1
|
Kim KS, Lee JS, Han SS, Cho JY. Accurate Determination of Circulatory Lipids Using a Combination of HILIC-MRM and RPLC-PRM. Anal Chem 2025; 97:9713-9721. [PMID: 40315190 PMCID: PMC12079635 DOI: 10.1021/acs.analchem.4c06409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
Circulatory lipids are important markers for characterizing disease phenotypes; however, accurately determining lipid species remains a significant challenge in lipidomic analysis. Here, we present a novel analytical workflow for accurate lipidome characterization in human plasma using mass spectrometry (MS) through the integration of hydrophilic interaction liquid chromatography (HILIC) and reversed-phase liquid chromatography (RPLC). This workflow enables rapid screening of 1,966 lipid species across 18 lipid classes using HILIC-multiple reaction monitoring (MRM), which enables facile identification of lipid species by lipid class-based separations. In the NIST Standard Reference Material for Human Plasma (SRM 1950), 489 lipid species were identified using HILIC-MRM and subsequently analyzed with RPLC-parallel reaction monitoring (PRM) to resolve potential lipid isobars within the same lipid class. Notably, RPLC-PRM identified 70 additional lipidomic features in SRM 1950 that were not detectable with HILIC-MRM. Furthermore, a high correlation (Pearson correlation coefficient = 0.81) was observed regarding the concentrations of lipid species not carrying isobaric interferences in between HILIC-MRM and RPLC-PRM, indicating that the individual lipid concentrations measured by each platform can be integrated. The workflow was further applied to a cohort of 284 human plasma samples from chronic kidney disease (CKD) patients, successfully profiling lipidomic phenotypes across CKD subtypes. These findings demonstrate that combining HILIC-MRM and RPLC-PRM as complementary platforms enhances the accuracy and comprehensiveness of lipidomic analysis.
Collapse
Affiliation(s)
- Kyeong-Seog Kim
- Department
of Biomedical Sciences, Seoul National University
College of Medicine, Seoul 03080, Republic
of Korea
- Department
of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Republic of Korea
- Seoul
National University, Seoul 08826, Republic
of Korea
| | - Jae-Seung Lee
- Department
of Biomedical Sciences, Seoul National University
College of Medicine, Seoul 03080, Republic
of Korea
- Department
of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Republic of Korea
- Seoul
National University, Seoul 08826, Republic
of Korea
| | - Seung Seok Han
- Department
of Internal Medicine, Seoul National University
College of Medicine, Seoul 03080, Republic
of Korea
| | - Joo-Youn Cho
- Department
of Biomedical Sciences, Seoul National University
College of Medicine, Seoul 03080, Republic
of Korea
- Department
of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Republic of Korea
- Seoul
National University, Seoul 08826, Republic
of Korea
- Kidney
Research Institute, Seoul National University
Medical Research Center, Seoul 03080, Republic
of Korea
| |
Collapse
|
2
|
Fougère L, Grison M, Laquel P, Montrazi M, Cordelières F, Fernández-Monreal M, Poujol C, Uemura T, Nakano A, Ito Y, Boutté Y. ER-to-Golgi trafficking through a dynamic intermediate cis-Golgi tubular network in Arabidopsis. Nat Cell Biol 2025; 27:424-437. [PMID: 40000850 DOI: 10.1038/s41556-025-01624-x] [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: 11/24/2023] [Accepted: 01/14/2025] [Indexed: 02/27/2025]
Abstract
Endoplasmic reticulum (ER)-to-Golgi trafficking is a central process of the secretory system of eukaryotic cells that ensures proper spatiotemporal sorting of proteins and lipids. However, the nature of the ER-Golgi intermediate compartments (ERGICs) and the molecular mechanisms mediating the transition between ERGICs and the Golgi, as well as the universality of these processes among eukaryotes, remain undiscovered. Here we identify a reticulated tubulo-vesicular network, labelled by MEMBRIN proteins, that is mostly independent of the Golgi, highly dynamic at the ER-Golgi interface and crossed by ER-induced released luminal cargos. We find that plant ERGICs become stabilized by the interaction they establish with pre-existing Golgi and gradually mature into Golgi cisternae, this process being dependent on C24-ceramide sphingolipids. Our study is a major twist in the understanding of the Golgi, as it identifies that the ERGICs in plants comprise a Golgi-independent and highly dynamic tubular network from which arise more stable Golgi-associated pre-cisternae structures.
Collapse
Affiliation(s)
- Louise Fougère
- Laboratoire de Biogenèse Membranaire, Université de Bordeaux, CNRS UMR5200, Villenave d'Ornon, France
| | - Magali Grison
- Laboratoire de Biogenèse Membranaire, Université de Bordeaux, CNRS UMR5200, Villenave d'Ornon, France
| | - Patricia Laquel
- Laboratoire de Biogenèse Membranaire, Université de Bordeaux, CNRS UMR5200, Villenave d'Ornon, France
| | - Matheus Montrazi
- Laboratoire de Biogenèse Membranaire, Université de Bordeaux, CNRS UMR5200, Villenave d'Ornon, France
| | - Fabrice Cordelières
- Bordeaux Imaging Center (BIC), Université de Bordeaux, INSERM, CNRS UAR3420, Bordeaux, France
| | | | - Christel Poujol
- Bordeaux Imaging Center (BIC), Université de Bordeaux, INSERM, CNRS UAR3420, Bordeaux, France
| | - Tomohiro Uemura
- Faculty of Core Research, Natural Science Division, Ochanomizu University, Tokyo, Japan
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama, Japan
| | - Yoko Ito
- Institute for Human Life Science, Ochanomizu University, Tokyo, Japan
| | - Yohann Boutté
- Laboratoire de Biogenèse Membranaire, Université de Bordeaux, CNRS UMR5200, Villenave d'Ornon, France.
| |
Collapse
|
3
|
Roffay C, García-Arcos JM, Chapuis P, López-Andarias J, Schneider F, Colom A, Tomba C, Di Meglio I, Barrett K, Dunsing V, Matile S, Roux A, Mercier V. Tutorial: fluorescence lifetime microscopy of membrane mechanosensitive Flipper probes. Nat Protoc 2024; 19:3457-3469. [PMID: 39210094 DOI: 10.1038/s41596-024-01027-6] [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: 02/28/2023] [Accepted: 05/21/2024] [Indexed: 09/04/2024]
Abstract
Measuring forces within living cells remains a technical challenge. In this Tutorial, we cover the development of hydrophobic mechanosensing fluorescent probes called Flippers, whose fluorescence lifetime depends on lipid packing. Flipper probes can therefore be used as reporters for membrane tension via the measurement of changes in their fluorescence lifetime. We describe the technical optimization of the probe for imaging and provide working examples for their characterizations in a variety of biological and in vitro systems. We further provide a guideline to measure biophysical parameters of cellular membranes by fluorescence lifetime imaging microscopy using Flipper probes, providing evidence that flippers can report long range forces in cells, tissues and organs.
Collapse
Affiliation(s)
- Chloé Roffay
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | | | - Pierrik Chapuis
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Javier López-Andarias
- Department of Organic Chemistry, University of Geneva, Geneva, Switzerland
- National Center of Competence in Research in Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Falk Schneider
- Translational Imaging Center, University of Southern California, Los Angeles, CA, USA
| | - Adai Colom
- Biofisika Institute (CSIC, UPV/EHU), Leioa, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, Campus Universitario, University of the Basque Country (UPV/EHU), Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Caterina Tomba
- CNRS, INSA Lyon, Ecole Centrale de Lyon, Universite Claude Bernard Lyon 1, CPE Lyon, INL, Villeurbanne, France
| | - Ilaria Di Meglio
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Katia Barrett
- Aix-Marseille Université & CNRS, IBDM-UMR7288 & Turing Centre for Living Systems, Parc Scientifique de Luminy, Marseille, France
| | - Valentin Dunsing
- Aix-Marseille Université & CNRS, IBDM-UMR7288 & Turing Centre for Living Systems, Parc Scientifique de Luminy, Marseille, France
| | - Stefan Matile
- Department of Organic Chemistry, University of Geneva, Geneva, Switzerland
- National Center of Competence in Research in Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, Geneva, Switzerland.
- National Center of Competence in Research in Chemical Biology, University of Geneva, Geneva, Switzerland.
| | - Vincent Mercier
- Department of Biochemistry, University of Geneva, Geneva, Switzerland.
- National Center of Competence in Research in Chemical Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
4
|
Sah S, Bifarin OO, Moore SG, Gaul DA, Chung H, Kwon SY, Cho H, Cho CH, Kim JH, Kim J, Fernández FM. Serum Lipidome Profiling Reveals a Distinct Signature of Ovarian Cancer in Korean Women. Cancer Epidemiol Biomarkers Prev 2024; 33:681-693. [PMID: 38412029 PMCID: PMC11061607 DOI: 10.1158/1055-9965.epi-23-1293] [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: 10/19/2023] [Revised: 01/11/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Distinguishing ovarian cancer from other gynecological malignancies is crucial for patient survival yet hindered by non-specific symptoms and limited understanding of ovarian cancer pathogenesis. Accumulating evidence suggests a link between ovarian cancer and deregulated lipid metabolism. Most studies have small sample sizes, especially for early-stage cases, and lack racial/ethnic diversity, necessitating more inclusive research for improved ovarian cancer diagnosis and prevention. METHODS Here, we profiled the serum lipidome of 208 ovarian cancer, including 93 early-stage patients with ovarian cancer and 117 nonovarian cancer (other gynecological malignancies) patients of Korean descent. Serum samples were analyzed with a high-coverage liquid chromatography high-resolution mass spectrometry platform, and lipidome alterations were investigated via statistical and machine learning (ML) approaches. RESULTS We found that lipidome alterations unique to ovarian cancer were present in Korean women as early as when the cancer is localized, and those changes increase in magnitude as the diseases progresses. Analysis of relative lipid abundances revealed specific patterns for various lipid classes, with most classes showing decreased abundance in ovarian cancer in comparison with other gynecological diseases. ML methods selected a panel of 17 lipids that discriminated ovarian cancer from nonovarian cancer cases with an AUC value of 0.85 for an independent test set. CONCLUSIONS This study provides a systemic analysis of lipidome alterations in human ovarian cancer, specifically in Korean women. IMPACT Here, we show the potential of circulating lipids in distinguishing ovarian cancer from nonovarian cancer conditions.
Collapse
Affiliation(s)
- Samyukta Sah
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia
- Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Olatomiwa O. Bifarin
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia
| | - Samuel G. Moore
- Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - David A. Gaul
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia
- Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Hyewon Chung
- Department of Obstetrics and Gynecology, School of Medicine, Keimyung University, Daegu Republic of Korea
| | - Sun Young Kwon
- Department of Pathology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chi-Heum Cho
- Department of Obstetrics and Gynecology, School of Medicine, Keimyung University, Daegu Republic of Korea
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaeyeon Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Facundo M. Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia
- Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
5
|
Fougère L, Mongrand S, Boutté Y. The function of sphingolipids in membrane trafficking and cell signaling in plants, in comparison with yeast and animal cells. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159463. [PMID: 38281556 DOI: 10.1016/j.bbalip.2024.159463] [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: 05/16/2023] [Revised: 12/04/2023] [Accepted: 01/23/2024] [Indexed: 01/30/2024]
Abstract
Sphingolipids are essential membrane components involved in a wide range of cellular, developmental and signaling processes. Sphingolipids are so essential that knock-out mutation often leads to lethality. In recent years, conditional or weak allele mutants as well as the broadening of the pharmacological catalog allowed to decipher sphingolipid function more precisely in a less invasive way. This review intends to provide a discussion and point of view on the function of sphingolipids with a main focus on endomembrane trafficking, Golgi-mediated protein sorting, cell polarity, cell-to-cell communication and cell signaling at the plasma membrane. While our main angle is the plant field research, we will constantly refer to and compare with the advances made in the yeast and animal field. In this review, we will emphasize the role of sphingolipids not only as a membrane component, but also as a key player at a center of homeostatic regulatory networks involving direct or indirect interaction with other lipids, proteins and ion fluxes.
