1
|
Zhou Q, You Y, Zhao Y, Xiao S, Song Z, Huang C, Qian J, Lu W, Tong H, Zhang Y, Wang Z, Li W, Zhang C, Guo X, Luo R, Hou Y, Cui J, Lu L, Zhou Y. TRPV4 drives the progression of leiomyosarcoma by promoting ECM1 generation and co-activating the FAK/PI3K/AKT/GSK3β pathway. Cell Oncol (Dordr) 2025; 48:455-470. [PMID: 39612152 PMCID: PMC11996984 DOI: 10.1007/s13402-024-01008-7] [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] [Accepted: 10/22/2024] [Indexed: 11/30/2024] Open
Abstract
PURPOSE Leiomyosarcoma (LMS) is an aggressive mesenchymal malignant tumor with poor therapeutic options, but the molecular mechanisms underlying LMS remain largely unknown. Increasing evidence indicates that transient receptor potential vanilloid 4 (TRPV4) levels are closely related to the advancement of various malignant tumors through diverse molecular mechanisms. However, the roles and regulatory mechanisms of TRPV4 in LMS progression remain unclear. METHODS Immunohistochemistry, Western blot, and immunofluorescence were used to investigate the relationship between TRPV4 expression and LMS. Survival analysis was conducted to evaluate the association between TRPV4 levels and prognosis in LMS patients. Intracellular Ca2+ measurement, colony formation, CCK-8, wound healing and Transwell assays and peritoneal metastasis mouse model were used to verify the effect of TRPV4 activity and expression on LMS proliferation and metastasis. RNA-seq and proteomics were performed to explore the underlying mechanism. RESULTS TRPV4 was upregulated in LMS tissues and cells and served as a novel prognostic factor. Moreover, TRPV4 overexpression enhanced cell proliferation, cell migration and invasion of LMS cells in vitro, as well as promoted tumor metastasis in vivo, which could be blocked by HC067047 intervention or TRPV4 knockdown. Combined RNA-seq and proteomics analysis of KEGG pathway indicated that ECM receptor interaction was obviously activated. Extracellular matrix protein 1 (ECM1) was identified as downstream gene of TRPV4. Mechanistically, TRPV4 overexpression increased ECM1 level and activated the FAK/PI3K/AKT/GSK3β pathway, which could be reversed by TRPV4 knockdown or LY294002 treatment. Moreover, ECM1 overexpression enhanced the activation of FAK/PI3K/AKT/GSK3β pathway. And simultaneous overexpression of TRPV4 and ECM1 synergistically activated this pathway. CONCLUSION Our findings provide a novel mechanism by which TRPV4 directly activates Ca2+/FAK/PI3K/AKT/GSK3β pathway and further indirectly enhances the FAK/PI3K/AKT/GSK3β pathway through the promotion and secretion of ECM1 to promote LMS malignant progression. Targeting the TRPV4/FAK axis might be a promising potential strategy for prognosis and treatment of LMS.
Collapse
Affiliation(s)
- Qiwen Zhou
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yang You
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yingying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Shuxiu Xiao
- Clinical Centre for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhengqing Song
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chuxin Huang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jiali Qian
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Weiqi Lu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hanxing Tong
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yong Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhiming Wang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Li
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chenlu Zhang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xi Guo
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Lili Lu
- Clinical Centre for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Xiamen Key Laboratory of Biotherapy, Xiamen, 361000, China.
| | - Yuhong Zhou
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Clinical Centre for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Gollowitzer A, Pein H, Rao Z, Waltl L, Bereuter L, Loeser K, Meyer T, Jafari V, Witt F, Winkler R, Su F, Große S, Thürmer M, Grander J, Hotze M, Harder S, Espada L, Magnutzki A, Gstir R, Weinigel C, Rummler S, Bonn G, Pachmayr J, Ermolaeva M, Harayama T, Schlüter H, Kosan C, Heller R, Thedieck K, Schmitt M, Shimizu T, Popp J, Shindou H, Kwiatkowski M, Koeberle A. Attenuated growth factor signaling during cell death initiation sensitizes membranes towards peroxidation. Nat Commun 2025; 16:1774. [PMID: 40000627 PMCID: PMC11861335 DOI: 10.1038/s41467-025-56711-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Cell death programs such as apoptosis and ferroptosis are associated with aberrant redox homeostasis linked to lipid metabolism and membrane function. Evidence for cross-talk between these programs is emerging. Here, we show that cytotoxic stress channels polyunsaturated fatty acids via lysophospholipid acyltransferase 12 into phospholipids that become susceptible to peroxidation under additional redox stress. This reprogramming is associated with altered acyl-CoA synthetase isoenzyme expression and caused by a decrease in growth factor receptor tyrosine kinase (RTK)-phosphatidylinositol-3-kinase signaling, resulting in suppressed fatty acid biosynthesis, for specific stressors via impaired Akt-SREBP1 activation. The reduced availability of de novo synthesized fatty acids favors the channeling of polyunsaturated fatty acids into phospholipids. Growth factor withdrawal by serum starvation mimics this phenotype, whereas RTK ligands counteract it. We conclude that attenuated RTK signaling during cell death initiation increases cells' susceptibility to oxidative membrane damage at the interface of apoptosis and alternative cell death programs.
Collapse
Affiliation(s)
- André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Helmut Pein
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Zhigang Rao
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Lorenz Waltl
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Leonhard Bereuter
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Konstantin Loeser
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Tobias Meyer
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Vajiheh Jafari
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Finja Witt
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - René Winkler
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
- Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916, Badalona, Spain
| | - Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Silke Große
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Maria Thürmer
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Madlen Hotze
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Sönke Harder
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lilia Espada
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Alexander Magnutzki
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Ronald Gstir
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Christina Weinigel
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Silke Rummler
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Günther Bonn
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Johanna Pachmayr
- Institute of Pharmacy, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Maria Ermolaeva
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Takeshi Harayama
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur - CNRS UMR7275 - Inserm U1323, 06560, Valbonne, France
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
| | - Regine Heller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Kathrin Thedieck
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
- Department Metabolism, Senescence and Autophagy, Research Center One Health Ruhr, University Alliance Ruhr & University Hospital Essen, University Duisburg-Essen, 45141, Essen, Germany
- Freiburg Materials Research Center FMF, Albert-Ludwigs-University of Freiburg, 79104, Freiburg, Germany
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
- German Cancer Consortium (DKTK), partner site Essen/Duesseldorf, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147, Essen, Germany
| | - Michael Schmitt
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Institute of Microbial Chemistry, Tokyo 141-0021, Japan
| | - Jürgen Popp
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Hideo Shindou
- Department of Lipid Life Science, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Marcel Kwiatkowski
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany.
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria.
| |
Collapse
|
3
|
Koeberle SC, Thürmer M, Su F, Werner M, Grander J, Hofer L, Gollowitzer A, Xuan LL, Benscheid FJ, Bonyadi Rad E, Zarrelli A, Di Fabio G, Werz O, Romanucci V, Lupp A, Koeberle A. Silybin A from Silybum marianum reprograms lipid metabolism to induce a cell fate-dependent class switch from triglycerides to phospholipids. Theranostics 2025; 15:2006-2034. [PMID: 39897559 PMCID: PMC11780512 DOI: 10.7150/thno.99562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/25/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Silybum marianum is used to protect against degenerative liver damage. The molecular mechanisms of its bioactive component, silybin, remained enigmatic, although membrane-stabilizing properties, modulation of membrane protein function, and metabolic regulation have been discussed for decades. Methods: Experiments were performed with hepatocyte cell lines and primary monocytes in vitro under both basal and stressed conditions, and in mice in vivo. Quantitative lipidomics was used to detect changes in phospholipids and triglycerides. Key findings were confirmed by Western blotting, quantitative PCR, microscopy, enzyme activity assays, metabolic flux studies, and functional relationships were investigated using selective inhibitors. Results: We show that specifically the stereoisomer silybin A decreases triglyceride levels and lipid droplet content, while enriching major phospholipid classes and maintaining a homeostatic phospholipid composition in human hepatocytes in vitro and in mouse liver in vivo under normal and pre-disease conditions. Conversely, in cell-based disease models of lipid overload and lipotoxic stress, silybin treatment primarily depletes triglycerides. Mechanistically, silymarin/silybin suppresses phospholipid-degrading enzymes, induces phospholipid biosynthesis to varying degrees depending on the conditions, and down-regulates triglyceride remodeling/biosynthesis, while inducing complex changes in sterol and fatty acid metabolism. Structure-activity relationship studies highlight the importance of the 1,4-benzodioxane ring configuration of silybin A in triglyceride reduction and the saturated 2,3-bond of the flavanonol moiety in phospholipid accumulation. Enrichment of hepatic phospholipids and intracellular membrane expansion are associated with a heightened biotransformation capacity. Conclusion: Our study deciphers the structural features of silybin contributing to hepatic lipid remodeling and suggests that silymarin/silybin protects the liver in individuals with mild metabolic dysregulation, involving a lipid class switch from triglycerides to phospholipids, whereas it may be less effective in disease states associated with severe metabolic dysregulation.
Collapse
Affiliation(s)
- Solveigh C. Koeberle
- Institute of Pharmaceutical Sciences/Pharmacognosy and Excellence Field BioHealth, University of Graz, 8010 Graz, Austria
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Maria Thürmer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Fengting Su
- Institute of Pharmaceutical Sciences/Pharmacognosy and Excellence Field BioHealth, University of Graz, 8010 Graz, Austria
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Markus Werner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Laura Hofer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Loc Le Xuan
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Felix J. Benscheid
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Ehsan Bonyadi Rad
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Armando Zarrelli
- Department of Chemical Sciences, University of Napoli Federico II, I-80126 Naples, Italy
| | - Giovanni Di Fabio
- Department of Chemical Sciences, University of Napoli Federico II, I-80126 Naples, Italy
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Valeria Romanucci
- Department of Chemical Sciences, University of Napoli Federico II, I-80126 Naples, Italy
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Andreas Koeberle
- Institute of Pharmaceutical Sciences/Pharmacognosy and Excellence Field BioHealth, University of Graz, 8010 Graz, Austria
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
4
|
Su F, Descher H, Bui-Hoang M, Stuppner H, Skvortsova I, Rad EB, Ascher C, Weiss A, Rao Z, Hohloch S, Koeberle SC, Gust R, Koeberle A. Iron(III)-salophene catalyzes redox cycles that induce phospholipid peroxidation and deplete cancer cells of ferroptosis-protecting cofactors. Redox Biol 2024; 75:103257. [PMID: 38955113 PMCID: PMC11263665 DOI: 10.1016/j.redox.2024.103257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Ferroptosis, a lipid peroxidation-driven cell death program kept in check by glutathione peroxidase 4 and endogenous redox cycles, promises access to novel strategies for treating therapy-resistant cancers. Chlorido [N,N'-disalicylidene-1,2-phenylenediamine]iron (III) complexes (SCs) have potent anti-cancer properties by inducing ferroptosis, apoptosis, or necroptosis through still poorly understood molecular mechanisms. Here, we show that SCs preferentially induce ferroptosis over other cell death programs in triple-negative breast cancer cells (LC50 ≥ 0.07 μM) and are particularly effective against cell lines with acquired invasiveness, chemo- or radioresistance. Redox lipidomics reveals that initiation of cell death is associated with extensive (hydroper)oxidation of arachidonic acid and adrenic acid in membrane phospholipids, specifically phosphatidylethanolamines and phosphatidylinositols, with SCs outperforming established ferroptosis inducers. Mechanistically, SCs effectively catalyze one-electron transfer reactions, likely via a redox cycle involving the reduction of Fe(III) to Fe(II) species and reversible formation of oxo-bridged dimeric complexes, as supported by cyclic voltammetry. As a result, SCs can use hydrogen peroxide to generate organic radicals but not hydroxyl radicals and oxidize membrane phospholipids and (membrane-)protective factors such as NADPH, which is depleted from cells. We conclude that SCs catalyze specific redox reactions that drive membrane peroxidation while interfering with the ability of cells, including therapy-resistant cancer cells, to detoxify phospholipid hydroperoxides.
Collapse
Affiliation(s)
- Fengting Su
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Hubert Descher
- Institute of Pharmacy/Pharmaceutical Chemistry, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Minh Bui-Hoang
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria; Unit of Pharmacognosy, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Hermann Stuppner
- Unit of Pharmacognosy, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Ira Skvortsova
- EXTRO-Lab, Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ehsan Bonyadi Rad
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Claudia Ascher
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Alexander Weiss
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Zhigang Rao
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Stephan Hohloch
- Institute for General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Solveigh C Koeberle
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Ronald Gust
- Institute of Pharmacy/Pharmaceutical Chemistry, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
5
|
Schwab A, Rao Z, Zhang J, Gollowitzer A, Siebenkäs K, Bindel N, D'Avanzo E, van Roey R, Hajjaj Y, Özel E, Armstark I, Bereuter L, Su F, Grander J, Bonyadi Rad E, Groenewoud A, Engel FB, Bell GW, Henry WS, Angeli JPF, Stemmler MP, Brabletz S, Koeberle A, Brabletz T. Zeb1 mediates EMT/plasticity-associated ferroptosis sensitivity in cancer cells by regulating lipogenic enzyme expression and phospholipid composition. Nat Cell Biol 2024; 26:1470-1481. [PMID: 39009641 PMCID: PMC11392809 DOI: 10.1038/s41556-024-01464-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 06/20/2024] [Indexed: 07/17/2024]
Abstract
Therapy resistance and metastasis, the most fatal steps in cancer, are often triggered by a (partial) activation of the epithelial-mesenchymal transition (EMT) programme. A mesenchymal phenotype predisposes to ferroptosis, a cell death pathway exerted by an iron and oxygen-radical-mediated peroxidation of phospholipids containing polyunsaturated fatty acids. We here show that various forms of EMT activation, including TGFβ stimulation and acquired therapy resistance, increase ferroptosis susceptibility in cancer cells, which depends on the EMT transcription factor Zeb1. We demonstrate that Zeb1 increases the ratio of phospholipids containing pro-ferroptotic polyunsaturated fatty acids over cyto-protective monounsaturated fatty acids by modulating the differential expression of the underlying crucial enzymes stearoyl-Co-A desaturase 1 (SCD), fatty acid synthase (FASN), fatty acid desaturase 2 (FADS2), elongation of very long-chain fatty acid 5 (ELOVL5) and long-chain acyl-CoA synthetase 4 (ACSL4). Pharmacological inhibition of selected lipogenic enzymes (SCD and FADS2) allows the manipulation of ferroptosis sensitivity preferentially in high-Zeb1-expressing cancer cells. Our data are of potential translational relevance and suggest a combination of ferroptosis activators and SCD inhibitors for the treatment of aggressive cancers expressing high Zeb1.
