1
|
Migni A, Bartolini D, Varfaj I, Moscardini IF, Sardella R, Garetto S, Lucci J, Galli F. Lipidomics reveals different therapeutic potential for natural and synthetic vitamin D formulations in hepatocyte lipotoxicity. Biomed Pharmacother 2025; 187:118068. [PMID: 40300390 DOI: 10.1016/j.biopha.2025.118068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/01/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
Natural sources of vitamin D (VD), have been proposed to represent an alternative to synthetic vitamin D in nutritional interventions, also holding therapeutic potential in non-alcoholic fatty liver disease (NAFLD). In this study lipidomics was used to comparatively investigate the molecular mechanisms behind the therapeutic effects of a natural VD formulation consisting of a Shitake mushroom extracts (NVD) and a synthetic cholecalciferol formulation (SVD) in HepaRG human hepatocytes exposed to free fatty acid (FFA)-induced lipotoxicity. The results demonstrate that the two VD formulations prevent lipotoxicity with similar efficacy, but different lipidomic fingerprints. Differentially expressed lipids in NVD' in vitro therapeutic effect indicated a reduced synthesis of cellular triglycerides; combined with a marked reshaping of glycerophospholipid metabolism and characteristic changes of the chain length and number of double bonds in the phosphatidylcholine pool that were absent in SVD treatment. Bioinformatics interpretation of lipidomics data associated NVD therapeutic properties to an enhanced insulin function and glycerophospholipid metabolism, whereas SVD was primarily associated with the inflammatory signaling and death pathways of the liver cell. These differences between the two VD formulations were further highlighted matching lipidomics data with gene microarray (transcriptomics) data available from previous studies on this experimental model; the resulting multiomics data identified lipid metabolism nodes specific for the multimolecular mechanisms of the two formulations which may deserve further pre-clinical investigation in the treatment of hepatocyte lipotoxicity.
Collapse
Affiliation(s)
- Anna Migni
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Umbria, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Umbria, Italy
| | - Ina Varfaj
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Umbria, Italy
| | | | - Roccaldo Sardella
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Umbria, Italy
| | - Stefano Garetto
- Bios-Therapy, Physiological Systems For Health S.p.A, Loc. Aboca 20, Sansepolcro, Arezzo, Italy
| | - Jacopo Lucci
- Bios-Therapy, Physiological Systems For Health S.p.A, Loc. Aboca 20, Sansepolcro, Arezzo, Italy
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Umbria, Italy.
| |
Collapse
|
2
|
Sastourné-Haletou R, Marynberg S, Pereira A, Su F, Chen M, Valet G, Sindikubwabo F, Cañeque T, Müller S, Colombeau L, Solier S, Gaillet C, Guianvarc'h D, Biot C, Karoyan P, Gueroui Z, Arimondo P, Klausen M, Vauzeilles B, Cossy J, Fontecave M, Gasser G, Policar C, Gautier A, Johannes L, Rodriguez R. PSL Chemical Biology Symposia: The Increasing Impact of Chemistry in Life Sciences. Chembiochem 2025; 26:e202500231. [PMID: 40195606 DOI: 10.1002/cbic.202500231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Indexed: 04/09/2025]
Abstract
This symposium is the 6th Paris Sciences & Lettres (PSL) Chemical Biology meeting (2015, 2016, 2019, 2023, 2024, 2025) being held at Institut Curie. This initiative originally started in 2013 at Institut de Chimie des Substances Naturelles (ICSN) in Gif-sur-Yvette and was mostly focused on organic synthesis. It was then exported at Institut Curie to cover a larger scope, before becoming the official French Chemical Biology meeting. This year, around 200 participants had the opportunity to meet world leaders in chemistry and biology who described their latest innovations and future trends covering topics as diverse as prebiotic chemistry, activity-based protein profiling, high-resolution cell imaging, nanotechnologies, bio-orthogonal chemistry, metal ion signaling, ferroptosis, and biocatalysis.