Collapse
Affiliation(s)
- Louise Fougère
- Laboratoire de Biogenèse Membranaire, Univ. Bordeaux, UMR 5200 CNRS, Villenave d'Ornon, France
| | - Sebastien Mongrand
- Laboratoire de Biogenèse Membranaire, Univ. Bordeaux, UMR 5200 CNRS, Villenave d'Ornon, France
| | - Yohann Boutté
- Laboratoire de Biogenèse Membranaire, Univ. Bordeaux, UMR 5200 CNRS, Villenave d'Ornon, France.
| |
Collapse
|
6
|
Girik V, van Ek L, Dentand Quadri I, Azam M, Cruz Cobo M, Mandavit M, Riezman I, Riezman H, Gavin AC, Nunes-Hasler P. Development of Genetically Encoded Fluorescent KSR1-Based Probes to Track Ceramides during Phagocytosis. Int J Mol Sci 2024; 25:2996. [PMID: 38474242 DOI: 10.3390/ijms25052996] [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: 01/20/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Ceramides regulate phagocytosis; however, their exact function remains poorly understood. Here, we sought (1) to develop genetically encoded fluorescent tools for imaging ceramides, and (2) to use them to examine ceramide dynamics during phagocytosis. Fourteen enhanced green fluorescent protein (EGFP) fusion constructs based on four known ceramide-binding domains were generated and screened. While most constructs localized to the nucleus or cytosol, three based on the CA3 ceramide-binding domain of kinase suppressor of ras 1 (KSR1) localized to the plasma membrane or autolysosomes. C-terminally tagged CA3 with a vector-based (C-KSR) or glycine-serine linker (C-KSR-GS) responded sensitively and similarly to ceramide depletion and accumulation using a panel of ceramide modifying drugs, whereas N-terminally tagged CA3 (N-KSR) responded differently to a subset of treatments. Lipidomic and liposome microarray analysis suggested that, instead, N-KSR may preferentially bind glucosyl-ceramide. Additionally, the three probes showed distinct dynamics during phagocytosis. Despite partial autolysosomal degradation, C-KSR and C-KSR-GS accumulated at the plasma membrane during phagocytosis, whereas N-KSR did not. Moreover, the weak recruitment of C-KSR-GS to the endoplasmic reticulum and phagosomes was enhanced through overexpression of the endoplasmic reticulum proteins stromal interaction molecule 1 (STIM1) and Sec22b, and was more salient in dendritic cells. The data suggest these novel probes can be used to analyze sphingolipid dynamics and function in living cells.
Collapse
Affiliation(s)
- Vladimir Girik
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Larissa van Ek
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Isabelle Dentand Quadri
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Maral Azam
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - María Cruz Cobo
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Marion Mandavit
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Isabelle Riezman
- Department of Biochemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland
| | - Howard Riezman
- Department of Biochemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland
| | - Anne-Claude Gavin
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Department of Biochemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland
| | - Paula Nunes-Hasler
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
7
|
Ma X, Fernández FM. Advances in mass spectrometry imaging for spatial cancer metabolomics. MASS SPECTROMETRY REVIEWS 2024; 43:235-268. [PMID: 36065601 PMCID: PMC9986357 DOI: 10.1002/mas.21804] [Citation(s) in RCA: 56] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 05/09/2023]
Abstract
Mass spectrometry (MS) has become a central technique in cancer research. The ability to analyze various types of biomolecules in complex biological matrices makes it well suited for understanding biochemical alterations associated with disease progression. Different biological samples, including serum, urine, saliva, and tissues have been successfully analyzed using mass spectrometry. In particular, spatial metabolomics using MS imaging (MSI) allows the direct visualization of metabolite distributions in tissues, thus enabling in-depth understanding of cancer-associated biochemical changes within specific structures. In recent years, MSI studies have been increasingly used to uncover metabolic reprogramming associated with cancer development, enabling the discovery of key biomarkers with potential for cancer diagnostics. In this review, we aim to cover the basic principles of MSI experiments for the nonspecialists, including fundamentals, the sample preparation process, the evolution of the mass spectrometry techniques used, and data analysis strategies. We also review MSI advances associated with cancer research in the last 5 years, including spatial lipidomics and glycomics, the adoption of three-dimensional and multimodal imaging MSI approaches, and the implementation of artificial intelligence/machine learning in MSI-based cancer studies. The adoption of MSI in clinical research and for single-cell metabolomics is also discussed. Spatially resolved studies on other small molecule metabolites such as amino acids, polyamines, and nucleotides/nucleosides will not be discussed in the context.
Collapse
Affiliation(s)
- Xin Ma
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Facundo M Fernández
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Chang JK, Teo G, Pewzner-Jung Y, Cuthbertson DJ, Futerman AH, Wenk MR, Choi H, Torta F. Q-RAI data-independent acquisition for lipidomic quantitative profiling. Sci Rep 2023; 13:19281. [PMID: 37935746 PMCID: PMC10630469 DOI: 10.1038/s41598-023-46312-8] [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: 01/31/2023] [Accepted: 10/30/2023] [Indexed: 11/09/2023] Open
Abstract
Untargeted lipidomics has been increasingly adopted for hypothesis generation in a biological context or discovery of disease biomarkers. Most of the current liquid chromatography mass spectrometry (LC-MS) based untargeted methodologies utilize a data dependent acquisition (DDA) approach in pooled samples for identification and MS-only acquisition for semi-quantification in individual samples. In this study, we present for the first time an untargeted lipidomic workflow that makes use of the newly implemented Quadrupole Resolved All-Ions (Q-RAI) acquisition function on the Agilent 6546 quadrupole time-of-flight (Q-TOF) mass spectrometer to acquire MS2 spectra in data independent acquisition (DIA) mode. This is followed by data processing and analysis on MetaboKit, a software enabling DDA-based spectral library construction and extraction of MS1 and MS2 peak areas, for reproducible identification and quantification of lipids in DIA analysis. This workflow was tested on lipid extracts from human plasma and showed quantification at MS1 and MS2 levels comparable to multiple reaction monitoring (MRM) targeted analysis of the same samples. Analysis of serum from Ceramide Synthase 2 (CerS2) null mice using the Q-RAI DIA workflow identified 88 lipid species significantly different between CerS2 null and wild type mice, including well-characterized changes previously associated with this phenotype. Our results show the Q-RAI DIA as a reliable option to perform simultaneous identification and reproducible relative quantification of lipids in exploratory biological studies.
Collapse
Affiliation(s)
- Jing Kai Chang
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Guoshou Teo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yael Pewzner-Jung
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Markus R Wenk
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Hyungwon Choi
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Federico Torta
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
9
|
Huang F, Cai F, Dahabieh MS, Gunawardena K, Talebi A, Dehairs J, El-Turk F, Park JY, Li M, Goncalves C, Gagnon N, Su J, LaPierre JH, Gaub P, Joyal JS, Mitchell JJ, Swinnen JV, Miller WH, del Rincón SV. Peroxisome disruption alters lipid metabolism and potentiates antitumor response with MAPK-targeted therapy in melanoma. J Clin Invest 2023; 133:e166644. [PMID: 37616051 PMCID: PMC10575734 DOI: 10.1172/jci166644] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 08/22/2023] [Indexed: 08/25/2023] Open
Abstract
Melanomas reprogram their metabolism to rapidly adapt to therapy-induced stress conditions, allowing them to persist and ultimately develop resistance. We report that a subpopulation of melanoma cells tolerate MAPK pathway inhibitors (MAPKis) through a concerted metabolic reprogramming mediated by peroxisomes and UDP-glucose ceramide glycosyltransferase (UGCG). Compromising peroxisome biogenesis, by repressing PEX3 expression, potentiated the proapoptotic effects of MAPKis via an induction of ceramides, an effect limited by UGCG-mediated ceramide metabolism. Cotargeting PEX3 and UGCG selectively eliminated a subset of metabolically active, drug-tolerant CD36+ melanoma persister cells, thereby sensitizing melanoma to MAPKis and delaying resistance. Increased levels of peroxisomal genes and UGCG were found in patient-derived MAPKi-relapsed melanomas, and simultaneously inhibiting PEX3 and UGCG restored MAPKi sensitivity in multiple models of therapy resistance. Finally, combination therapy consisting of a newly identified inhibitor of the PEX3-PEX19 interaction, a UGCG inhibitor, and MAPKis demonstrated potent antitumor activity in preclinical melanoma models, thus representing a promising approach for melanoma treatment.
Collapse
Affiliation(s)
- Fan Huang
- Lady Davis Institute
- Department of Experimental Medicine, and
| | - Feiyang Cai
- Lady Davis Institute
- Department of Experimental Medicine, and
| | | | | | - Ali Talebi
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Farah El-Turk
- McGill University Health Centre, Montreal, Quebec, Canada
- Centre Hospitalier Universitaire Sainte Justine, Montreal, Quebec, Canada
| | - Jae Yeon Park
- McGill University Health Centre, Montreal, Quebec, Canada
| | - Mengqi Li
- Lady Davis Institute
- Department of Experimental Medicine, and
| | | | | | | | | | - Perrine Gaub
- Centre de Recherche, CHU St. Justine, Montréal, Quebec, Canada
| | | | | | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Wilson H. Miller
- Lady Davis Institute
- Department of Experimental Medicine, and
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Sonia V. del Rincón
- Lady Davis Institute
- Department of Experimental Medicine, and
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Enkler L, Szentgyörgyi V, Pennauer M, Prescianotto-Baschong C, Riezman I, Wiesyk A, Avraham RE, Spiess M, Zalckvar E, Kucharczyk R, Riezman H, Spang A. Arf1 coordinates fatty acid metabolism and mitochondrial homeostasis. Nat Cell Biol 2023; 25:1157-1172. [PMID: 37400497 PMCID: PMC10415182 DOI: 10.1038/s41556-023-01180-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/05/2023] [Indexed: 07/05/2023]
Abstract
Lipid mobilization through fatty acid β-oxidation is a central process essential for energy production during nutrient shortage. In yeast, this catabolic process starts in the peroxisome from where β-oxidation products enter mitochondria and fuel the tricarboxylic acid cycle. Little is known about the physical and metabolic cooperation between these organelles. Here we found that expression of fatty acid transporters and of the rate-limiting enzyme involved in β-oxidation is decreased in cells expressing a hyperactive mutant of the small GTPase Arf1, leading to an accumulation of fatty acids in lipid droplets. Consequently, mitochondria became fragmented and ATP synthesis decreased. Genetic and pharmacological depletion of fatty acids phenocopied the arf1 mutant mitochondrial phenotype. Although β-oxidation occurs in both mitochondria and peroxisomes in mammals, Arf1's role in fatty acid metabolism is conserved. Together, our results indicate that Arf1 integrates metabolism into energy production by regulating fatty acid storage and utilization, and presumably organelle contact sites.