Collapse
Affiliation(s)
- Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Zhigang Rao
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Jie Zhang
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Katharina Siebenkäs
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Nino Bindel
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Elisabetta D'Avanzo
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Yussuf Hajjaj
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ece Özel
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Isabell Armstark
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Leonhard Bereuter
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Ehsan Bonyadi Rad
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Arwin Groenewoud
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - George W Bell
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Whitney S Henry
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Dept. of Biology, MIT, Cambridge, MA, USA
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria.
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| |
Collapse
|
6
|
Hu R, Fan W, Li S, Zhang G, Zang L, Qin L, Li R, Chen R, Zhang L, Gu W, Zhang Y, Rajagopalan S, Sun Q, Liu C. PM 2.5-induced cellular senescence drives brown adipose tissue impairment in middle-aged mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116423. [PMID: 38705039 PMCID: PMC12070286 DOI: 10.1016/j.ecoenv.2024.116423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Airborne fine particulate matter (PM2.5) exposure is closely associated with metabolic disturbance, in which brown adipose tissue (BAT) is one of the main contributing organs. However, knowledge of the phenotype and mechanism of PM2.5 exposure-impaired BAT is quite limited. In the study, male C57BL/6 mice at three different life phases (young, adult, and middle-aged) were simultaneously exposed to concentrated ambient PM2.5 or filtered air for 8 weeks using a whole-body inhalational exposure system. H&E staining and high-resolution respirometry were used to assess the size of adipocytes and mitochondrial function. Transcriptomics was performed to determine the differentially expressed genes in BAT. Quantitative RT-PCR, immunohistochemistry staining, and immunoblots were performed to verify the transcriptomics and explore the mechanism for BAT mitochondrial dysfunction. Firstly, PM2.5 exposure caused altered BAT morphology and mitochondrial dysfunction in middle-aged but not young or adult mice. Furthermore, PM2.5 exposure increased cellular senescence in BAT of middle-aged mice, accompanied by cell cycle arrest, impaired DNA replication, and inhibited AKT signaling pathway. Moreover, PM2.5 exposure disrupted apoptosis and autophagy homeostasis in BAT of middle-aged mice. Therefore, BAT in middle-aged mice was more vulnerable to PM2.5 exposure, and the cellular senescence-initiated apoptosis, autophagy, and mitochondrial dysfunction may be the mechanism of PM2.5 exposure-induced BAT impairment.
Collapse
Affiliation(s)
- Renjie Hu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Wenjun Fan
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Sanduo Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Guoqing Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Lu Zang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Li Qin
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Rucheng Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Lu Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Weijia Gu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Yunhui Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200433, China
| | - Sanjay Rajagopalan
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China.
| |
Collapse
|
7
|
Bielawiec P, Dziemitko S, Konstantynowicz-Nowicka K, Sztolsztener K, Chabowski A, Harasim-Symbor E. Cannabigerol-A useful agent restoring the muscular phospholipids milieu in obese and insulin-resistant Wistar rats? Front Mol Biosci 2024; 11:1401558. [PMID: 38919749 PMCID: PMC11196617 DOI: 10.3389/fmolb.2024.1401558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Numerous strategies have been proposed to minimize obesity-associated health effects, among which phytocannabinoids appear to be effective and safe compounds. In particular, cannabigerol (CBG) emerges as a potent modulator of the composition of membrane phospholipids (PLs), which plays a critical role in the development of insulin resistance. Therefore, here we consider the role of CBG treatment on the composition of PLs fraction with particular emphasis on phospholipid subclasses (e.g., phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS), and phosphatidylinositol (PI)) in the red gastrocnemius muscle of Wistar rats fed the standard or high-fat, high-sucrose (HFHS) diet. The intramuscular PLs content was determined by gas-liquid chromatography and based on the composition of individual FAs, we assessed the stearoyl-CoA desaturase 1 (SCD1) index as well as the activity of n-3 and n-6 polyunsaturated fatty acids (PUFAs) pathways. Expression of various proteins engaged in the inflammatory pathway, FAs elongation, and desaturation processes was measured using Western blotting. Our research has demonstrated the important association of obesity with alterations in the composition of muscular PLs, which was significantly improved by CBG supplementation, enriching the lipid pools in n-3 PUFAs and decreasing the content of arachidonic acid (AA), which in turn influenced the activity of PUFAs pathways in various PLs subclasses. CBG also inhibited the local inflammation development and profoundly reduced the SCD1 activity. Collectively, restoring the PLs homeostasis of the myocyte membrane by CBG indicates its new potential medical application in the treatment of obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Patrycja Bielawiec
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | | | | | | | | | | |
Collapse
|
8
|
Su F, Koeberle A. Regulation and targeting of SREBP-1 in hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:673-708. [PMID: 38036934 PMCID: PMC11156753 DOI: 10.1007/s10555-023-10156-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is an increasing burden on global public health and is associated with enhanced lipogenesis, fatty acid uptake, and lipid metabolic reprogramming. De novo lipogenesis is under the control of the transcription factor sterol regulatory element-binding protein 1 (SREBP-1) and essentially contributes to HCC progression. Here, we summarize the current knowledge on the regulation of SREBP-1 isoforms in HCC based on cellular, animal, and clinical data. Specifically, we (i) address the overarching mechanisms for regulating SREBP-1 transcription, proteolytic processing, nuclear stability, and transactivation and (ii) critically discuss their impact on HCC, taking into account (iii) insights from pharmacological approaches. Emphasis is placed on cross-talk with the phosphatidylinositol-3-kinase (PI3K)-protein kinase B (Akt)-mechanistic target of rapamycin (mTOR) axis, AMP-activated protein kinase (AMPK), protein kinase A (PKA), and other kinases that directly phosphorylate SREBP-1; transcription factors, such as liver X receptor (LXR), peroxisome proliferator-activated receptors (PPARs), proliferator-activated receptor γ co-activator 1 (PGC-1), signal transducers and activators of transcription (STATs), and Myc; epigenetic mechanisms; post-translational modifications of SREBP-1; and SREBP-1-regulatory metabolites such as oxysterols and polyunsaturated fatty acids. By carefully scrutinizing the role of SREBP-1 in HCC development, progression, metastasis, and therapy resistance, we shed light on the potential of SREBP-1-targeting strategies in HCC prevention and treatment.
Collapse
Affiliation(s)
- Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
9
|
Wibel R, van Hoogevest P, Drescher S. The role of phospholipids in drug delivery formulations - Recent advances presented at the Researcher's Day 2023 Conference of the Phospholipid Research Center Heidelberg. Eur J Pharm Biopharm 2024; 197:114215. [PMID: 38350530 DOI: 10.1016/j.ejpb.2024.114215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/15/2024]
Abstract
This Focus on Meetings contribution summarizes recent advances in the research on phospholipids and their applications for drug delivery and analytical purposes that have been presented at the hybrid Researcher's Day 2023 Conference of the Phospholipid Research Center (PRC), held on July 3-5, 2023, in Bad Dürkheim, Germany. The PRC is a non-profit organization focused on expanding and sharing scientific and technological knowledge of phospholipids in pharmaceutical and other applications. This is accomplished by, e.g., funding doctoral and postdoctoral research projects. The progress made with these projects is presented at the Researcher's Day Conference every two years. Four main topics were presented and discussed in various lectures: (1) formulation of phospholipid-based nanocarriers, (2) therapeutic applications of phospholipids and phospholipid-based nanocarriers, (3) phospholipids as excipients in oral, dermal, and parenteral dosage forms, and (4) interactions of phospholipids and phospholipid-based vesicles in biological environment and their use as analytical platforms.
Collapse
Affiliation(s)
- Richard Wibel
- Phospholipid Research Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Peter van Hoogevest
- Phospholipid Research Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Simon Drescher
- Phospholipid Research Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany.
| |
Collapse
|
10
|
Sharifi S, Chaudhari P, Martirosyan A, Eberhardt AO, Witt F, Gollowitzer A, Lange L, Woitzat Y, Okoli EM, Li H, Rahnis N, Kirkpatrick J, Werz O, Ori A, Koeberle A, Bierhoff H, Ermolaeva M. Reducing the metabolic burden of rRNA synthesis promotes healthy longevity in Caenorhabditis elegans. Nat Commun 2024; 15:1702. [PMID: 38402241 PMCID: PMC10894287 DOI: 10.1038/s41467-024-46037-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
Ribosome biogenesis is initiated by RNA polymerase I (Pol I)-mediated synthesis of pre-ribosomal RNA (pre-rRNA). Pol I activity was previously linked to longevity, but the underlying mechanisms were not studied beyond effects on nucleolar structure and protein translation. Here we use multi-omics and functional tests to show that curtailment of Pol I activity remodels the lipidome and preserves mitochondrial function to promote longevity in Caenorhabditis elegans. Reduced pre-rRNA synthesis improves energy homeostasis and metabolic plasticity also in human primary cells. Conversely, the enhancement of pre-rRNA synthesis boosts growth and neuromuscular performance of young nematodes at the cost of accelerated metabolic decline, mitochondrial stress and premature aging. Moreover, restriction of Pol I activity extends lifespan more potently than direct repression of protein synthesis, and confers geroprotection even when initiated late in life, showcasing this intervention as an effective longevity and metabolic health treatment not limited by aging.
Collapse
Affiliation(s)
- Samim Sharifi
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, Jena, 07745, Germany
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
- Matter Bio, Inc., Brooklyn, NY, 11237, USA
| | - Prerana Chaudhari
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Asya Martirosyan
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Joseph-Stelzmann-Straße 26, 50931, Cologne, Germany
| | - Alexander Otto Eberhardt
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, Jena, 07745, Germany
| | - Finja Witt
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Lisa Lange
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, Jena, 07745, Germany
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Yvonne Woitzat
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | | | - Huahui Li
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, PR China
| | - Norman Rahnis
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Joanna Kirkpatrick
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
- Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Holger Bierhoff
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, Jena, 07745, Germany.
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany.
| | - Maria Ermolaeva
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
11
|
Lang L, Zheng J, Liang S, Zhang F, Fu Y, Deng K, Li F, Yang X, Wang J, Luo Y, Zhang S, Zhu X, Wang L, Gao P, Zhu C, Shu G, Xi Q, Zhang Y, Jiang Q, Wang S. Browning of Mammary Fat Suppresses Pubertal Mammary Gland Development of Mice via Elevation of Serum Phosphatidylcholine and Inhibition of PI3K/Akt Pathway. Int J Mol Sci 2023; 24:16171. [PMID: 38003364 PMCID: PMC10671055 DOI: 10.3390/ijms242216171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Mammary fat plays a profound role in the postnatal development of mammary glands. However, the specific types (white, brown, or beige) of adipocytes in mammary fat and their potential regulatory effects on modulating mammary gland development remain poorly understood. This study aimed to investigate the role of the browning of mammary fat on pubertal mammary gland development and explore the underlying mechanisms. Thus, the mammary gland development and the serum lipid profile were evaluated in mice treated with CL316243, a β3-adrenoceptor agonist, to induce mammary fat browning. In addition, the proliferation of HC11 cells co-cultured with brown adipocytes or treated with the altered serum lipid metabolite was determined. Our results showed that the browning of mammary fat by injection of CL316243 suppressed the pubertal development of mice mammary glands, accompanied by the significant elevation of serum dioleoylphosphocholine (DOPC). In addition, the proliferation of HC11 was repressed when co-cultured with brown adipocytes or treated with DOPC. Furthermore, DOPC suppressed the activation of the PI3K/Akt pathway, while the DOPC-inhibited HC11 proliferation was reversed by SC79, an Akt activator, suggesting the involvement of the PI3K/Akt pathway in the DOPC-inhibited proliferation of HC11. Together, the browning of mammary fat suppressed the development of the pubertal mammary gland, which was associated with the elevated serum DOPC and the inhibition of the PI3K/Akt pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (L.L.); (J.Z.); (S.L.); (F.Z.); (Y.F.); (K.D.); (F.L.); (X.Y.); (J.W.); (Y.L.); (S.Z.); (X.Z.); (L.W.); (P.G.); (C.Z.); (G.S.); (Q.X.); (Y.Z.); (Q.J.)
| |
Collapse
|
12
|
Liao S, Gollowitzer A, Börmel L, Maier C, Gottschalk L, Werz O, Wallert M, Koeberle A, Lorkowski S. α-Tocopherol-13'-Carboxychromanol Induces Cell Cycle Arrest and Cell Death by Inhibiting the SREBP1-SCD1 Axis and Causing Imbalance in Lipid Desaturation. Int J Mol Sci 2023; 24:ijms24119229. [PMID: 37298183 DOI: 10.3390/ijms24119229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
α-Tocopherol-13'-carboxychromanol (α-T-13'-COOH) is an endogenously formed bioactive α-tocopherol metabolite that limits inflammation and has been proposed to exert lipid metabolism-regulatory, pro-apoptotic, and anti-tumoral properties at micromolar concentrations. The mechanisms underlying these cell stress-associated responses are, however, poorly understood. Here, we show that the induction of G0/G1 cell cycle arrest and apoptosis in macrophages triggered by α-T-13'-COOH is associated with the suppressed proteolytic activation of the lipid anabolic transcription factor sterol regulatory element-binding protein (SREBP)1 and with decreased cellular levels of stearoyl-CoA desaturase (SCD)1. In turn, the fatty acid composition of neutral lipids and phospholipids shifts from monounsaturated to saturated fatty acids, and the concentration of the stress-preventive, pro-survival lipokine 1,2-dioleoyl-sn-glycero-3-phospho-(1'-myo-inositol) [PI(18:1/18:1)] decreases. The selective inhibition of SCD1 mimics the pro-apoptotic and anti-proliferative activity of α-T-13'-COOH, and the provision of the SCD1 product oleic acid (C18:1) prevents α-T-13'-COOH-induced apoptosis. We conclude that micromolar concentrations of α-T-13'-COOH trigger cell death and likely also cell cycle arrest by suppressing the SREBP1-SCD1 axis and depleting cells of monounsaturated fatty acids and PI(18:1/18:1).