Collapse
Affiliation(s)
- Romain Sastourné-Haletou
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Sacha Marynberg
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Arthur Pereira
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Fubao Su
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Mengnuo Chen
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Gaspard Valet
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Fabien Sindikubwabo
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Tatiana Cañeque
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Sebastian Müller
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Ludovic Colombeau
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Stéphanie Solier
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Christine Gaillet
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | | | - Christophe Biot
- UGSF, Université de Lille, UMR 8576 CNRS, 59655, Villeneuve d'Ascq, France
| | - Philippe Karoyan
- CPCV, École Normale Supérieure, Sorbonne Université, UMR 8228 CNRS, 75005, Paris, France
| | - Zoher Gueroui
- CPCV, École Normale Supérieure, Sorbonne Université, UMR 8228 CNRS, 75005, Paris, France
| | - Paola Arimondo
- Chimie Biologique Épigénétique, Institut Pasteur, UMR 3523 CNRS, 75724, Paris, France
| | - Maxime Klausen
- ICB, Chimie ParisTech, UMR 8060 CNRS, 75005, Paris, France
| | - Boris Vauzeilles
- ICSN, Université Paris-Saclay, UPR 2301 CNRS, 91190, Gif-sur-Yvette, France
| | | | - Marc Fontecave
- LCPB, Collège de France, UMR 8229 CNRS, 75005, Paris, France
| | - Gilles Gasser
- ICB, Chimie ParisTech, UMR 8060 CNRS, 75005, Paris, France
| | - Clotilde Policar
- CPCV, École Normale Supérieure, Sorbonne Université, UMR 8228 CNRS, 75005, Paris, France
| | - Arnaud Gautier
- CPCV, École Normale Supérieure, Sorbonne Université, UMR 8228 CNRS, 75005, Paris, France
| | - Ludger Johannes
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Raphaël Rodriguez
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| |
Collapse
|
3
|
Xu X, Xu C, Zhang W, Liu Z, Wei Y, Yang K, Yuan B. Single-Lipid Diffusion Behaviors in Cell Membranes Modulated by Cholesterol-Based Heterogeneity. J Phys Chem B 2025. [PMID: 40418728 DOI: 10.1021/acs.jpcb.5c01625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Over a century after the proposal of Fluid Mosaic Model, the relationship between functionally related multiple-scale spatial heterogeneity of the cell membrane and mobility of component molecules, both inherent features of cell membrane, remains elusive. Single-lipid tracking enables the analysis of structural heterogeneity at different spatial scales within the cell membrane from a lipid diffusion perspective. Herein, specifically designed cholesterol (Chol)-based membrane systems were utilized to investigate the distinct impacts of molecular-level interactions between diverse membrane components and micrometer-scale spatial confinement on lipid diffusion. The results demonstrate that the incorporation of Chol into 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) membranes decelerates lipid diffusion, with a positive correlation observed between the degree of deceleration and the mole ratio of Chol molecules. Across all these systems, lipid diffusion consistently adheres to the continuous time random walk (CTRW) model, indicating lipid entrapment resulting from specific molecular interactions. Conversely, micrometer-scale spatial confinement induced by phase separation not only reduces the diffusion rate of DOPC molecules but also triggers a transition from CTRW to fractional Brownian motion (fBM) or random walk on a fractal (RWF) mode within a confinement width range of 6.3-5.4 μm, suggesting a crowded microenvironment. In living cell membranes, this transformation in lipid diffusion is observed following Chol depletion, implying that lipid raft disruption leads to increased crowding within the lipid microenvironment. This study enhances our understanding of the relationship between lipid diffusion and membrane microenvironment across different spatial scales while providing insights into characterizing spatially heterogeneous structures within cell membranes from the perspective of lipid diffusion.
Collapse
Affiliation(s)
- Xiao Xu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu 215006, China
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| | - Cheng Xu
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| | - Wanting Zhang
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zhiheng Liu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu 215006, China
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| | - Yushuang Wei
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu 215006, China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| |
Collapse
|
4
|
Doktorova M, Symons JL, Zhang X, Wang HY, Schlegel J, Lorent JH, Heberle FA, Sezgin E, Lyman E, Levental KR, Levental I. Cell membranes sustain phospholipid imbalance via cholesterol asymmetry. Cell 2025; 188:2586-2602.e24. [PMID: 40179882 DOI: 10.1016/j.cell.2025.02.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/05/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025]
Abstract
Membranes are molecular interfaces that compartmentalize cells to control the flow of nutrients and information. These functions are facilitated by diverse collections of lipids, nearly all of which are distributed asymmetrically between the two bilayer leaflets. Most models of biomembrane structure and function include the implicit assumption that these leaflets have similar abundances of phospholipids. Here, we show that this assumption is generally invalid and investigate the consequences of lipid abundance imbalances in mammalian plasma membranes (PMs). Using lipidomics, we report that cytoplasmic leaflets of human erythrocyte membranes have >50% overabundance of phospholipids compared with exoplasmic leaflets. This imbalance is enabled by an asymmetric interleaflet distribution of cholesterol, which regulates cellular cholesterol homeostasis. These features produce unique functional characteristics, including low PM permeability and resting tension in the cytoplasmic leaflet that regulates protein localization.