Collapse
Affiliation(s)
| | | | | | | | - Isabelle Riezman
- Department of Biochemistry, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Aneta Wiesyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Reut Ester Avraham
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Einat Zalckvar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Roza Kucharczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Howard Riezman
- Department of Biochemistry, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Anne Spang
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
11
|
Melero A, Jiménez-Rojo N. Cracking the membrane lipid code. Curr Opin Cell Biol 2023; 83:102203. [PMID: 37437490 DOI: 10.1016/j.ceb.2023.102203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/01/2023] [Accepted: 06/16/2023] [Indexed: 07/14/2023]
Abstract
Why has nature acquired such a huge lipid repertoire? Although it would be theoretically possible to make a lipid bilayer fulfilling barrier functions with only one glycerophospholipid, there are diverse and numerous different lipid species. Lipids are heterogeneously distributed across the evolutionary tree with lipidomes evolving in parallel to organismal complexity. Moreover, lipids are different between organs and tissues and even within the same cell, different organelles have characteristic lipid signatures. At the molecular level, membranes are asymmetric and laterally heterogeneous. This lipid asymmetry at different scales indicates that these molecules may play very specific molecular functions in biology. Some of these roles have been recently uncovered: lipids have been shown to be essential in processes such as hypoxia and ferroptosis or in protein sorting and trafficking but many of them remain still unknown. In this review we will discuss the importance of understanding lipid diversity in biology across scales and we will share a toolbox with some of the emerging technologies that are helping us to uncover new lipid molecular functions in cell biology and, step by step, crack the membrane lipid code.
Collapse
Affiliation(s)
- Alejandro Melero
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Noemi Jiménez-Rojo
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain; Instituto Biofisika (UPV/EHU, CSIC), 48940, Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
12
|
Álvarez D, Sapia J, Vanni S. Computational modeling of membrane trafficking processes: From large molecular assemblies to chemical specificity. Curr Opin Cell Biol 2023; 83:102205. [PMID: 37451175 DOI: 10.1016/j.ceb.2023.102205] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023]
Abstract
In the last decade, molecular dynamics (MD) simulations have become an essential tool to investigate the molecular properties of membrane trafficking processes, often in conjunction with experimental approaches. The combination of MD simulations with recent developments in structural biology, such as cryo-electron microscopy and artificial intelligence-based structure determination, opens new, exciting possibilities for future investigations. However, the full potential of MD simulations to provide a molecular view of the complex and dynamic processes involving membrane trafficking can only be realized if certain limitations are addressed, and especially those concerning the quality of coarse-grain models, which, despite recent successes in describing large-scale systems, still suffer from far-from-ideal chemical accuracy. In this review, we will highlight recent success stories of MD simulations in the investigation of membrane trafficking processes, their implications for future research, and the challenges that lie ahead in this specific research domain.
Collapse
Affiliation(s)
- Daniel Álvarez
- Department of Biology, University of Fribourg, Switzerland; Departamento de Química Física y Analítica, Universidad de Oviedo, Spain
| | - Jennifer Sapia
- Department of Biology, University of Fribourg, Switzerland
| | - Stefano Vanni
- Department of Biology, University of Fribourg, Switzerland; Université Côte d'Azur, Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.
| |
Collapse
|
13
|
Aguilera-Romero A, Lucena R, Sabido-Bozo S, Muñiz M. Impact of sphingolipids on protein membrane trafficking. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159334. [PMID: 37201864 DOI: 10.1016/j.bbalip.2023.159334] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/28/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
Membrane trafficking is essential to maintain the spatiotemporal control of protein and lipid distribution within membrane systems of eukaryotic cells. To achieve their functional destination proteins are sorted and transported into lipid carriers that construct the secretory and endocytic pathways. It is an emerging theme that lipid diversity might exist in part to ensure the homeostasis of these pathways. Sphingolipids, a chemical diverse type of lipids with special physicochemical characteristics have been implicated in the selective transport of proteins. In this review, we will discuss current knowledge about how sphingolipids modulate protein trafficking through the endomembrane systems to guarantee that proteins reach their functional destination and the proposed underlying mechanisms.
Collapse
Affiliation(s)
- Auxiliadora Aguilera-Romero
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain.
| | - Rafael Lucena
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Susana Sabido-Bozo
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Manuel Muñiz
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain.
| |
Collapse
|
14
|
Lackner K, Sailer S, van Klinken JB, Wever E, Pras-Raves ML, Dane AD, Honsho M, Abe Y, Keller MA, Golderer G, Werner-Felmayer G, Fujiki Y, Vaz FM, Werner ER, Watschinger K. Alterations in ether lipid metabolism and the consequences for the mouse lipidome. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159285. [PMID: 36690320 DOI: 10.1016/j.bbalip.2023.159285] [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: 09/13/2022] [Revised: 12/18/2022] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Alkylglycerol monooxygenase (AGMO) and plasmanylethanolamine desaturase (PEDS1) are enzymes involved in ether lipid metabolism. While AGMO degrades plasmanyl lipids by oxidative cleavage of the ether bond, PEDS1 exclusively synthesizes a specific subclass of ether lipids, the plasmalogens, by introducing a vinyl ether double bond into plasmanylethanolamine phospholipids. Ether lipids are characterized by an ether linkage at the sn-1 position of the glycerol backbone and they are found in membranes of different cell types. Decreased plasmalogen levels have been associated with neurological diseases like Alzheimer's disease. Agmo-deficient mice do not present an obvious phenotype under unchallenged conditions. In contrast, Peds1 knockout mice display a growth phenotype. To investigate the molecular consequences of Agmo and Peds1 deficiency on the mouse lipidome, five tissues from each mouse model were isolated and subjected to high resolution mass spectrometry allowing the characterization of up to 2013 lipid species from 42 lipid subclasses. Agmo knockout mice moderately accumulated plasmanyl and plasmenyl lipid species. Peds1-deficient mice manifested striking changes characterized by a strong reduction of plasmenyl lipids and a concomitant massive accumulation of plasmanyl lipids resulting in increased total ether lipid levels in the analyzed tissues except for the class of phosphatidylethanolamines where total levels remained remarkably constant also in Peds1 knockout mice. The rate-limiting enzyme in ether lipid metabolism, FAR1, was not upregulated in Peds1-deficient mice, indicating that the selective loss of plasmalogens is not sufficient to activate the feedback mechanism observed in total ether lipid deficiency.
Collapse
Affiliation(s)
- Katharina Lackner
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria.
| | - Sabrina Sailer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria; Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Strasse 1, 6020 Innsbruck, Austria.
| | - Jan-Bert van Klinken
- Amsterdam UMC location University of Amsterdam, Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Department of Human Genetics, Leiden University Medical Center (LUMC), Einthovenweg 20, Leiden, 2333, ZC, the Netherlands.
| | - Eric Wever
- Amsterdam UMC location University of Amsterdam, Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Bioinformatics Laboratory, Department of Epidemiology & Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands.
| | - Mia L Pras-Raves
- Amsterdam UMC location University of Amsterdam, Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Bioinformatics Laboratory, Department of Epidemiology & Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands.
| | - Adrie D Dane
- Amsterdam UMC location University of Amsterdam, Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Bioinformatics Laboratory, Department of Epidemiology & Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands.
| | - Masanori Honsho
- Department of Neuroinflammation and Brain Fatigue Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan.
| | - Yuichi Abe
- Faculty of Arts and Science, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan.
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Strasse 1, 6020 Innsbruck, Austria.
| | - Georg Golderer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria.
| | - Gabriele Werner-Felmayer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria.
| | - Yukio Fujiki
- Institute of Rheological Functions of Food, Kyushu University Collaboration Program, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan; Graduate School of Science, University of Hyogo, Hyogo, Japan.
| | - Frédéric M Vaz
- Amsterdam UMC location University of Amsterdam, Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, 1105, AZ, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam 1105, AZ, The Netherlands.
| | - Ernst R Werner
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria.
| | - Katrin Watschinger
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria.
| |
Collapse
|
15
|
Shaw JL, Pablo JL, Greka A. Mechanisms of Protein Trafficking and Quality Control in the Kidney and Beyond. Annu Rev Physiol 2023; 85:407-423. [PMID: 36763970 DOI: 10.1146/annurev-physiol-031522-100639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Numerous trafficking and quality control pathways evolved to handle the diversity of proteins made by eukaryotic cells. However, at every step along the biosynthetic pathway, there is the potential for quality control system failure. This review focuses on the mechanisms of disrupted proteostasis. Inspired by diseases caused by misfolded proteins in the kidney (mucin 1 and uromodulin), we outline the general principles of protein biosynthesis, delineate the recognition and degradation pathways targeting misfolded proteins, and discuss the role of cargo receptors in protein trafficking and lipid homeostasis. We also discuss technical approaches including live-cell fluorescent microscopy, chemical screens to elucidate trafficking mechanisms, multiplexed single-cell CRISPR screening platforms to systematically delineate mechanisms of proteostasis, and the advancement of novel tools to degrade secretory and membrane-associated proteins. By focusing on components of trafficking that go awry, we highlight ongoing efforts to understand fundamental mechanisms of disrupted proteostasis and implications for the treatment of human proteinopathies.
Collapse
Affiliation(s)
- Jillian L Shaw
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; .,Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Juan Lorenzo Pablo
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; .,Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; .,Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Tran ML, Kim Y, von Blume J. Quantification of Protein Exit at the Trans-Golgi Network. Methods Mol Biol 2023; 2557:583-594. [PMID: 36512239 DOI: 10.1007/978-1-0716-2639-9_35] [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] [Indexed: 12/15/2022]
Abstract
With one-third of all newly synthesized proteins entering the secretory pathway, correct protein sorting is essential for cellular homeostasis. In the last three decades, researchers have developed numerous biochemical, genetic, and cell biological approaches to study protein export and sorting from the trans-Golgi network (TGN). However, accurately quantifying protein transport from one compartment to the next in the secretory pathway has been challenging. The Retention Using Selective Hooks (RUSH) system is a method that allows monitoring trafficking of a protein of interest in real time, similar to a pulse-chase experiment but without the need of radiolabeling. Accurate calculations, however, are necessary and currently lacking. Here, we combine the RUSH system with live cell imaging to quantify and calculate half lives. We exemplify our approach using a soluble secreted protein (LyzC). This system will benefit membrane trafficking researchers by adding numbers to protein export and comparing the export kinetics of different cargoes and variating conditions.
Collapse
Affiliation(s)
- Mai Ly Tran
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Yeongho Kim
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Julia von Blume
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
17
|
Griess K, Rieck M, Müller N, Karsai G, Hartwig S, Pelligra A, Hardt R, Schlegel C, Kuboth J, Uhlemeyer C, Trenkamp S, Jeruschke K, Weiss J, Peifer-Weiss L, Xu W, Cames S, Yi X, Cnop M, Beller M, Stark H, Kondadi AK, Reichert AS, Markgraf D, Wammers M, Häussinger D, Kuss O, Lehr S, Eizirik D, Lickert H, Lammert E, Roden M, Winter D, Al-Hasani H, Höglinger D, Hornemann T, Brüning JC, Belgardt BF. Sphingolipid subtypes differentially control proinsulin processing and systemic glucose homeostasis. Nat Cell Biol 2023; 25:20-29. [PMID: 36543979 PMCID: PMC9859757 DOI: 10.1038/s41556-022-01027-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 10/11/2022] [Indexed: 12/24/2022]
Abstract
Impaired proinsulin-to-insulin processing in pancreatic β-cells is a key defective step in both type 1 diabetes and type 2 diabetes (T2D) (refs. 1,2), but the mechanisms involved remain to be defined. Altered metabolism of sphingolipids (SLs) has been linked to development of obesity, type 1 diabetes and T2D (refs. 3-8); nonetheless, the role of specific SL species in β-cell function and demise is unclear. Here we define the lipid signature of T2D-associated β-cell failure, including an imbalance of specific very-long-chain SLs and long-chain SLs. β-cell-specific ablation of CerS2, the enzyme necessary for generation of very-long-chain SLs, selectively reduces insulin content, impairs insulin secretion and disturbs systemic glucose tolerance in multiple complementary models. In contrast, ablation of long-chain-SL-synthesizing enzymes has no effect on insulin content. By quantitatively defining the SL-protein interactome, we reveal that CerS2 ablation affects SL binding to several endoplasmic reticulum-Golgi transport proteins, including Tmed2, which we define as an endogenous regulator of the essential proinsulin processing enzyme Pcsk1. Our study uncovers roles for specific SL subtypes and SL-binding proteins in β-cell function and T2D-associated β-cell failure.
