51
|
Rodencal J, Kim N, Li VL, He A, Lange M, He J, Tarangelo A, Schafer ZT, Olzmann JA, Sage J, Long JZ, Dixon SJ. A Cell Cycle-Dependent Ferroptosis Sensitivity Switch Governed by EMP2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549715. [PMID: 37502927 PMCID: PMC10370086 DOI: 10.1101/2023.07.19.549715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Ferroptosis is a non-apoptotic form of cell death characterized by iron-dependent lipid peroxidation. Ferroptosis can be induced by system xc- cystine/glutamate antiporter inhibition or by direct inhibition of the phospholipid hydroperoxidase glutathione peroxidase 4 (GPX4). The regulation of ferroptosis in response to system xc- inhibition versus direct GPX4 inhibition may be distinct. Here, we show that cell cycle arrest enhances sensitivity to ferroptosis triggered by GPX4 inhibition but not system xc- inhibition. Arrested cells have increased levels of oxidizable polyunsaturated fatty acid-containing phospholipids, which drives sensitivity to GPX4 inhibition. Epithelial membrane protein 2 (EMP2) expression is reduced upon cell cycle arrest and is sufficient to enhance ferroptosis in response to direct GPX4 inhibition. An orally bioavailable GPX4 inhibitor increased markers of ferroptotic lipid peroxidation in vivo in combination with a cell cycle arresting agent. Thus, responses to different ferroptosis-inducing stimuli can be regulated by cell cycle state.
Collapse
Affiliation(s)
- Jason Rodencal
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Nathan Kim
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Veronica L. Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew He
- Departments of Pediatrics and Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mike Lange
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jianping He
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Amy Tarangelo
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Zachary T. Schafer
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - James A. Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Julien Sage
- Departments of Pediatrics and Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jonathan Z. Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Scott J. Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Lead contact
| |
Collapse
|
52
|
Mishima E, Wahida A, Seibt T, Conrad M. Diverse biological functions of vitamin K: from coagulation to ferroptosis. Nat Metab 2023:10.1038/s42255-023-00821-y. [PMID: 37337123 DOI: 10.1038/s42255-023-00821-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/12/2023] [Indexed: 06/21/2023]
Abstract
Vitamin K is essential for several physiological processes, such as blood coagulation, in which it serves as a cofactor for the conversion of peptide-bound glutamate to γ-carboxyglutamate in vitamin K-dependent proteins. This process is driven by the vitamin K cycle facilitated by γ-carboxyglutamyl carboxylase, vitamin K epoxide reductase and ferroptosis suppressor protein-1, the latter of which was recently identified as the long-sought-after warfarin-resistant vitamin K reductase. In addition, vitamin K has carboxylation-independent functions. Akin to ubiquinone, vitamin K acts as an electron carrier for ATP production in some organisms and prevents ferroptosis, a type of cell death hallmarked by lipid peroxidation. In this Perspective, we provide an overview of the diverse functions of vitamin K in physiology and metabolism and, at the same time, offer a perspective on its role in ferroptosis together with ferroptosis suppressor protein-1. A comparison between vitamin K and ubiquinone, from an evolutionary perspective, may offer further insights into the manifold roles of vitamin K in biology.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany.
- Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Tobias Seibt
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
53
|
Shen N, Li Y, Liu Y, Liu Y, Xin H, Cui Y. Gypsophila oldhamiana leaves as a potential industrial resource of lipids, alkaloids, flavonoids and anti-osteoporosis components. INDUSTRIAL CROPS AND PRODUCTS 2023; 196:116510. [DOI: 10.1016/j.indcrop.2023.116510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
|
54
|
Sarparast M, Pourmand E, Hinman J, Vonarx D, Reason T, Zhang F, Paithankar S, Chen B, Borhan B, Watts JL, Alan J, Lee KSS. Dihydroxy-Metabolites of Dihomo-γ-linolenic Acid Drive Ferroptosis-Mediated Neurodegeneration. ACS CENTRAL SCIENCE 2023; 9:870-882. [PMID: 37252355 PMCID: PMC10214511 DOI: 10.1021/acscentsci.3c00052] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Indexed: 05/31/2023]
Abstract
Even after decades of research, the mechanism of neurodegeneration remains understudied, hindering the discovery of effective treatments for neurodegenerative diseases. Recent reports suggest that ferroptosis could be a novel therapeutic target for neurodegenerative diseases. While polyunsaturated fatty acid (PUFA) plays an important role in neurodegeneration and ferroptosis, how PUFAs may trigger these processes remains largely unknown. PUFA metabolites from cytochrome P450 and epoxide hydrolase metabolic pathways may modulate neurodegeneration. Here, we test the hypothesis that specific PUFAs regulate neurodegeneration through the action of their downstream metabolites by affecting ferroptosis. We find that the PUFA dihomo-γ-linolenic acid (DGLA) specifically induces ferroptosis-mediated neurodegeneration in dopaminergic neurons. Using synthetic chemical probes, targeted metabolomics, and genetic mutants, we show that DGLA triggers neurodegeneration upon conversion to dihydroxyeicosadienoic acid through the action of CYP-EH (CYP, cytochrome P450; EH, epoxide hydrolase), representing a new class of lipid metabolites that induce neurodegeneration via ferroptosis.
Collapse
Affiliation(s)
- Morteza Sarparast
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Elham Pourmand
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jennifer Hinman
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Derek Vonarx
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Tommy Reason
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Fan Zhang
- Department
of Pharmacology and Toxicology, Michigan
State University, East Lansing, Michigan 48824, United States
| | - Shreya Paithankar
- Department
of Pediatrics and Human Development, Michigan
State University, Grand Rapids, Michigan 49503, United States
| | - Bin Chen
- Department
of Pharmacology and Toxicology, Michigan
State University, East Lansing, Michigan 48824, United States
- Department
of Pediatrics and Human Development, Michigan
State University, Grand Rapids, Michigan 49503, United States
| | - Babak Borhan
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jennifer L. Watts
- School
of Molecular Biosciences, Washington State
University, Pullman, Washington 99164, United States
| | - Jamie Alan
- Department
of Pharmacology and Toxicology, Michigan
State University, East Lansing, Michigan 48824, United States
| | - Kin Sing Stephen Lee
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Pharmacology and Toxicology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
55
|
Wang Y, Wu W, Gong J. Live or death in cells: from micronutrition metabolism to cell fate. Front Cell Dev Biol 2023; 11:1185989. [PMID: 37250891 PMCID: PMC10213646 DOI: 10.3389/fcell.2023.1185989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
Micronutrients and cell death have a strong relationship and both are essential for human to maintain good body health. Dysregulation of any micronutrients causes metabolic or chronic diseases, including obesity, cardiometabolic condition, neurodegeneration, and cancer. The nematode Caenorhabditis elegans is an ideal genetic organism for researching the mechanisms of micronutrients in metabolism, healthspan, and lifespan. For example, C. elegans is a haem auxotroph, and the research of this special haem trafficking pathway contributes important reference to mammal study. Also, C. elegans characteristics including anatomy simply, clear cell lineage, well-defined genetics, and easily differentiated cell forms make it a powerful tool for studying the mechanisms of cell death including apoptosis, necrosis, autophagy, and ferroptosis. Here, we describe the understanding of micronutrient metabolism currently and also sort out the fundamental mechanisms of different kinds of cell death. A thorough understanding of these physiological processes not only builds a foundation for developing better treatments for various micronutrient disorders but also provides key insights into human health and aging.
Collapse
Affiliation(s)
- Yuting Wang
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianke Gong
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
56
|
Papsdorf K, Miklas JW, Hosseini A, Cabruja M, Morrow CS, Savini M, Yu Y, Silva-García CG, Haseley NR, Murphy LM, Yao P, de Launoit E, Dixon SJ, Snyder MP, Wang MC, Mair WB, Brunet A. Lipid droplets and peroxisomes are co-regulated to drive lifespan extension in response to mono-unsaturated fatty acids. Nat Cell Biol 2023; 25:672-684. [PMID: 37127715 PMCID: PMC10185472 DOI: 10.1038/s41556-023-01136-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/23/2023] [Indexed: 05/03/2023]
Abstract
Dietary mono-unsaturated fatty acids (MUFAs) are linked to longevity in several species. But the mechanisms by which MUFAs extend lifespan remain unclear. Here we show that an organelle network involving lipid droplets and peroxisomes is critical for MUFA-induced longevity in Caenorhabditis elegans. MUFAs upregulate the number of lipid droplets in fat storage tissues. Increased lipid droplet number is necessary for MUFA-induced longevity and predicts remaining lifespan. Lipidomics datasets reveal that MUFAs also modify the ratio of membrane lipids and ether lipids-a signature associated with decreased lipid oxidation. In agreement with this, MUFAs decrease lipid oxidation in middle-aged individuals. Intriguingly, MUFAs upregulate not only lipid droplet number but also peroxisome number. A targeted screen identifies genes involved in the co-regulation of lipid droplets and peroxisomes, and reveals that induction of both organelles is optimal for longevity. Our study uncovers an organelle network involved in lipid homeostasis and lifespan regulation, opening new avenues for interventions to delay aging.
Collapse
Affiliation(s)
| | - Jason W Miklas
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Amir Hosseini
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Matias Cabruja
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Christopher S Morrow
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Marzia Savini
- Department of Molecular and Human Genetics, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Yong Yu
- Department of Molecular and Human Genetics, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Carlos G Silva-García
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Pallas Yao
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Meng C Wang
- Department of Molecular and Human Genetics, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - William B Mair
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA, USA.
- Wu Tsai Institute of Neurosciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
57
|
Wang X, Zhou Y, Min J, Wang F. Zooming in and out of ferroptosis in human disease. Front Med 2023; 17:173-206. [PMID: 37121959 DOI: 10.1007/s11684-023-0992-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/12/2023] [Indexed: 05/02/2023]
Abstract
Ferroptosis is defined as an iron-dependent regulated form of cell death driven by lipid peroxidation. In the past decade, it has been implicated in the pathogenesis of various diseases that together involve almost every organ of the body, including various cancers, neurodegenerative diseases, cardiovascular diseases, lung diseases, liver diseases, kidney diseases, endocrine metabolic diseases, iron-overload-related diseases, orthopedic diseases and autoimmune diseases. Understanding the underlying molecular mechanisms of ferroptosis and its regulatory pathways could provide additional strategies for the management of these disease conditions. Indeed, there are an expanding number of studies suggesting that ferroptosis serves as a bona-fide target for the prevention and treatment of these diseases in relevant pre-clinical models. In this review, we summarize the progress in the research into ferroptosis and its regulatory mechanisms in human disease, while providing evidence in support of ferroptosis as a target for the treatment of these diseases. We also discuss our perspectives on the future directions in the targeting of ferroptosis in human disease.