Collapse
Affiliation(s)
- Milka Doktorova
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA; Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 17165 Solna, Sweden.
| | - Jessica L Symons
- Department of Integrative Biology & Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoxuan Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Jan Schlegel
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| | - Joseph H Lorent
- Department of Cellular and Molecular Pharmacology, TFAR, LDRI, UCLouvain, Avenue Mounier 73, B1.73.05, 1200 Brussels, Belgium
| | - Frederick A Heberle
- Department of Chemistry, University of Tennessee Knoxville, Knoxville, TN 37916, USA
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| | - Edward Lyman
- Department of Physics and Astronomy, Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA.
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
5
|
Wang HY, Rumin A, Doktorova M, Sputay D, Chan SH, Wehman AM, Levental KR, Levental I. Loss of lipid asymmetry facilitates plasma membrane blebbing by decreasing membrane lipid packing. Proc Natl Acad Sci U S A 2025; 122:e2417145122. [PMID: 40324083 DOI: 10.1073/pnas.2417145122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/21/2025] [Indexed: 05/07/2025] Open
Abstract
Membrane blebs have important roles in cell migration, apoptosis, and intercellular communication through extracellular vesicles (EVs). While plasma membranes (PM) typically maintain phosphatidylserine (PS) on their cytoplasmic leaflet, most blebs have PS exposed on their outer leaflet, revealing that loss of steady-state lipid asymmetry often accompanies PM blebbing. How these changes in PM lipid organization regulate membrane properties and affect bleb formation remains unknown. We confirmed that lipid scrambling through the scramblase TMEM16F is essential for chemically induced membrane blebbing across cell types, with the kinetics of PS exposure being tightly coupled to the kinetics of bleb formation. Measurement of lipid packing with environment-sensitive probes revealed that lipid scrambling changes the physical properties of the PM, reducing lipid packing and facilitating the bilayer bending required for bleb formation. Accordingly, reducing lipid packing of the PM through cholesterol extraction, elevated temperature, or treatment with biological amphiphiles promoted blebbing in the absence of TMEM16F. Consistent with these cellular observations, blebbing in Caenorhabditis elegans embryos measured via EV production was significantly reduced by depleting the TMEM16-homolog ANOH-2. Our findings suggest that changing membrane biophysical properties by lipid scrambling is an important contributor to the formation of blebs and EVs and potentially other cellular processes involving PM deformation.
Collapse
Affiliation(s)
- Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903
| | - Alissa Rumin
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, CO 80210
| | - Milka Doktorova
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna 171 65, Sweden
| | - Daryna Sputay
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903
| | - Sze Ham Chan
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903
| | - Ann M Wehman
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, CO 80210
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
6
|
Sputay D, Doktorova M, Chan SH, Guo EH, Wang HY, Lorent JH, Levental I, Levental KR. Immune cell activation produces locally scrambled foci of plasma membrane lipids. Faraday Discuss 2025. [PMID: 40351229 PMCID: PMC12067185 DOI: 10.1039/d4fd00205a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/11/2025] [Indexed: 05/14/2025]
Abstract
Most eukaryotic cells maintain a large disparity in lipid compositions between the cytosolic and external leaflets of the plasma membrane (PM) bilayer. This lipid asymmetry is maintained by energy-consuming flippase enzymes that selectively drive phospholipids into the cytosolic leaflet, often against large concentration gradients. Scramblases, activated by intracellular Ca2+ or apoptotic signaling, shuttle phospholipids down their concentration gradient to release lipid asymmetry. Such scrambling is typically evidenced by exposure of phosphatidylserine (PS) to the external leaflet and is associated with many physiological processes, most notably blood clotting and cell death, but also activation of immune cells. Here, we show that both PS and phosphatidylethanolamine (PE) appear on the PM external leaflet following immune receptor-mediated activation of mast cells. We also observe similar effects in T cells. Importantly, in contrast to wholesale release of PM asymmetry induced by calcium ionophores or apoptosis, we show that scrambling in activated immune cells is focal, with small, stable regions of surface exposed PS. These scrambled foci are calcium dependent, have lower lipid packing than their surrounding outer leaflet, and are reversible. These observations of local, transient scrambling during physiological activation of healthy immune cells suggest important roles for the lateral and transbilayer organization of membrane lipids.