Collapse
Affiliation(s)
- Kerstin Griess
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Michael Rieck
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Nadine Müller
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Gergely Karsai
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
- Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Sonja Hartwig
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Angela Pelligra
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Robert Hardt
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Caroline Schlegel
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Jennifer Kuboth
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Celina Uhlemeyer
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Sandra Trenkamp
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Kay Jeruschke
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jürgen Weiss
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Leon Peifer-Weiss
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Weiwei Xu
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Sandra Cames
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Mathias Beller
- Institute for Mathematical Modeling of Biological Systems and Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Arun Kumar Kondadi
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Markgraf
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marianne Wammers
- Department of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oliver Kuss
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Centre for Health and Society, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefan Lehr
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Decio Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
- Welbio, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Heiko Lickert
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Eckhard Lammert
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Hadi Al-Hasani
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Doris Höglinger
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Thorsten Hornemann
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
- Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Bengt-Frederik Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
| |
Collapse
|
18
|
Feng MW, Delneri D, Millar CB, O'Keefe RT. Eisosome disruption by noncoding RNA deletion increases protein secretion in yeast. PNAS NEXUS 2022; 1:pgac241. [PMID: 36712349 PMCID: PMC9802208 DOI: 10.1093/pnasnexus/pgac241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Noncoding RNAs (ncRNAs) regulate many aspects of gene expression. We investigated how ncRNAs affected protein secretion in yeast by large-scale screening for improved endogenous invertase secretion in ncRNA deletion strains with deletion of stable unannotated transcripts (SUTs), cryptic unstable transcripts (CUTs), tRNAs, or snRNAs. We identified three candidate ncRNAs, SUT418, SUT390, and SUT125, that improved endogenous invertase secretion when deleted. As SUTs can affect expression of nearby genes, we quantified adjacent gene transcription and found that the PIL1 gene was down-regulated in the SUT125 deletion strain. Pil1 is a core component of eisosomes, nonmobile invaginations found throughout the plasma membrane. PIL1 knockout alone, or in combination with eisosome components LSP1 or SUR7, resulted in further increased secretion of invertase. Secretion of heterologous GFP was also increased upon PIL1 deletion, but this increase was signal sequence dependent. To reveal the potential for increased biopharmaceutical production, secretion of monoclonal antibody Pexelizumab scFv peptide was increased by PIL1 deletion. Global analysis of secreted proteins revealed that approximately 20% of secreted proteins, especially serine-enriched secreted proteins, including invertase, were increased upon eisosome disruption. Eisosomes are enriched with APC transporters and sphingolipids, which are essential components for secretory vesicle formation and protein sorting. Sphingolipid and serine biosynthesis pathways were up-regulated upon PIL1 deletion. We propose that increased secretion of endogenous and heterologous proteins upon PIL1 deletion resulted from sphingolipid redistribution in the plasma membrane and up-regulated sphingolipid biosynthesis. Overall, a new pathway to improve protein secretion in yeast via eisosome disruption has been identified.
Collapse
Affiliation(s)
- Matthew Wenjie Feng
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Daniela Delneri
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PT, UK,Manchester Institute of Biotechnology, Faculty of Biology Medicine and Health, The University of Manchester, 131 Princess street, Manchester, M1 7DN, UK
| | - Catherine B Millar
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | | |
Collapse
|
19
|
Anwar MU, Sergeeva OA, Abrami L, Mesquita FS, Lukonin I, Amen T, Chuat A, Capolupo L, Liberali P, D'Angelo G, van der Goot FG. ER-Golgi-localized proteins TMED2 and TMED10 control the formation of plasma membrane lipid nanodomains. Dev Cell 2022; 57:2334-2346.e8. [PMID: 36174556 DOI: 10.1016/j.devcel.2022.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/24/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022]
Abstract
To promote infections, pathogens exploit host cell machineries such as structural elements of the plasma membrane. Studying these interactions and identifying molecular players are ideal for gaining insights into the fundamental biology of the host cell. Here, we used the anthrax toxin to screen a library of 1,500 regulatory, cell-surface, and membrane trafficking genes for their involvement in the intoxication process. We found that endoplasmic reticulum (ER)-Golgi-localized proteins TMED2 and TMED10 are required for toxin oligomerization at the plasma membrane of human cells, an essential step dependent on localization to cholesterol-rich lipid nanodomains. Biochemical, morphological, and mechanistic analyses showed that TMED2 and TMED10 are essential components of a supercomplex that operates the exchange of both cholesterol and ceramides at ER-Golgi membrane contact sites. Overall, this study of anthrax intoxication led to the discovery that lipid compositional remodeling at ER-Golgi interfaces fully controls the formation of functional membrane nanodomains at the cell surface.
Collapse
Affiliation(s)
- Muhammad U Anwar
- Global Health Institute, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Oksana A Sergeeva
- Global Health Institute, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Laurence Abrami
- Global Health Institute, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Francisco S Mesquita
- Global Health Institute, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Ilya Lukonin
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; University of Basel, 4056 Basel, Switzerland
| | - Triana Amen
- Global Health Institute, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Audrey Chuat
- Global Health Institute, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Laura Capolupo
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; University of Basel, 4056 Basel, Switzerland
| | - Giovanni D'Angelo
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland.
| | - F Gisou van der Goot
- Global Health Institute, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland.
| |
Collapse
|
20
|
Petrenko V, Sinturel F, Loizides-Mangold U, Montoya JP, Chera S, Riezman H, Dibner C. Type 2 diabetes disrupts circadian orchestration of lipid metabolism and membrane fluidity in human pancreatic islets. PLoS Biol 2022; 20:e3001725. [PMID: 35921354 PMCID: PMC9348689 DOI: 10.1371/journal.pbio.3001725] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/24/2022] [Indexed: 11/18/2022] Open
Abstract
Recent evidence suggests that circadian clocks ensure temporal orchestration of lipid homeostasis and play a role in pathophysiology of metabolic diseases in humans, including type 2 diabetes (T2D). Nevertheless, circadian regulation of lipid metabolism in human pancreatic islets has not been explored. Employing lipidomic analyses, we conducted temporal profiling in human pancreatic islets derived from 10 nondiabetic (ND) and 6 T2D donors. Among 329 detected lipid species across 8 major lipid classes, 5% exhibited circadian rhythmicity in ND human islets synchronized in vitro. Two-time point-based lipidomic analyses in T2D human islets revealed global and temporal alterations in phospho- and sphingolipids. Key enzymes regulating turnover of sphingolipids were rhythmically expressed in ND islets and exhibited altered levels in ND islets bearing disrupted clocks and in T2D islets. Strikingly, cellular membrane fluidity, measured by a Nile Red derivative NR12S, was reduced in plasma membrane of T2D diabetic human islets, in ND donors’ islets with disrupted circadian clockwork, or treated with sphingolipid pathway modulators. Moreover, inhibiting the glycosphingolipid biosynthesis led to strong reduction of insulin secretion triggered by glucose or KCl, whereas inhibiting earlier steps of de novo ceramide synthesis resulted in milder inhibitory effect on insulin secretion by ND islets. Our data suggest that circadian clocks operative in human pancreatic islets are required for temporal orchestration of lipid homeostasis, and that perturbation of temporal regulation of the islet lipid metabolism upon T2D leads to altered insulin secretion and membrane fluidity. These phenotypes were recapitulated in ND islets bearing disrupted clocks.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Flore Sinturel
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Ursula Loizides-Mangold
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Jonathan Paz Montoya
- Proteomics Core Facility, EPFL, Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Simona Chera
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
- * E-mail:
| |
Collapse
|
21
|
Xue M, Zhang T, Cheng ZJ, Guo B, Zeng Y, Lin R, Zheng P, Liu M, Hu F, Li F, Zhang W, Li L, Zhao Q, Sun B, Tang X. Effect of a Functional Phospholipid Metabolome-Protein Association Pathway on the Mechanism of COVID-19 Disease Progression. Int J Biol Sci 2022; 18:4618-4628. [PMID: 35874944 PMCID: PMC9305269 DOI: 10.7150/ijbs.72450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
This study aimed to explore the clinical practice of phospholipid metabolic pathways in COVID-19. In this study, 48 COVID-19 patients and 17 healthy controls were included. Patients were divided into mild (n=40) and severe (n=8) according to their severity. Phospholipid metabolites, TCA circulating metabolites, eicosanoid metabolites, and closely associated enzymes and transfer proteins were detected in the plasma of all individuals using metabolomics and proteomics assays, respectively. 30 of the 33 metabolites found differed significantly (P<0.05) between patients and healthy controls (P<0.05), with D-dimmer significantly correlated with all of the lysophospholipid metabolites (LysoPE, LysoPC, LysoPI and LPA). In particular, we found that phosphatidylinositol (PI) and phosphatidylcholine (PC) could identify patients from healthy controls (AUC 0.771 and 0.745, respectively) and that the severity of the patients could be determined (AUC 0.663 and 0.809, respectively). The last measurement before discharge also revealed significant changes in both PI and PC. For the first time, our study explores the significance of the phospholipid metabolic system in COVID-19 patients. Based on molecular pathway mechanisms, three important phospholipid pathways related to Ceramide-Malate acid (Cer-SM), Lysophospholipid (LPs), and membrane function were established. Clinical values discovered included the role of Cer in maintaining the inflammatory internal environment, the modulation of procoagulant LPA by upstream fibrinolytic metabolites, and the role of PI and PC in predicting disease aggravation.