Collapse
Affiliation(s)
- Xue Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Ningbo First Hospital, Ningbo, 315000, China
| | - Junxia Min
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Fudi Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
58
|
Pan B, Yuan S, Mayernik L, Yap YT, Moin K, Chung CS, Maddipati K, Krawetz SA, Zhang Z, Hess RA, Chen X. Disrupted intercellular bridges and spermatogenesis in fatty acyl-CoA reductase 1 knockout mice: A new model of ether lipid deficiency. FASEB J 2023; 37:e22908. [PMID: 37039784 PMCID: PMC10150578 DOI: 10.1096/fj.202201848r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/10/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023]
Abstract
Peroxisomal fatty acyl-CoA reductase 1 (FAR1) is a rate-limiting enzyme for ether lipid (EL) synthesis. Gene mutations in FAR1 cause a rare human disease. Furthermore, altered EL homeostasis has also been associated with various prevalent human diseases. Despite their importance in human health, the exact cellular functions of FAR1 and EL are not well-understood. Here, we report the generation and initial characterization of the first Far1 knockout (KO) mouse model. Far1 KO mice were subviable and displayed growth retardation. The adult KO male mice had smaller testes and were infertile. H&E and immunofluorescent staining showed fewer germ cells in seminiferous tubules. Round spermatids were present but no elongated spermatids or spermatozoa were observed, suggesting a spermatogenesis arrest at this stage. Large multi-nucleated giant cells (MGC) were found lining the lumen of seminiferous tubules with many of them undergoing apoptosis. The immunofluorescent signal of TEX14, an essential component of intercellular bridges (ICB) between developing germ cells, was greatly reduced and mislocalized in KO testis, suggesting the disrupted ICBs as an underlying cause of MGC formation. Integrative analysis of our total testis RNA-sequencing results and published single-cell RNA-sequencing data unveiled cell type-specific molecular alterations underlying the spermatogenesis arrest. Many genes essential for late germ cell development showed dramatic downregulation, whereas genes essential for extracellular matrix dynamics and cell-cell interactions were among the most upregulated genes. Together, this work identified the cell type-specific requirement of ELs in spermatogenesis and suggested a critical role of Far1/ELs in the formation/maintenance of ICB during meiosis.
Collapse
Affiliation(s)
- Bo Pan
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Shuo Yuan
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
- Department of Occupational and Environmental Medicine, School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Linda Mayernik
- Department of Pharmacology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Yi Tian Yap
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Kamiar Moin
- Department of Pharmacology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Charles S. Chung
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Krishnarao Maddipati
- Department of Pathology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Stephen A. Krawetz
- Department of Obstetrics & Gynecology, Wayne State University, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics & Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Rex A. Hess
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Xuequn Chen
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
59
|
Koeberle SC, Kipp AP, Stuppner H, Koeberle A. Ferroptosis-modulating small molecules for targeting drug-resistant cancer: Challenges and opportunities in manipulating redox signaling. Med Res Rev 2023; 43:614-682. [PMID: 36658724 PMCID: PMC10947485 DOI: 10.1002/med.21933] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023]
Abstract
Ferroptosis is an iron-dependent cell death program that is characterized by excessive lipid peroxidation. Triggering ferroptosis has been proposed as a promising strategy to fight cancer and overcome drug resistance in antitumor therapy. Understanding the molecular interactions and structural features of ferroptosis-inducing compounds might therefore open the door to efficient pharmacological strategies against aggressive, metastatic, and therapy-resistant cancer. We here summarize the molecular mechanisms and structural requirements of ferroptosis-inducing small molecules that target central players in ferroptosis. Focus is placed on (i) glutathione peroxidase (GPX) 4, the only GPX isoenzyme that detoxifies complex membrane-bound lipid hydroperoxides, (ii) the cystine/glutamate antiporter system Xc - that is central for glutathione regeneration, (iii) the redox-protective transcription factor nuclear factor erythroid 2-related factor (NRF2), and (iv) GPX4 repression in combination with induced heme degradation via heme oxygenase-1. We deduce common features for efficient ferroptotic activity and highlight challenges in drug development. Moreover, we critically discuss the potential of natural products as ferroptosis-inducing lead structures and provide a comprehensive overview of structurally diverse biogenic and bioinspired small molecules that trigger ferroptosis via iron oxidation, inhibition of the thioredoxin/thioredoxin reductase system or less defined modes of action.
Collapse
Affiliation(s)
- Solveigh C. Koeberle
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckTirolInnsbruckAustria
- Department of Molecular Nutritional Physiology, Institute of Nutritional SciencesFriedrich Schiller University JenaThüringenJenaGermany
| | - Anna P. Kipp
- Department of Molecular Nutritional Physiology, Institute of Nutritional SciencesFriedrich Schiller University JenaThüringenJenaGermany
| | - Hermann Stuppner
- Unit of Pharmacognosy, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckTirolInnsbruckAustria
| | - Andreas Koeberle
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckTirolInnsbruckAustria
| |
Collapse
|
60
|
Costa I, Barbosa DJ, Silva V, Benfeito S, Borges F, Remião F, Silva R. Research Models to Study Ferroptosis's Impact in Neurodegenerative Diseases. Pharmaceutics 2023; 15:pharmaceutics15051369. [PMID: 37242612 DOI: 10.3390/pharmaceutics15051369] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Ferroptosis is a type of regulated cell death promoted by the appearance of oxidative perturbations in the intracellular microenvironment constitutively controlled by glutathione peroxidase 4 (GPX4). It is characterized by increased production of reactive oxygen species, intracellular iron accumulation, lipid peroxidation, inhibition of system Xc-, glutathione depletion, and decreased GPX4 activity. Several pieces of evidence support the involvement of ferroptosis in distinct neurodegenerative diseases. In vitro and in vivo models allow a reliable transition to clinical studies. Several in vitro models, including differentiated SH-SY5Y and PC12 cells, among others, have been used to investigate the pathophysiological mechanisms of distinct neurodegenerative diseases, including ferroptosis. In addition, they can be useful in the development of potential ferroptosis inhibitors that can be used as disease-modifying drugs for the treatment of such diseases. On the other hand, in vivo models based on the manipulation of rodents and invertebrate animals, such as Drosophila melanogaster, Caenorhabditis elegans, and zebrafish, have been increasingly used for research in neurodegeneration. This work provides an up-to-date review of the main in vitro and in vivo models that can be used to evaluate ferroptosis in the most prevalent neurodegenerative diseases, and to explore potential new drug targets and novel drug candidates for effective disease-modifying therapies.
Collapse
Affiliation(s)
- Inês Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Vera Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Sofia Benfeito
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
61
|
Schiavi A, Salveridou E, Brinkmann V, Shaik A, Menzel R, Kalyanasundaram S, Nygård S, Nilsen H, Ventura N. Mitochondria hormesis delays aging and associated diseases in Caenorhabditis elegans impacting on key ferroptosis players. iScience 2023; 26:106448. [PMID: 37020951 PMCID: PMC10067770 DOI: 10.1016/j.isci.2023.106448] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 11/28/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Excessive iron accumulation or deficiency leads to a variety of pathologies in humans and developmental arrest in the nematode Caenorhabditis elegans. Instead, sub-lethal iron depletion extends C. elegans lifespan. Hypoxia preconditioning protects against severe hypoxia-induced neuromuscular damage across species but it has low feasible application. In this study, we assessed the potential beneficial effects of genetic and chemical interventions acting via mild iron instead of oxygen depletion. We show that limiting iron availability in C. elegans through frataxin silencing or the iron chelator bipyridine, similar to hypoxia preconditioning, protects against hypoxia-, age-, and proteotoxicity-induced neuromuscular deficits. Mechanistically, our data suggest that the beneficial effects elicited by frataxin silencing are in part mediated by counteracting ferroptosis, a form of non-apoptotic cell death mediated by iron-induced lipid peroxidation. This is achieved by impacting on different key ferroptosis players and likely via gpx-independent redox systems. We thus point to ferroptosis inhibition as a novel potential strategy to promote healthy aging.
Collapse
Affiliation(s)
- Alfonso Schiavi
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Eva Salveridou
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Vanessa Brinkmann
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Anjumara Shaik
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | | | - Sumana Kalyanasundaram
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ståle Nygård
- Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Natascia Ventura
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
62
|
Chen Z, Ho IL, Soeung M, Yen EY, Liu J, Yan L, Rose JL, Srinivasan S, Jiang S, Edward Chang Q, Feng N, Gay JP, Wang Q, Wang J, Lorenzi PL, Veillon LJ, Wei B, Weinstein JN, Deem AK, Gao S, Genovese G, Viale A, Yao W, Lyssiotis CA, Marszalek JR, Draetta GF, Ying H. Ether phospholipids are required for mitochondrial reactive oxygen species homeostasis. Nat Commun 2023; 14:2194. [PMID: 37069167 PMCID: PMC10110566 DOI: 10.1038/s41467-023-37924-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/04/2023] [Indexed: 04/19/2023] Open
Abstract
Mitochondria are hubs where bioenergetics, redox homeostasis, and anabolic metabolism pathways integrate through a tightly coordinated flux of metabolites. The contributions of mitochondrial metabolism to tumor growth and therapy resistance are evident, but drugs targeting mitochondrial metabolism have repeatedly failed in the clinic. Our study in pancreatic ductal adenocarcinoma (PDAC) finds that cellular and mitochondrial lipid composition influence cancer cell sensitivity to pharmacological inhibition of electron transport chain complex I. Profiling of patient-derived PDAC models revealed that monounsaturated fatty acids (MUFAs) and MUFA-linked ether phospholipids play a critical role in maintaining ROS homeostasis. We show that ether phospholipids support mitochondrial supercomplex assembly and ROS production; accordingly, blocking de novo ether phospholipid biosynthesis sensitized PDAC cells to complex I inhibition by inducing mitochondrial ROS and lipid peroxidation. These data identify ether phospholipids as a regulator of mitochondrial redox control that contributes to the sensitivity of PDAC cells to complex I inhibition.
Collapse
Affiliation(s)
- Ziheng Chen
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - I-Lin Ho
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melinda Soeung
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Er-Yen Yen
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jintan Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liang Yan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Johnathon L Rose
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanjana Srinivasan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shan Jiang
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Q Edward Chang
- The Oncology Research for Biologics and Immunotherapy Translation (ORBIT), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ningping Feng
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason P Gay
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lucas J Veillon
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bo Wei
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John N Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Angela K Deem
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sisi Gao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giannicola Genovese
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wantong Yao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joseph R Marszalek
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
63
|
Dixon SJ, Pratt DA. Ferroptosis: A flexible constellation of related biochemical mechanisms. Mol Cell 2023; 83:1030-1042. [PMID: 36977413 PMCID: PMC10081971 DOI: 10.1016/j.molcel.2023.03.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/22/2023] [Accepted: 03/03/2023] [Indexed: 03/29/2023]
Abstract
It is common to think about and depict biological processes as being governed by fixed pathways with specific components interconnected by concrete positive and negative interactions. However, these models may fail to effectively capture the regulation of cell biological processes that are driven by chemical mechanisms that do not rely absolutely on specific metabolites or proteins. Here, we discuss how ferroptosis, a non-apoptotic cell death mechanism with emerging links to disease, may be best understood as a highly flexible mechanism that can be executed and regulated by many functionally related metabolites and proteins. The inherent plasticity of ferroptosis has implications for how to define and study this mechanism in healthy and diseased cells and organisms.
Collapse
Affiliation(s)
- Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA.