Collapse
Affiliation(s)
- Daryna Sputay
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Milka Doktorova
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Sweden
| | - Sze Ham Chan
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Emma Han Guo
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Joseph H Lorent
- Cellular and Molecular Pharmacology (FACM), Louvain Drug Research Institute, UCLouvain, Belgium
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| |
Collapse
|
7
|
Pareek CS, Sachajko M, Kalra G, Sultana S, Szostak A, Chalaskiewicz K, Kepka-Borkowska K, Poławska E, Ogłuszka M, Pierzchała D, Starzyński R, Taniguchi H, Juszczuk-Kubiak E, Lepczyński A, Ślaska B, Kozera W, Czarnik U, Wysocki P, Kadarmideen HN, Te Pas MFW, Szyda J, Pierzchała M. Identification of trait-associated microRNA modules in liver transcriptome of pig fed with PUFAs-enriched supplementary diet. J Appl Genet 2025; 66:389-407. [PMID: 39546271 PMCID: PMC12000271 DOI: 10.1007/s13353-024-00912-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/16/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024]
Abstract
Dietary lipids provide energy, are cellular structural components, and are involved in physiological processes. Lipids are the dietary source in supplementary diet experiments in pigs. This study aims to investigate the dietary effects of PUFAs on the hepatic transcriptome and physiological pathways of two diets on two pig breeds. Polish Landrace (PL: n = 6) and six PLxDuroc (PLxD: n = 6) pigs were fed with a normal diet (n = 3) or PUFAs-enriched healthy diet (n = 3), and the hepatic miRNA profiles were studied for weighted gene co-expression network analysis biological interactions between gene networks and metabolic pathways of DE miRNA genes. The study identified trait-associated modules that were significantly associated with four phenotypic traits in the dietary groups of PL and PLxD: meat colour (a*), shoulder subcutaneous fat thickness, conductivity 24 h post-mortem (PE24), and ashes. Trait-wise, a large set of co-expressed miRNAs of porcine liver were identified in these trait-associated significant modules (9, 7, 2, and 8) in PL and PLxD. Each module is represented by a module eigengene (ME). Forty-four miRNAs out of 94 miRNAs interacted with 6719 statistically significant target genes with a target score > 90. The GO/pathway analysis showed association with pathways including regulation of metallopeptidase activity, sebaceous gland development, collagen fibril organization, WNT signalling, epithelial tube morphogenesis, etc. The study showed the differences in miRNA expression between the dietary groups of PL and PLxD breeds. Hub genes of discovered miRNA clusters can be considered predicted miRNA genes associated with PE24, meat colour, shoulder subcutaneous fat thickness, and ashes. Discovered target genes for miRNA clusters play significant roles in biological functions such as (i) muscle and body growth development, (ii) different cellular processes and developments, (iii) system development, and (iv) metabolic processes.
