study, we describe for the first time the presence of ACSL6 mRNA in human skeletal muscle and the role that ACSL6 plays in lipid synthesis in both rodent and human skeletal muscle. ACSL6 mRNA was observed to be up-regulated by acute high-fat meal ingestion in both rodents and humans. In rats, we also demonstrated that fasting and chronic aerobic training negatively modulated the ACSL6 mRNA and other genes of lipid synthesis. Similar results were obtained following ACSL6 knockdown in rat myotubes, which was associated with a decreased accumulation of TAGs and lipid droplets. Under the same knockdown condition, we further demonstrate an increase in fatty acid content, p-AMPK, mitochondrial content, mitochondrial respiratory rates and palmitate oxidation. These results were associated with increased PGC-1α, UCP2 and UCP3 mRNA and decreased reactive oxygen species production. In human myotubes, ACSL6 overexpression reduced palmitate oxidation and PGC-1α mRNA. In conclusion, ACSL6 drives acyl-CoA toward lipid synthesis and its downregulation improves mitochondrial biogenesis, respiratory capacity and lipid oxidation. These outcomes are associated with the activation of the AMPK/PGC1-α pathway.
Collapse
Affiliation(s)
- Bruno G Teodoro
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.,Federal Institute of Education Science and Technology of São Paulo, Sertãozinho, São Paulo, Brazil
| | - Igor H Sampaio
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Lucas H M Bomfim
- Department of Structural and Functional Biology, Institute of Biology
| | - André L Queiroz
- Department of Structural and Functional Biology, Institute of Biology
| | | | - Anderson O Souza
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Anna M A P Fernandes
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Marcos N Eberlin
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | | - Donghai Zheng
- Department of Kinesiology.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - P Darrell Neufer
- Department of Physiology.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Ronald N Cortright
- Department of Kinesiology.,Department of Physiology.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Luciane C Alberici
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
44
|
Huang Y, Huang S, Lam SM, Liu Z, Shui G, Zhang YQ. Acsl, the Drosophila ortholog of intellectual-disability-related ACSL4, inhibits synaptic growth by altered lipids. J Cell Sci 2016; 129:4034-4045. [PMID: 27656110 DOI: 10.1242/jcs.195032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/16/2016] [Indexed: 12/17/2022] Open
Abstract
Nervous system development and function are tightly regulated by metabolic processes, including the metabolism of lipids such as fatty acids. Mutations in long-chain acyl-CoA synthetase 4 (ACSL4) are associated with non-syndromic intellectual disabilities. We previously reported that Acsl, the Drosophila ortholog of mammalian ACSL3 and ACSL4, inhibits neuromuscular synapse growth by suppressing bone morphogenetic protein (BMP) signaling. Here, we report that Acsl regulates the composition of fatty acids and membrane lipids, which in turn affects neuromuscular junction (NMJ) synapse development. Acsl mutant brains had a decreased abundance of C16:1 fatty acyls; restoration of Acsl expression abrogated NMJ overgrowth and the increase in BMP signaling. A lipidomic analysis revealed that Acsl suppressed the levels of three lipid raft components in the brain, including mannosyl glucosylceramide (MacCer), phosphoethanolamine ceramide and ergosterol. The MacCer level was elevated in Acsl mutant NMJs and, along with sterol, promoted NMJ overgrowth, but was not associated with the increase in BMP signaling in the mutants. These findings suggest that Acsl inhibits NMJ growth by stimulating C16:1 fatty acyl production and concomitantly suppressing raft-associated lipid levels.