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
64
|
Zhu J, Meng W, Man Lam S, Shui G, Huang X. Phosphatidylcholine deficiency increases ferroptosis susceptibility in the C. elegans germline. J Genet Genomics 2023; 50:318-329. [PMID: 36933794 DOI: 10.1016/j.jgg.2023.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/15/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023]
Abstract
Ferroptosis, a regulated and iron-dependent form of cell death characterized by peroxidation of membrane phospholipids, has tremendous potential for the therapy of human diseases. The causal link between phospholipid homeostasis and ferroptosis is incompletely understood. Here, we reveal that spin-4, a previously identified regulator of the "B12-one-carbon cycle-phosphatidylcholine (PC)" pathway, sustains germline development and fertility by ensuring PC sufficiency in the nematode Caenorhabditis elegans. Mechanistically, SPIN-4 regulates lysosomal activity which is required for B12-associated PC synthesis. PC deficiency-induced sterility can be rescued by reducing the levels of polyunsaturated fatty acids (PUFAs), reactive oxygen species (ROS) , and redox-active iron, which indicates that the sterility is mediated by germline ferroptosis. These results highlight the critical role of PC homeostasis in ferroptosis susceptibility and offer a new target for pharmacological approaches.
Collapse
Affiliation(s)
- Jinglin Zhu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
65
|
The Emerging Role of Heat Shock Factor 1 (HSF1) and Heat Shock Proteins (HSPs) in Ferroptosis. PATHOPHYSIOLOGY 2023; 30:63-82. [PMID: 36976734 PMCID: PMC10057451 DOI: 10.3390/pathophysiology30010007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Cells employ a well-preserved physiological stress response mechanism, termed the heat shock response, to activate a certain type of molecular chaperone called heat shock proteins (HSPs). HSPs are activated by transcriptional activators of heat shock genes known as heat shock factors (HSFs). These molecular chaperones are categorized as the HSP70 superfamily, which includes HSPA (HSP70) and HSPH (HSP110) families; the DNAJ (HSP40) family; the HSPB family (small heat shock proteins (sHSPs)); chaperonins and chaperonin-like proteins; and other heat-inducible protein families. HSPs play a critical role in sustaining proteostasis and protecting cells against stressful stimuli. HSPs participate in folding newly synthesized proteins, holding folded proteins in their native conformation, preventing protein misfolding and accumulation, and degrading denatured proteins. Ferroptosis is a recently identified type of oxidative iron-dependent cell demise. It was coined recently in 2012 by Stockwell Lab members, who described a special kind of cell death induced by erastin or RSL3. Ferroptosis is characterized by alterations in oxidative status resulting from iron accumulation, increased oxidative stress, and lipid peroxidation, which are mediated by enzymatic and non-enzymatic pathways. The process of ferroptotic cell death is regulated at multiple, and it is involved in several pathophysiological conditions. Much research has emerged in recent years demonstrating the involvement of HSPs and their regulator heat shock factor 1 (HSF1) in ferroptosis regulation. Understanding the machinery controlling HSF1 and HSPs in ferroptosis can be employed in developing therapeutic interventions for ferroptosis occurrence in a number of pathological conditions. Therefore, this review comprehensively summarized the basic characteristics of ferroptosis and the regulatory functions of HSF1 and HSPs in ferroptosis.
Collapse
|
66
|
Mortensen MS, Ruiz J, Watts JL. Polyunsaturated Fatty Acids Drive Lipid Peroxidation during Ferroptosis. Cells 2023; 12:804. [PMID: 36899940 PMCID: PMC10001165 DOI: 10.3390/cells12050804] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Ferroptosis is a form of regulated cell death that is intricately linked to cellular metabolism. In the forefront of research on ferroptosis, the peroxidation of polyunsaturated fatty acids has emerged as a key driver of oxidative damage to cellular membranes leading to cell death. Here, we review the involvement of polyunsaturated fatty acids (PUFAs), monounsaturated fatty acids (MUFAs), lipid remodeling enzymes and lipid peroxidation in ferroptosis, highlighting studies revealing how using the multicellular model organism Caenorhabditis elegans contributes to the understanding of the roles of specific lipids and lipid mediators in ferroptosis.
Collapse
Affiliation(s)
| | | | - Jennifer L. Watts
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
67
|
Rodencal J, Dixon SJ. A tale of two lipids: Lipid unsaturation commands ferroptosis sensitivity. Proteomics 2023; 23:e2100308. [PMID: 36398995 DOI: 10.1002/pmic.202100308] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
Membrane lipids play important roles in the regulation of cell fate, including the execution of ferroptosis. Ferroptosis is a non-apoptotic cell death mechanism defined by iron-dependent membrane lipid peroxidation. Phospholipids containing polyunsaturated fatty acids (PUFAs) are highly vulnerable to peroxidation and are essential for ferroptosis execution. By contrast, the incorporation of less oxidizable monounsaturated fatty acids (MUFAs) in membrane phospholipids protects cells from ferroptosis. The enzymes and pathways that govern PUFA and MUFA metabolism therefore play a critical role in determining cellular sensitivity to ferroptosis. Here, we review three lipid metabolic processes-fatty acid biosynthesis, ether lipid biosynthesis, and phospholipid remodeling-that can govern ferroptosis sensitivity by regulating the balance of PUFAs and MUFAs in membrane phospholipids.
Collapse
Affiliation(s)
- Jason Rodencal
- Department of Biology, Stanford University, Stanford, California, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, California, USA
| |
Collapse
|
68
|
Novichkova E, Nayak S, Boussiba S, Gopas J, Zilberg D, Khozin-Goldberg I. Dietary Application of the Microalga Lobosphaera incisa P127 Reduces Severity of Intestinal Inflammation, Modulates Gut-Associated Gene Expression, and Microbiome in the Zebrafish Model of IBD. Mol Nutr Food Res 2023; 67:e2200253. [PMID: 36683256 DOI: 10.1002/mnfr.202200253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 11/30/2022] [Indexed: 01/24/2023]
Abstract
SCOPE Microalgae are an emerging nutritional resource of biomolecules with potential to alleviate gut inflammation. The study explores the anti-inflammatory and immunomodulatory potential of the microalga Lobosphaera incisa P127, which accumulates a rare omega-6 LC-PUFA dihomo-ɣ-linolenic acid (DGLA) under nitrogen starvation. The therapeutic potential of dietary supplementation with P127 is investigated in the zebrafish model of IBD (TNBS-induced colitis). METHODS AND RESULTS Guts are sampled from zebrafish fed experimental diets for 4 weeks, before and 24 h after TNBS challenge. Diets containing 15% non-starved (Ns) and 7.5% and 15% N-starved (St) algal biomass significantly attenuate the severity of gut injury and goblet cell depletion. In contrast, diets containing 7.5% Ns and DGLA ethyl ester have no effect on gut condition. Fish fed 15% St, high-DGLA biomass, have the fewest individuals with pathological alterations in the gut. Dietary inclusion of Ns and St distinctly modulates gut-associated expression of the immune and inflammatory genes. Fish fed 15% Ns biomass display a coordinated boost in immune gene expression and show major changes in the gut microbiome prior challenge. CONCLUSION Dietary inclusion of L. incisa biomass at two physiological states, ameliorates TNBS-induced gut inflammation, suggesting the synergistic beneficial effects of biomass components not limited to DGLA.
Collapse
Affiliation(s)
- Ekaterina Novichkova
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
- The Albert Katz International School for Desert Studies, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| | - Sagar Nayak
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
- The Jacob Blaustein Center for Scientific Cooperation, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| | - Sammy Boussiba
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| | - Jacob Gopas
- Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8400501, Israel
| | - Dina Zilberg
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| | - Inna Khozin-Goldberg
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| |
Collapse
|
69
|
Lee J, Shin D, Roh JL. Lipid metabolism alterations and ferroptosis in cancer: Paving the way for solving cancer resistance. Eur J Pharmacol 2023; 941:175497. [PMID: 36621602 DOI: 10.1016/j.ejphar.2023.175497] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/22/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
Cancer often perturbs lipid metabolism, which leads to the alteration of metabolism intermediates, contributing to their deregulated growth and metastasis. Alteration of lipid metabolism shifting to contain more polyunsaturated fatty acids (PUFAs) in membrane phospholipids (PLs) also leads to cancer therapy resistance. High amounts of PL-PUFAs render cancer cells more vulnerable to lipid peroxidation (LPO), predisposing them towards ferroptosis, a new form of iron-dependent oxidative regulated cell death. The commitment of cancer undergoing ferroptotic cell death depends on the adaptive lipidome remodeling, LPO patterns, and LPO scavenging ability in heterogeneous cancer cells. Ferroptosis is receiving attention in cancer research as treating cancers, altering membrane lipid homeostasis, and refractory from conventional therapies. Therefore, a better understanding of the molecular underpinning of lipid metabolism alterations may provide new opportunities for solving cancer resistance. This review intends to understand altered lipid metabolism in cancers and discuss lipid composition and metabolic processes associated with ferroptosis induction in cancers.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Daiha Shin
- Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
70
|
Jordan JM, Webster AK, Chen J, Chitrakar R, Ryan Baugh L. Early-life starvation alters lipid metabolism in adults to cause developmental pathology in Caenorhabditis elegans. Genetics 2023; 223:iyac172. [PMID: 36449523 PMCID: PMC9910403 DOI: 10.1093/genetics/iyac172] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 03/25/2022] [Accepted: 10/31/2022] [Indexed: 12/02/2022] Open
Abstract
Early-life malnutrition increases adult disease risk in humans, but the causal changes in gene regulation, signaling, and metabolism are unclear. In the roundworm Caenorhabditis elegans, early-life starvation causes well-fed larvae to develop germline tumors and other gonad abnormalities as adults. Furthermore, reduced insulin/IGF signaling during larval development suppresses these starvation-induced abnormalities. How early-life starvation and insulin/IGF signaling affect adult pathology is unknown. We show that early-life starvation has pervasive effects on adult gene expression which are largely reversed by reduced insulin/IGF signaling following recovery from starvation. Early-life starvation increases adult fatty-acid synthetase fasn-1 expression in daf-2 insulin/IGF signaling receptor-dependent fashion, and fasn-1/FASN promotes starvation-induced abnormalities. Lipidomic analysis reveals increased levels of phosphatidylcholine in adults subjected to early-life starvation, and supplementation with unsaturated phosphatidylcholine during development suppresses starvation-induced abnormalities. Genetic analysis of fatty-acid desaturases reveals positive and negative effects of desaturation on development of starvation-induced abnormalities. In particular, the ω3 fatty-acid desaturase fat-1 and the Δ5 fatty-acid desaturase fat-4 inhibit and promote development of abnormalities, respectively. fat-4 is epistatic to fat-1, suggesting that arachidonic acid-containing lipids promote development of starvation-induced abnormalities, and supplementation with ARA enhanced development of abnormalities. This work shows that early-life starvation and insulin/IGF signaling converge on regulation of adult lipid metabolism, affecting stem-cell proliferation and tumor formation.