Collapse
Affiliation(s)
- Mingshan Xue
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China.,Guangzhou Laboratory, XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Teng Zhang
- MoE Frontiers Science Center for Precision Oncology, Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau. Taipa, Macau, China
| | - Zhangkai J Cheng
- Guangzhou Laboratory, XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Baojun Guo
- Guangzhou Laboratory, XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Yifeng Zeng
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Runpei Lin
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Peiyan Zheng
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Mingtao Liu
- Guangzhou Laboratory, XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Fengyu Hu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Feng Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Wensheng Zhang
- Institue of automation Chinese Academy of Sciences, Beijing, China
| | - Lu Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau. Taipa, Macau, China
| | - Baoqing Sun
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Xiaoping Tang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China.,Guangzhou Laboratory, XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| |
Collapse
|
22
|
Mir SA, Chen L, Burugupalli S, Burla B, Ji S, Smith AAT, Narasimhan K, Ramasamy A, Tan KML, Huynh K, Giles C, Mei D, Wong G, Yap F, Tan KH, Collier F, Saffery R, Vuillermin P, Bendt AK, Burgner D, Ponsonby AL, Lee YS, Chong YS, Gluckman PD, Eriksson JG, Meikle PJ, Wenk MR, Karnani N. Population-based plasma lipidomics reveals developmental changes in metabolism and signatures of obesity risk: a mother-offspring cohort study. BMC Med 2022; 20:242. [PMID: 35871677 PMCID: PMC9310480 DOI: 10.1186/s12916-022-02432-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Lipids play a vital role in health and disease, but changes to their circulating levels and the link with obesity remain poorly characterized in expecting mothers and their offspring in early childhood. METHODS LC-MS/MS-based quantitation of 480 lipid species was performed on 2491 plasma samples collected at 4 time points in the mother-offspring Asian cohort GUSTO (Growing Up in Singapore Towards healthy Outcomes). These 4 time points constituted samples collected from mothers at 26-28 weeks of gestation (n=752) and 4-5 years postpartum (n=650), and their offspring at birth (n=751) and 6 years of age (n=338). Linear regression models were used to identify the pregnancy and developmental age-specific variations in the plasma lipidomic profiles, and their association with obesity risk. An independent birth cohort (n=1935), the Barwon Infant Study (BIS), comprising mother-offspring dyads of Caucasian origin was used for validation. RESULTS Levels of 36% of the profiled lipids were significantly higher (absolute fold change > 1.5 and Padj < 0.05) in antenatal maternal circulation as compared to the postnatal phase, with phosphatidylethanolamine levels changing the most. Compared to antenatal maternal lipids, cord blood showed lower concentrations of most lipid species (79%) except lysophospholipids and acylcarnitines. Changes in lipid concentrations from birth to 6 years of age were much higher in magnitude (log2FC=-2.10 to 6.25) than the changes observed between a 6-year-old child and an adult (postnatal mother) (log2FC=-0.68 to 1.18). Associations of cord blood lipidomic profiles with birth weight displayed distinct trends compared to the lipidomic profiles associated with child BMI at 6 years. Comparison of the results between the child and adult BMI identified similarities in association with consistent trends (R2=0.75). However, large number of lipids were associated with BMI in adults (67%) compared to the children (29%). Pre-pregnancy BMI was specifically associated with decrease in the levels of phospholipids, sphingomyelin, and several triacylglycerol species in pregnancy. CONCLUSIONS In summary, our study provides a detailed landscape of the in utero lipid environment provided by the gestating mother to the growing fetus, and the magnitude of changes in plasma lipidomic profiles from birth to early childhood. We identified the effects of adiposity on the circulating lipid levels in pregnant and non-pregnant women as well as offspring at birth and at 6 years of age. Additionally, the pediatric vs maternal overlap of the circulating lipid phenotype of obesity risk provides intergenerational insights and early opportunities to track and intervene the onset of metabolic adversities. CLINICAL TRIAL REGISTRATION This birth cohort is a prospective observational study, which was registered on 1 July 2010 under the identifier NCT01174875 .
Collapse
Affiliation(s)
- Sartaj Ahmad Mir
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.,Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Li Chen
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore
| | - Satvika Burugupalli
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Bo Burla
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Shanshan Ji
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Adam Alexander T Smith
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Kothandaraman Narasimhan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore
| | - Adaikalavan Ramasamy
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore
| | - Karen Mei-Ling Tan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Corey Giles
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Ding Mei
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Gerard Wong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore
| | - Fabian Yap
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Kok Hian Tan
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Fiona Collier
- School of Medicine, Deakin University, Geelong, Australia.,Child Health Research Unit, Barwon Health, Geelong, Australia.,Murdoch Children's Research Institute, University of Melbourne, Parkville, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, University of Melbourne, Parkville, Australia.,The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Peter Vuillermin
- School of Medicine, Deakin University, Geelong, Australia.,Child Health Research Unit, Barwon Health, Geelong, Australia.,Murdoch Children's Research Institute, University of Melbourne, Parkville, Australia
| | - Anne K Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - David Burgner
- Murdoch Children's Research Institute, University of Melbourne, Parkville, Australia.,The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, University of Melbourne, Parkville, Australia.,The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore.,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore.,Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Folkhalsan Research Center, Helsinki, Finland.,Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia.
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore. .,Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Neerja Karnani
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore. .,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore, 117609, Singapore. .,DataHub Division, Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
23
|
Scrima S, Tiberti M, Campo A, Corcelle-Termeau E, Judith D, Foged MM, Clemmensen KKB, Tooze SA, Jäättelä M, Maeda K, Lambrughi M, Papaleo E. Unraveling membrane properties at the organelle-level with LipidDyn. Comput Struct Biotechnol J 2022; 20:3604-3614. [PMID: 35860415 PMCID: PMC9283888 DOI: 10.1016/j.csbj.2022.06.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 12/22/2022] Open
Abstract
Cellular membranes are formed from different lipids in various amounts and proportions depending on the subcellular localization. The lipid composition of membranes is sensitive to changes in the cellular environment, and its alterations are linked to several diseases. Lipids not only form lipid-lipid interactions but also interact with other biomolecules, including proteins. Molecular dynamics (MD) simulations are a powerful tool to study the properties of cellular membranes and membrane-protein interactions on different timescales and resolutions. Over the last few years, software and hardware for biomolecular simulations have been optimized to routinely run long simulations of large and complex biological systems. On the other hand, high-throughput techniques based on lipidomics provide accurate estimates of the composition of cellular membranes at the level of subcellular compartments. Lipidomic data can be analyzed to design biologically relevant models of membranes for MD simulations. Similar applications easily result in a massive amount of simulation data where the bottleneck becomes the analysis of the data. In this context, we developed LipidDyn, a Python-based pipeline to streamline the analyses of MD simulations of membranes of different compositions. Once the simulations are collected, LipidDyn provides average properties and time series for several membrane properties such as area per lipid, thickness, order parameters, diffusion motions, lipid density, and lipid enrichment/depletion. The calculations exploit parallelization, and the pipeline includes graphical outputs in a publication-ready form. We applied LipidDyn to different case studies to illustrate its potential, including membranes from cellular compartments and transmembrane protein domains. LipidDyn is available free of charge under the GNU General Public License from https://github.com/ELELAB/LipidDyn.
Collapse
Affiliation(s)
- Simone Scrima
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, 2800 Lyngby, Denmark
| | - Matteo Tiberti
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Alessia Campo
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Elisabeth Corcelle-Termeau
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Delphine Judith
- Institut Cochin, Inserm U1016-CNRS, UMR8104, Université de Paris, Paris, France
| | - Mads Møller Foged
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | | | - Sharon A. Tooze
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Marja Jäättelä
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kenji Maeda
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Matteo Lambrughi
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Elena Papaleo
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, 2800 Lyngby, Denmark
| |
Collapse
|
24
|
A set of gene knockouts as a resource for global lipidomic changes. Sci Rep 2022; 12:10533. [PMID: 35732804 PMCID: PMC9218125 DOI: 10.1038/s41598-022-14690-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/10/2022] [Indexed: 11/14/2022] Open
Abstract
Enzyme specificity in lipid metabolic pathways often remains unresolved at the lipid species level, which is needed to link lipidomic molecular phenotypes with their protein counterparts to construct functional pathway maps. We created lipidomic profiles of 23 gene knockouts in a proof-of-concept study based on a CRISPR/Cas9 knockout screen in mammalian cells. This results in a lipidomic resource across 24 lipid classes. We highlight lipid species phenotypes of multiple knockout cell lines compared to a control, created by targeting the human safe-harbor locus AAVS1 using up to 1228 lipid species and subspecies, charting lipid metabolism at the molecular level. Lipid species changes are found in all knockout cell lines, however, some are most apparent on the lipid class level (e.g., SGMS1 and CEPT1), while others are most apparent on the fatty acid level (e.g., DECR2 and ACOT7). We find lipidomic phenotypes to be reproducible across different clones of the same knockout and we observed similar phenotypes when two enzymes that catalyze subsequent steps of the long-chain fatty acid elongation cycle were targeted.
Collapse
|
25
|
Girik V, Feng S, Hariri H, Henne WM, Riezman H. Vacuole-Specific Lipid Release for Tracking Intracellular Lipid Metabolism and Transport in Saccharomyces cerevisiae. ACS Chem Biol 2022; 17:1485-1494. [PMID: 35667650 PMCID: PMC9207805 DOI: 10.1021/acschembio.2c00120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/02/2022] [Indexed: 11/30/2022]
Abstract
Lipid metabolism is spatiotemporally regulated within cells, yet intervention into lipid functions at subcellular resolution remains difficult. Here, we report a method that enables site-specific release of sphingolipids and cholesterol inside the vacuole in Saccharomyces cerevisiae. Using this approach, we monitored real-time sphingolipid metabolic flux out of the vacuole by mass spectrometry and found that the endoplasmic reticulum-vacuole-tethering protein Mdm1 facilitated the metabolism of sphingoid bases into ceramides. In addition, we showed that cholesterol, once delivered into yeast using our method, could restore cell proliferation induced by ergosterol deprivation, overcoming the previously described sterol-uptake barrier under aerobic conditions. Together, these data define a new way to study intracellular lipid metabolism and transport from the vacuole in yeast.
Collapse
Affiliation(s)
- Vladimir Girik
- Department
of Biochemistry, University of Geneva, Geneva 1205, Switzerland
| | - Suihan Feng
- Department
of Biochemistry, University of Geneva, Geneva 1205, Switzerland
- National
Centre of Competence in Research (NCCR) in Chemical Biology, University of Geneva, Geneva 1205, Switzerland
| | - Hanaa Hariri
- Department
of Cell Biology, UT Southwestern Medical
Center, Dallas, Texas 75390-9039 United States
| | - W. Mike Henne
- Department
of Cell Biology, UT Southwestern Medical
Center, Dallas, Texas 75390-9039 United States
| | - Howard Riezman
- Department
of Biochemistry, University of Geneva, Geneva 1205, Switzerland
| |
Collapse
|
26
|
Sailer S, Lackner K, Pras-Raves ML, Wever EJM, van Klinken JB, Dane AD, Geley S, Koch J, Golderer G, Werner-Felmayer G, Keller MA, Zwerschke W, Vaz FM, Werner ER, Watschinger K. Adaptations of the 3T3-L1 adipocyte lipidome to defective ether lipid catabolism upon Agmo knockdown. J Lipid Res 2022; 63:100222. [PMID: 35537527 PMCID: PMC9192799 DOI: 10.1016/j.jlr.2022.100222] [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: 12/02/2021] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 10/24/2022] Open
Abstract
Little is known about the physiological role of alkylglycerol monooxygenase (AGMO), the only enzyme capable of cleaving the 1-O-alkyl ether bond of ether lipids. Expression and enzymatic activity of this enzyme can be detected in a variety of tissues including adipose tissue. This labile lipolytic membrane-bound protein uses tetrahydrobiopterin as a cofactor, and mice with reduced tetrahydrobiopterin levels have alterations in body fat distribution and blood lipid concentrations. In addition, manipulation of AGMO in macrophages led to significant changes in the cellular lipidome, and alkylglycerolipids, the preferred substrates of AGMO, were shown to accumulate in mature adipocytes. Here, we investigated the roles of AGMO in lipid metabolism by studying 3T3-L1 adipogenesis. AGMO activity was induced over 11 days using an adipocyte differentiation protocol. We show that RNA interference-mediated knockdown of AGMO did not interfere with adipocyte differentiation or affect lipid droplet formation. Furthermore, lipidomics revealed that plasmalogen phospholipids were preferentially accumulated upon Agmo knockdown, and a significant shift toward longer and more polyunsaturated acyl side chains of diacylglycerols and triacylglycerols could be detected by mass spectrometry. Our results indicate that alkylglycerol catabolism has an influence not only on ether-linked species but also on the degree of unsaturation in the massive amounts of triacylglycerols formed during in vitro 3T3-L1 adipocyte differentiation.