Collapse
Affiliation(s)
- C S Pareek
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100, Toruń, Poland
- Division of Functional Genomics in Biological and Biomedical Research, Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100, Torun, Poland
| | - M Sachajko
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100, Toruń, Poland
| | - G Kalra
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100, Toruń, Poland
| | - S Sultana
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100, Toruń, Poland
| | - A Szostak
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - K Chalaskiewicz
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - K Kepka-Borkowska
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - E Poławska
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - M Ogłuszka
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - D Pierzchała
- Maria Sklodowska-Curie National Research Institute of Oncology, W.K. Roentgena 5 Str, 02-781, Warsaw, Poland
| | - R Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - H Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
- African Genome Center, Mohammed VI Polytechnic University, UM6P, Lot 660, Hay Moulay Rachid Ben Guerir, 43150, Morocco
| | - E Juszczuk-Kubiak
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology Prof. Wacław, Dąbrowski Institute of Agriculture and Food Biotechnology - State Research Institute (IBPRS-PIB), Rakowiecka 36 Str, 02-532, Warsaw, Poland
| | - A Lepczyński
- Department of Physiology, Cytobiology and Proteomics, West Pomeranian University of Technology, K. Janickiego 32 Str, 71-270, Szczecin, Poland
| | - B Ślaska
- Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13 Str, 20-950, Lublin, Poland
| | - W Kozera
- Department of Pig Breeding, Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bio-Engineering, University of Warmia and Mazury in Olsztyn, Ul. M. Oczapowskiego 5 Str, 10-719, Olsztyn, Poland
| | - U Czarnik
- Department of Pig Breeding, Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bio-Engineering, University of Warmia and Mazury in Olsztyn, Ul. M. Oczapowskiego 5 Str, 10-719, Olsztyn, Poland
| | - P Wysocki
- Department of Pig Breeding, Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bio-Engineering, University of Warmia and Mazury in Olsztyn, Ul. M. Oczapowskiego 5 Str, 10-719, Olsztyn, Poland
| | - H N Kadarmideen
- Department of Animal and Veterinary Sciences, Aarhus University, Blichers Alle 20, 8830, Tjele, Denmark
| | - M F W Te Pas
- Wageningen Livestock Research, Wageningen University and Research, 6708 WD, Wageningen, The Netherlands
| | - J Szyda
- Biostatistics Group, Department of Genetics, Wrocław University of Environmental and Life Sciences, Kozuchowska 7, 51-631, Wrocław, Poland
| | - M Pierzchała
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland.
| |
Collapse
|
8
|
Bertrand B, Munoz-Garay C. Unlocking the power of membrane biophysics: enhancing the study of antimicrobial peptides activity and selectivity. Biophys Rev 2025; 17:605-625. [PMID: 40376398 PMCID: PMC12075066 DOI: 10.1007/s12551-025-01312-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/26/2025] [Indexed: 05/18/2025] Open
Abstract
The application of membrane-active antimicrobial peptides (AMPs) is considered to be a viable alternative to conventional antibiotics for treating infections caused by multidrug-resistant pathogenic microorganisms. In vitro and in silico biophysical approaches are indispensable for understanding the underlying molecular mechanisms of membrane-active AMPs. Lipid bilayer models are widely used to mimic and study the implication of various factors affecting these bio-active molecules, and their relationship with the physical parameters of the different membranes themselves. The quality and resemblance of these models to their target is crucial for elucidating how these AMPs work. Unfortunately, over the last few decades, no notable efforts have been made to improve or refine membrane mimetics, as it pertains to the elucidation of AMPs molecular mechanisms. In this review, we discuss the importance of improving the quality and resemblance of target membrane models, in terms of lipid composition and distribution, which ultimately directly influence physical parameters such as charge, fluidity, and thickness. In conjunction, membrane and peptide properties determine the global effect of selectivity, activity, and potency. It is therefore essential to define these interactions, and to do so, more refined lipid models are necessary. In this review, we focus on the significant advancements in promoting biomimetic membranes that closely resemble native ones, for which thorough biophysical characterization is key. This includes utilizing more complex lipid compositions that mimic various cell types. Additionally, we discuss important considerations to be taken into account when working with more complex systems.