Collapse
Affiliation(s)
- James M Jordan
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Amy K Webster
- Department of Biology, Duke University, Durham, NC 27708, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA
| | - Jingxian Chen
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Rojin Chitrakar
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - L Ryan Baugh
- Department of Biology, Duke University, Durham, NC 27708, USA
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA
| |
Collapse
|
71
|
Abstract
It has been 10 years since the concept of ferroptosis was put forward and research focusing on ferroptosis has been increasing continuously. Ferroptosis is driven by iron-dependent lipid peroxidation, which can be antagonized by glutathione peroxidase 4 (GPX4), ferroptosis inhibitory protein 1 (FSP1), dihydroorotate dehydrogenase (DHODH) and Fas-associated factor 1 (FAF1). Various cellular metabolic events, including lipid metabolism, can modulate ferroptosis sensitivity. It is worth noting that the reprogramming of lipid metabolism in cancer cells can promote the occurrence and development of tumors. The metabolic flexibility of cancer cells opens the possibility for the coordinated targeting of multiple lipid metabolic pathways to trigger cancer cells ferroptosis. In addition, cancer cells must obtain immortality, escape from programmed cell death including ferroptosis, to promote cancer progression, which provides new perspectives for improving cancer therapy. Targeting the vulnerability of ferroptosis has received attention as one of the significant possible strategies to treat cancer given its role in regulating tumor cell survival. We review the impact of iron and lipid metabolism on ferroptosis and the potential role of the crosstalk of lipid metabolism reprogramming and ferroptosis in antitumor immunity and sum up agents targeting lipid metabolism and ferroptosis for cancer therapy.
Collapse
|
72
|
Sarparast M, Pourmand E, Hinman J, Vonarx D, Reason T, Zhang F, Paithankar S, Chen B, Borhan B, Watts JL, Alan J, Lee KSS. Dihydroxy-Metabolites of Dihomo-gamma-linolenic Acid Drive Ferroptosis-Mediated Neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522933. [PMID: 36711920 PMCID: PMC9881903 DOI: 10.1101/2023.01.05.522933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Even after decades of research, the mechanism of neurodegeneration remains understudied, hindering the discovery of effective treatments for neurodegenerative diseases. Recent reports suggest that ferroptosis could be a novel therapeutic target for neurodegenerative diseases. While polyunsaturated fatty acid (PUFA) plays an important role in neurodegeneration and ferroptosis, how PUFAs may trigger these processes remains largely unknown. PUFA metabolites from cytochrome P450 and epoxide hydrolase metabolic pathways may modulate neurodegeneration. Here, we test the hypothesis that specific PUFAs regulate neurodegeneration through the action of their downstream metabolites by affecting ferroptosis. We find that the PUFA, dihomo gamma linolenic acid (DGLA), specifically induces ferroptosis-mediated neurodegeneration in dopaminergic neurons. Using synthetic chemical probes, targeted metabolomics, and genetic mutants, we show that DGLA triggers neurodegeneration upon conversion to dihydroxyeicosadienoic acid through the action of CYP-EH, representing a new class of lipid metabolite that induces neurodegeneration via ferroptosis.
Collapse
Affiliation(s)
- Morteza Sarparast
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Elham Pourmand
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Jennifer Hinman
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Derek Vonarx
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Tommy Reason
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Fan Zhang
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Shreya Paithankar
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, USA
| | - Bin Chen
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA,Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, USA
| | - Babak Borhan
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Jennifer L. Watts
- School of Molecular Biosciences, Washington State University, Pullman, WA, USA
| | - Jamie Alan
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA,Corresponding Authors
| | - Kin Sing Stephen Lee
- Department of Chemistry, Michigan State University, East Lansing, MI, USA,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA,Corresponding Authors
| |
Collapse
|
73
|
Organelle-Specific Mechanisms in Crosstalk between Apoptosis and Ferroptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3400147. [PMID: 36644574 PMCID: PMC9836800 DOI: 10.1155/2023/3400147] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 01/07/2023]
Abstract
Apoptosis has been extensively studied, whereas ferroptosis is a newly discovered form of regulated cell death that involves iron-dependent accumulations of lipid hydroperoxides. While these two cell death mechanisms were initially believed to be mutually exclusive, recent studies have revealed cellular contexts requiring a balanced interaction between them. Numerous subcellular sites and signaling molecules within these sites are involved in both processes, either as modules or switches that allow cells to choose on how to proceed. The close relationships between apoptosis and ferroptosis, as well as the possibility of switching from one to the other, are described in this review. To understand the crosstalk between apoptosis and ferroptosis, various organelle-specific mechanisms must be analyzed and compared. The ability to switch apoptosis to ferroptosis by targeting cellular organelles has a great potential in cancer therapy.
Collapse
|
74
|
Turchi R, Tortolici F, Benvenuto M, Punziano C, De Luca A, Rufini S, Faraonio R, Bei R, Lettieri-Barbato D, Aquilano K. Low Sulfur Amino Acid, High Polyunsaturated Fatty Acid Diet Inhibits Breast Cancer Growth. Int J Mol Sci 2022; 24:ijms24010249. [PMID: 36613691 PMCID: PMC9820692 DOI: 10.3390/ijms24010249] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer cells may acquire resistance to stress signals and reprogram metabolism to meet the energetic demands to support their high proliferation rate and avoid death. Hence, targeting nutrient dependencies of cancer cells has been suggested as a promising anti-cancer strategy. We explored the possibility of killing breast cancer (BC) cells by modifying nutrient availability. We used in vitro models of BC (MCF7 and MDA-MB-231) that were maintained with a low amount of sulfur amino acids (SAAs) and a high amount of oxidizable polyunsatured fatty acids (PUFAs). Treatment with anti-apoptotic, anti-ferroptotic and antioxidant drugs were used to determine the modality of cell death. We reproduced these conditions in vivo by feeding BC-bearing mice with a diet poor in proteins and SAAs and rich in PUFAs (LSAA/HPUFA). Western blot analysis, qPCR and histological analyses were used to assess the anti-cancer effects and the molecular pathways involved. We found that BC cells underwent oxidative damage to DNA and proteins and both apoptosis and ferroptosis were induced. Along with caspases-mediated PARP1 cleavage, we found a lowering of the GSH-GPX4 system and an increase of lipid peroxides. A LSAA/HPUFA diet reduced tumor mass and its vascularization and immune cell infiltration, and induced apoptosis and ferroptotic hallmarks. Furthermore, mitochondrial mass was found to be increased, and the buffering of mitochondrial reactive oxygen species limited GPX4 reduction and DNA damage. Our results suggest that administration of custom diets, targeting the dependency of cancer cells on certain nutrients, can represent a promising complementary option for anti-cancer therapy.
Collapse
Affiliation(s)
- Riccardo Turchi
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Flavia Tortolici
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Monica Benvenuto
- Departmental Faculty of Medicine, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carolina Punziano
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
| | - Anastasia De Luca
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Stefano Rufini
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Daniele Lettieri-Barbato
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- IRCCS Santa Lucia, 00179 Rome, Italy
- Correspondence: (D.L.-B.); (K.A.)
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- Correspondence: (D.L.-B.); (K.A.)
| |
Collapse
|
75
|
Identification and Validation of Ferroptosis-Related DNA Methylation Signature for Predicting the Prognosis and Guiding the Treatment in Cutaneous Melanoma. Int J Mol Sci 2022; 23:ijms232415677. [PMID: 36555319 PMCID: PMC9778758 DOI: 10.3390/ijms232415677] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Cutaneous melanoma (CM) is one of the most aggressive skin tumors with a poor prognosis. Ferroptosis is a newly discovered form of regulated cell death that is closely associated with cancer development and immunotherapy. The aim of this study was to establish and validate a ferroptosis-related gene (FRG) DNA methylation signature to predict the prognosis of CM patients using data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database. A reliable four-FRG DNA methylation prognostic signature was constructed via Cox regression analysis based on TCGA database. Kaplan-Meier analysis showed that patients in the high-risk group tended to have a shorter overall survival (OS) than the low-risk group in both training TCGA and validation GEO cohorts. Time-dependent receiver operating characteristic (ROC) analysis showed the areas under the curve (AUC) at 1, 3, and 5 years were 0.738, 0.730, and 0.770 in TCGA cohort and 0.773, 0.775, and 0.905 in the validation cohort, respectively. Univariate and multivariate Cox regression analyses indicated that the signature was an independent prognostic indicator of OS in patients with CM. Immunogenomic profiling showed the low-risk group of patients had a higher immunophenoscore, and most immune checkpoints were negatively associated with the risk signature. Functional enrichment analysis revealed that immune response and immune-related pathways were enriched in the low-risk group. In conclusion, we established and validated a four-FRG DNA methylation signature that independently predicts prognosis in CM patients. This signature was strongly correlated with the immune landscape, and may serve as a biomarker to guide clinicians in making more precise and personalized treatment decisions for CM patients.
Collapse
|
76
|
Liu Y, Xin H, Li W, Shen N, Cui Y. Ginkgo biloba endopleura: An unexplored industrial waste as a potential source of flavonoids, lipids and anti-lung cancer compounds. INDUSTRIAL CROPS AND PRODUCTS 2022; 189:115851. [DOI: 10.1016/j.indcrop.2022.115851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
|
77
|
Li L, Yu XJ, Gao L, Cheng L, Sun B, Wang G. Diabetic Ferroptosis and Pancreatic Cancer: Foe or Friend? Antioxid Redox Signal 2022; 37:1206-1221. [PMID: 35996983 DOI: 10.1089/ars.2022.0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Pancreatic cancer and diabetes have a reciprocal causation relationship. As a potential risk factor, diabetes increases morbidity and promotes pancreatic cancer progression. The main mechanisms include islet dysfunction-induced systemic metabolic disorder, pancreatic stellate cell activation, and immunosuppression. Ferroptosis is regarded as regulated cell death, which participates in chemotherapy resistance and is refractory to radiation therapy and immunotherapy. Diabetes-induced ferroptosis causes many complications, but the underlying mechanism of diabetes-related ferroptosis in pancreatic cancer has not been discussed. Recent Advances: Ferroptosis alleviates pancreatic intraepithelial neoplasia (PanIN) progression by activating chronic inflammation. The specific drugs that cause ferroptosis achieve tumor suppression by inducing lipid peroxidation. Ferroptosis plays pro and con roles in cancer. Both the ferroptosis inhibitor and inducer exhibit antitumor effects through killing cancer cells or directly affecting tumor growth. Diabetes-induced ferroptosis contributes to tumor cell death by different components, including tumor cells, fibroblasts, immune cells, and adipocytes. A better understanding of its role in modulating the tumor microenvironment will reveal diabetes-associated ferroptotic features in cancer development, which can be used to figure out possible treatment strategies for cancer patients with hyperglycemia. Critical Issues: We demonstrate the potential roles of diabetes-related ferroptosis in pancreatic cancer progression and discuss ferroptosis-related antitumor effects and therapeutics for pancreatic cancer treatment. Future Directions: Further studies are required to highlight mechanisms of diabetes-mediated ferroptosis in pancreatic cancer tumorigenesis and progression. The antitumor effects of ferroptosis regulators combined with chemotherapy, targeted therapy, or immunotherapy in diabetic patients should be investigated. We hope that pancreatic cancer patients with diabetes will benefit from ferroptosis-related therapies. Antioxid. Redox Signal. 37, 1206-1221.