Collapse
Affiliation(s)
- Sabrina Sailer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Lackner
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Mia L Pras-Raves
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry and Pediatrics, Emma Children's Hospital, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Bioinformatics Laboratory, Department of Epidemiology & Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Eric J M Wever
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry and Pediatrics, Emma Children's Hospital, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Bioinformatics Laboratory, Department of Epidemiology & Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Jan B van Klinken
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry and Pediatrics, Emma Children's Hospital, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan D Dane
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry and Pediatrics, Emma Children's Hospital, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Bioinformatics Laboratory, Department of Epidemiology & Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Stephan Geley
- Institute of Molecular Pathophysiology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob Koch
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Golderer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Werner-Felmayer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Werner Zwerschke
- Division of Cell Metabolism and Differentiation Research, Research Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry and Pediatrics, Emma Children's Hospital, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, The Netherlands
| | - Ernst R Werner
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Katrin Watschinger
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
27
|
Rodriguez-Gallardo S, Sabido-Bozo S, Ikeda A, Araki M, Okazaki K, Nakano M, Aguilera-Romero A, Cortes-Gomez A, Lopez S, Waga M, Nakano A, Kurokawa K, Muñiz M, Funato K. Quality-controlled ceramide-based GPI-anchored protein sorting into selective ER exit sites. Cell Rep 2022; 39:110768. [PMID: 35508142 DOI: 10.1016/j.celrep.2022.110768] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/23/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Glycosylphosphatidylinositol-anchored proteins (GPI-APs) exit the endoplasmic reticulum (ER) through a specialized export pathway in the yeast Saccharomyces cerevisiae. We have recently shown that a very-long acyl chain (C26) ceramide present in the ER membrane drives clustering and sorting of GPI-APs into selective ER exit sites (ERES). Now, we show that this lipid-based ER sorting also involves the C26 ceramide as a lipid moiety of GPI-APs, which is incorporated into the GPI anchor through a lipid-remodeling process after protein attachment in the ER. Moreover, we also show that a GPI-AP with a C26 ceramide moiety is monitored by the GPI-glycan remodelase Ted1, which, in turn, is required for receptor-mediated export of GPI-APs. Therefore, our study reveals a quality-control system that ensures lipid-based sorting of GPI-APs into selective ERESs for differential ER export, highlighting the physiological need for this specific export pathway.
Collapse
Affiliation(s)
- Sofia Rodriguez-Gallardo
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Susana Sabido-Bozo
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Atsuko Ikeda
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Misako Araki
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Kouta Okazaki
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Auxiliadora Aguilera-Romero
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Alejandro Cortes-Gomez
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Sergio Lopez
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Miho Waga
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan
| | - Kazuo Kurokawa
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan.
| | - Manuel Muñiz
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain.
| | - Kouichi Funato
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan.
| |
Collapse
|
28
|
Space- and Time-Resolved Metabolomics of a High-Grade Serous Ovarian Cancer Mouse Model. Cancers (Basel) 2022; 14:cancers14092262. [PMID: 35565391 PMCID: PMC9104348 DOI: 10.3390/cancers14092262] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/10/2022] Open
Abstract
The dismally low survival rate of ovarian cancer patients diagnosed with high-grade serous carcinoma (HGSC) emphasizes the lack of effective screening strategies. One major obstacle is the limited knowledge of the underlying mechanisms of HGSC pathogenesis at very early stages. Here, we present the first 10-month time-resolved serum metabolic profile of a triple mutant (TKO) HGSC mouse model, along with the spatial lipidome profile of its entire reproductive system. A high-coverage liquid chromatography mass spectrometry-based metabolomics approach was applied to longitudinally collected serum samples from both TKO (n = 15) and TKO control mice (n = 15), tracking metabolome and lipidome changes from premalignant stages to tumor initiation, early stages, and advanced stages until mouse death. Time-resolved analysis showed specific temporal trends for 17 lipid classes, amino acids, and TCA cycle metabolites, associated with HGSC progression. Spatial lipid distributions within the reproductive system were also mapped via ultrahigh-resolution matrix-assisted laser desorption/ionization (MALDI) mass spectrometry and compared with serum lipid profiles for various lipid classes. Altogether, our results show that the remodeling of lipid and fatty acid metabolism, amino acid biosynthesis, TCA cycle and ovarian steroidogenesis are critical components of HGSC onset and development. These metabolic alterations are accompanied by changes in energy metabolism, mitochondrial and peroxisomal function, redox homeostasis, and inflammatory response, collectively supporting tumorigenesis.
Collapse
|
29
|
Blomme A, Peter C, Mui E, Rodriguez Blanco G, An N, Mason LM, Jamieson LE, McGregor GH, Lilla S, Ntala C, Patel R, Thiry M, Kung SHY, Leclercq M, Ford CA, Rushworth LK, McGarry DJ, Mason S, Repiscak P, Nixon C, Salji MJ, Markert E, MacKay GM, Kamphorst JJ, Graham D, Faulds K, Fazli L, Gleave ME, Avezov E, Edwards J, Yin H, Sumpton D, Blyth K, Close P, Murphy DJ, Zanivan S, Leung HY. THEM6-mediated reprogramming of lipid metabolism supports treatment resistance in prostate cancer. EMBO Mol Med 2022; 14:e14764. [PMID: 35014179 PMCID: PMC8899912 DOI: 10.15252/emmm.202114764] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/21/2022] Open
Abstract
Despite the clinical benefit of androgen-deprivation therapy (ADT), the majority of patients with advanced prostate cancer (PCa) ultimately develop lethal castration-resistant prostate cancer (CRPC). In this study, we identified thioesterase superfamily member 6 (THEM6) as a marker of ADT resistance in PCa. THEM6 deletion reduces in vivo tumour growth and restores castration sensitivity in orthograft models of CRPC. Mechanistically, we show that the ER membrane-associated protein THEM6 regulates intracellular levels of ether lipids and is essential to trigger the induction of the ER stress response (UPR). Consequently, THEM6 loss in CRPC cells significantly alters ER function, reducing de novo sterol biosynthesis and preventing lipid-mediated activation of ATF4. Finally, we demonstrate that high THEM6 expression is associated with poor survival and correlates with high levels of UPR activation in PCa patients. Altogether, our results highlight THEM6 as a novel driver of therapy resistance in PCa as well as a promising target for the treatment of CRPC.
Collapse
Affiliation(s)
| | | | - Ernest Mui
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | | | - Ning An
- Laboratory of Cancer SignalingGIGA‐InstituteUniversity of LiègeLiègeBelgium
| | | | - Lauren E Jamieson
- Centre for Molecular NanometrologyDepartment of Pure and Applied ChemistryTechnology and Innovation CentreUniversity of StrathclydeGlasgowUK
| | - Grace H McGregor
- CRUK Beatson InstituteGarscube EstateGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | | | - Chara Ntala
- CRUK Beatson InstituteGarscube EstateGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | | | - Marc Thiry
- GIGA‐NeurosciencesUnit of Cell and Tissue BiologyUniversity of LiègeLiègeBelgium
| | - Sonia H Y Kung
- Department of Urologic SciencesFaculty of MedicineUniversity of British ColumbiaVancouverBCCanada
- Vancouver Prostate CentreVancouverBCCanada
| | - Marine Leclercq
- Laboratory of Cancer SignalingGIGA‐InstituteUniversity of LiègeLiègeBelgium
| | | | - Linda K Rushworth
- CRUK Beatson InstituteGarscube EstateGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | | | - Susan Mason
- CRUK Beatson InstituteGarscube EstateGlasgowUK
| | | | - Colin Nixon
- CRUK Beatson InstituteGarscube EstateGlasgowUK
| | - Mark J Salji
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | - Elke Markert
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | | | - Jurre J Kamphorst
- CRUK Beatson InstituteGarscube EstateGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | - Duncan Graham
- Centre for Molecular NanometrologyDepartment of Pure and Applied ChemistryTechnology and Innovation CentreUniversity of StrathclydeGlasgowUK
| | - Karen Faulds
- Centre for Molecular NanometrologyDepartment of Pure and Applied ChemistryTechnology and Innovation CentreUniversity of StrathclydeGlasgowUK
| | - Ladan Fazli
- Department of Urologic SciencesFaculty of MedicineUniversity of British ColumbiaVancouverBCCanada
- Vancouver Prostate CentreVancouverBCCanada
| | - Martin E Gleave
- Department of Urologic SciencesFaculty of MedicineUniversity of British ColumbiaVancouverBCCanada
- Vancouver Prostate CentreVancouverBCCanada
| | - Edward Avezov
- UK Dementia Research Institute at University of CambridgeDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Joanne Edwards
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | - Huabing Yin
- School of EngineeringUniversity of GlasgowGlasgowUK
| | | | - Karen Blyth
- CRUK Beatson InstituteGarscube EstateGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | - Pierre Close
- Laboratory of Cancer SignalingGIGA‐InstituteUniversity of LiègeLiègeBelgium
| | - Daniel J Murphy
- CRUK Beatson InstituteGarscube EstateGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | - Sara Zanivan
- CRUK Beatson InstituteGarscube EstateGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| | - Hing Y Leung
- CRUK Beatson InstituteGarscube EstateGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGarscube EstateGlasgowUK
| |
Collapse
|
30
|
Need for more focus on lipid species in studies of biological and model membranes. Prog Lipid Res 2022; 86:101160. [DOI: 10.1016/j.plipres.2022.101160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 03/06/2022] [Indexed: 11/23/2022]
|
31
|
Körner C, Fröhlich F. Compartmentation and functions of sphingolipids. Curr Opin Cell Biol 2022; 74:104-111. [DOI: 10.1016/j.ceb.2022.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/13/2022] [Accepted: 01/22/2022] [Indexed: 02/08/2023]
|
32
|
Plasma membrane effects of sphingolipid-synthesis inhibition by myriocin in CHO cells: a biophysical and lipidomic study. Sci Rep 2022; 12:955. [PMID: 35046440 PMCID: PMC8770663 DOI: 10.1038/s41598-021-04648-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/08/2021] [Indexed: 11/08/2022] Open
Abstract
Suppression of a specific gene effect can be achieved by genetic as well as chemical methods. Each approach may hide unexpected drawbacks, usually in the form of side effects. In the present study, the specific inhibitor myriocin was used to block serine palmitoyltransferase (SPT), the first enzyme in the sphingolipid synthetic pathway, in CHO cells. The subsequent biophysical changes in plasma membranes were measured and compared with results obtained with a genetically modified CHO cell line containing a defective SPT (the LY-B cell line). Similar effects were observed with both approaches: sphingomyelin values were markedly decreased in myriocin-treated CHO cells and, in consequence, their membrane molecular order (measured as laurdan general polarization) and mechanical resistance (AFM-measured breakthrough force values) became lower than in the native, non-treated cells. Cells treated with myriocin reacted homeostatically to maintain membrane order, synthesizing more fully saturated and less polyunsaturated GPL than the non-treated ones, although they achieved it only partially, their plasma membranes remaining slightly more fluid and more penetrable than those from the control cells. The good agreement between results obtained with very different tools, such as genetically modified and chemically treated cells, reinforces the use of both methods and demonstrates that both are adequate for their intended use, i.e. the complete and specific inhibition of sphingolipid synthesis in CHO cells, without apparent unexpected effects.