Collapse
Affiliation(s)
- Sijia Liao
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743 Jena, Germany
| | - André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Lisa Börmel
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743 Jena, Germany
| | - Charlotte Maier
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Luisa Gottschalk
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Maria Wallert
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743 Jena, Germany
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743 Jena, Germany
| |
Collapse
|
13
|
Yang N, Pang J, Huang Z, Zhang Q, Wang Z, Sun D. Enantioselective toxicity effect and mechanism of hexaconazole enantiomers to human breast cancer cells. Food Chem Toxicol 2023; 173:113612. [PMID: 36681264 DOI: 10.1016/j.fct.2023.113612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/03/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023]
Abstract
The toxicity effects of chiral pesticides on living organisms have attracted an increasing public attention. This study aims to investigate the toxicity effect and mechanism of hexaconazole (HEX) to human breast cancer cell (MCF-7) at enantiomer levels. HEX exposure obviously inhibited cells activities in a dose-dependent manner. Under the conditions of VIP >1 and p < 0.05, a total of 255 and 177 differential metabolites (DMs), 17 and 15 amino acid- and lipid-related metabolic pathways were disturbed after (+)-HEX and (-)-HEX exposure, respectively. HEX exposure may affect cell membrane function, signal transduction, and cell differentiation. We further investigated the mechanism of enantioselective differences by using molecular docking which showed that CYP17A1 was the main enzyme that leading to endocrine disrupting effects with the binding energy of -6.30 and -6.08 kcal/mol compared to CYP19A1 enzyme which were -5.81 and -5.93 kcal/mol for (+)-HEX and (-)-HEX, respectively. The docking results explained the reasons why (+)-HEX achieved higher cytotoxicity and induced more seriously metabolic profiles than its antipode. These findings could provide a new insight to understand the enantioselective cytotoxicity effect and mechanism of HEX and will be conducive to assessing its risk to human health at enantiomer levels.
Collapse
Affiliation(s)
- Na Yang
- School of Public Health/the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Junxiao Pang
- Key Laboratory of Critical Technology for Degradation of Pesticide Residues in Agro-products in Guizhou Ecological Environment, Food and Pharmaceutical Engineering Institute, Guiyang University, Guiyang, 550005, China
| | - Zhoubing Huang
- School of Public Health/the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Qinghai Zhang
- School of Public Health/the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Zelan Wang
- School of Public Health/the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Dali Sun
- School of Public Health/the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China.
| |
Collapse
|
14
|
Untargeted Lipidomics of Erythrocytes under Simulated Microgravity Conditions. Int J Mol Sci 2023; 24:ijms24054379. [PMID: 36901810 PMCID: PMC10002504 DOI: 10.3390/ijms24054379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Lipidomics and metabolomics are nowadays widely used to provide promising insights into the pathophysiology of cellular stress disorders. Our study expands, with the use of a hyphenated ion mobility mass spectrometric platform, the understanding of the cellular processes and stress due to microgravity. By lipid profiling of human erythrocytes, we annotated complex lipids such as oxidized phosphocholines, phosphocholines bearing arachidonic in their moiety, as well as sphingomyelins and hexosyl ceramides associated with microgravity conditions. Overall, our findings give an insight into the molecular alterations and identify erythrocyte lipidomics signatures associated with microgravity conditions. If the present results are confirmed in future studies, they may help to develop suitable treatments for astronauts after return to Earth.
Collapse
|
15
|
Chen H, Liao C, Yang X, Zhou H, Wu Y, Sun Q, Li S, Zhang W. Multi-omics analysis revealed the role of CYP1A2 in the induction of mechanical allodynia in type 1 diabetes. Front Genet 2023; 14:1151340. [PMID: 37035728 PMCID: PMC10076588 DOI: 10.3389/fgene.2023.1151340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Background: Mechanical allodynia (MA) is one of the leading clinical symptoms of painful diabetic peripheral neuropathy (PDPN), which is a primary reason for non-traumatic amputations, foot ulceration, and gait abnormalities in patients with diabetes. However, the pathogenic mechanisms of MA have not yet been fully elucidated, and there is no effective treatment. This study aims to study the potential pathogenetic mechanisms of MA and to provide targets for the therapy of MA. Methods: A single intraperitoneal injection of streptozotocin induced type 1 diabetes in rat models. Subsequently, rats were divided into the control group, the diabetic group without MA, and the diabetic group with MA based on weekly behavioral assays. The differentially expressed lipids in the sciatic nerve of each group were detected using untargeted lipidomics, and the differentially expressed genes in the sciatic nerve of each group were detected by transcriptomics. The pathogenesis of MA was predicted using integrated analysis and validated by immunofluorescence staining and transmission electron microscopy. Results: Untargeted lipidomics revealed the accumulation of a more severe lipid in MA rats. Transcriptomics results suggested that differentially expressed genes in MA rats were primarily related to lipid droplets and myelin sheath. Integrated analysis results indicated that the downregulation of Cytochrome P450 1A2 (CYP1A2) expression was closely linked to lipid metabolism disorders. Immunofluorescence staining demonstrated that down-regulation of CYP1A2 expression occurred in MA rats. Transmission electron microscopy results showed that more severe lipid droplet accumulation and myelin sheath degeneration occurred in MA rats. Conclusion: Our findings imply that the downregulation of CYP1A2 expression leads to disorders of lipid metabolism and further leads to lipid droplet accumulation and myelin sheath degeneration, which might ultimately lead to the development of MA. Therefore, our study contributes to promoting the understanding of the molecular mechanisms of MA and providing potential targets for the clinical treatment of MA.
Collapse
|
16
|
Zivko C, Witt F, Koeberle A, Fuhrmann G, Luciani P. Formulating elafibranor and obeticholic acid with phospholipids decreases drug-induced association of SPARC to extracellular vesicles from LX-2 human hepatic stellate cells. Eur J Pharm Biopharm 2023; 182:32-40. [PMID: 36470521 DOI: 10.1016/j.ejpb.2022.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/17/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
Chronic hepatic diseases often compromise liver function and are directly responsible for up to two million yearly deaths world-wide. There are yet no treatment options to solve this global medical need. Experimental drugs elafibranor (Ela) and obeticholic acid (OA) appeared promising in numerous earlier studies, but they recently struggled to show significant benefits in patients. Little is known on the drugs' impact on hepatic stellate cells (HSCs), key players in liver fibrogenesis. We recently reported a beneficial effect of polyenylphosphatidylcholines (PPCs)-rich formulations in reverting fibrogenic features of HSCs, including differences in their extracellular vesicles (EVs). Here, we newly formulated Ela and OA in PPC liposomes and evaluated their performance on the LX-2 (human HSC) cell line through our rigorous methods of EV-analysis, now expanded to include lipidomics. We show that direct treatments with Ela and OA increase EV-associated secreted protein acidic and cysteine rich (SPARC), a matricellular protein overexpressed in fibrogenesis. However, our results suggest that this potentially damaging drugs' action to HSCs could be mitigated when delivering them with lipid-based formulations, most notably with a PPC-rich phospholipid inducing specific changes in the cellular and EV phospholipid composition. Thus, EV analysis substantially deepens evaluations of drug performances and delivery strategies.
Collapse
Affiliation(s)
- Cristina Zivko
- Institute of Pharmacy, Friedrich Schiller University of Jena, Jena, Germany; Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Finja Witt
- Michael Popp Institute, University of Innsbruck, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute, University of Innsbruck, Innsbruck, Austria
| | - Gregor Fuhrmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Department of Pharmacy, Saarland University, Saarbrücken, Germany; Department of Biology, Friedrich-Alexander-University Erlangen, Erlangen, Germany.
| | - Paola Luciani
- Institute of Pharmacy, Friedrich Schiller University of Jena, Jena, Germany; Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
17
|
Lpcat3 deficiency promotes palmitic acid-induced 3T3-L1 mature adipocyte inflammation through enhanced ROS generation. Acta Biochim Biophys Sin (Shanghai) 2022; 55:117-130. [PMID: 36331295 PMCID: PMC10157521 DOI: 10.3724/abbs.2022161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphatidylcholines (PCs) are major phospholipids in the mammalian cell membrane. Structural remodeling of PCs is associated with many biological processes. Lysophosphatidylcholine acyltransferase 3 (Lpcat3), which catalyzes the incorporation of polyunsaturated fatty acyl chains into the sn-2 site of PCs, plays an important role in maintaining plasma membrane fluidity. Adipose tissue is one of the main distribution organs of Lpcat3, while the relationship between Lpcat3 and adipose tissue dysfunction during overexpansion remains unknown. In this study, we reveal that both polyunsaturated PC content and Lpcat3 expression are increased in abdominal adipose tissues of high-fat diet-fed mice when compared with chow-diet-fed mice, indicating that Lpcat3 is involved in adipose tissue overexpansion and dysfunction. Our experiments in 3T3-L1 adipocytes show that inhibition of Lpcat3 does not change triglyceride accumulation but increases palmitic acid-induced inflammation and lipolysis. Conversely, Lpcat3 overexpression exhibits anti-inflammatory and anti-lipolytic effects. Furthermore, mechanistic studies demonstrate that Lpcat3 deficiency promotes reactive oxygen species (ROS) generation by increasing NOX enzyme activity by facilitating the translocation of NOX4 to lipid rafts, thereby aggregating 3T3-L1 adipocyte inflammation induced by palmitic acid. Moreover, overexpression of Lpcat3 exhibits the opposite effects. These findings suggest that Lpcat3 protects adipocytes from inflammation during adipose tissue overexpansion by reducing ROS generation. In conclusion, our study demonstrates that Lpcat3 deficiency promotes palmitic acid-induced inflammation in 3T3-L1 adipocytes by enhancing ROS generation.
Collapse
|
18
|
PI(18:1/18:1) is a SCD1-derived lipokine that limits stress signaling. Nat Commun 2022; 13:2982. [PMID: 35624087 PMCID: PMC9142606 DOI: 10.1038/s41467-022-30374-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/27/2022] [Indexed: 02/07/2023] Open
Abstract
Cytotoxic stress activates stress-activated kinases, initiates adaptive mechanisms, including the unfolded protein response (UPR) and autophagy, and induces programmed cell death. Fatty acid unsaturation, controlled by stearoyl-CoA desaturase (SCD)1, prevents cytotoxic stress but the mechanisms are diffuse. Here, we show that 1,2-dioleoyl-sn-glycero-3-phospho-(1’-myo-inositol) [PI(18:1/18:1)] is a SCD1-derived signaling lipid, which inhibits p38 mitogen-activated protein kinase activation, counteracts UPR, endoplasmic reticulum-associated protein degradation, and apoptosis, regulates autophagy, and maintains cell morphology and proliferation. SCD1 expression and the cellular PI(18:1/18:1) proportion decrease during the onset of cell death, thereby repressing protein phosphatase 2 A and enhancing stress signaling. This counter-regulation applies to mechanistically diverse death-inducing conditions and is found in multiple human and mouse cell lines and tissues of Scd1-defective mice. PI(18:1/18:1) ratios reflect stress tolerance in tumorigenesis, chemoresistance, infection, high-fat diet, and immune aging. Together, PI(18:1/18:1) is a lipokine that links fatty acid unsaturation with stress responses, and its depletion evokes stress signaling. Fatty acid unsaturation by stearoyl-CoA desaturase 1 (SCD1) protects against cellular stress through unclear mechanisms. Here the authors show 1,2-dioleoyl-sn-glycero-3-phospho-(1’-myo-inositol) is an SCD1-derived signaling lipid that regulates stress-adaption, protects against cell death and promotes proliferation.