Collapse
Affiliation(s)
- Yan Huang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Sheng Huang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Sin Man Lam
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhihua Liu
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guanghou Shui
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Q Zhang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
45
|
Seeger DR, Murphy CC, Murphy EJ. Astrocyte arachidonate and palmitate uptake and metabolism is differentially modulated by dibutyryl-cAMP treatment. Prostaglandins Leukot Essent Fatty Acids 2016; 110:16-26. [PMID: 27255639 DOI: 10.1016/j.plefa.2016.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 12/29/2022]
Abstract
Astrocytes play a vital role in brain lipid metabolism; however the impact of the phenotypic shift in astrocytes to a reactive state on arachidonic acid metabolism is unknown. Therefore, we determined the impact of dibutyryl-cAMP (dBcAMP) treatment on radiolabeled arachidonic acid ([1-(14)C]20:4n-6) and palmitic acid ([1-(14)C]16:0) uptake and metabolism in primary cultured murine cortical astrocytes. In dBcAMP treated astrocytes, total [1-(14)C]20:4n-6 uptake was increased 1.9-fold compared to control, while total [1-(14)C]16:0 uptake was unaffected. Gene expression of long-chain acyl-CoA synthetases (Acsl), acyl-CoA hydrolase (Acot7), fatty acid binding protein(s) (Fabp) and alpha-synuclein (Snca) were determined using qRT-PCR. dBcAMP treatment increased expression of Acsl3 (4.8-fold) and Acsl4 (1.3-fold), which preferentially use [1-(14)C]20:4n-6 and are highly expressed in astrocytes, consistent with the increase in [1-(14)C]20:4n-6 uptake. However, expression of Fabp5 and Fabp7 were significantly reduced by 25% and 45%, respectively. Acot7 (20%) was also reduced, suggesting dBcAMP treatment favors acyl-CoA formation. dBcAMP treatment enhanced [1-(14)C]20:4n-6 (2.2-fold) and [1-(14)C]16:0 (1.6-fold) esterification into total phospholipids, but the greater esterification of [1-(14)C]20:4n-6 is consistent with the observed uptake through increased Acsl, but not Fabp expression. Although total [1-(14)C]16:0 uptake was not affected, there was a dramatic decrease in [1-(14)C]16:0 in the free fatty acid pool as esterification into the phospholipid pool was increased, which is consistent with the increase in Acsl3 and Acsl4 expression. In summary, our data demonstrates that dBcAMP treatment increases [1-(14)C]20:4n-6 uptake in astrocytes and this increase appears to be due to increased expression of Acsl3 and Acsl4 coupled with a reduction in Acot7 expression.
Collapse
Affiliation(s)
- D R Seeger
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - C C Murphy
- Department of Nutrition, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - E J Murphy
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58203, USA.
| |
Collapse
|
46
|
Andersen MK, Jørsboe E, Sandholt CH, Grarup N, Jørgensen ME, Færgeman NJ, Bjerregaard P, Pedersen O, Moltke I, Hansen T, Albrechtsen A. Identification of Novel Genetic Determinants of Erythrocyte Membrane Fatty Acid Composition among Greenlanders. PLoS Genet 2016; 12:e1006119. [PMID: 27341449 PMCID: PMC4920407 DOI: 10.1371/journal.pgen.1006119] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/20/2016] [Indexed: 11/25/2022] Open
Abstract
Fatty acids (FAs) are involved in cellular processes important for normal body function, and perturbation of FA balance has been linked to metabolic disturbances, including type 2 diabetes. An individual’s level of FAs is affected by diet, lifestyle, and genetic variation. We aimed to improve the understanding of the mechanisms and pathways involved in regulation of FA tissue levels, by identifying genetic loci associated with inter-individual differences in erythrocyte membrane FA levels. We assessed the levels of 22 FAs in the phospholipid fraction of erythrocyte membranes from 2,626 Greenlanders in relation to single nucleotide polymorphisms genotyped on the MetaboChip or imputed. We identified six independent association signals. Novel loci were identified on chromosomes 5 and 11 showing strongest association with oleic acid (rs76430747 in ACSL6, beta (SE): -0.386% (0.034), p = 1.8x10-28) and docosahexaenoic acid (rs6035106 in DTD1, 0.137% (0.025), p = 6.4x10-8), respectively. For a missense variant (rs80356779) in CPT1A, we identified a number of novel FA associations, the strongest with 11-eicosenoic acid (0.473% (0.035), p = 2.6x10-38), and for variants in FADS2 (rs174570), LPCAT3 (rs2110073), and CERS4 (rs11881630) we replicated known FA associations. Moreover, we observed metabolic implications of the ACSL6 (rs76430747) and CPT1A (rs80356779) variants, which both were associated with altered HbA1c (0.051% (0.013), p = 5.6x10-6 and -0.034% (0.016), p = 3.1x10-4, respectively). The latter variant was also associated with reduced insulin resistance (HOMA-IR, -0.193 (0.050), p = 3.8x10-6), as well as measures of smaller body size, including weight (-2.676 kg (0.523), p = 2.4x10-7), lean mass (-1.200 kg (0.271), p = 1.7x10-6), height (-0.966 cm (0.230), p = 2.0x10-5), and BMI (-0.638 kg/m2 (0.181), p = 2.8x10-4). In conclusion, we have identified novel genetic determinants of FA composition in phospholipids in erythrocyte membranes, and have shown examples of links between genetic variants associated with altered FA membrane levels and changes in metabolic traits. Disruption of fatty-acid balance has in several previous studies been linked to human health conditions, including the metabolic syndrome, type 2 diabetes, and insulin resistance. Composition of fatty acids in lipid membranes is influenced, not only by diet and lifestyle, but also by genetic variation. By identifying genes linked to changes in the level of specific fatty acids, it may be possible to identify biological mechanisms and pathways central to regulation of fatty-acid composition in lipid membranes. We therefore aimed at finding such genes by studying Greenlanders. We identified six genomic regions harboring variants, which were associated with the level of at least one of 22 assessed erythrocyte membrane fatty acids, including two novel regions not previously linked to fatty acid levels. Moreover, we showed that two of the identified variants were associated with altered levels of glycosylated hemoglobin, and one of these variants was associated with reduced insulin resistance and decreased measures of body size. These results contribute to our understanding of fatty acid metabolism, and support a link between fatty acid balance and metabolic health.