Collapse
Affiliation(s)
- Brandt Bertrand
- Instituto de Ciencias Físicas (ICF), Universidad Nacional Autónoma de México (UNAM), Avenida Universidad 2001, Chamilpa, 62210 Cuernavaca, Morelos México
| | - Carlos Munoz-Garay
- Instituto de Ciencias Físicas (ICF), Universidad Nacional Autónoma de México (UNAM), Avenida Universidad 2001, Chamilpa, 62210 Cuernavaca, Morelos México
| |
Collapse
|
9
|
Lancaster GI, Murphy AJ. Do physiological changes in fatty acid composition alter cellular ferroptosis susceptibility and influence cell function? J Lipid Res 2025; 66:100765. [PMID: 40021010 PMCID: PMC11981815 DOI: 10.1016/j.jlr.2025.100765] [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: 12/19/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Ferroptosis is an iron-dependent form of cell death driven by the excessive peroxidation of poly-unsaturated fatty acids (PUFAs) within membrane phospholipids. Ferroptosis is a hallmark of many diseases and preventing or inducing ferroptosis has considerable therapeutic potential. Like other forms of cell death, the pathological importance and therapeutic potential of ferroptosis is well appreciated. However, while cell death modalities such as apoptosis and necroptosis have critical physiological roles, such as in development and tissue homeostasis, whether ferroptosis has important physiological roles is largely unknown. In this regard, key questions for field are as follows: Is ferroptosis used for physiological processes? Are certain cell-types purposely adapted to be either resistant or sensitive to ferroptosis to be able to function optimally? Do physiological perturbations such as aging and diet impact ferroptosis susceptibility? Herein, we have reviewed emerging evidence that supports the idea that being able to selectively and controllably induce or resist ferroptosis is essential for development and cell function. While several factors regulate ferroptosis, it appears that the ability of cells and tissues to control their lipid composition, specifically the abundance of phospholipids containing PUFAs, is crucial for cells to be able to either resist or be sensitized to ferroptosis. Finally, aging and diets enriched in specific PUFAs lead to an increase in cellular PUFA levels which may sensitize cells to ferroptosis. Therefore, changes in dietary PUFAs or againg may impact the pathogenesis of diseases where ferroptosis is involved.
Collapse
Affiliation(s)
- Graeme I Lancaster
- Department of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Immunology, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| | - Andrew J Murphy
- Department of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Immunology, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
10
|
Giblin A, Cammack AJ, Blomberg N, Anoar S, Mikheenko A, Carcolé M, Atilano ML, Hull A, Shen D, Wei X, Coneys R, Zhou L, Mohammed Y, Olivier-Jimenez D, Wang LY, Kinghorn KJ, Niccoli T, Coyne AN, van der Kant R, Lashley T, Giera M, Partridge L, Isaacs AM. Neuronal polyunsaturated fatty acids are protective in ALS/FTD. Nat Neurosci 2025; 28:737-747. [PMID: 40000803 PMCID: PMC11976277 DOI: 10.1038/s41593-025-01889-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 01/07/2025] [Indexed: 02/27/2025]
Abstract
Here we report a conserved transcriptomic signature of reduced fatty acid and lipid metabolism gene expression in a Drosophila model of C9orf72 repeat expansion, the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), and in human postmortem ALS spinal cord. We performed lipidomics on C9 ALS/FTD Drosophila, induced pluripotent stem (iPS) cell neurons and postmortem FTD brain tissue. This revealed a common and specific reduction in phospholipid species containing polyunsaturated fatty acids (PUFAs). Feeding C9 ALS/FTD flies PUFAs yielded a modest increase in survival. However, increasing PUFA levels specifically in neurons of C9 ALS/FTD flies, by overexpressing fatty acid desaturase enzymes, led to a substantial extension of lifespan. Neuronal overexpression of fatty acid desaturases also suppressed stressor-induced neuronal death in iPS cell neurons of patients with both C9 and TDP-43 ALS/FTD. These data implicate neuronal fatty acid saturation in the pathogenesis of ALS/FTD and suggest that interventions to increase neuronal PUFA levels may be beneficial.
Collapse
Affiliation(s)
- Ashling Giblin
- UK Dementia Research Institute, UCL, London, UK
- Institute of Healthy Ageing, UCL, London, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Alexander J Cammack
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Niek Blomberg
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Alla Mikheenko
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Mireia Carcolé
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | - Alex Hull
- Institute of Healthy Ageing, UCL, London, UK
| | - Dunxin Shen
- Institute of Healthy Ageing, UCL, London, UK
| | - Xiaoya Wei
- Institute of Healthy Ageing, UCL, London, UK
| | - Rachel Coneys
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Lele Zhou
- UK Dementia Research Institute, UCL, London, UK
- Institute of Healthy Ageing, UCL, London, UK
| | - Yassene Mohammed
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Damien Olivier-Jimenez
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Lian Y Wang
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Alyssa N Coyne
- Department of Neurology, Johns Hopkins University, Baltimore, MA, USA
- Brain Science Institute, Johns Hopkins University, Baltimore, MA, USA
| | - Rik van der Kant
- Alzheimer Center Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Martin Giera
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Adrian M Isaacs
- UK Dementia Research Institute, UCL, London, UK.