Collapse
|
19
|
Defries D, Curtis K, Petkau JC, Shariati-Ievari S, Blewett H, Aliani M. Patterns of Alpha-Linolenic Acid Incorporation into Phospholipids in H4IIE Cells. J Nutr Biochem 2022; 106:109014. [PMID: 35461904 DOI: 10.1016/j.jnutbio.2022.109014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/15/2021] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
Alpha linolenic acid (ALA) is an 18-carbon essential fatty acid found in plant-based foods and oils. While much attention has been placed on conversion of ALA to long chain polyunsaturated fatty acids, alternative routes of ALA metabolism exist and may lead to formation of other bioactive metabolites of ALA. The current study employed a non-targeted metabolomics approach to profile ALA metabolites that are significantly upregulated by ALA treatment. H4IIE hepatoma cells (n=3 samples per time point) were treated with 60 μM ALA or vehicle for 0, 0.25, 0.5, 1, 2, 3, 4, 6, 8, and 12 hours. Samples were then extracted with methanol and analyzed using high-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. We observed selective changes in ALA incorporation into phospholipid classes and subclasses over the 12 hours following ALA treatment. While levels of specific molecular species of ALA-containing phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, and lysophospholipids were elevated with ALA treatment, others were not affected. Of the phospholipids that were increased, some [e.g. PC(18:3/18:1), PC(18:3/18:4), PE(18:3/18:2), PE(18:3/18:3)] were elevated almost immediately after exposure to ALA, while others (e.g. PE(18:1/18:3) PA(18:3/22:6), and PA(18:3/18:2)] were not elevated until several hours after ALA treatment. Overall, these results suggest that ALA incorporation into phospholipids is selective and support a metabolic hierarchy for ALA incorporation into specific phospholipids. Given the functionality of phospholipids based on their fatty acid composition, future studies will need to investigate the implications of ALA incorporation into specific phospholipids on cell function.
Collapse
Affiliation(s)
- Danielle Defries
- Department of Kinesiology and Applied Health, University of Winnipeg, 3D09 Duckworth Building, 515 Portage Avenue, Winnipeg, Manitoba, Canada, R3B 2E9.
| | - Kayla Curtis
- Department of Food and Human Nutritional Sciences, University of Manitoba, Room 209 Human Ecology Building, 35 Chancellor's Circle, Winnipeg, Manitoba, Canada, R3T 2N2
| | - Jay C Petkau
- Canadian Centre for Agri-food Research in Health and Medicine (CCARM), St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Avenue, Winnipeg, Manitoba, Canada, R2H 2A6; Morden Research and Development Centre, Agriculture and Agri-food Canada, Route 100, Unit 100-101 Morden, Manitoba, Canada, R6M 1Y5
| | - Shiva Shariati-Ievari
- Department of Food and Human Nutritional Sciences, University of Manitoba, Room 209 Human Ecology Building, 35 Chancellor's Circle, Winnipeg, Manitoba, Canada, R3T 2N2; Canadian Centre for Agri-food Research in Health and Medicine (CCARM), St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Avenue, Winnipeg, Manitoba, Canada, R2H 2A6
| | - Heather Blewett
- Department of Food and Human Nutritional Sciences, University of Manitoba, Room 209 Human Ecology Building, 35 Chancellor's Circle, Winnipeg, Manitoba, Canada, R3T 2N2; Canadian Centre for Agri-food Research in Health and Medicine (CCARM), St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Avenue, Winnipeg, Manitoba, Canada, R2H 2A6; Morden Research and Development Centre, Agriculture and Agri-food Canada, Route 100, Unit 100-101 Morden, Manitoba, Canada, R6M 1Y5
| | - Michel Aliani
- Department of Food and Human Nutritional Sciences, University of Manitoba, Room 209 Human Ecology Building, 35 Chancellor's Circle, Winnipeg, Manitoba, Canada, R3T 2N2; Canadian Centre for Agri-food Research in Health and Medicine (CCARM), St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Avenue, Winnipeg, Manitoba, Canada, R2H 2A6; Division of Neurodegenerative Disorders (DND), St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Avenue, Winnipeg, Manitoba, Canada, R2H 2A6.
| |
Collapse
|
20
|
Rabel M, Warncke P, Thürmer M, Grüttner C, Bergemann C, Kurland HD, Müller FA, Koeberle A, Fischer D. The differences of the impact of a lipid and protein corona on the colloidal stability, toxicity, and degradation behavior of iron oxide nanoparticles. NANOSCALE 2021; 13:9415-9435. [PMID: 34002735 DOI: 10.1039/d0nr09053k] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
AIM In this study, the influence of a serum albumin (SA) and human plasma (HP) derived protein- and lipid molecule corona on the toxicity and biodegradability of different iron oxide nanoparticles (IONP) was investigated. METHODS IONP were synthesized and physicochemically characterized regarding size, charge, and colloidal stability. The adsorbed proteins were quantified and separated by gel electrophoresis. Adsorbed lipids were profiled by ultraperformance liquid chromatography-ESI-tandem mass spectrometry. The biocompatibility was investigated using isolated erythrocytes and a shell-less hen's egg model. The biodegradability was assessed by iron release studies in artificial body fluids. RESULTS The adsorption patterns of proteins and lipids varied depending on the surface characteristics of the IONP like charge and hydrophobicity. The biomolecule corona modified IONP displayed favorable colloidal stability and toxicological profile compared to IONP without biomolecule coronas, reducing erythrocyte aggregation and hemolysis in vitro as well as the corresponding effects ex ovo/in vivo. The coronas decreased the degradation speed of all tested IONP compared to bare particles, but, whereas all IONP degraded at the same rate for the SA corona, substantial differences were evident for IONP with HP-derived corona depending on the lipid adsorption profile. CONCLUSION In this study the impact of the proteins and lipids in the biomolecule corona on the entire IONP application cycle from the injection process to the degradation was demonstrated.
Collapse
Affiliation(s)
- Martin Rabel
- Pharmaceutical Technology and Biopharmacy, Friedrich-Schiller-University Jena, Lessingstraße 8, 07743 Jena, Germany
| | - Paul Warncke
- Pharmaceutical Technology and Biopharmacy, Friedrich-Schiller-University Jena, Lessingstraße 8, 07743 Jena, Germany
| | - Maria Thürmer
- Department of Pharmaceutical and Medical Chemistry, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Cordula Grüttner
- micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany
| | | | - Heinz-Dieter Kurland
- Otto Schott Institute of Materials Research (OSIM), Friedrich-Schiller-University Jena, Löbdergraben 32, 07743 Jena, Germany
| | - Frank A Müller
- Otto Schott Institute of Materials Research (OSIM), Friedrich-Schiller-University Jena, Löbdergraben 32, 07743 Jena, Germany
| | - Andreas Koeberle
- Department of Pharmaceutical and Medical Chemistry, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany and Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Mitterweg 24, 6020 Innsbruck, Austria
| | - Dagmar Fischer
- Department of Chemistry and Pharmacy, Friedrich-Alexander-University Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany.
| |
Collapse
|
21
|
Wolfgang MJ. Remodeling glycerophospholipids affects obesity-related insulin signaling in skeletal muscle. J Clin Invest 2021; 131:148176. [PMID: 33855969 DOI: 10.1172/jci148176] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
It has long been known that fatty acids can either adversely or positively affect insulin signaling in skeletal muscle, depending on chain length or saturation, and can therefore be primary drivers of systemic insulin sensitivity. However, the detailed mechanisms linking fatty acids to insulin signaling in skeletal muscle have been elusive. In this issue of the JCI, Ferrara et al. suggest a model whereby membrane lipid remodeling mediates skeletal muscle insulin sensitivity. The authors demonstrate that membrane glycerophospholipid fatty acid remodeling by lysophosphatidylcholine acyltransferase 3 (LPCAT3) in skeletal muscle from subjects with obesity was induced, suppressing insulin signaling and glucose tolerance. Loss or gain of LPCAT3 function in mouse models showed that Lpcat3 was both required and sufficient for high-fat diet-induced muscle insulin resistance. These results suggest that the physiochemical properties of muscle cell membranes may drive insulin sensitivity and, therefore, systemic glucose intolerance.
Collapse
Affiliation(s)
- Michael J Wolfgang
- Department of Biological Chemistry.,Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Ferrara PJ, Rong X, Maschek JA, Verkerke AR, Siripoksup P, Song H, Green TD, Krishnan KC, Johnson JM, Turk J, Houmard JA, Lusis AJ, Drummond MJ, McClung JM, Cox JE, Shaikh SR, Tontonoz P, Holland WL, Funai K. Lysophospholipid acylation modulates plasma membrane lipid organization and insulin sensitivity in skeletal muscle. J Clin Invest 2021; 131:135963. [PMID: 33591957 DOI: 10.1172/jci135963] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/11/2021] [Indexed: 01/09/2023] Open
Abstract
Aberrant lipid metabolism promotes the development of skeletal muscle insulin resistance, but the exact identity of lipid-mediated mechanisms relevant to human obesity remains unclear. A comprehensive lipidomic analysis of primary myocytes from individuals who were insulin-sensitive and lean (LN) or insulin-resistant with obesity (OB) revealed several species of lysophospholipids (lyso-PLs) that were differentially abundant. These changes coincided with greater expression of lysophosphatidylcholine acyltransferase 3 (LPCAT3), an enzyme involved in phospholipid transacylation (Lands cycle). Strikingly, mice with skeletal muscle-specific knockout of LPCAT3 (LPCAT3-MKO) exhibited greater muscle lysophosphatidylcholine/phosphatidylcholine, concomitant with improved skeletal muscle insulin sensitivity. Conversely, skeletal muscle-specific overexpression of LPCAT3 (LPCAT3-MKI) promoted glucose intolerance. The absence of LPCAT3 reduced phospholipid packing of cellular membranes and increased plasma membrane lipid clustering, suggesting that LPCAT3 affects insulin receptor phosphorylation by modulating plasma membrane lipid organization. In conclusion, obesity accelerates the skeletal muscle Lands cycle, whose consequence might induce the disruption of plasma membrane organization that suppresses muscle insulin action.
Collapse
Affiliation(s)
- Patrick J Ferrara
- Diabetes and Metabolism Research Center and.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes and Obesity Institute and.,Human Performance Laboratory, East Carolina University, Greenville, North Carolina, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Xin Rong
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, California, USA
| | - J Alan Maschek
- Diabetes and Metabolism Research Center and.,Metabolomics, Mass Spectrometry, and Proteomics Core and
| | - Anthony Rp Verkerke
- Diabetes and Metabolism Research Center and.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes and Obesity Institute and.,Human Performance Laboratory, East Carolina University, Greenville, North Carolina, USA
| | - Piyarat Siripoksup
- Diabetes and Metabolism Research Center and.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Haowei Song
- Division of Endocrinology Metabolism and Lipid Research, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | - Jordan M Johnson
- Diabetes and Metabolism Research Center and.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes and Obesity Institute and.,Human Performance Laboratory, East Carolina University, Greenville, North Carolina, USA
| | - John Turk
- Division of Endocrinology Metabolism and Lipid Research, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Joseph A Houmard
- East Carolina Diabetes and Obesity Institute and.,Human Performance Laboratory, East Carolina University, Greenville, North Carolina, USA
| | - Aldons J Lusis
- Cardiology Division, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Micah J Drummond
- Diabetes and Metabolism Research Center and.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | | | - James E Cox
- Diabetes and Metabolism Research Center and.,Metabolomics, Mass Spectrometry, and Proteomics Core and.,Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - Saame Raza Shaikh
- East Carolina Diabetes and Obesity Institute and.,Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, California, USA
| | - William L Holland
- Diabetes and Metabolism Research Center and.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center and.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes and Obesity Institute and.,Human Performance Laboratory, East Carolina University, Greenville, North Carolina, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
23
|
Deb S, Felix DA, Koch P, Deb MK, Szafranski K, Buder K, Sannai M, Groth M, Kirkpatrick J, Pietsch S, Gollowitzer A, Groß A, Riemenschneider P, Koeberle A, González‐Estévez C, Rudolph KL. Tnfaip2/exoc3-driven lipid metabolism is essential for stem cell differentiation and organ homeostasis. EMBO Rep 2021; 22:e49328. [PMID: 33300287 PMCID: PMC7788457 DOI: 10.15252/embr.201949328] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/21/2020] [Accepted: 10/29/2020] [Indexed: 01/07/2023] Open
Abstract
Lipid metabolism influences stem cell maintenance and differentiation but genetic factors that control these processes remain to be delineated. Here, we identify Tnfaip2 as an inhibitor of reprogramming of mouse fibroblasts into induced pluripotent stem cells. Tnfaip2 knockout impairs differentiation of embryonic stem cells (ESCs), and knockdown of the planarian para-ortholog, Smed-exoc3, abrogates in vivo tissue homeostasis and regeneration-processes that are driven by somatic stem cells. When stimulated to differentiate, Tnfaip2-deficient ESCs fail to induce synthesis of cellular triacylglycerol (TAG) and lipid droplets (LD) coinciding with reduced expression of vimentin (Vim)-a known inducer of LD formation. Smed-exoc3 depletion also causes a strong reduction of TAGs in planarians. The study shows that Tnfaip2 acts epistatically with and upstream of Vim in impairing cellular reprogramming. Supplementing palmitic acid (PA) and palmitoyl-L-carnitine (the mobilized form of PA) restores the differentiation capacity of Tnfaip2-deficient ESCs and organ maintenance in Smed-exoc3-depleted planarians. Together, these results identify a novel role of Tnfaip2 and exoc3 in controlling lipid metabolism, which is essential for ESC differentiation and planarian organ maintenance.