Collapse
Affiliation(s)
- Mette Korre Andersen
- Section for Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emil Jørsboe
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Helene Sandholt
- Section for Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Section for Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Nils Joakim Færgeman
- Villum Center for Bioanalytical Sciences, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Peter Bjerregaard
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
- Greenland Centre for Health Research, University of Greenland, Nuuk, Greenland
| | - Oluf Pedersen
- Section for Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida Moltke
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (IM); (TH); (AA)
| | - Torben Hansen
- Section for Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- * E-mail: (IM); (TH); (AA)
| | - Anders Albrechtsen
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (IM); (TH); (AA)
| |
Collapse
|
47
|
Kolahi K, Louey S, Varlamov O, Thornburg K. Real-Time Tracking of BODIPY-C12 Long-Chain Fatty Acid in Human Term Placenta Reveals Unique Lipid Dynamics in Cytotrophoblast Cells. PLoS One 2016; 11:e0153522. [PMID: 27124483 PMCID: PMC4849650 DOI: 10.1371/journal.pone.0153522] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/30/2016] [Indexed: 01/10/2023] Open
Abstract
While the human placenta must provide selected long-chain fatty acids to support the developing fetal brain, little is known about the mechanisms underlying the transport process. We tracked the movement of the fluorescently labeled long-chain fatty acid analogue, BODIPY-C12, across the cell layers of living explants of human term placenta. Although all layers took up the fatty acid, rapid esterification of long-chain fatty acids and incorporation into lipid droplets was exclusive to the inner layer cytotrophoblast cells rather than the expected outer syncytiotrophoblast layer. Cytotrophoblast is a progenitor cell layer previously relegated to a repair role. As isolated cytotrophoblasts differentiated into syncytialized cells in culture, they weakened their lipid processing capacity. Syncytializing cells suppress previously active genes that regulate fatty-acid uptake (SLC27A2/FATP2, FABP4, ACSL5) and lipid metabolism (GPAT3, LPCAT3). We speculate that cytotrophoblast performs a previously unrecognized role in regulating placental fatty acid uptake and metabolism.
Collapse
Affiliation(s)
- Kevin Kolahi
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, United States of America
- Center for Developmental Health, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Samantha Louey
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Oleg Varlamov
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Kent Thornburg
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, United States of America
- Center for Developmental Health, Oregon Health and Science University, Portland, Oregon, United States of America
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
48
|
Alves-Bezerra M, Klett EL, De Paula IF, Ramos IB, Coleman RA, Gondim KC. Long-chain acyl-CoA synthetase 2 knockdown leads to decreased fatty acid oxidation in fat body and reduced reproductive capacity in the insect Rhodnius prolixus. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:650-62. [PMID: 27091636 DOI: 10.1016/j.bbalip.2016.04.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/16/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022]
Abstract
Long-chain acyl-CoA esters are important intermediates in lipid metabolism and are synthesized from fatty acids by long-chain acyl-CoA synthetases (ACSL). The hematophagous insect Rhodnius prolixus, a vector of Chagas' disease, produces glycerolipids in the midgut after a blood meal, which are stored as triacylglycerol in the fat body and eggs. We identified twenty acyl-CoA synthetase genes in R. prolixus, two encoding ACSL isoforms (RhoprAcsl1 and RhoprAcsl2). RhoprAcsl1 transcripts increased in posterior midgut on the second day after feeding, and RhoprAcsl2 was highly transcribed on the tenth day. Both enzymes were expressed in Escherichia coli. Recombinant RhoprACSL1 and RhoprACSL2 had broad pH optima (7.5-9.5 and 6.5-9.5, respectively), were inhibited by triacsin C, and were rosiglitazone-insensitive. Both showed similar apparent Km for palmitic and oleic acid (2-6 μM), but different Km for arachidonic acid (0.5 and 6 μM for RhoprACSL1-Flag and RhoprACSL2-Flag, respectively). The knockdown of RhoprAcsl1 did not result in noticeable phenotypes. However, RhoprACSL2 deficient insects exhibited a 2.5-fold increase in triacylglycerol content in the fat body, and 90% decrease in fatty acid β-oxidation. RhoprAcsl2 knockdown also resulted in 20% increase in lifespan, delayed digestion, 30% reduced oviposition, and 50% reduction in egg hatching. Laid eggs and hatched nymphs showed remarkable alterations in morphology. In summary, R. prolixus ACSL isoforms have distinct roles on lipid metabolism. Although RhoprACSL1 functions remain unclear, we propose that RhoprACSL2 is the main contributor for the formation of the intracellular acyl-CoA pool channeled for β-oxidation in the fat body, and is also required for normal reproduction.