Collapse
Affiliation(s)
- Le Li
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xing-Jia Yu
- Department of Centric Operating Room, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Gao
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Long Cheng
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
78
|
Kim J, Jo Y, Cho D, Ryu D. L-threonine promotes healthspan by expediting ferritin-dependent ferroptosis inhibition in C. elegans. Nat Commun 2022; 13:6554. [PMID: 36323683 PMCID: PMC9628521 DOI: 10.1038/s41467-022-34265-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022] Open
Abstract
The pathways that impact longevity in the wake of dietary restriction (DR) remain still ill-defined. Most studies have focused on nutrient limitation and perturbations of energy metabolism. We showed that the L-threonine was elevated in Caenorhabditis elegans under DR, and that L-threonine supplementation increased its healthspan. Using metabolic and transcriptomic profiling in worms that were fed with RNAi to induce loss of key candidate mediators. L-threonine supplementation and loss-of-threonine dehydrogenaseincreased the healthspan by attenuating ferroptosis in a ferritin-dependent manner. Transcriptomic analysis showed that FTN-1 encoding ferritin was elevated, implying FTN-1 is an essential mediator of longevity promotion. Organismal ferritin levels were positively correlated with chronological aging and L-threonine supplementation protected against age-associated ferroptosis through the DAF-16 and HSF-1 pathways. Our investigation uncovered the role of a distinct and universal metabolite, L-threonine, in DR-mediated improvement in organismal healthspan, suggesting it could be an effective intervention for preventing senescence progression and age-induced ferroptosis.
Collapse
Affiliation(s)
- Juewon Kim
- Basic Research & Innovation Division, Amorepacific R&D Center, Yongin, Korea
| | - Yunju Jo
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Donghyun Cho
- Basic Research & Innovation Division, Amorepacific R&D Center, Yongin, Korea
| | - Dongryeol Ryu
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
79
|
Chen J, Wang Y, Han L, Wang R, Gong C, Yang G, Li Z, Gao S, Yuan Y. A ferroptosis-inducing biomimetic nanocomposite for the treatment of drug-resistant prostate cancer. Mater Today Bio 2022; 17:100484. [PMID: 36388460 PMCID: PMC9649379 DOI: 10.1016/j.mtbio.2022.100484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/14/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Second-generation androgen receptor (AR) inhibitors such as enzalutamide are the first-line treatments for castration-resistant prostate cancer (CRPC). Resistance to enzalutamide will greatly increase the difficulty of prostate cancer treatment and reduce the survival time of patients. However, drug-resistant cancer cells seem to be more sensitive to ferroptosis. Therefore, we constructed a biomimetic tumor-targeting magnetic lipid nanoparticle (t-ML) to codeliver dihomo-γ-linolenic acid (DGLA) and 2,4-dienoyl-CoA reductase 1 (DECR1) siRNA (t-ML@DGLA/siDECR1). DGLA is a dietary polyunsaturated fatty acid (PUFA), while DECR1 is overexpressed in prostate cancer and can inhibit the generation of PUFAs. The combination of DGLA and siDECR1 can efficiently induce ferroptosis by peroxidation of PUFAs, which has been verified both in vitro and in vivo. With the assistance of an external magnet, t-ML showed good tumor targeting ability and biocompatibility, and t-ML@DGLA/siDECR1 exhibited significant ferroptosis induction and tumor suppression capabilities. Moreover, in a nude mouse model of prostate cancer fed on a high-fat diet (HFD), there was no distant organ metastasis when the tumor-bearing mice were treated with t-ML@DGLA/siDECR1 and an external magnet, with upregulated PUFAs and downregulated monounsaturated fatty acids (MUFAs). Hence, this study has broadened the way of treating drug-resistant prostate cancer based on ferroptosis induction.
Collapse
Affiliation(s)
- Jiyuan Chen
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Yujie Wang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Lu Han
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Rong Wang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Chunai Gong
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Gang Yang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Zhe Li
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Shen Gao
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Corresponding author.
| |
Collapse
|
80
|
Klasson TD, LaGory EL, Zhao H, Huynh SK, Papandreou I, Moon EJ, Giaccia AJ. ACSL3 regulates lipid droplet biogenesis and ferroptosis sensitivity in clear cell renal cell carcinoma. Cancer Metab 2022; 10:14. [PMID: 36192773 PMCID: PMC9528056 DOI: 10.1186/s40170-022-00290-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC), the predominant subtype of kidney cancer, possesses characteristic alterations to multiple metabolic pathways, including the accumulation of cytosolic lipid droplets. However, the pathways that drive lipid droplet accumulation in ccRCC cells and their importance to cancer biology remain poorly understood. METHODS We sought to identify the carbon sources necessary for lipid droplet accumulation using Oil red O staining and isotope-tracing lipidomics. The role of the acyl-CoA synthetase (ACSL) family members, an important group of lipid metabolic enzymes, was investigated using siRNA and drug mediated inhibition. CTB and XTT assays were performed to determine the effect of ACSL3 knockdown and lipid starvation on ccRCC cell viability and shRNA was used to study the effect of ACSL3 in an orthotopic mouse model. The relationship between ferroptosis susceptibility of ccRCC and ACSL3 controlled lipid metabolism was examined using CTB and FACS-based assays. The importance of 5-LOX in ferroptosis susceptibility in ccRCC was shown with XTT survival assays, and the expression level and predictive value of 5-LOX in TCGA ccRCC data was assessed. RESULTS We found that ccRCC cells obtain the necessary substrates for lipid droplet accumulation by metabolizing exogenous serum derived lipids and not through de novo lipogenesis. We show that this metabolism of exogenous fatty acids into lipid droplets requires the enzyme acyl-CoA synthetase 3 (ACSL3) and not other ACSL family proteins. Importantly, genetic or pharmacologic suppression of ACSL3 is cytotoxic to ccRCC cells in vitro and causes a reduction of tumor weight in an orthotopic mouse model. Conversely, ACSL3 inhibition decreases the susceptibility of ccRCC cells to ferroptosis, a non-apoptotic form of cell death involving lipid peroxidation. The sensitivity of ccRCC to ferroptosis is also highly dependent on the composition of exogenous fatty acids and on 5-lipoxygenase (5-LOX), a leukotriene producing enzyme which produces lipid peroxides that have been implicated in other cancers but not in ccRCC. CONCLUSIONS ACSL3 regulates the accumulation of lipid droplets in ccRCC and is essential for tumor growth. In addition, ACSL3 also modulates ferroptosis sensitivity in a manner dependent on the composition of exogenous fatty acids. Both functions of ACSL3 could be exploited for ccRCC therapy.
Collapse
Affiliation(s)
- Timothy D Klasson
- Department of Radiation Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Edward L LaGory
- Department of Radiation Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Hongjuan Zhao
- Department of Radiation Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Star K Huynh
- Department of Radiation Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Ioanna Papandreou
- Department of Radiation Oncology, The Ohio State Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Eui Jung Moon
- Department of Radiation Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, 94305, USA
- Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building (ORCRB), Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford School of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building (ORCRB), Roosevelt Drive, Oxford, OX3 7DQ, UK.
| |
Collapse
|
81
|
Guo S, Chen J, Yi X, Lu Z, Guo W. Identification and validation of ferroptosis-related lncRNA signature as a prognostic model for skin cutaneous melanoma. Front Immunol 2022; 13:985051. [PMID: 36248853 PMCID: PMC9556814 DOI: 10.3389/fimmu.2022.985051] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Melanoma is a type of skin cancer, which originates from the malignant transformation of epidermal melanocytes, with extremely high lethality. Ferroptosis has been documented to be highly related to cancer pathogenesis and the effect of immunotherapy. In addition, the dysregulation of lncRNAs is greatly implicated in melanoma progression and ferroptosis regulation. However, the significance of ferroptosis-related lncRNA in melanoma treatment and the prognosis of melanoma patients remains elusive. Methods Via Least Absolute Shrinkage Selection Operator (LASSO) regression analysis in the TCGA SKCM database, a cutaneous melanoma risk model was established based on differentially-expressed ferroptosis-related lncRNAs (DEfrlncRNAs). The nomogram, receiver operating characteristic (ROC) curves, and calibration plots were conducted to examine the predictive performance of this model. Sequentially, we continued to analyze the differences between the high- and low-risk groups, in terms of clinical characteristics, immune cell infiltration, immune-related functions, and chemotherapy drug sensitivity. Moreover, the expressions of DEfrlncRNAs, PD-L1, and CD8 were also examined by qRT-PCR and immunohistochemical staining in melanoma tissues to further confirm the potential clinical implication of DEfrlncRNAs in melanoma immunotherapy. Results 16 DEfrlncRNAs were identified, and a representative risk score for patient survival was constructed based on these 16 genes. The risk score was found to be an independent prognostic factor for the survival of melanoma patients. In addition, the low-risk group of patients had higher immune cell infiltration in the melanoma lesions, higher sensitivity to chemotherapeutic agents, and a better survival prognosis. Besides, the high expression of the identified 5 DEfrlncRNA in the low-risk group might suggest a higher possibility to benefit from immune checkpoint blockade therapy in the treatment of melanoma. Conclusion The DEfrlncRNA risk prediction model related to ferroptosis genes can independently predict the prognosis of patients with melanoma and provide a basis for evaluating the response of clinical treatment in melanoma.
Collapse
Affiliation(s)
- Sen Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jianru Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiuli Yi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zifan Lu
- Department of Biopharmaceuticals, School of Pharmacy, Fourth Military Medical University, Xi’an, China
- *Correspondence: Weinan Guo, ; Zifan Lu,
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Weinan Guo, ; Zifan Lu,
| |
Collapse
|
82
|
Bacterial diet modulates tamoxifen-induced death via host fatty acid metabolism. Nat Commun 2022; 13:5595. [PMID: 36151093 PMCID: PMC9508336 DOI: 10.1038/s41467-022-33299-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 09/06/2022] [Indexed: 11/18/2022] Open
Abstract
Tamoxifen is a selective estrogen receptor (ER) modulator that is used to treat ER-positive breast cancer, but that at high doses kills both ER-positive and ER-negative breast cancer cells. We recapitulate this off-target effect in Caenorhabditis elegans, which does not have an ER ortholog. We find that different bacteria dramatically modulate tamoxifen toxicity in C. elegans, with a three-order of magnitude difference between animals fed Escherichia coli, Comamonas aquatica, and Bacillus subtilis. Remarkably, host fatty acid (FA) biosynthesis mitigates tamoxifen toxicity, and different bacteria provide the animal with different FAs, resulting in distinct FA profiles. Surprisingly these bacteria modulate tamoxifen toxicity by different death mechanisms, some of which are modulated by FA supplementation and others by antioxidants. Together, this work reveals a complex interplay between microbiota, FA metabolism and tamoxifen toxicity that may provide a blueprint for similar studies in more complex mammals. Here, Diot et al. use the nematode Caenorhabditis elegans as a model to identify off-target toxicity mechanisms for tamoxifen, and find that these include fatty acid metabolism and cell death, which can be modulated by different bacterial species.