Collapse
Affiliation(s)
- Sarmistha Deb
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Daniel A Felix
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Philipp Koch
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | | | - Karol Szafranski
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Katrin Buder
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Mara Sannai
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Marco Groth
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | | | - Stefan Pietsch
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - André Gollowitzer
- Institute of PharmacyFriedrich‐Schiller‐UniversityJenaGermany
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Alexander Groß
- Institute of Medical Systems BiologyUlm UniversityUlmGermany
| | | | - Andreas Koeberle
- Institute of PharmacyFriedrich‐Schiller‐UniversityJenaGermany
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | | | - Karl Lenhard Rudolph
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
- University Hospital JenaFriedrich Schiller UniversityJenaGermany
| |
Collapse
|
24
|
Drescher S, van Hoogevest P. The Phospholipid Research Center: Current Research in Phospholipids and Their Use in Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12121235. [PMID: 33353254 PMCID: PMC7766331 DOI: 10.3390/pharmaceutics12121235] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the research on phospholipids and their use for drug delivery related to the Phospholipid Research Center Heidelberg (PRC). The focus is on projects that have been approved by the PRC since 2017 and are currently still ongoing or have recently been completed. The different projects cover all facets of phospholipid research, from basic to applied research, including the use of phospholipids in different administration forms such as liposomes, mixed micelles, emulsions, and extrudates, up to industrial application-oriented research. These projects also include all routes of administration, namely parenteral, oral, and topical. With this review we would like to highlight possible future research directions, including a short introduction into the world of phospholipids.
Collapse
|
25
|
Espada L, Dakhovnik A, Chaudhari P, Martirosyan A, Miek L, Poliezhaieva T, Schaub Y, Nair A, Döring N, Rahnis N, Werz O, Koeberle A, Kirkpatrick J, Ori A, Ermolaeva MA. Loss of metabolic plasticity underlies metformin toxicity in aged Caenorhabditis elegans. Nat Metab 2020; 2:1316-1331. [PMID: 33139960 DOI: 10.1038/s42255-020-00307-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022]
Abstract
Current clinical trials are testing the life-extending benefits of the diabetes drug metformin in healthy individuals without diabetes. However, the metabolic response of a non-diabetic cohort to metformin treatment has not been studied. Here, we show in C. elegans and human primary cells that metformin shortens lifespan when provided in late life, contrary to its positive effects in young organisms. We find that metformin exacerbates ageing-associated mitochondrial dysfunction, causing respiratory failure. Age-related failure to induce glycolysis and activate the dietary-restriction-like mobilization of lipid reserves in response to metformin result in lethal ATP exhaustion in metformin-treated aged worms and late-passage human cells, which can be rescued by ectopic stabilization of cellular ATP content. Metformin toxicity is alleviated in worms harbouring disruptions in insulin-receptor signalling, which show enhanced resilience to mitochondrial distortions at old age. Together, our data show that metformin induces deleterious changes of conserved metabolic pathways in late life, which could bring into question its benefits for older individuals without diabetes.
Collapse
Affiliation(s)
- Lilia Espada
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | | | - Prerana Chaudhari
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Asya Martirosyan
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Laura Miek
- Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | | | - Yvonne Schaub
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Ashish Nair
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Nadia Döring
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Norman Rahnis
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Oliver Werz
- Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Andreas Koeberle
- Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
- Michael Popp Research Institute, University of Innsbruck, Innsbruck, Austria
| | | | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Maria A Ermolaeva
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
| |
Collapse
|
26
|
Zaharia OP, Strassburger K, Knebel B, Kupriyanova Y, Karusheva Y, Wolkersdorfer M, Bódis K, Markgraf DF, Burkart V, Hwang JH, Kotzka J, Al-Hasani H, Szendroedi J, Roden M. Role of Patatin-Like Phospholipase Domain-Containing 3 Gene for Hepatic Lipid Content and Insulin Resistance in Diabetes. Diabetes Care 2020; 43:2161-2168. [PMID: 32910776 DOI: 10.2337/dc20-0329] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/14/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The rs738409(G) single nucleotide polymorphism (SNP) in the patatin-like phospholipase domain-containing 3 (PNPLA3) gene associates with increased risk and progression of nonalcoholic fatty liver disease (NAFLD). As the recently described severe insulin-resistant diabetes (SIRD) cluster specifically relates to NAFLD, this study examined whether this SNP differently associates with hepatic lipid content (hepatocellular lipids [HCL]) and insulin sensitivity in recent-onset diabetes. RESEARCH DESIGN AND METHODS A total of 917 participants in the German Diabetes Study (GDS) underwent genotyping, hyperinsulinemic-euglycemic clamps with stable isotopic tracer dilution, and MRS. RESULTS The G allele associated positively with HCL (β = 0.36, P < 0.01), independent of age, sex, and BMI across the whole cohort, but not in the individual clusters. Those with SIRD exhibited lowest whole-body insulin sensitivity compared with those with severe insulin-deficient (SIDD), moderate obesity-related (MOD), moderate age-related (MARD), and severe autoimmune diabetes (SAID) clusters (all P < 0.001). Interestingly, the SIRD group presented with higher prevalence of the rs738409(G) SNP compared with other clusters and the glucose-tolerant control group (P < 0.05). HCL was higher in the SIRD group (median 13.6% [1st quartile 5.8; 3rd quartile 19.1] compared with the MOD (6.4 % [2.1; 12.4], P < 0.05), MARD (3.0% [1.0; 7.9], P < 0.001), SAID (0.4% [0.0; 1.5], P < 0.001), and glucose-tolerant (0.9% [0.4; 4.9), P < 0.001) group. Although the PNPLA3 polymorphism did not directly associate with whole-body insulin sensitivity in SIRD, the G-allele carriers had higher circulating free fatty acid concentrations and greater adipose tissue insulin resistance compared with noncarriers (both P < 0.001). CONCLUSIONS Members of the SIRD cluster are more frequently carriers of the rs738409(G) variant. The SNP-associated adipose tissue insulin resistance and excessive lipolysis may contribute to their NAFLD.
Collapse
Affiliation(s)
- Oana P Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany.,Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Birgit Knebel
- German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Yanislava Karusheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | | | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel F Markgraf
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Jong-Hee Hwang
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Jörg Kotzka
- German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hadi Al-Hasani
- German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | | |
Collapse
|
27
|
Ishibashi K, Takeda Y, Nakata L, Hakuno F, Takahashi SI, Atsumi GI. Elaidate, a trans fatty acid, suppresses insulin signaling for glucose uptake in a manner distinct from that of stearate. Biochimie 2020; 177:98-107. [PMID: 32822725 DOI: 10.1016/j.biochi.2020.07.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 07/15/2020] [Accepted: 07/30/2020] [Indexed: 12/19/2022]
Abstract
The dietary intake of elaidate (elaidic acid), a trans-fatty acid, is associated with the development of various diseases. Since elaidate is a C18 unsaturated fatty acid with a steric structure similar to that of a C18 saturated fatty acid (stearate), we previously revealed that insulin-dependent glucose uptake was impaired in adipocytes exposed to elaidate prior to and during differentiation similar to stearate. However, it is still unknown whether the mechanism of impairment of insulin-dependent glucose uptake due to elaidate is similar to that of stearate. Here, we indicate that persistent exposure to elaidate has particular effects on insulin signaling and GLUT4 dynamics. Insulin-induced accumulation of Akt at the plasma membrane (PM) and elevations of phosphorylated Akt and AS160 levels in whole cells were suppressed in adipocytes persistently exposed to 50 μM elaidate. Interestingly, persistent exposure to the same concentration of stearate has no effect on the phosphorylated Akt and AS160 levels. When cells were exposed to these fatty acids, elaidate suppressed insulin-induced fusion, but not translocation, of GLUT4 storage vesicles in the PM, whereas stearate did not suppress the fusion and translocation of GLUT4 storage, indicating that elaidate has suppressive effects on the accumulation of Akt and fusion of GLUT4 storage vesicles and that both elaidate and stearate vary in the mechanisms by which they impair insulin-dependent glucose uptake.
Collapse
Affiliation(s)
- Kenichi Ishibashi
- Department of Molecular Physiology and Pathology, Faculty of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Yoshihiro Takeda
- Department of Molecular Physiology and Pathology, Faculty of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Lisa Nakata
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Fumihiko Hakuno
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Shin-Ichiro Takahashi
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Gen-Ichi Atsumi
- Department of Molecular Physiology and Pathology, Faculty of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo, 173-8605, Japan.
| |
Collapse
|
28
|
Meßner M, Schmitt S, Ardelt MA, Fröhlich T, Müller M, Pein H, Huber-Cantonati P, Ortler C, Koenig LM, Zobel L, Koeberle A, Arnold GJ, Rothenfußer S, Kiemer AK, Gerbes AL, Zischka H, Vollmar AM, Pachmayr J. Metabolic implication of tigecycline as an efficacious second-line treatment for sorafenib-resistant hepatocellular carcinoma. FASEB J 2020; 34:11860-11882. [PMID: 32652772 DOI: 10.1096/fj.202001128r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
Sorafenib represents the current standard of care for patients with advanced-stage hepatocellular carcinoma (HCC). However, acquired drug resistance occurs frequently during therapy and is accompanied by rapid tumor regrowth after sorafenib therapy termination. To identify the mechanism of this therapy-limiting growth resumption, we established robust sorafenib resistance HCC cell models that exhibited mitochondrial dysfunction and chemotherapeutic crossresistance. We found a rapid relapse of tumor cell proliferation after sorafenib withdrawal, which was caused by renewal of mitochondrial structures alongside a metabolic switch toward high electron transport system (ETS) activity. The translation-inhibiting antibiotic tigecycline impaired the biogenesis of mitochondrial DNA-encoded ETS subunits and limited the electron acceptor turnover required for glutamine oxidation. Thereby, tigecycline prevented the tumor relapse in vitro and in murine xenografts in vivo. These results offer a promising second-line therapeutic approach for advanced-stage HCC patients with progressive disease undergoing sorafenib therapy or treatment interruption due to severe adverse events.
Collapse
Affiliation(s)
- Martina Meßner
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.,Institute of Pharmacy, Center for Public Health, Paracelsus Medical University, Salzburg, Austria
| | - Sabine Schmitt
- School of Medicine, Institute of Toxicology and Environmental Hygiene, Technical University Munich, Munich, Germany
| | - Maximilian A Ardelt
- Institute of Pharmacy, Center for Public Health, Paracelsus Medical University, Salzburg, Austria
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Centre, LMU Munich, Munich, Germany
| | - Martin Müller
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Helmut Pein
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Petra Huber-Cantonati
- Institute of Pharmacy, Center for Public Health, Paracelsus Medical University, Salzburg, Austria
| | - Carina Ortler
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Lars M Koenig
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
| | - Lena Zobel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Andreas Koeberle
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany.,Michael Popp Research Institute, University of Innsbruck, Innsbruck, Austria
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Centre, LMU Munich, Munich, Germany
| | - Simon Rothenfußer
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Alexander L Gerbes
- Department of Medicine 2, Liver Center Munich, University Hospital, LMU Munich, Munich, Germany
| | - Hans Zischka
- School of Medicine, Institute of Toxicology and Environmental Hygiene, Technical University Munich, Munich, Germany.,Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Johanna Pachmayr
- Institute of Pharmacy, Center for Public Health, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
29
|
Phospholipid profiling enables to discriminate tumor- and non-tumor-derived human colon epithelial cells: Phospholipidome similarities and differences in colon cancer cell lines and in patient-derived cell samples. PLoS One 2020; 15:e0228010. [PMID: 31999740 PMCID: PMC6992008 DOI: 10.1371/journal.pone.0228010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 01/05/2020] [Indexed: 01/10/2023] Open
Abstract
Identification of changes of phospholipid (PL) composition occurring during colorectal cancer (CRC) development may help us to better understand their roles in CRC cells. Here, we used LC-MS/MS-based PL profiling of cell lines derived from normal colon mucosa, or isolated at distinct stages of CRC development, in order to study alterations of PL species potentially linked with cell transformation. We found that a detailed evaluation of phosphatidylinositol (PI) and phosphatidylserine (PS) classes allowed us to cluster the studied epithelial cell lines according to their origin: i) cells originally derived from normal colon tissue (NCM460, FHC); ii) cell lines derived from colon adenoma or less advanced differentiating adenocarcinoma cells (AA/C1, HT-29); or, iii) cells obtained by in vitro transformation of adenoma cells and advanced colon adenocarcinoma cells (HCT-116, AA/C1/SB10, SW480, SW620). Although we tentatively identified several PS and PI species contributing to cell line clustering, full PI and PS profiles appeared to be a key to the successful cell line discrimination. In parallel, we compared PL composition of primary epithelial (EpCAM-positive) cells, isolated from tumor and adjacent non-tumor tissues of colon cancer patients, with PL profiles of cell lines derived from normal colon mucosa (NCM460) and from colon adenocarcinoma (HCT-116, SW480) cells, respectively. In general, higher total levels of all PL classes were observed in tumor cells. The overall PL profiles of the cell lines, when compared with the respective patient-derived cells, exhibited similarities. Nevertheless, there were also some notable differences in levels of individual PL species. This indicated that epithelial cell lines, derived either from normal colon tissue or from CRC cells, could be employed as models for functional lipidomic analyses of colon cells, albeit with some caution. The biological significance of the observed PL deregulation, or their potential links with specific CRC stages, deserve further investigation.