Collapse
|
33
|
Assies L, García-Calvo J, Piazzolla F, Sanchez S, Kato T, Reymond L, Goujon A, Colom A, López-Andarias J, Straková K, Mahecic D, Mercier V, Riggi M, Jiménez-Rojo N, Roffay C, Licari G, Tsemperouli M, Neuhaus F, Fürstenberg A, Vauthey E, Hoogendoorn S, Gonzalez-Gaitan M, Zumbuehl A, Sugihara K, Gruenberg J, Riezman H, Loewith R, Manley S, Roux A, Winssinger N, Sakai N, Pitsch S, Matile S. Flipper Probes for the Community. Chimia (Aarau) 2021; 75:1004-1011. [PMID: 34920768 DOI: 10.2533/chimia.2021.1004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
This article describes four fluorescent membrane tension probes that have been designed, synthesized, evaluated, commercialized and applied to current biology challenges in the context of the NCCR Chemical Biology. Their names are Flipper-TR®, ER Flipper-TR®, Lyso Flipper-TR®, and Mito Flipper-TR®. They are available from Spirochrome.
Collapse
Affiliation(s)
- Lea Assies
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - José García-Calvo
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Francesca Piazzolla
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Samantha Sanchez
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Takehiro Kato
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Luc Reymond
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Spirochrome AG, Chalberwiesenstrasse 4, CH-8260 Stein am Rhein, Switzerland
| | - Antoine Goujon
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Adai Colom
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva
| | - Javier López-Andarias
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Karolína Straková
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Dora Mahecic
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; École Polytechnique Fédérale de Lausanne - EPFL, SB Cubotron 427, CH-1015 Lausanne, Switzerland
| | - Vincent Mercier
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva
| | - Margot Riggi
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva; Department of Molecular Biology, University of Geneva
| | - Noemi Jiménez-Rojo
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva
| | - Chloé Roffay
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva
| | | | - Maria Tsemperouli
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Chemistry, University of Fribourg, 9 Chemin du Musée, CH-1700 Fribourg, Switzerland
| | - Frederik Neuhaus
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Chemistry, University of Fribourg, 9 Chemin du Musée, CH-1700 Fribourg, Switzerland
| | - Alexandre Fürstenberg
- Department of Physical Chemistry, University of Geneva; Department of Inorganic and Analytical Chemistry, University of Geneva
| | - Eric Vauthey
- Department of Physical Chemistry, University of Geneva
| | - Sascha Hoogendoorn
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Marcos Gonzalez-Gaitan
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva
| | - Andreas Zumbuehl
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Chemistry, University of Fribourg, 9 Chemin du Musée, CH-1700 Fribourg, Switzerland
| | - Kaori Sugihara
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Physical Chemistry, University of Geneva
| | - Jean Gruenberg
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva
| | - Howard Riezman
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva
| | - Robbie Loewith
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Molecular Biology, University of Geneva
| | - Suliana Manley
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; École Polytechnique Fédérale de Lausanne - EPFL, SB Cubotron 427, CH-1015 Lausanne, Switzerland
| | - Aurelien Roux
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Biochemistry, University of Geneva
| | - Nicolas Winssinger
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Naomi Sakai
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland
| | - Stefan Pitsch
- Spirochrome AG, Chalberwiesenstrasse 4, CH-8260 Stein am Rhein, Switzerland
| | - Stefan Matile
- National Centre of Competence in Research (NCCR) Chemical Biology, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland; Department of Organic Chemistry, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 CH-Geneva, Switzerland;,
| |
Collapse
|
34
|
Perez-Valle A, Abad-García B, Fresnedo O, Barreda-Gómez G, Aspichueta P, Asumendi A, Astigarraga E, Fernández JA, Boyano MD, Ochoa B. A UHPLC-Mass Spectrometry View of Human Melanocytic Cells Uncovers Potential Lipid Biomarkers of Melanoma. Int J Mol Sci 2021; 22:12061. [PMID: 34769491 PMCID: PMC8585039 DOI: 10.3390/ijms222112061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer due to its ability to colonize distant sites and initiate metastasis. Although these processes largely depend on the lipid-based cell membrane scaffold, our understanding of the melanoma lipid phenotype lags behind most other aspects of this tumor cell. Here, we examined a panel of normal human epidermal and nevus melanocytes and primary and metastatic melanoma cell lines to determine whether distinctive cell-intrinsic lipidomes can discern non-neoplastic from neoplastic melanocytes and define their metastatic potential. Lipidome profiles were obtained by UHPLC-ESI mass-spectrometry, and differences in the signatures were analyzed by multivariate statistical analyses. Significant and highly specific changes in more than 30 lipid species were annotated in the initiation of melanoma, whereas less numerous changes were associated with melanoma progression and the non-malignant transformation of nevus melanocytes. Notably, the "malignancy lipid signature" features marked drops in pivotal membrane lipids, like sphingomyelins, and aberrant elevation of ether-type lipids and phosphatidylglycerol and phosphatidylinositol variants, suggesting a previously undefined remodeling of sphingolipid and glycerophospholipid metabolism. Besides broadening the molecular definition of this neoplasm, the different lipid profiles identified may help improve the clinical diagnosis/prognosis and facilitate therapeutic interventions for cutaneous melanoma.
Collapse
Affiliation(s)
- Arantza Perez-Valle
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.P.-V.); (A.A.)
| | - Beatriz Abad-García
- Central Analysis Service, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| | - Olatz Fresnedo
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (O.F.); (P.A.)
| | - Gabriel Barreda-Gómez
- IMG Pharma Biotech S.L., Bizkaia Technological Park, 48160 Derio, Spain; (G.B.-G.); (E.A.)
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (O.F.); (P.A.)
- Biocruces-Bizkaia Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Aintzane Asumendi
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.P.-V.); (A.A.)
- Biocruces-Bizkaia Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Egoitz Astigarraga
- IMG Pharma Biotech S.L., Bizkaia Technological Park, 48160 Derio, Spain; (G.B.-G.); (E.A.)
| | - José A. Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| | - María Dolores Boyano
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.P.-V.); (A.A.)
- Biocruces-Bizkaia Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Begoña Ochoa
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (O.F.); (P.A.)
| |
Collapse
|
35
|
Lange M, Angelidou G, Ni Z, Criscuolo A, Schiller J, Blüher M, Fedorova M. AdipoAtlas: A reference lipidome for human white adipose tissue. Cell Rep Med 2021; 2:100407. [PMID: 34755127 PMCID: PMC8561168 DOI: 10.1016/j.xcrm.2021.100407] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/29/2021] [Accepted: 08/26/2021] [Indexed: 01/16/2023]
Abstract
Obesity, characterized by expansion and metabolic dysregulation of white adipose tissue (WAT), has reached pandemic proportions and acts as a primer for a wide range of metabolic disorders. Remodeling of WAT lipidome in obesity and associated comorbidities can explain disease etiology and provide valuable diagnostic and prognostic markers. To support understanding of WAT lipidome remodeling at the molecular level, we provide in-depth lipidomics profiling of human subcutaneous and visceral WAT of lean and obese individuals. We generate a human WAT reference lipidome by performing tissue-tailored preanalytical and analytical workflows, which allow accurate identification and semi-absolute quantification of 1,636 and 737 lipid molecular species, respectively. Deep lipidomic profiling allows identification of main lipid (sub)classes undergoing depot-/phenotype-specific remodeling. Previously unanticipated diversity of WAT ceramides is now uncovered. AdipoAtlas reference lipidome serves as a data-rich resource for the development of WAT-specific high-throughput methods and as a scaffold for systems medicine data integration.
Collapse
Affiliation(s)
- Mike Lange
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Georgia Angelidou
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Zhixu Ni
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Angela Criscuolo
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
- Thermo Fisher Scientific, Dreieich, Germany
| | - Jürgen Schiller
- Institute of Medical Physics and Biophysics, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
36
|
Monasterio BG, Jiménez-Rojo N, García-Arribas AB, Riezman H, Goñi FM, Alonso A. CHO/LY-B cell growth under limiting sphingolipid supply: Correlation between lipid composition and biophysical properties of sphingolipid-restricted cell membranes. FASEB J 2021; 35:e21657. [PMID: 34010474 DOI: 10.1096/fj.202001879rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 04/09/2021] [Accepted: 04/26/2021] [Indexed: 11/11/2022]
Abstract
Sphingolipids (SL) are ubiquitous in mammalian cell membranes, yet there is little data on the behavior of cells under SL-restriction conditions. LY-B cells derive from a CHO linein whichserine palmitoyl transferase (SPT), thus de novo SL synthesis, is suppressed, while maintaining the capacity of taking up and metabolizing exogenous sphingoid bases from the culture medium. In this study, LY-B cells were adapted to grow in a fetal bovine serum (FBS)-deficient medium to avoid external uptake of lipids. The lowest FBS concentration that allowed LY-B cell growth, though at a slow rate, under our conditions was 0.04%, that is, 250-fold less than the standard (10%) concentration. Cells grown under limiting SL concentrations remained viable for at least 72 hours. Enriching with sphingomyelin the SL-deficient medium allowed the recovery of growth rates analogous to those of control LY-B cells. Studies including whole cells, plasma membrane preparations, and derived lipid vesicles were carried out. Laurdan fluorescence was recorded to measure membrane molecular order, showing a significant decrease in the rigidity of LY-B cells, not only in plasma membrane but also in whole cell lipid extract, as a result of SL limitation in the growth medium. Plasma membrane preparations and whole cell lipid extracts were also studied using atomic force microscopy in the force spectroscopy mode. Force measurements demonstrated that lower breakthrough forces were required to penetrate samples obtained from SL-poor LY-B cells than those obtained from control cells. Mass-spectroscopic analysis was also a helpful tool to understand the rearrangement undergone by the LY-B cell lipid metabolism. The most abundant SL in LY-B cells, sphingomyelin, decreased by about 85% as a result of SL limitation in the medium, the bioactive lipid ceramide and the ganglioside precursor hexosylceramide decreased similarly, together with cholesterol. Quantitative SL analysis showed that a 250-fold reduction in sphingolipid supply to LY-B cells led only to a sixfold decrease in membrane sphingolipids, underlining the resistance to changes in composition of these cells. Plasma membrane compositions exhibited similar changes, at least qualitatively, as the whole cells with SL restriction. A linear correlation was observed between the sphingomyelin concentration in the membranes, the degree of lipid order as measured by laurdan fluorescence, and membrane breakthrough forces assessed by atomic force microscopy. Smaller, though significant, changes were also detected in glycerophospholipids under SL-restriction conditions.
Collapse
Affiliation(s)
- Bingen G Monasterio
- Instituto Biofisika (CSIC, UPV/EHU), Universidad del País Vasco, Leioa, Spain.,Departamento de Bioquímica, Universidad del País Vasco, Leioa, Spain
| | - Noemi Jiménez-Rojo
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Aritz B García-Arribas
- Instituto Biofisika (CSIC, UPV/EHU), Universidad del País Vasco, Leioa, Spain.,Departamento de Bioquímica, Universidad del País Vasco, Leioa, Spain
| | - Howard Riezman
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Félix M Goñi
- Instituto Biofisika (CSIC, UPV/EHU), Universidad del País Vasco, Leioa, Spain.,Departamento de Bioquímica, Universidad del País Vasco, Leioa, Spain
| | - Alicia Alonso
- Instituto Biofisika (CSIC, UPV/EHU), Universidad del País Vasco, Leioa, Spain.,Departamento de Bioquímica, Universidad del País Vasco, Leioa, Spain
| |
Collapse
|
37
|
Aldrovandi M, Fedorova M, Conrad M. Juggling with lipids, a game of Russian roulette. Trends Endocrinol Metab 2021; 32:463-473. [PMID: 33985897 DOI: 10.1016/j.tem.2021.04.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 01/13/2023]
Abstract
Lipid peroxidation (LPO) is the molecular mechanism involved in oxidative damage of cellular membranes and the hallmark of a nonapoptotic form of cell death, known as ferroptosis. This iron-dependent cell death is an emerging strategy in cancer treatment and one of the central cell death mechanisms accounting for early cell loss and organ dysfunction in both neurodegenerative disease and ischemia-reperfusion injury. Although the biological roles of LPO products have attracted considerable attention, not only for their pathological mechanisms but also for their potential clinical application as biomarkers, the existence of a common lethal lipid death signal generated during ferroptosis remains poorly explored. A better understanding of the LPO process, however, may unleash unprecedented opportunities for therapeutic intervention of as-yet incurable diseases.