Collapse
|
83
|
Qin S, Wang Y, Li L, Liu J, Xiao C, Duan D, Hao W, Qin C, Chen J, Yao L, Zhang R, You J, Zheng JS, Shen E, Wu L. Early-life vitamin B12 orchestrates lipid peroxidation to ensure reproductive success via SBP-1/SREBP1 in Caenorhabditis elegans. Cell Rep 2022; 40:111381. [PMID: 36130518 DOI: 10.1016/j.celrep.2022.111381] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/05/2022] [Accepted: 08/27/2022] [Indexed: 11/03/2022] Open
Abstract
Vitamin B12 (B12) deficiency is a critical problem worldwide. Such deficiency in infants has long been known to increase the propensity to develop obesity and diabetes later in life through unclear mechanisms. Here, we establish a Caenorhabditis elegans model to study how early-life B12 impacts adult health. We find that early-life B12 deficiency causes increased lipogenesis and lipid peroxidation in adult worms, which in turn induces germline defects through ferroptosis. Mechanistically, we show the central role of the methionine cycle-SBP-1/SREBP1-lipogenesis axis in programming adult traits by early-life B12. Moreover, SBP-1/SREBP1 participates in a crucial feedback loop with NHR-114/HNF4 to maintain cellular B12 homeostasis. Inhibition of SBP-1/SREBP1-lipogenesis signaling and ferroptosis later in life can reverse disorders in adulthood when B12 cannot. Overall, this study provides mechanistic insights into the life-course effects of early-life B12 on the programming of adult health and identifies potential targets for future interventions for adiposity and infertility.
Collapse
Affiliation(s)
- Shenlu Qin
- Fudan University, Shanghai, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yihan Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Lili Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Junli Liu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Congmei Xiao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Duo Duan
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Wanyu Hao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Chunxia Qin
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jie Chen
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Luxia Yao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Runshuai Zhang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jia You
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Ju-Sheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Enzhi Shen
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Lianfeng Wu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
84
|
Magtanong L, Mueller GD, Williams KJ, Billmann M, Chan K, Armenta DA, Pope LE, Moffat J, Boone C, Myers CL, Olzmann JA, Bensinger SJ, Dixon SJ. Context-dependent regulation of ferroptosis sensitivity. Cell Chem Biol 2022; 29:1409-1418.e6. [PMID: 35809566 PMCID: PMC9481678 DOI: 10.1016/j.chembiol.2022.06.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 04/11/2022] [Accepted: 06/15/2022] [Indexed: 02/08/2023]
Abstract
Ferroptosis is an important mediator of pathophysiological cell death and an emerging target for cancer therapy. Whether ferroptosis sensitivity is governed by a single regulatory mechanism is unclear. Here, based on the integration of 24 published chemical genetic screens combined with targeted follow-up experimentation, we find that the genetic regulation of ferroptosis sensitivity is highly variable and context-dependent. For example, the lipid metabolic gene acyl-coenzyme A (CoA) synthetase long chain family member 4 (ACSL4) appears far more essential for ferroptosis triggered by direct inhibition of the lipid hydroperoxidase glutathione peroxidase 4 (GPX4) than by cystine deprivation. Despite this, distinct pro-ferroptotic stimuli converge upon a common lethal effector mechanism: accumulation of lipid peroxides at the plasma membrane. These results indicate that distinct genetic mechanisms regulate ferroptosis sensitivity, with implications for the initiation and analysis of this process in vivo.
Collapse
Affiliation(s)
- Leslie Magtanong
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Grace D Mueller
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Kevin J Williams
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Lipidomics Laboratory, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Maximilian Billmann
- Department of Computer Science and Engineering, University of Minnesota-Twin Cities, 200 Union Street, Minneapolis, MN 55455, USA
| | - Katherine Chan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - David A Armenta
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Lauren E Pope
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jason Moffat
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Chad L Myers
- Department of Computer Science and Engineering, University of Minnesota-Twin Cities, 200 Union Street, Minneapolis, MN 55455, USA; Program in Biomedical Informatics and Computational Biology, University of Minnesota-Twin Cities, 200 Union Street, Minneapolis, MN 55455, USA
| | - James A Olzmann
- Departments of Molecular and Cell Biology and Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Steven J Bensinger
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Lipidomics Laboratory, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
85
|
Trautenberg LC, Brankatschk M, Shevchenko A, Wigby S, Reinhardt K. Ecological lipidology. eLife 2022; 11:79288. [PMID: 36069772 PMCID: PMC9451535 DOI: 10.7554/elife.79288] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Dietary lipids (DLs), particularly sterols and fatty acids, are precursors for endogenous lipids that, unusually for macronutrients, shape cellular and organismal function long after ingestion. These functions – cell membrane structure, intracellular signalling, and hormonal activity – vary with the identity of DLs, and scale up to influence health, survival, and reproductive fitness, thereby affecting evolutionary change. Our Ecological Lipidology approach integrates biochemical mechanisms and molecular cell biology into evolution and nutritional ecology. It exposes our need to understand environmental impacts on lipidomes, the lipid specificity of cell functions, and predicts the evolution of lipid-based diet choices. Broad interdisciplinary implications of Ecological Lipidology include food web alterations, species responses to environmental change, as well as sex differences and lifestyle impacts on human nutrition, and opportunities for DL-based therapies.
Collapse
Affiliation(s)
| | - Marko Brankatschk
- Biotechnology Center (BIOTEC), Technische Universität Dresden, Dresden, Germany
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Stuart Wigby
- Applied Zoology, Technische Universität Dresden, Dresden, Germany.,Department of Evolution, Ecology and Behaviour, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Klaus Reinhardt
- Applied Zoology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
86
|
Chen L, Yang CS, Chen SD, Zhou QX, Wang GQ, Cai SL, Li WH, Luo HZ. Multi-omics characterization of the unsaturated fatty acid biosynthesis pathway in colon cancer. Am J Cancer Res 2022; 12:3985-4000. [PMID: 36119831 PMCID: PMC9442000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/30/2022] [Indexed: 06/15/2023] Open
Abstract
The biosynthesis of unsaturated fatty acids is involved in the initiation and progression of colon adenocarcinoma (COAD). In this study, we aimed to investigate the multi-omics characteristics of unsaturated fatty acid biosynthesis-related genes and explore their prognostic value in colon cancer by analyzing the data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. An unsaturated fatty acid biosynthesis pathway related-genes enrichment score (BUFAS) was constructed utilizing the single sample gene set enrichment analysis (ssGSEA). We discovered that a high BUFAS was associated with longer overall survival (OS) in both the training and the validation sets. Multivariable analysis including the clinical characteristics further verified the independent prognostic value of the BUFAS in both the TCGA-COAD and the GSE39582 datasets. In addition, GSEA analysis revealed that BUFAS was positively associated with several signaling pathways, including MTORC1, peroxisome, and pathways related to fatty acid metabolism, while was negatively associated with other signaling pathways, such as hedgehog, NOTCH, and Wnt/beta-catenin pathway. Furthermore, in the COAD cell lines of the Genomics of Drug Sensitivity in Cancer (GDSC) database, we found that BUFAS was positively correlated with the drug sensitivities of cisplatin, gemcitabine, camptothecin, lapatinib, and afatinib, while was negatively correlated with that of ponatinib. Moreover, in the COAD single-cell transcriptomic dataset (GSE146771), the BUFAS varied among different cell types and was enriched in mast cells and fibroblasts. Taken together, the BUFAS we constructed could be used as an independent prognostic signature in predicting the OS and drug resistance of colon cancer. Unsaturated fatty acid biosynthesis pathway might serve as potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Ling Chen
- Department of Gastrointestinal Surgery, Xiangya HospitalNo. 87 Xiangya Road, Changsha, Hunan, China
| | - Chang-Shun Yang
- Department of Surgical Oncology, Shengli Clinical Medical College of Fujian Medical UniversityNo. 134 East Street, Fuzhou, Fujian, China
| | - Si-Dong Chen
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Qiao-Xia Zhou
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Guo-Qiang Wang
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Shang-Li Cai
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Wei-Hua Li
- Department of Surgical Oncology, Shengli Clinical Medical College of Fujian Medical UniversityNo. 134 East Street, Fuzhou, Fujian, China
| | - Hong-Zhi Luo
- Department of Tumor Surgery, Zhongshan City People’s HospitalNo. 2 Sunwen Middle Road, Zhongshan, Guangdong, China
| |
Collapse
|
87
|
Chen Y, Liao X, Jing P, Hu L, Yang Z, Yao Y, Liao C, Zhang S. Linoleic Acid-Glucosamine Hybrid for Endogenous Iron-Activated Ferroptosis Therapy in High-Grade Serous Ovarian Cancer. Mol Pharm 2022; 19:3187-3198. [PMID: 35939328 DOI: 10.1021/acs.molpharmaceut.2c00333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
As the most common subtype in ovarian malignancies, high-grade serous ovarian cancer (HGSOC) made less therapeutic progress in past decades due to the lack of effective drug-able targets. Herein, an effective linoleic acid (LA) and glucosamine (GlcN) hybrid (LA-GlcN) was synthesized for the treatment of HGSOC. The GlcN was introduced to recognize the glucose transporter 1 (GLUT 1) overexpressed in tumor cells to enhance the uptake of LA-GlcN, and the unsaturated LA was employed to trigger ferroptosis by iron-dependent lipid peroxidation. Since the iron content of HGSOC was ∼5 and 2 times, respectively, higher than that of the normal ovarian cells and low-grade serous ovarian cancer cells, these excess irons make them a good target to enhance the ferroptosis of LA-GlcN. The in vitro study demonstrated that LA-GlcN could selectively kill HGSOC cells without affecting normal cells; the in vivo study revealed that LA-GlcN at the dose of 50 mg kg-1 achieved a comparable tumor inhibition as doxorubicin hydrochloride (4 mg kg-1) while the overall survival of mice was extended largely due to the low toxicity, and when the dose was increased to 100 mg kg-1, the therapeutic outcomes could be improved further. This dietary hybrid which targets the excess endogenous iron to activate ferroptosis represents a promising drug for HGSOC treatment.
Collapse
Affiliation(s)
- Ying Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & College of Pharmacy, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China.,College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Xiaoming Liao
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Pei Jing
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Liangkui Hu
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Zengqiu Yang
- The State Key Laboratory of Functions and Applications of Medicinal Plants & College of Pharmacy, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Yongchao Yao
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital Sichuan University, Chengdu 610041, China
| | - Chunyan Liao
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Shiyong Zhang
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| |
Collapse
|
88
|
NADPH and Mitochondrial Quality Control as Targets for a Circadian-Based Fasting and Exercise Therapy for the Treatment of Parkinson's Disease. Cells 2022; 11:cells11152416. [PMID: 35954260 PMCID: PMC9367803 DOI: 10.3390/cells11152416] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Dysfunctional mitochondrial quality control (MQC) is implicated in the pathogenesis of Parkinson's disease (PD). The improper selection of mitochondria for mitophagy increases reactive oxygen species (ROS) levels and lowers ATP levels. The downstream effects include oxidative damage, failure to maintain proteostasis and ion gradients, and decreased NAD+ and NADPH levels, resulting in insufficient energy metabolism and neurotransmitter synthesis. A ketosis-based metabolic therapy that increases the levels of (R)-3-hydroxybutyrate (BHB) may reverse the dysfunctional MQC by partially replacing glucose as an energy source, by stimulating mitophagy, and by decreasing inflammation. Fasting can potentially raise cytoplasmic NADPH levels by increasing the mitochondrial export and cytoplasmic metabolism of ketone body-derived citrate that increases flux through isocitrate dehydrogenase 1 (IDH1). NADPH is an essential cofactor for nitric oxide synthase, and the nitric oxide synthesized can diffuse into the mitochondrial matrix and react with electron transport chain-synthesized superoxide to form peroxynitrite. Excessive superoxide and peroxynitrite production can cause the opening of the mitochondrial permeability transition pore (mPTP) to depolarize the mitochondria and activate PINK1-dependent mitophagy. Both fasting and exercise increase ketogenesis and increase the cellular NAD+/NADH ratio, both of which are beneficial for neuronal metabolism. In addition, both fasting and exercise engage the adaptive cellular stress response signaling pathways that protect neurons against the oxidative and proteotoxic stress implicated in PD. Here, we discuss how intermittent fasting from the evening meal through to the next-day lunch together with morning exercise, when circadian NAD+/NADH is most oxidized, circadian NADP+/NADPH is most reduced, and circadian mitophagy gene expression is high, may slow the progression of PD.