Collapse
|
30
|
Wang R, Li B, Lam SM, Shui G. Integration of lipidomics and metabolomics for in-depth understanding of cellular mechanism and disease progression. J Genet Genomics 2019; 47:69-83. [PMID: 32178981 DOI: 10.1016/j.jgg.2019.11.009] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022]
Abstract
Mass spectrometry (MS)-based omics technologies are now widely used to profile small molecules in multiple matrices to confer comprehensive snapshots of cellular metabolic phenotypes. The metabolomes of cells, tissues, and organisms comprise a variety of molecules including lipids, amino acids, sugars, organic acids, and so on. Metabolomics mainly focus on the hydrophilic classes, while lipidomics has emerged as an independent omics owing to the complexities of the organismal lipidomes. The potential roles of lipids and small metabolites in disease pathogenesis have been widely investigated in various human diseases, but system-level understanding is largely lacking, which could be partly attributed to the insufficiency in terms of metabolite coverage and quantitation accuracy in current analytical technologies. While scientists are continuously striving to develop high-coverage omics approaches, integration of metabolomics and lipidomics is becoming an emerging approach to mechanistic investigation. Integration of metabolome and lipidome offers a complete atlas of the metabolic landscape, enabling comprehensive network analysis to identify critical metabolic drivers in disease pathology, facilitating the study of interconnection between lipids and other metabolites in disease progression. In this review, we summarize omics-based findings on the roles of lipids and metabolites in the pathogenesis of selected major diseases threatening public health. We also discuss the advantages of integrating lipidomics and metabolomics for in-depth understanding of molecular mechanism in disease pathogenesis.
Collapse
Affiliation(s)
- Raoxu Wang
- 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, 100101, China
| | - Bowen Li
- Lipidall Technologies Company Limited, Changzhou, 213000, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; Lipidall Technologies Company Limited, Changzhou, 213000, 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, 100101, China.
| |
Collapse
|
31
|
Zhao X, Zhang W, Zhang D, Liu X, Cao W, Chen Q, Ouyang Z, Xia Y. A lipidomic workflow capable of resolving sn- and C[double bond, length as m-dash]C location isomers of phosphatidylcholines. Chem Sci 2019; 10:10740-10748. [PMID: 32153749 PMCID: PMC7020929 DOI: 10.1039/c9sc03521d] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/04/2019] [Indexed: 01/01/2023] Open
Abstract
As a major class of mammalian lipids, phosphatidylcholines (PCs) often contain mixtures of structural isomers, resulting from different lipogenesis pathways. Profiling PCs at the isomer level, however, remains challenging in lipidomic settings, especially for characterizing the positions of fatty acyls on the glycerol backbone (sn-positions) and the locations of carbon-carbon double bonds (C[double bond, length as m-dash]Cs) in unsaturated acyl chains. In this work, we have developed a workflow for profiling PCs down to sn- and C[double bond, length as m-dash]C locations at high coverage and sensitivity. This capability is enabled by radical-directed fragmentation, forming sn-1 specific fragment ions upon collision-induced dissociation (CID) of bicarbonate anion adducts of PCs ([M + HCO3]-) inside a mass spectrometer. This new tandem mass spectrometry (MS/MS) method can be simply incorporated into liquid chromatography by employing ammonium bicarbonate in the mobile phase without any instrument modification needed. It is also compatible with the online Paternò-Büchì reaction and subsequent MS/MS for the assignment of C[double bond, length as m-dash]C locations in sn-1 fatty acyl chains of unsaturated PCs. The analytical performance of the workflow is manifested by identification of 82 distinct PC molecular species from the polar extract of bovine liver, including quantification of 19 pairs of sn-isomers. Finally, we demonstrate that five pairs of PC sn-isomers show significant compositional changes in tissue samples of human breast cancer relative to controls, suggesting a potential for monitoring PC sn-isomers for biomedical applications.
Collapse
Affiliation(s)
- Xue Zhao
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biological , Department of Chemistry , Tsinghua University , Beijing 100084 , China .
| | - Wenpeng Zhang
- Department of Chemistry , Purdue University , West Lafayette , IN 47907 , USA
| | - Donghui Zhang
- State Key Laboratory of Precision Measurement Technology and Instruments , Department of Precision Instrument , Tsinghua University , Beijing , 100084 , China
| | - Xinwei Liu
- State Key Laboratory of Precision Measurement Technology and Instruments , Department of Precision Instrument , Tsinghua University , Beijing , 100084 , China
| | - Wenbo Cao
- State Key Laboratory of Precision Measurement Technology and Instruments , Department of Precision Instrument , Tsinghua University , Beijing , 100084 , China
| | - Qinhua Chen
- Affiliated Dongfeng Hospital , Hubei University of Medicine , Shiyan , Hubei Province 442000 , China
| | - Zheng Ouyang
- State Key Laboratory of Precision Measurement Technology and Instruments , Department of Precision Instrument , Tsinghua University , Beijing , 100084 , China
| | - Yu Xia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biological , Department of Chemistry , Tsinghua University , Beijing 100084 , China .
| |
Collapse
|
32
|
Njeru SN, Kraus J, Meena JK, Lechel A, Katz SF, Kumar M, Knippschild U, Azoitei A, Wezel F, Bolenz C, Leithäuser F, Gollowitzer A, Omrani O, Hoischen C, Koeberle A, Kestler HA, Günes C, Rudolph KL. Aneuploidy-inducing gene knockdowns overlap with cancer mutations and identify Orp3 as a B-cell lymphoma suppressor. Oncogene 2019; 39:1445-1465. [PMID: 31659255 DOI: 10.1038/s41388-019-1073-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/25/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022]
Abstract
Aneuploidy can instigate tumorigenesis. However, mutations in genes that control chromosome segregation are rare in human tumors as these mutations reduce cell fitness. Screening experiments indicate that the knockdown of multiple classes of genes that are not directly involved in chromosome segregation can lead to aneuploidy induction. The possible contribution of these genes to cancer formation remains yet to be defined. Here we identified gene knockdowns that lead to an increase in aneuploidy in checkpoint-deficient human cancer cells. Computational analysis revealed that the identified genes overlap with recurrent mutations in human cancers. The knockdown of the three strongest selected candidate genes (ORP3, GJB3, and RXFP1) enhances the malignant transformation of human fibroblasts in culture. Furthermore, the knockout of Orp3 results in an aberrant expansion of lymphoid progenitor cells and a high penetrance formation of chromosomal instable, pauci-clonal B-cell lymphoma in aging mice. At pre-tumorous stages, lymphoid cells from the animals exhibit deregulated phospholipid metabolism and an aberrant induction of proliferation regulating pathways associating with increased aneuploidy in hematopoietic progenitor cells. Together, these results support the concept that aneuploidy-inducing gene deficiencies contribute to cellular transformation and carcinogenesis involving the deregulation of various molecular processes such as lipid metabolism, proliferation, and cell survival.
Collapse
Affiliation(s)
- Sospeter N Njeru
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany.,Paul-Ehrlich-Institute, Division Immunology, 63225, Langen, Germany
| | - Johann Kraus
- Institute of Medical Systems Biology, Ulm University, 89081, Ulm, Germany
| | - Jitendra K Meena
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany.,Baylor College of Medicine, Houston, TX, USA
| | - André Lechel
- Department of Internal Medicine I, Ulm University Hospital, 89081, Ulm, Germany
| | - Sarah-Fee Katz
- Department of Internal Medicine I, Ulm University Hospital, 89081, Ulm, Germany
| | - Mukesh Kumar
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, 89081, Ulm, Germany
| | - Anca Azoitei
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany
| | - Felix Wezel
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany
| | - Christian Bolenz
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany
| | | | - André Gollowitzer
- Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Omid Omrani
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany
| | - Christian Hoischen
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany
| | - Andreas Koeberle
- Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany.,Michael Popp Research Institute, University of Innsbruck, Innsbruck, Austria
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, 89081, Ulm, Germany.
| | - Cagatay Günes
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany.
| | - K Lenhard Rudolph
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany.
| |
Collapse
|
33
|
Knebel B, Fahlbusch P, Dille M, Wahlers N, Hartwig S, Jacob S, Kettel U, Schiller M, Herebian D, Koellmer C, Lehr S, Müller-Wieland D, Kotzka J. Fatty Liver Due to Increased de novo Lipogenesis: Alterations in the Hepatic Peroxisomal Proteome. Front Cell Dev Biol 2019; 7:248. [PMID: 31709254 PMCID: PMC6823594 DOI: 10.3389/fcell.2019.00248] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/08/2019] [Indexed: 12/15/2022] Open
Abstract
In non-alcoholic fatty liver disease (NAFLD) caused by ectopic lipid accumulation, lipotoxicity is a crucial molecular risk factor. Mechanisms to eliminate lipid overflow can prevent the liver from functional complications. This may involve increased secretion of lipids or metabolic adaptation to ß-oxidation in lipid-degrading organelles such as mitochondria and peroxisomes. In addition to dietary factors, increased plasma fatty acid levels may be due to increased triglyceride synthesis, lipolysis, as well as de novo lipid synthesis (DNL) in the liver. In the present study, we investigated the impact of fatty liver caused by elevated DNL, in a transgenic mouse model with liver-specific overexpression of human sterol regulatory element-binding protein-1c (alb-SREBP-1c), on hepatic gene expression, on plasma lipids and especially on the proteome of peroxisomes by omics analyses, and we interpreted the results with knowledge-based analyses. In summary, the increased hepatic DNL is accompanied by marginal gene expression changes but massive changes in peroxisomal proteome. Furthermore, plasma phosphatidylcholine (PC) as well as lysoPC species were altered. Based on these observations, it can be speculated that the plasticity of organelles and their functionality may be directly affected by lipid overflow.
Collapse
Affiliation(s)
- Birgit Knebel
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Pia Fahlbusch
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Matthias Dille
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Natalie Wahlers
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Sonja Hartwig
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Sylvia Jacob
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Ulrike Kettel
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Martina Schiller
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Children’s Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Cornelia Koellmer
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Stefan Lehr
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| | - Dirk Müller-Wieland
- Department of Internal Medicine I, Clinical Research Centre, University Hospital Aachen, Aachen, Germany
| | - Jorg Kotzka
- Leibniz Center for Diabetes Research, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, Düsseldorf, Germany
| |
Collapse
|
34
|
van der Veen JN, Lingrell S, McCloskey N, LeBlond ND, Galleguillos D, Zhao YY, Curtis JM, Sipione S, Fullerton MD, Vance DE, Jacobs RL. A role for phosphatidylcholine and phosphatidylethanolamine in hepatic insulin signaling. FASEB J 2019; 33:5045-5057. [PMID: 30615497 DOI: 10.1096/fj.201802117r] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Phosphatidylethanolamine N-methyltransferase (PEMT) is an important enzyme in hepatic phosphatidylcholine (PC) biosynthesis. Pemt-/- mice fed a high-fat diet are protected from obesity and whole-body insulin resistance. However, Pemt-/- mice develop severe nonalcoholic steatohepatitis (NASH). Because NASH is often associated with hepatic insulin resistance, we investigated whether the increased insulin sensitivity in Pemt-/- mice was restricted to nonhepatic tissues or whether the liver was also insulin sensitive. Strikingly, the livers of Pemt-/- mice compared with those of Pemt+/+ mice were not insulin resistant, despite elevated levels of hepatic triacylglycerols and diacylglycerols, as well as increased hepatic inflammation and fibrosis. Endogenous glucose production was lower in Pemt-/- mice under both basal and hyperinsulinemic conditions. Experiments in primary hepatocytes and hepatoma cells revealed improved insulin signaling in the absence of PEMT, which was not due to changes in diacylglycerols, ceramides, or gangliosides. On the other hand, the phospholipid composition in hepatocytes seems critically important for insulin signaling such that lowering the PC:phosphatidylethanolamine (PE) ratio improves insulin signaling. Thus, treatments to reduce the PC:PE ratio in liver may protect against the development of hepatic insulin resistance.-Van der Veen, J. N., Lingrell, S., McCloskey, N., LeBlond, N. D., Galleguillos, D., Zhao, Y. Y., Curtis, J. M., Sipione, S., Fullerton, M. D., Vance, D. E., Jacobs, R. L. A role for phosphatidylcholine and phosphatidylethanolamine in hepatic insulin signaling.
Collapse
Affiliation(s)
- Jelske N van der Veen
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada.,Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Susanne Lingrell
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada.,Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nicholas McCloskey
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Nicholas D LeBlond
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Danny Galleguillos
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada.,Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada; and
| | - Yuan Y Zhao
- Department of Agricultural, Food, and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Jonathan M Curtis
- Department of Agricultural, Food, and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Simonetta Sipione
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada.,Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada; and
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Dennis E Vance
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada.,Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - René L Jacobs
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada.,Department of Agricultural, Food, and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
35
|
Mariniello K, Min Y, Ghebremeskel K. Phosphorylation of protein kinase B, the key enzyme in insulin-signaling cascade, is enhanced in linoleic and arachidonic acid–treated HT29 and HepG2 cells. Nutrition 2019; 57:52-58. [DOI: 10.1016/j.nut.2018.05.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 11/30/2022]
|
36
|
Chu KO, Chan KP, Chan SO, Ng TK, Jhanji V, Wang CC, Pang CP. Metabolomics of Green-Tea Catechins on Vascular-Endothelial-Growth-Factor-Stimulated Human-Endothelial-Cell Survival. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:12866-12875. [PMID: 30406651 DOI: 10.1021/acs.jafc.8b05998] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Neovascularization causes serious oculopathy related to upregulation of vascular-endothelial-growth factor (VEGF) causing new capillary growth via endothelial cells. Green-tea-extract (GTE) constituents possess antiangiogenesis properties. We used VEGF to induce human umbilical-vein endothelial cells (HUVECs) and applied GTE, epigallocatechin gallate (EGCG), and mixtures of different compositions of purified catechins (M1 and M2) to evaluate their efficacies of inhibition and their underlying mechanisms using cell-cycle analysis and untargeted metabolomics techniques. GTE, EGCG, M1, and M2 induced HUVEC apoptosis by 22.1 ± 2, 20.0 ± 0.7, 50.7 ± 8.5, and 69.8 ± 4.1%, respectively. GTE exerted a broad, balanced metabolomics spectrum, involving suppression of the biosynthesis of cellular building blocks and oxidative-phosphorylation metabolites as well as promotion of the biosynthesis of membrane lipids and growth factors. M2 mainly induced mechanisms associated with energy and biosynthesis suppression. Therefore, GTE exerted mechanisms involving both promotion and suppression activities, whereas purified catechins induced extensive apoptosis. GTE could be a more promising antineovascularization remedy for ocular treatment.