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
11
|
Yaghmour MH, Sajeevan T, Thiele C, Kuerschner L. Phosphatidylcholine synthesis and remodeling in brain endothelial cells. J Lipid Res 2025; 66:100773. [PMID: 40074037 PMCID: PMC12002869 DOI: 10.1016/j.jlr.2025.100773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/21/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Mammalian cells synthesize hundreds of different variants of their prominent membrane lipid phosphatidylcholine (PC), all differing in the side chain composition. This batch is constantly remodeled by the Lands cycle, a metabolic pathway replacing one chain at a time. Using the alkyne lipid lyso-phosphatidylpropargylcholine (LpPC), a precursor and intermediate in PC synthesis and remodeling, we study both processes in brain endothelial bEND3 cells. A novel method for multiplexed sample analysis by mass spectrometry is developed that offers high throughput and molecular species resolution of the propargyl-labeled PC lipids. Their time-resolved profiles and kinetic parameters of metabolism demonstrate the plasticity of the PC pool and the acute handling of lipid influx in endothelial cells differs from that in hepatocytes. Side chain remodeling as a form of lipid cycling adapts the PC pool to the cell's need and maintains lipid homeostasis. We estimate that endothelial cells possess the theoretical capacity to remodel up to 99% of their PC pool within 3.5 h using the Lands cycle. However, PC species are not subjected stochastically to this remodeling pathway as different species containing duplets of saturated, omega-3, and omega-6 side chains show different decay kinetics. Our findings emphasize the essential function of Lands cycling for monitoring and adapting the side chain composition of PC in endothelial cells.
Collapse
Affiliation(s)
- Mohamed H Yaghmour
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Theja Sajeevan
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Christoph Thiele
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Lars Kuerschner
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany.
| |
Collapse
|
12
|
Solano LE, Keshet U, Reinschmidt A, Chavez Y, Hulsy WD, Fiehn O, Nikolaidis N. Dynamic Lipidome Reorganization in Response to Heat Shock Stress. Int J Mol Sci 2025; 26:2843. [PMID: 40243420 PMCID: PMC11989226 DOI: 10.3390/ijms26072843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
The heat shock response (HSR) is a conserved cellular mechanism critical for adaptation to environmental and physiological stressors, with broad implications for cell survival, immune responses, and cancer biology. While the HSR has been extensively studied at the proteomic and transcriptomic levels, the role of lipid metabolism and membrane reorganization remains underexplored. Here, we integrate mass spectrometry-based lipidomics with RNA sequencing to characterize global lipidomic and transcriptomic changes in HeLa cells exposed to three conditions: control, heat shock (HS), and HS with eight hours of recovery. Heat shock-induced extensive lipid remodeling, including significant increases in fatty acids, glycerophospholipids, and sphingolipids, with partial normalization during recovery. Transcriptomic analysis identified over 2700 upregulated and 2300 downregulated genes under heat shock, with GO enrichment suggesting potential transcriptional contributions to lipid metabolism. However, transcriptional changes alone did not fully explain the observed lipidomic shifts, suggesting additional layers of regulation. Joint pathway analysis revealed enrichment in glycerophospholipid and sphingolipid metabolism, while network analysis identified lipid transport regulators (STAB2, APOB), stress-linked metabolic nodes (KNG1), and persistent sphingolipid enrichment during recovery. These findings provide a comprehensive framework for understanding lipid-mediated mechanisms of the HSR and highlight the importance of multi-omics integration in stress adaptation and disease biology.
Collapse
Affiliation(s)
- Luis E. Solano
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA 92831, USA; (L.E.S.); (A.R.); (Y.C.); (W.D.H.)
| | - Uri Keshet
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (U.K.); (O.F.)
| | - Andrew Reinschmidt
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA 92831, USA; (L.E.S.); (A.R.); (Y.C.); (W.D.H.)
| | - Yonny Chavez
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA 92831, USA; (L.E.S.); (A.R.); (Y.C.); (W.D.H.)
| | - William Drew Hulsy
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA 92831, USA; (L.E.S.); (A.R.); (Y.C.); (W.D.H.)