Collapse
|
89
|
Wang W, Pan F, Lin X, Yuan J, Tao C, Wang R. Ferroptosis-Related Hub Genes in Hepatocellular Carcinoma: Prognostic Signature, Immune-Related, and Drug Resistance Analysis. Front Genet 2022; 13:907331. [PMID: 35938001 PMCID: PMC9355705 DOI: 10.3389/fgene.2022.907331] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/20/2022] [Indexed: 01/10/2023] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is the most prevalent type of primary liver cancer with a high fatality rate and dismal prognosis because of frequent recurrence and lack of efficient therapies. Ferroptosis is a recently recognized iron-dependent cell death distinct from necroptosis and apoptosis. The relationship between ferroptosis-related hub gene expression and prognosis in HCC remains to be further elucidated. Methods: Ferroptosis-related genes from the FerrDb database and the mRNA sequencing data and clinical information of HCC patients were obtained from The Cancer Genome Atlas (TCGA) database. The least absolute shrinkage and selection operator (LASSO) Cox regression was applied to identify a prognostic signature consisting of five ferroptosis-related hub genes in the TCGA cohort. The International Cancer Genome Consortium (ICGC) database was utilized to validate the reliability of the signature. Functional enrichment and immune-related analysis, including single-sample gene set enrichment analysis (ssGSEA), immune checkpoints, TIP-related genes, tumor stemness, and m6A-related genes, were performed to analyze the underlying mechanism. Additionally, the correlations between ferroptosis and drug resistance were evaluated using the NCI-60 database. Results: A 5–hub-gene signature associated with ferroptosis was constructed by multivariate Cox regression analysis to stratify patients into two risk groups. Patients with high risk had worse prognosis than those with low risk. Multivariate Cox regression analysis uncovered that the risk score was an independent prognostic indicator. We also proved the signature’s predictive capacity using the Kaplan–Meier method and receiver operating characteristic (ROC) curve analysis. Functional analysis showed that nuclear division and the cell cycle were enriched. Immune-related analysis revealed that the signature was enriched in immune-related pathways. Moreover, the risk signature was significantly associated with immune cell infiltration, immune checkpoints, TIP-related genes, tumor stem cells, as well as m6A-related genes. Furthermore, these genes were crucial regulators of drug resistance. Conclusion: We identified and validated a novel hub gene signature that is closely associated with ferroptosis as a new and efficient biomarker with favorable potential for predicting the prognosis of HCC patients. In addition, it also offers new insights into the molecular mechanisms of HCC and provides an effective approach for the treatment of HCC. Further studies are necessary to validate the results of our study.
Collapse
Affiliation(s)
- Wei Wang
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Fan Pan
- Department of Medical Oncology, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
| | - Xinrong Lin
- Department of Medical Oncology, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
| | - Jiakai Yuan
- Department of Medical Oncology, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
| | - Chunyu Tao
- Department of Medical Oncology, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Medical Oncology, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
- *Correspondence: Rui Wang,
| |
Collapse
|
90
|
An Q, Lin R, Wang D, Wang C. Emerging roles of fatty acid metabolism in cancer and their targeted drug development. Eur J Med Chem 2022; 240:114613. [PMID: 35853429 DOI: 10.1016/j.ejmech.2022.114613] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022]
Abstract
Metabolic reprogramming is now considered as one of hallmark of tumor cells and provides them with a selective survival/growth advantage to resist harsh micro-environmental stress. Fatty acid (FA) metabolism of tumor cells supports the biosynthetic needs and provides fuel sources for energy supply. Since FA metabolic reprogramming is a critical link in tumor metabolism, its various roles in tumors have attracted increasing interest. Herein, we review the mechanisms through which cancer cells rewire their FA metabolism with a focus on the pathway of FA metabolism and its targeting drug development. The failure and successful cases of targeting tumor FA metabolism are expected to bypass the metabolic vulnerability and improve the efficacy of targeted therapy.
Collapse
Affiliation(s)
- Qi An
- Scientific Research and Teaching Department, Public Health Clinical Center of Chengdu, 377 Jingming Road, Jinjiang District, Chengdu, Sichuan, 610061, China
| | - Rui Lin
- Scientific Research and Teaching Department, Public Health Clinical Center of Chengdu, 377 Jingming Road, Jinjiang District, Chengdu, Sichuan, 610061, China
| | - Dongmei Wang
- Scientific Research and Teaching Department, Public Health Clinical Center of Chengdu, 377 Jingming Road, Jinjiang District, Chengdu, Sichuan, 610061, China
| | - Chuan Wang
- Scientific Research and Teaching Department, Public Health Clinical Center of Chengdu, 377 Jingming Road, Jinjiang District, Chengdu, Sichuan, 610061, China.
| |
Collapse
|
91
|
Andreani C, Bartolacci C, Scaglioni PP. Ferroptosis: A Specific Vulnerability of RAS-Driven Cancers? Front Oncol 2022; 12:923915. [PMID: 35912247 PMCID: PMC9337859 DOI: 10.3389/fonc.2022.923915] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/03/2022] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis has emerged as a new type of programmed cell death that can be harnessed for cancer therapy. The concept of ferroptosis was for the first time proposed in in the early 2000s, as an iron-dependent mode of regulated cell death caused by unrestricted lipid peroxidation (LPO) and subsequent plasma membrane rupture. Since the discovery and characterization of ferroptosis, a wealth of research has improved our understanding of the main pathways regulating this process, leading to both the repurposing and the development of small molecules. However, ferroptosis is still little understood and several aspects remain to be investigated. For instance, it is unclear whether specific oncogenes, cells of origin or tumor niches impose specific susceptibility/resistance to ferroptosis or if there are some ferroptosis-related genes that may be used as bona fide pan-cancer targetable dependencies. In this context, even though RAS-driven cancer cell lines seemed to be selectively sensitive to ferroptosis inducers, subsequent studies have questioned these results, indicating that in some cases mutant RAS is necessary, but not sufficient to induce ferroptosis. In this perspective, based on publicly available genomic screening data and the literature, we discuss the relationship between RAS-mutation and ferroptosis susceptibility in cancer.
Collapse
Affiliation(s)
| | | | - Pier Paolo Scaglioni
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
92
|
Stockwell BR. Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell 2022; 185:2401-2421. [PMID: 35803244 PMCID: PMC9273022 DOI: 10.1016/j.cell.2022.06.003] [Citation(s) in RCA: 1495] [Impact Index Per Article: 498.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/17/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023]
Abstract
Ferroptosis, a form of cell death driven by iron-dependent lipid peroxidation, was identified as a distinct phenomenon and named a decade ago. Ferroptosis has been implicated in a broad set of biological contexts, from development to aging, immunity, and cancer. This review describes key regulators of this form of cell death within a framework of metabolism, ROS biology, and iron biology. Key concepts and major unanswered questions in the ferroptosis field are highlighted. The next decade promises to yield further breakthroughs in the mechanisms governing ferroptosis and additional ways of harnessing ferroptosis for therapeutic benefit.
Collapse
Affiliation(s)
- Brent R Stockwell
- Department of Biological Sciences, Department of Chemistry, Herbert Irving Comprehensive Cancer Center, Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA.
| |
Collapse
|
93
|
Abstract
Ferroptosis is a type of regulated cell death characterized by an excessive lipid peroxidation of cellular membranes caused by the disruption of the antioxidant defense system and/or an imbalanced cellular metabolism. Ferroptosis differentiates from other forms of regulated cell death in that several metabolic pathways and nutritional aspects, including endogenous antioxidants (such as coenzyme Q10, vitamin E, and di/tetrahydrobiopterin), iron handling, energy sensing, selenium utilization, amino acids, and fatty acids, directly regulate the cells' sensitivity to lipid peroxidation and ferroptosis. As hallmarks of ferroptosis have been documented in a variety of diseases, including neurodegeneration, acute organ injury, and therapy-resistant tumors, the modulation of ferroptosis using pharmacological tools or by metabolic reprogramming holds great potential for the treatment of ferroptosis-associated diseases and cancer therapy. Hence, this review focuses on the regulation of ferroptosis by metabolic and nutritional cues and discusses the potential of nutritional interventions for therapy by targeting ferroptosis. Expected final online publication date for the Annual Review of Nutrition, Volume 42 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany; .,Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany; .,Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
94
|
Zhang C, Yu J, Yang C, Shang S, Lv X, Cui B, Hua F. Crosstalk between ferroptosis and stress-Implications in cancer therapeutic responses. CANCER INNOVATION 2022; 1:92-113. [PMID: 38089453 PMCID: PMC10686180 DOI: 10.1002/cai2.7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/15/2022] [Indexed: 07/05/2024]
Abstract
Ferroptosis is a newly discovered form of cell death that is characterized by the accumulation of iron-dependent lipid peroxidation. Research on ferroptosis has seen exponential growth over the past few years. Tumor cells are strongly dependent on iron for their growth, which makes them develop mechanisms to increase iron uptake and inhibit iron output, thereby completing iron accumulation. Ferroptosis can be induced or inhibited by various stresses through multiple mechanisms, making it stands at the crossroads of stresses related cancer cell fate determination. In this review, we give a brief summary of ferroptosis hallmarks and provide a systematic analysis of the current molecular mechanisms and regulatory networks of diverse stress conditions on ferroptosis. We also discuss the relationships between ferroptosis and cancer therapy responses to further understand potential targets and therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Cheng Zhang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Jiao‐jiao Yu
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Chen Yang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Shuang Shang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xiao‐xi Lv
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Bing Cui
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Fang Hua
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
95
|
Wang G, Wang JJ, Xu XN, Shi F, Fu XL. Targeting cellular energy metabolism- mediated ferroptosis by small molecule compounds for colorectal cancer therapy. J Drug Target 2022; 30:819-832. [PMID: 35481396 DOI: 10.1080/1061186x.2022.2071909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Alterations in cellular energy metabolism, including glycolysis, glutamine and lipid metabolism that affects ferroptosis in the tumour microenvironment (TME), play a critical role in the development and progression of colorectal cancer (CRC) and offer evolutionary advantages to tumour cells and even enhance their aggressive phenotype. This review summarises the findings on the dysregulated energy metabolism pathways, including lipid and fatty acid metabolism especially for regulating the ferroptosis in TME. Moreover, the cellular energy metabolism and tumour ferroptosis to be regulated by small molecule compounds, which targeting the different aspects of metabolic pathways of energy production as well as metabolic enzymes that connect with the tumour cell growth and ferroptosis in CRC are also discussed. In this review, we will provide a comprehensive summary on small molecule compounds regulatory function of different energy metabolic routes on ferroptosis in tumour cells and discuss those metabolic vulnerabilities for the development of potential ferroptosis-based tumour therapies for colorectal cancer.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Xiao-Na Xu
- Department of Medicine, Jiangsu University, Zhenjiang City, China
| | - Feng Shi
- Department of Medicine, Jiangsu University, Zhenjiang City, China
| | - Xing-Li Fu
- Department of Medicine, Jiangsu University, Zhenjiang City, China
| |
Collapse
|
96
|
Martins AC, Virgolini MB, Tinkov AA, Skalny AV, Tirumala RP, Farina M, Santamaria A, Lu R, Aschner M. Iron overload and neurodegenerative diseases: What can we learn from Caenorhabditis elegans? TOXICOLOGY RESEARCH AND APPLICATION 2022; 6. [PMID: 35990536 PMCID: PMC9390093 DOI: 10.1177/23978473221091852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Iron (Fe) is an essential trace element required for several physiological processes. It plays important roles in mitochondrial function, synthesis, and metabolism of the neurotransmitter, as well as oxygen transport. However, excess Fe can cause toxicity. Particularly, Fe overload may result in neurotoxicity, contributing to the development and progression of neurodegenerative diseases, although the molecular mechanisms underlying Fe-induced neurodegeneration have yet to be entirely understood. Alternative (non-rodent) experimental models have been pointed as important approaches to elucidate molecular and physiological events mediating Fe-induced pathology. Among such alternative strategies, an advantageous experimental worm-model system, Caenorhabditis elegans ( C. elegans), has been used to investigate Fe-induced neurotoxicity and neurodegenerative disorders. Its genome has been fully sequenced, corroborating that it shares significant homology with mammalians, and has approximately 40% of human disease-related genes. As part of this review, we discuss studies using the C. elegans model to study molecular mechanisms such as oxidative stress, mitochondrial dysfunction, disturbed homeostasis, and its potential contribution to the study of metal-induced neurodegenerative diseases such as Parkinson’s disease (PD) and Alzheimer’s disease (AD).