Collapse
Affiliation(s)
- Kai On Chu
- Department of Ophthalmology and Visual Sciences , The Chinese University of Hong Kong, Hong Kong Eye Hospital , Kowloon , Hong Kong
- School of Biomedical Sciences , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong
| | - Kwok Ping Chan
- Department of Ophthalmology and Visual Sciences , The Chinese University of Hong Kong, Hong Kong Eye Hospital , Kowloon , Hong Kong
| | - Sun On Chan
- School of Biomedical Sciences , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong
| | - Tsz Kin Ng
- Department of Ophthalmology and Visual Sciences , The Chinese University of Hong Kong, Hong Kong Eye Hospital , Kowloon , Hong Kong
| | - Vishal Jhanji
- Department of Ophthalmology and Visual Sciences , The Chinese University of Hong Kong, Hong Kong Eye Hospital , Kowloon , Hong Kong
| | - Chi Chiu Wang
- School of Biomedical Sciences , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong
- Department of Obstetrics and Gynaecology , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong
- Li Ka Shing Institute of Health Science , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences , The Chinese University of Hong Kong, Hong Kong Eye Hospital , Kowloon , Hong Kong
| |
Collapse
|
37
|
Abstract
Phospholipids are major constituents of biological membranes. The fatty acyl chain composition of phospholipids determines the biophysical properties of membranes and thereby affects their impact on biological processes. The composition of fatty acyl chains is also actively regulated through a deacylation and reacylation pathway called Lands' cycle. Recent studies of mouse genetic models have demonstrated that lysophosphatidylcholine acyltransferases (LPCATs), which catalyze the incorporation of fatty acyl chains into the sn-2 site of phosphatidylcholine, play important roles in pathophysiology. Two LPCAT family members, LPCAT1 and LPCAT3, have been particularly well studied. LPCAT1 is crucial for proper lung function due to its role in pulmonary surfactant biosynthesis. LPCAT3 maintains systemic lipid homeostasis by regulating lipid absorption in intestine, lipoprotein secretion, and de novo lipogenesis in liver. Mounting evidence also suggests that changes in LPCAT activity may be potentially involved in pathological conditions, including nonalcoholic fatty liver disease, atherosclerosis, viral infections, and cancer. Pharmacological manipulation of LPCAT activity and membrane phospholipid composition may provide new therapeutic options for these conditions.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90272, USA;
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90272, USA;
| |
Collapse
|
38
|
Abstract
Liver X receptors α and β (LXRα and LXRβ) are nuclear receptors with pivotal roles in the transcriptional control of lipid metabolism. Transcriptional activity of LXRs is induced in response to elevated cellular levels of cholesterol. LXRs bind to and regulate the expression of genes that encode proteins involved in cholesterol absorption, transport, efflux, excretion and conversion to bile acids. The coordinated, tissue-specific actions of the LXR pathway maintain systemic cholesterol homeostasis and regulate immune and inflammatory responses. LXRs also regulate fatty acid metabolism by controlling the lipogenic transcription factor sterol regulatory element-binding protein 1c and regulate genes that encode proteins involved in fatty acid elongation and desaturation. LXRs exert important effects on the metabolism of phospholipids, which, along with cholesterol, are major constituents of cellular membranes. LXR activation preferentially drives the incorporation of polyunsaturated fatty acids into phospholipids by inducing transcription of the remodelling enzyme lysophosphatidylcholine acyltransferase 3. The ability of the LXR pathway to couple cellular sterol levels with the saturation of fatty acids in membrane phospholipids has implications for several physiological processes, including lipoprotein production, dietary lipid absorption and intestinal stem cell proliferation. Understanding how LXRs regulate membrane composition and function might provide new therapeutic insight into diseases associated with dysregulated lipid metabolism, including atherosclerosis, diabetes mellitus and cancer.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
39
|
Jenkins CM, Yang K, Liu G, Moon SH, Dilthey BG, Gross RW. Cytochrome c is an oxidative stress-activated plasmalogenase that cleaves plasmenylcholine and plasmenylethanolamine at the sn-1 vinyl ether linkage. J Biol Chem 2018. [PMID: 29530984 DOI: 10.1074/jbc.ra117.001629] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Plasmalogens are phospholipids critical for cell function and signaling that contain a vinyl ether linkage at the sn-1 position and are highly enriched in arachidonic acid (AA) at the sn-2 position. However, the enzyme(s) responsible for the cleavage of the vinyl ether linkage in plasmalogens has remained elusive. Herein, we report that cytochrome c, in the presence of either cardiolipin (CL), O2 and H2O2, or oxidized CL and O2, catalyzes the oxidation of the plasmalogen vinyl ether linkage, promoting its hydrolytic cleavage and resultant production of 2-AA-lysolipids and highly reactive α-hydroxy fatty aldehydes. Using stable isotope labeling in synergy with strategic chemical derivatizations and high-mass-accuracy MS, we deduced the chemical mechanism underlying this long sought-after reaction. Specifically, labeling with either 18O2 or H218O, but not with H218O2, resulted in M + 2 isotopologues of the α-hydroxyaldehyde, whereas reactions with both 18O2 and H218O identified the M + 4 isotopologue. Furthermore, incorporation of 18O from 18O2 was predominantly located at the α-carbon. In contrast, reactions with H218O yielded 18O linked to the aldehyde carbon. Importantly, no significant labeling of 2-AA-lysolipids with 18O2, H218O, or H218O2 was present. Intriguingly, phosphatidylinositol phosphates (PIP2 and PIP3) effectively substituted for cardiolipin. Moreover, cytochrome c released from myocardial mitochondria subjected to oxidative stress cleaved plasmenylcholine in membrane bilayers, and this was blocked with a specific mAb against cytochrome c Collectively, these results identify the first plasmalogenase in biology, reveal the production of previously unanticipated signaling lipids by cytochrome c, and present new perspectives on cellular signaling during oxidative stress.
Collapse
Affiliation(s)
- Christopher M Jenkins
- From the Division of Bioorganic Chemistry and Molecular Pharmacology and.,Departments of Medicine and
| | - Kui Yang
- From the Division of Bioorganic Chemistry and Molecular Pharmacology and
| | - Gaoyuan Liu
- From the Division of Bioorganic Chemistry and Molecular Pharmacology and.,Departments of Medicine and.,the Department of Chemistry, Washington University, St. Louis, Missouri 63130
| | - Sung Ho Moon
- From the Division of Bioorganic Chemistry and Molecular Pharmacology and.,Departments of Medicine and
| | - Beverly G Dilthey
- From the Division of Bioorganic Chemistry and Molecular Pharmacology and.,Departments of Medicine and
| | - Richard W Gross
- From the Division of Bioorganic Chemistry and Molecular Pharmacology and .,Departments of Medicine and.,the Department of Chemistry, Washington University, St. Louis, Missouri 63130.,Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110 and
| |
Collapse
|
40
|
Wang B, Rong X, Palladino END, Wang J, Fogelman AM, Martín MG, Alrefai WA, Ford DA, Tontonoz P. Phospholipid Remodeling and Cholesterol Availability Regulate Intestinal Stemness and Tumorigenesis. Cell Stem Cell 2018; 22:206-220.e4. [PMID: 29395055 PMCID: PMC5807072 DOI: 10.1016/j.stem.2017.12.017] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/31/2017] [Accepted: 12/20/2017] [Indexed: 02/02/2023]
Abstract
Adequate availability of cellular building blocks, including lipids, is a prerequisite for cellular proliferation, but excess dietary lipids are linked to increased cancer risk. Despite these connections, specific regulatory relationships between membrane composition, intestinal stem cell (ISC) proliferation, and tumorigenesis are unclear. We reveal an unexpected link between membrane phospholipid remodeling and cholesterol biosynthesis and demonstrate that cholesterol itself acts as a mitogen for ISCs. Inhibition of the phospholipid-remodeling enzyme Lpcat3 increases membrane saturation and stimulates cholesterol biosynthesis, thereby driving ISC proliferation. Pharmacologic inhibition of cholesterol synthesis normalizes crypt hyperproliferation in Lpcat3-deficient organoids and mice. Conversely, increasing cellular cholesterol content stimulates crypt organoid growth, and providing excess dietary cholesterol or driving endogenous cholesterol synthesis through SREBP-2 expression promotes ISC proliferation in vivo. Finally, disruption of Lpcat3-dependent phospholipid and cholesterol homeostasis dramatically enhances tumor formation in Apcmin mice. These findings identify a critical dietary-responsive phospholipid-cholesterol axis regulating ISC proliferation and tumorigenesis.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Xin Rong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Elisa N D Palladino
- Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, St. Louis, MO, USA
| | - Jiafang Wang
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Martín G Martín
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Waddah A Alrefai
- Research and Development, Jesse Brown VA Medical Center, Chicago, IL, USA; Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - David A Ford
- Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, St. Louis, MO, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Zhang L, Tian Y, Yang J, Li J, Tang H, Wang Y. Colon Ascendens Stent Peritonitis (CASP) Induces Excessive Inflammation and Systemic Metabolic Dysfunction in a Septic Rat Model. J Proteome Res 2017; 17:680-688. [PMID: 29205045 DOI: 10.1021/acs.jproteome.7b00730] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The colon ascendens stent peritonitis (CASP) surgery induces a leakage of gut contents, causing polymicrobial sepsis related to post-operative multiple organ failure and death in surgical patient. To evaluate the effects of CASP on multiple organs, we analyzed the systemic metabolic consequences in liver, kidney, lung, and heart of rats after CASP by employing a combination of metabolomics, clinical chemistry, and biological assays. We found that CASP surgery after 18 h resulted in striking elevations of lipid, amino acids, acetate, choline, PC, and GPC in rat liver together with significant depletion of glucose and glycogen. Marked elevations of organic acids including lactate, acetate, and creatine and amino acids accompanied by decline of glucose, betaine, TMAO, choline metabolites (PC and GPC) nucleotides, and a range of organic osmolytes such as myo-inositol are observed in the kidney of 18 h post-operative rat. Furthermore, 18 h post-operative rats exhibited accumulations of lipid, amino acids, and depletions of taurine, myo-inositol, choline, PC, and GPC and some nucleotides including uridine, inosine, and adenosine in the lung. In addition, significant elevations of some amino acids, uracil, betaine, and choline metabolites, together with depletion of inosine-5'-monophosphate, were only observed in the heart of 18 h post-operative rats. These results provide new insights into pathological consequences of CASP surgery, which are important for timely prognosis of sepsis.
Collapse
Affiliation(s)
- Limin Zhang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS) , Wuhan 430071, China
| | - Yuan Tian
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS) , Wuhan 430071, China
| | - Jianfen Yang
- Research Institute of General Surgery, General Hospital of Nanjing Military Region , Nanjing, Jiangsu 210002, China
| | - Jieshou Li
- Research Institute of General Surgery, General Hospital of Nanjing Military Region , Nanjing, Jiangsu 210002, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Centre for Genetics and Development, Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, School of Life Sciences, Fudan University , Shanghai 200433, PR China
| | - Yulan Wang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS) , Wuhan 430071, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University , Hangzhou 310058, PR China
| |
Collapse
|
42
|
Glatzel DK, Koeberle A, Pein H, Löser K, Stark A, Keksel N, Werz O, Müller R, Bischoff I, Fürst R. Acetyl-CoA carboxylase 1 regulates endothelial cell migration by shifting the phospholipid composition. J Lipid Res 2017; 59:298-311. [PMID: 29208696 DOI: 10.1194/jlr.m080101] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/22/2017] [Indexed: 11/20/2022] Open
Abstract
The enzyme acetyl-CoA carboxylase (ACC) plays a crucial role in fatty acid metabolism. In recent years, ACC has been recognized as a promising drug target for treating different diseases. However, the role of ACC in vascular endothelial cells (ECs) has been neglected so far. To characterize the role of ACC, we used the ACC inhibitor, soraphen A, as a chemical tool, and also a gene silencing approach. We found that ACC1 was the predominant isoform in human umbilical vein ECs as well as in human microvascular ECs and that soraphen A reduced the levels of malonyl-CoA. We revealed that ACC inhibition shifted the lipid composition of EC membranes. Accordingly, membrane fluidity, filopodia formation, and migratory capacity were reduced. The antimigratory action of soraphen A depended on an increase in the cellular proportion of PUFAs and, most importantly, on a decreased level of phosphatidylglycerol. Our study provides a causal link between ACC, membrane lipid composition, and cell migration in ECs. Soraphen A represents a useful chemical tool to investigate the role of fatty acid metabolism in ECs and ACC inhibition offers a new and valuable therapeutic perspective for the treatment of EC migration-related diseases.