Collapse
Affiliation(s)
- Michele Alves-Bezerra
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eric L Klett
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Iron F De Paula
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Isabela B Ramos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rosalind A Coleman
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Katia C Gondim
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
49
|
Recuero-Checa MA, Sharma M, Lau C, Watkins PA, Gaydos CA, Dean D. Chlamydia trachomatis growth and development requires the activity of host Long-chain Acyl-CoA Synthetases (ACSLs). Sci Rep 2016; 6:23148. [PMID: 26988341 PMCID: PMC4796813 DOI: 10.1038/srep23148] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/25/2016] [Indexed: 11/17/2022] Open
Abstract
The obligate-intracellular pathogen Chlamydia trachomatis (Ct) has undergone considerable genome reduction with consequent dependence on host biosynthetic pathways, metabolites and enzymes. Long-chain acyl-CoA synthetases (ACSLs) are key host-cell enzymes that convert fatty acids (FA) into acyl-CoA for use in metabolic pathways. Here, we show that the complete host ACSL family [ACSL1 and ACSL3-6] translocates into the Ct membrane-bound vacuole, termed inclusion, and remains associated with membranes of metabolically active forms of Ct throughout development. We discovered that three different pharmacologic inhibitors of ACSL activity independently impede Ct growth in a dose-dependent fashion. Using an FA competition assay, host ACSLs were found to activate Ct branched-chain FAs, suggesting that one function of the ACSLs is to activate Ct FAs and host FAs (recruited from the cytoplasm) within the inclusion. Because the ACSL inhibitors can deplete lipid droplets (LD), we used a cell line where LD synthesis was switched off to evaluate whether LD deficiency affects Ct growth. In these cells, we found no effect on growth or on translocation of ACSLs into the inclusion. Our findings support an essential role for ACSL activation of host-cell and bacterial FAs within the inclusion to promote Ct growth and development, independent of LDs.
Collapse
Affiliation(s)
- Maria A. Recuero-Checa
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children’s Hospital Oakland Research Institute, Oakland, CA, 94609, USA
- Department of Infectious Disease, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Manu Sharma
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children’s Hospital Oakland Research Institute, Oakland, CA, 94609, USA
| | - Constance Lau
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children’s Hospital Oakland Research Institute, Oakland, CA, 94609, USA
| | - Paul A. Watkins
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Charlotte A. Gaydos
- Department of Infectious Disease, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Deborah Dean
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children’s Hospital Oakland Research Institute, Oakland, CA, 94609, USA
- Department of Bioengineering, University of California at Berkeley and San Francisco, CA, USA
| |
Collapse
|
50
|
Ye X, Zhang Y, Wang X, Li Y, Gao Y. Tumor-suppressive functions of long-chain acyl-CoA synthetase 4 in gastric cancer. IUBMB Life 2016; 68:320-7. [PMID: 26949059 DOI: 10.1002/iub.1486] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/01/2016] [Indexed: 12/19/2022]
Abstract
Long chain acyl CoA synthetase 4 (ACSL4) is a key enzyme in fatty acid metabolism with marked preference for arachidonic acid (AA). Recent reports have implicated its crucial roles in tumorigenesis. However in gastric cancer (GC), the expression and function of ACSL4 remain unclear. In the present study, we identified ACSL4 as a potential tumor suppressor in GC. The ACSL4 expression in GC samples was evaluated by real-time PCR and immunohistochemistry. The results indicated that the mRNA and protein levels of ACSL4 were frequently downregulated in cancer tissues compared with the adjacent non-cancerous mucosa control tissues. Cell-based functional assays exhibited that ectopic expression of ACSL4 inhibits cell growth, colony formation and cell migration, whereas ACSL4 knockdown enhanced these effects. In a nude mice model, ACSL4 knockdown also promoted subcutaneous xenografts' growth in vivo. Moreover, western blot analysis revealed that ACSL4 expression had a significant effect on FAK and P21 protein level. These findings suggest that ACSL4 plays a tumor-suppressive role and could be a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Xiaojuan Ye
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yi Zhang
- Department of Oncology, East Hospital, Dalian Medical University, Shanghai, 200120, China
| | - Xiao Wang
- Department of Oncology, East Hospital, Dalian Medical University, Shanghai, 200120, China
| | - Yandong Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| |
Collapse
|