Collapse
Affiliation(s)
- Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Miriam B Virgolini
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Técnicas (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, Russia
| | - Anatoly V. Skalny
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- K.G. Razumovsky Moscow State University of Technologies and Management, Moscow, Russia
| | | | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
97
|
Krümmel B, von Hanstein AS, Plötz T, Lenzen S, Mehmeti I. Differential effects of saturated and unsaturated free fatty acids on ferroptosis in rat β-cells. J Nutr Biochem 2022; 106:109013. [PMID: 35447320 DOI: 10.1016/j.jnutbio.2022.109013] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/21/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022]
Abstract
Elevated plasma concentrations of saturated free fatty acids (SFAs) are involved in pancreatic β-cell dysfunction and apoptosis, referred to as lipotoxicity. However, in contrast to apoptosis, the involvement of ferroptosis, as a distinct type of oxidative regulated cell death in β-cell lipotoxicity remains elusive. Therefore, the aim of this study was to determine the effects of various free fatty acids on ferroptosis induction in rat insulin-producing β-cells. Herein, rat insulin-producing β-cells underwent lipid peroxidation in the presence of long-chain SFAs and ω-6-polyunsaturated fatty acids (PUFAs), but only the latter induced ferroptosis. On the other hand, ω-3-polyunsaturated fatty acid α-linolenate did not induce ferroptosis but sensitized insulin-producing β-cells to SFA-mediated lipid peroxidation. While the monounsaturated fatty acid oleate, overexpression of glutathione peroxidase 4 (GPx4), and the specific ferroptosis inhibitor ferrostatin-1 significantly abrogated lipid peroxidation, neither GPx4 nor ferrostatin-1 affected palmitate-mediated toxicity. Site-specific expression of catalase in cytosol, mitochondria, and ER attenuated lipid peroxidation, indicating the contribution of metabolically generated H2O2 from all three subcellular compartments. These observations suggest that only ω-6-PUFAs reach the thresholds of lipid peroxidation required for ferroptosis, whereas SFAs favour apoptosis in β-cells. Hence, avoiding an excessive dietary intake of ω-6-PUFAs might be a crucial prerequisite for prevention of reactive oxygen species-mediated ferroptosis in insulin-producing cells.
Collapse
Affiliation(s)
- Bastian Krümmel
- Institute of Experimental Diabetes Research, Hannover Medical School, Hannover, Germany; Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Anna-Sophie von Hanstein
- Institute of Experimental Diabetes Research, Hannover Medical School, Hannover, Germany; Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Thomas Plötz
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Sigurd Lenzen
- Institute of Experimental Diabetes Research, Hannover Medical School, Hannover, Germany; Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Ilir Mehmeti
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
98
|
Li X, Zhou L, Lu T, Zhang L, Li Y, Xu J, Yin M, Long H. Constructing an immune- and ferroptosis-related lncRNA signature to predict the immune landscape of human bladder cancer. J Clin Lab Anal 2022; 36:e24389. [PMID: 35421267 PMCID: PMC9102655 DOI: 10.1002/jcla.24389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/25/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Background LncRNAs play a variety of roles in the tumor microenvironment and cancer immune responses. Determining the significance of bladder cancer (BLCA)‐related genes to predict the prognostic and therapeutic response of BLCA is important. Methods IrlncRNA/ frlncRNA pairs were determined using univariate analysis. The signature was constructed based on this pairs. Finally, analysis and internal validation were performed from several aspects. Results We identified 60 immune‐ and ferroptosis‐related lncRNA pairs, among which 12 were included in the Cox proportional hazards model. Patients in low‐risk group survived for significantly longer. Survival and riskScore analyses showed that the low‐risk group had a significantly better clinical outcome. ROC curve analysis showed that AUC of OS values were more than 0.75 in the training set and the whole cohort. As assessed using Cox analysis, the riskScore was an independent prognostic predictor in the training, testing set and the whole cohort. The areas under the multi‐index ROC in the training set, the testing set, and the whole cohort were 0.777, 0.692, and 0.748, respectively. High‐risk group was positively associated with most of tumor‐infiltrating immune cells. High‐risk Scores correlated positively with high expression of CD274, but not with PD‐1. Low riskScores correlated positively with high expression levels of the genes ERBB2 and nectin‐4. High‐risk Score was associated with a lower IC50 value for Docetaxel, cisplatin, and Pazopanib, while there was an opposite result for metformin. Conclusions The signature constructed by pairing irlncRNAs and frlncRNAs showed a notable clinical predictive value.
Collapse
Affiliation(s)
- Xing Li
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Libin Zhou
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Tefei Lu
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Lei Zhang
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Yanjun Li
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Jianting Xu
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Min Yin
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Huimin Long
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
99
|
Zhou A, Fang T, Chen K, Xu Y, Chen Z, Ning X. Biomimetic Activator of Sonodynamic Ferroptosis Amplifies Inherent Peroxidation for Improving the Treatment of Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106568. [PMID: 35092152 DOI: 10.1002/smll.202106568] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/24/2021] [Indexed: 06/14/2023]
Abstract
Ferroptosis is a type of nonapoptotic cell death and is gradually emerging as an important anticancer treatment. However, its therapeutic efficacy is impaired by low intracellular levels of reactive oxygen species (ROS) and long-chain polyunsaturated fatty acids, significantly limiting its therapeutic potential. Herein, a multimodal strategy to improve ferroptosis is presented, in which a state-of-art engineered erythrocyte, termed as sonodynamic amplified ferroptosis erythrocyte (SAFE), is developed for simultaneously activating ferroptosis and oxygen-riched sonodynamic therapy (SDT). SAFE is composed of internalizing RGD peptide and red blood cell membrane hybrid camouflaged nanocomplex of hemoglobin, perfluorocarbon, ferroptosis activator (dihomo-γ-linolenic acid, DGLA), and sonosensitizer verteporfin. It is identified that SAFE, under ultrasound stimulation, can not only substantially supply oxygen to overcome tumor hypoxia associated therapeutic resistance, but effectively activate ferroptosis through the coeffect of SDT triggered ROS production and DGLA mediated lipid peroxidation. In vivo studies reveal that SAFE selectively accumulates in tumor tissues and induces desirable anticancer effects under mild ultrasound stimulation. Importantly, SAFE can effectively inhibit tumor growth with minimal invasiveness, resulting in a prolonged survival period of mice. Therefore, a multimodal ferroptosis therapy driven by oxygen-riched sonodynamic peroxidation of lipids, significantly advancing synergistic cancer treatment, is presented.
Collapse
Affiliation(s)
- Anwei Zhou
- Jiangsu Province Nanjing, Qixia District, Xianlin Road No. 163, Nanjing, 210093, China
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, School of Physics, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Tianliang Fang
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-Sen University, Guangzhou, 510008, China
| | - Kerong Chen
- Jiangsu Province Nanjing, Qixia District, Xianlin Road No. 163, Nanjing, 210093, China
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Yurui Xu
- Jiangsu Province Nanjing, Qixia District, Xianlin Road No. 163, Nanjing, 210093, China
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Zhuo Chen
- Jiangsu Province Nanjing, Qixia District, Xianlin Road No. 163, Nanjing, 210093, China
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, School of Physics, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Xinghai Ning
- Jiangsu Province Nanjing, Qixia District, Xianlin Road No. 163, Nanjing, 210093, China
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
100
|
MS4A15 drives ferroptosis resistance through calcium-restricted lipid remodeling. Cell Death Differ 2022; 29:670-686. [PMID: 34663908 PMCID: PMC8901757 DOI: 10.1038/s41418-021-00883-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 01/07/2023] Open
Abstract
Ferroptosis is an iron-dependent form of cell death driven by biochemical processes that promote oxidation within the lipid compartment. Calcium (Ca2+) is a signaling molecule in diverse cellular processes such as migration, neurotransmission, and cell death. Here, we uncover a crucial link between ferroptosis and Ca2+ through the identification of the novel tetraspanin MS4A15. MS4A15 localizes to the endoplasmic reticulum, where it blocks ferroptosis by depleting luminal Ca2+ stores and reprogramming membrane phospholipids to ferroptosis-resistant species. Specifically, prolonged Ca2+ depletion inhibits lipid elongation and desaturation, driving lipid droplet dispersion and formation of shorter, more saturated ether lipids that protect phospholipids from ferroptotic reactive species. We further demonstrate that increasing luminal Ca2+ levels can preferentially sensitize refractory cancer cell lines. In summary, MS4A15 regulation of anti-ferroptotic lipid reservoirs provides a key resistance mechanism that is distinct from antioxidant and lipid detoxification pathways. Manipulating Ca2+ homeostasis offers a compelling strategy to balance cellular lipids and cell survival in ferroptosis-associated diseases.
Collapse
|