Collapse
Affiliation(s)
- Daniel K Glatzel
- Institute of Pharmaceutical Biology, Biocenter, Goethe University, Frankfurt, Germany
| | - Andreas Koeberle
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Helmut Pein
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Konstantin Löser
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Anna Stark
- Institute of Pharmaceutical Biology, Biocenter, Goethe University, Frankfurt, Germany
| | - Nelli Keksel
- Institute of Biochemistry and Molecular Biology, Rheinische Friedrich-Wilhelms-University of Bonn, Bonn, Germany
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarland University, Saarbrücken, Germany
| | - Iris Bischoff
- Institute of Pharmaceutical Biology, Biocenter, Goethe University, Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Biocenter, Goethe University, Frankfurt, Germany
| |
Collapse
|
43
|
Targeting de novo lipogenesis as a novel approach in anti-cancer therapy. Br J Cancer 2017; 118:43-51. [PMID: 29112683 PMCID: PMC5765225 DOI: 10.1038/bjc.2017.374] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/22/2017] [Accepted: 09/22/2017] [Indexed: 12/13/2022] Open
Abstract
Background: Although altered membrane physiology has been discussed within the context of cancer, targeting membrane characteristics by drugs being an attractive therapeutic strategy has received little attention so far. Methods: Various acetyl-CoA carboxylase 1 (ACC1), and fatty acid synthase (FASN) inhibitors (like Soraphen A and Cerulenin) as well as genetic knockdown approaches were employed to study the effects of disturbed phospholipid composition on membrane properties and its functional impact on cancer progression. By using state-of-the-art methodologies such as LC-MS/MS, optical tweezers measurements of giant plasma membrane vesicles and fluorescence recovery after photobleaching analysis, membrane characteristics were examined. Confocal laser scanning microscopy, proximity ligation assays, immunoblotting as well as migration, invasion and proliferation experiments unravelled the functional relevance of membrane properties in vitro and in vivo. Results: By disturbing the deformability and lateral fluidity of cellular membranes, the dimerisation, localisation and recycling of cancer-relevant transmembrane receptors is compromised. Consequently, impaired activation of growth factor receptor signalling cascades results in abrogated tumour growth and metastasis in different in vitro and in vivo models. Conclusions: This study highlights the field of membrane properties as a promising druggable cellular target representing an innovative strategy for development of anti-cancer agents.
Collapse
|
44
|
Furse S, Shearman GC. Do lipids shape the eukaryotic cell cycle? Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:9-19. [PMID: 28964796 DOI: 10.1016/j.bbalip.2017.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/07/2017] [Accepted: 09/24/2017] [Indexed: 10/18/2022]
Abstract
Successful passage through the cell cycle presents a number of structural challenges to the cell. Inceptive studies carried out in the last five years have produced clear evidence of modulations in the lipid profile (sometimes referred to as the lipidome) of eukaryotes as a function of the cell cycle. This mounting body of evidence indicates that lipids play key roles in the structural transformations seen across the cycle. The accumulation of this evidence coincides with a revolution in our understanding of how lipid composition regulates a plethora of biological processes ranging from protein activity through to cellular signalling and membrane compartmentalisation. In this review, we discuss evidence from biological, chemical and physical studies of the lipid fraction across the cell cycle that demonstrate that lipids are well-developed cellular components at the heart of the biological machinery responsible for managing progress through the cell cycle. Furthermore, we discuss the mechanisms by which this careful control is exercised.
Collapse
Affiliation(s)
- Samuel Furse
- NucReg Research Programme, Molekylærbiologisk institutt, Unversitetet i Bergen, Thormøhlens gate 55, 5008, Bergen, Norway; Core Metabolomics and Lipidomics Laboratory, Department of Biochemistry, University of Cambridge, c/o Level 4, Pathology Building, Addenbrookes Hospital, Cambridge, CB2 0QQ, United Kingdom..
| | - Gemma C Shearman
- Faculty of Science, Engineering and Computing, Penrhyn Road, Kingston upon Thames, Surrey KT1 2EE, United Kingdom
| |
Collapse
|
45
|
Effects of the dietary carbohydrate-fat ratio on plasma phosphatidylcholine profiles in human and mouse. J Nutr Biochem 2017; 50:83-94. [PMID: 29040839 DOI: 10.1016/j.jnutbio.2017.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/25/2017] [Accepted: 08/30/2017] [Indexed: 11/23/2022]
Abstract
Phosphatidylcholines (PCs), a major class of human plasma phospholipids, are composed of highly diverse fatty acids. Because the dietary carbohydrate-fat ratio alters the hepatic fatty acid metabolism, plasma fatty acids that bind PCs, which are secreted as lipoproteins from the liver, may be affected by long-term consumption of a high-carbohydrate diet or a high-fat diet. Therefore, in this study, we profiled the plasma PC species comprehensively in formulated dieting conditions to identify those phospholipid molecules that reflect the dietary carbohydrate-fat ratio. C57BL6J mice were fed diets containing different amounts of fat for 8 weeks, and plasma PC species were analyzed under fasting conditions using liquid chromatography-mass spectrometry. In addition, a cross-sectional study of 78 middle-aged Japanese men, who participated in health checkups, was conducted. Nutrient intakes were estimated by a brief self-administered diet-history questionnaire. The plasma PC profiles changed depending on the dietary carbohydrate-fat ratio. Especially, PC (16:0/16:1) and PC (16:0/18:1) levels increased as the dietary carbohydrate-fat ratio increased in human and mouse, suggesting that these PC species reflected the increase in de novo lipogenesis and might become useful biomarkers of the dietary carbohydrate-fat ratio. Since these PCs act as ligands for peroxisome proliferator-activated receptor α, PC species reflecting the dietary carbohydrate-fat ratio may influence metabolism of glucose and lipids.
Collapse
|
46
|
Pein H, Koeberle SC, Voelkel M, Schneider F, Rossi A, Thürmer M, Loeser K, Sautebin L, Morrison H, Werz O, Koeberle A. Vitamin A regulates Akt signaling through the phospholipid fatty acid composition. FASEB J 2017; 31:4566-4577. [PMID: 28687611 DOI: 10.1096/fj.201700078r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/19/2017] [Indexed: 01/04/2023]
Abstract
Protein kinases, including the serine/threonine kinase Akt, mediate manifold bioactivities of vitamin A, although the mechanisms behind the sustained kinase activation are diffuse. To investigate the role of cellular lipids as targetable factors in Akt signaling, we combined mass spectrometry-based lipidomics with immunologic detection of Akt (Ser473) phosphorylation. A screening campaign revealed retinol (vitamin A alcohol) and all-trans retinoic acid (vitamin A acid) (RA) as hits that time-dependently (≥24 h) deplete phosphatidylcholine-bound polyunsaturated fatty acids (PUFA-PCs) from NIH-3T3 mouse fibroblasts while inducing Akt activation (EC50 ≈ 0.1-1 µM). Other mitogenic and stress-regulated kinases were hardly affected. Organized in a coregulated phospholipid subcluster, PUFA-PCs compensated for the RA-induced loss of cellular PUFA-PCs and diminished Akt activation when supplemented. The counter-regulation of phospholipids and Akt by RA was mimicked by knockdown of lysophosphatidylcholine acyltransferase-3 or the selective retinoid X receptor (RXR) agonist bexarotene and prevented by the selective RXR antagonist Hx531. Treatment of mice with retinol decreased the tissue ratio of PUFA-PC and enhanced basal Akt activation preferentially in brain, which was attributed to astrocytes in dissociated cortical cultures. Together, our findings show that RA regulates the long-term activation of Akt by changes in the phospholipid composition.-Pein, H., Koeberle, S. C., Voelkel, M., Schneider, F., Rossi, A., Thürmer, M., Loeser, K., Sautebin, L., Morrison, H., Werz, O., Koeberle, A. Vitamin A regulates Akt signaling through the phospholipid fatty acid composition.
Collapse
Affiliation(s)
- Helmut Pein
- Institute of Pharmacy, Friedrich-Schiller-University, Jena, Germany
| | | | - Maria Voelkel
- Institute of Pharmacy, Friedrich-Schiller-University, Jena, Germany
| | - Freya Schneider
- Institute of Pharmacy, Friedrich-Schiller-University, Jena, Germany
| | - Antonietta Rossi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Maria Thürmer
- Institute of Pharmacy, Friedrich-Schiller-University, Jena, Germany
| | | | - Lidia Sautebin
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Helen Morrison
- Leibniz Institute of Age Research, Fritz-Lipmann-Institute, Jena, Germany
| | - Oliver Werz
- Institute of Pharmacy, Friedrich-Schiller-University, Jena, Germany
| | - Andreas Koeberle
- Institute of Pharmacy, Friedrich-Schiller-University, Jena, Germany;
| |
Collapse
|
47
|
Miscibility Transition Temperature Scales with Growth Temperature in a Zebrafish Cell Line. Biophys J 2017; 113:1212-1222. [PMID: 28552311 DOI: 10.1016/j.bpj.2017.04.052] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/23/2017] [Accepted: 04/05/2017] [Indexed: 11/22/2022] Open
Abstract
Cells can alter the lipid content of their plasma membranes upon changes in their environment to maintain and adjust membrane function. Recent work suggests that some membrane functions arise because cellular plasma membranes are poised close to a miscibility transition under growth conditions. Here we report experiments utilizing giant plasma membrane vesicles (GPMVs) to explore how membrane transition temperature varies with growth temperature in a zebrafish cell line (ZF4) that can be adapted for growth between 20 and 32°C. We find that GPMV transition temperatures adjust to be 16.7 ± 1.2°C below growth temperature for four growth temperatures investigated and that adjustment occurs over roughly 2 days when temperature is abruptly lowered from 28 to 20°C. We also find that GPMVs have slightly different lipidomes when isolated from cells adapted for growth at 28 and 20°C. Similar to past work in vesicles derived from mammalian cells, fluctuating domains are observed in ZF4-derived GPMVs, consistent with their having critical membrane compositions. Taken together, these experimental results suggest that cells in culture biologically tune their membrane composition in a way that maintains specific proximity to a critical miscibility transition.
Collapse
|
48
|
Craddock CP, Adams N, Kroon JT, Bryant FM, Hussey PJ, Kurup S, Eastmond PJ. Cyclin-dependent kinase activity enhances phosphatidylcholine biosynthesis in Arabidopsis by repressing phosphatidic acid phosphohydrolase activity. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2017; 89:3-14. [PMID: 27595588 PMCID: PMC5299491 DOI: 10.1111/tpj.13321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/22/2016] [Accepted: 08/24/2016] [Indexed: 05/21/2023]
Abstract
Coordination of endomembrane biogenesis with cell cycle progression is considered to be important in maintaining cell function during growth and development. We previously showed that the disruption of PHOSPHATIDIC ACID PHOSPHOHYDROLASE (PAH) activity in Arabidopsis thaliana stimulates biosynthesis of the major phospholipid phosphatidylcholine (PC) and causes expansion of the endoplasmic reticulum. Here we show that PC biosynthesis is repressed by disruption of the core cell cycle regulator CYCLIN-DEPENDENT KINASE A;1 (CDKA;1) and that this repression is reliant on PAH. Furthermore, we show that cyclin-dependent kinases (CDKs) phosphorylate PAH1 at serine 162, which reduces both its activity and membrane association. Expression of a CDK-insensitive version of PAH1 with a serine 162 to alanine substitution represses PC biosynthesis and also reduces the rate of cell division in early leaf development. Together our findings reveal a physiologically important mechanism that couples the rate of phospholipid biosynthesis and endomembrane biogenesis to cell cycle progression in Arabidopsis.
Collapse
Affiliation(s)
- Christian P. Craddock
- School of Life SciencesUniversity of WarwickCoventryCV4 7ALUK
- Present address: Center for Plant Cell BiologyDepartment of Botany and Plant SciencesUniversity of CaliforniaRiverside92521USA
| | - Nicolette Adams
- School of Life SciencesUniversity of WarwickCoventryCV4 7ALUK
- Present address: Centre for Proteomic and Genomic ResearchUpper LevelSt Peter's MallCorner Anzio and Main Road ObservatoryCape Town7925South Africa
| | - Johan T.M. Kroon
- School of Biological and Biomedical SciencesDurham UniversityDurhamDH1 3LEUK
| | - Fiona M. Bryant
- Department of Plant Biology and Crop ScienceRothamsted ResearchHarpendenHertfordshireAL5 2JQUK
- Present address: School of Biological and Biomedical SciencesDurham UniversityDurhamDH1 3LEUK
| | - Patrick J. Hussey
- School of Biological and Biomedical SciencesDurham UniversityDurhamDH1 3LEUK
| | - Smita Kurup
- Department of Plant Biology and Crop ScienceRothamsted ResearchHarpendenHertfordshireAL5 2JQUK
| | - Peter J. Eastmond
- Department of Plant Biology and Crop ScienceRothamsted ResearchHarpendenHertfordshireAL5 2JQUK
| |
Collapse
|
49
|
|
50
|
Stearoyl-CoA desaturase-1 and adaptive stress signaling. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1719-1726. [DOI: 10.1016/j.bbalip.2016.08.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/09/2016] [Accepted: 08/17/2016] [Indexed: 12/31/2022]
|