Collapse
Affiliation(s)
- Maceler Aldrovandi
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, 85764 Neuherberg, Germany
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig 04013, Germany; Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig 04013, Germany.
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, 85764 Neuherberg, Germany; Pirogov Russian National Research Medical University, Laboratory of Experimental Oncology, Moscow 117997, Russia.
| |
Collapse
|
38
|
The Protein Toxins Ricin and Shiga Toxin as Tools to Explore Cellular Mechanisms of Internalization and Intracellular Transport. Toxins (Basel) 2021; 13:toxins13060377. [PMID: 34070659 PMCID: PMC8227415 DOI: 10.3390/toxins13060377] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/22/2021] [Indexed: 12/18/2022] Open
Abstract
Protein toxins secreted by bacteria and found in plants can be threats to human health. However, their extreme toxicity can also be exploited in different ways, e.g., to produce hybrid toxins directed against cancer cells and to study transport mechanisms in cells. Investigations during the last decades have shown how powerful these molecules are as tools in cell biological research. Here, we first present a partly historical overview, with emphasis on Shiga toxin and ricin, of how such toxins have been used to characterize processes and proteins of importance for their trafficking. In the second half of the article, we describe how one can now use toxins to investigate the role of lipid classes for intracellular transport. In recent years, it has become possible to quantify hundreds of lipid species using mass spectrometry analysis. Thus, it is also now possible to explore the importance of lipid species in intracellular transport. The detailed analyses of changes in lipids seen under conditions of inhibited toxin transport reveal previously unknown connections between syntheses of lipid classes and demonstrate the ability of cells to compensate under given conditions.
Collapse
|
39
|
Parton RG, Tillu V, McMahon KA, Collins BM. Key phases in the formation of caveolae. Curr Opin Cell Biol 2021; 71:7-14. [PMID: 33677149 DOI: 10.1016/j.ceb.2021.01.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 12/20/2022]
Abstract
Caveolae are abundant plasma membrane pits formed by the coordinated action of peripheral and integral membrane proteins and membrane lipids. Here, we discuss recent studies that are starting to provide a glimpse of how filamentous cavin proteins, membrane-embedded caveolin proteins, and specific plasma membrane lipids are brought together to make the unique caveola surface domain. Protein assembly involves multiple low-affinity interactions that are dependent on 'fuzzy' charge-dependent interactions mediated in part by disordered cavin and caveolin domains. We propose that cavins help generate a lipid domain conducive to full insertion of caveolin into the bilayer to promote caveola formation. The synergistic assembly of these dynamic protein complexes supports the formation of a metastable membrane domain that can be readily disassembled both in response to cellular stress and during endocytic trafficking. We present a mechanistic model for generation of caveolae based on these new insights.
Collapse
Affiliation(s)
- Robert G Parton
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, 4072, Australia; The University of Queensland, Centre for Microscopy and Microanalysis, Brisbane, Queensland, 4072, Australia.
| | - Vikas Tillu
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, 4072, Australia
| | - Kerrie-Ann McMahon
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, 4072, Australia
| | - Brett M Collins
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
40
|
López-Andarias J, Straková K, Martinent R, Jiménez-Rojo N, Riezman H, Sakai N, Matile S. Genetically Encoded Supramolecular Targeting of Fluorescent Membrane Tension Probes within Live Cells: Precisely Localized Controlled Release by External Chemical Stimulation. JACS AU 2021; 1:221-232. [PMID: 34467286 PMCID: PMC8395630 DOI: 10.1021/jacsau.0c00069] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Indexed: 05/12/2023]
Abstract
To image membrane tension in selected membranes of interest (MOI) inside living systems, the field of mechanobiology requires increasingly elaborated small-molecule chemical tools. We have recently introduced HaloFlipper, i.e., a mechanosensitive flipper probe that can localize in the MOI using HaloTag technology to report local membrane tension changes using fluorescence lifetime imaging microscopy. However, the linker tethering the probe to HaloTag hampers the lateral diffusion of the probe in all the lipid domains of the MOI. For a more global membrane tension measurement in any MOI, we present here a supramolecular chemistry strategy for selective localization and controlled release of flipper into the MOI, using a genetically encoded supramolecular tag. SupraFlippers, functionalized with a desthiobiotin ligand, can selectively accumulate in the organelle having expressed streptavidin. The addition of biotin as a biocompatible external stimulus with a higher affinity for Sav triggers the release of the probe, which spontaneously partitions into the MOI. Freed in the lumen of endoplasmic reticulum (ER), SupraFlippers report the membrane orders along the secretory pathway from the ER over the Golgi apparatus to the plasma membrane. Kinetics of the process are governed by both the probe release and the transport through lipid domains. The concentration of biotin can control the former, while the expression level of a transmembrane protein (Sec12) involved in the stimulation of the vesicular transport from ER to Golgi influences the latter. Finally, the generation of a cell-penetrating and fully functional Sav-flipper complex using cyclic oligochalcogenide (COC) transporters allows us to combine the SupraFlipper strategy and HaloTag technology.
Collapse
|
41
|
Sailer S, Keller MA, Werner ER, Watschinger K. The Emerging Physiological Role of AGMO 10 Years after Its Gene Identification. Life (Basel) 2021; 11:life11020088. [PMID: 33530536 PMCID: PMC7911779 DOI: 10.3390/life11020088] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023] Open
Abstract
The gene encoding alkylglycerol monooxygenase (AGMO) was assigned 10 years ago. So far, AGMO is the only known enzyme capable of catalysing the breakdown of alkylglycerols and lyso-alkylglycerophospholipids. With the knowledge of the genetic information, it was possible to relate a potential contribution for mutations in the AGMO locus to human diseases by genome-wide association studies. A possible role for AGMO was implicated by genetic analyses in a variety of human pathologies such as type 2 diabetes, neurodevelopmental disorders, cancer, and immune defence. Deficient catabolism of stored lipids carrying an alkyl bond by an absence of AGMO was shown to impact on the overall lipid composition also outside the ether lipid pool. This review focuses on the current evidence of AGMO in human diseases and summarises experimental evidence for its role in immunity, energy homeostasis, and development in humans and several model organisms. With the progress in lipidomics platform and genetic identification of enzymes involved in ether lipid metabolism such as AGMO, it is now possible to study the consequence of gene ablation on the global lipid pool and further on certain signalling cascades in a variety of model organisms in more detail.
Collapse
Affiliation(s)
- Sabrina Sailer
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
| | - Markus A. Keller
- Institute of Human Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Ernst R. Werner
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
| | - Katrin Watschinger
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
- Correspondence: ; Tel.: +43-512-9003-70344
| |
Collapse
|
42
|
Rodriguez-Gallardo S, Kurokawa K, Sabido-Bozo S, Cortes-Gomez A, Ikeda A, Zoni V, Aguilera-Romero A, Perez-Linero AM, Lopez S, Waga M, Araki M, Nakano M, Riezman H, Funato K, Vanni S, Nakano A, Muñiz M. Ceramide chain length-dependent protein sorting into selective endoplasmic reticulum exit sites. SCIENCE ADVANCES 2020; 6:6/50/eaba8237. [PMID: 33310842 PMCID: PMC7732199 DOI: 10.1126/sciadv.aba8237] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 10/30/2020] [Indexed: 05/05/2023]
Abstract
Protein sorting in the secretory pathway is crucial to maintain cellular compartmentalization and homeostasis. In addition to coat-mediated sorting, the role of lipids in driving protein sorting during secretory transport is a longstanding fundamental question that still remains unanswered. Here, we conduct 3D simultaneous multicolor high-resolution live imaging to demonstrate in vivo that newly synthesized glycosylphosphatidylinositol-anchored proteins having a very long chain ceramide lipid moiety are clustered and sorted into specialized endoplasmic reticulum exit sites that are distinct from those used by transmembrane proteins. Furthermore, we show that the chain length of ceramide in the endoplasmic reticulum membrane is critical for this sorting selectivity. Our study provides the first direct in vivo evidence for lipid chain length-based protein cargo sorting into selective export sites of the secretory pathway.
Collapse
Affiliation(s)
- Sofia Rodriguez-Gallardo
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Kazuo Kurokawa
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Saitama, Japan.
| | - Susana Sabido-Bozo
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Alejandro Cortes-Gomez
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Atsuko Ikeda
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Valeria Zoni
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Auxiliadora Aguilera-Romero
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Ana Maria Perez-Linero
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Sergio Lopez
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Miho Waga
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Saitama, Japan
| | - Misako Araki
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Howard Riezman
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Kouichi Funato
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Stefano Vanni
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Saitama, Japan
| | - Manuel Muñiz
- Department of Cell Biology, Faculty of Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain.
| |
Collapse
|
43
|
Dichlberger A, Zhou K, Bäck N, Nyholm T, Backman A, Mattjus P, Ikonen E, Blom T. LAPTM4B controls the sphingolipid and ether lipid signature of small extracellular vesicles. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158855. [PMID: 33181324 DOI: 10.1016/j.bbalip.2020.158855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 01/08/2023]
Abstract
Lysosome Associated Protein Transmembrane 4B (LAPTM4B) is a four-membrane spanning ceramide interacting protein that regulates mTORC1 signaling. Here, we show that LAPTM4B is sorted into intraluminal vesicles (ILVs) of multivesicular endosomes (MVEs) and released in small extracellular vesicles (sEVs) into conditioned cell culture medium and human urine. Efficient sorting of LAPTM4B into ILV membranes depends on its third transmembrane domain containing a sphingolipid interaction motif (SLim). Unbiased lipidomic analysis reveals a strong enrichment of glycosphingolipids in sEVs secreted from LAPTM4B knockout cells and from cells expressing a SLim-deficient LAPTM4B mutant. The altered sphingolipid profile is accompanied by a distinct SLim-dependent co-modulation of ether lipid species. The changes in the lipid composition of sEVs derived from LAPTM4B knockout cells is reflected by an increased stability of membrane nanodomains of sEVs. These results identify LAPTM4B as a determinant of the glycosphingolipid profile and membrane properties of sEVs.
Collapse
Affiliation(s)
- Andrea Dichlberger
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Kecheng Zhou
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Nils Bäck
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Thomas Nyholm
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
| | - Anders Backman
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
| | - Peter Mattjus
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
| | - Elina Ikonen
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Tomas Blom
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| |
Collapse
|
44
|
Abstract
Biological membranes consist of a surprisingly high number of different lipid species. Little is known about how individual lipids cooperate in modulating cellular functions. A new study suggests an intricate interplay of sphingolipids with ether lipids in vesicular transport.
Collapse
Affiliation(s)
- Britta Brügger
- Heidelberg University Biochemistry Center (BZH), Im Neuenheimer Feld 328, 69120 Heidelberg, Germany.
| |
Collapse
|