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (U.K.); (O.F.)
| | - Nikolas Nikolaidis
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA 92831, USA; (L.E.S.); (A.R.); (Y.C.); (W.D.H.)
| |
Collapse
|
13
|
Solano L, Keshet U, Reinschmidt A, Chavez Y, Hulsy WD, Fiehn O, Nikolaidis N. Dynamic Lipidome Reorganization in Response to Heat Shock Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638884. [PMID: 40027697 PMCID: PMC11870493 DOI: 10.1101/2025.02.18.638884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The heat shock response (HSR) is a conserved cellular mechanism critical for adaptation to environmental and physiological stressors, with broad implications for cell survival, immune responses, and cancer biology. While the HSR has been extensively studied at the proteomic and transcriptomic levels, the role of lipid metabolism and membrane reorganization remains underexplored. Here, we integrate mass spectrometry-based lipidomics with RNA sequencing to characterize global lipidomic and transcriptomic changes in HeLa cells exposed to three conditions: control, heat shock (HS), and HS with eight hours of recovery. Heat shock-induced extensive lipid remodeling, including significant increases in fatty acids, glycerophospholipids, and sphingolipids, with partial normalization during recovery. Transcriptomic analysis identified over 2,700 upregulated and 2,300 downregulated genes under heat shock, with GO enrichment suggesting potential transcriptional contributions to lipid metabolism. However, transcriptional changes alone did not fully explain the observed lipidomic shifts, suggesting additional layers of regulation. Joint pathway analysis revealed enrichment in glycerophospholipid and sphingolipid metabolism, while network analysis identified lipid transport regulators (STAB2, APOB), stress-linked metabolic nodes (KNG1), and persistent sphingolipid enrichment during recovery. These findings provide a comprehensive framework for understanding lipid-mediated mechanisms of the HSR and highlight the importance of multi-omics integration in stress adaptation and disease biology.
Collapse
Affiliation(s)
- Luis Solano
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Uri Keshet
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA
| | - Andrew Reinschmidt
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Yonny Chavez
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - William Drew Hulsy
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA
| | - Nikolas Nikolaidis
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| |
Collapse
|
14
|
Gao F, Chen Q, Sun H, Zhang W, Shi B. Konjac glucomannan and κ-carrageenan improve hepatic fatty acid metabolism and colonic microbiota in suckling piglet. Int J Biol Macromol 2025; 288:138790. [PMID: 39675607 DOI: 10.1016/j.ijbiomac.2024.138790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Konjac glucomannan (KGM) and κ-carrageenan are polysaccharides that have garnered attention for their potential health benefits. This study aimed to evaluate the maternal supplementation of KGM and κ-carrageenan (SF) during later gestation and lactation on the effect of hepatic lipid metabolism and colonic microflora in offspring. Regarding antioxidant and inflammatory factors in the suckling piglet liver, our results showed that nuclear factor erythroid 2-related factor 2 (Nrf2) and interleukin (IL)-10 levels were significantly increased in the SF group (P < 0.05). In liver mitochondrial function, the mRNA levels of voltage-dependent anion channel 1 (VDAC1), fission 1 (Fis1), and peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) were significantly up-regulated in the SF group compared to the control (Con) group (P < 0.05). The mRNA level of peroxisome proliferator-activated receptor alpha (PPARα) was remarkably down-regulated in the SF group (P < 0.05). In the colonic microflora of suckling piglets, we found that the SF group increased the abundance of Megasphaera and reduced the abundance of Erysipelotrichaceae_unclassified. The occludin level was significantly increased in the SF group than in the Con group (P < 0.05). In summary, maternal supplementation with SF improves hepatic lipid metabolism and colonic microflora in suckling piglets.
Collapse
Affiliation(s)
- Feng Gao
- Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences Center for Agricultural Technology, Harbin 150081, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Qinrui Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Haowen Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Wentao Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| |
Collapse
|
15
|
Raskovic D, Alvarado G, Hines KM, Xu L, Gatto C, Wilkinson BJ, Pokorny A. Growth of Staphylococcus aureus in the presence of oleic acid shifts the glycolipid fatty acid profile and increases resistance to antimicrobial peptides. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184395. [PMID: 39500386 DOI: 10.1016/j.bbamem.2024.184395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/27/2024] [Accepted: 10/23/2024] [Indexed: 11/10/2024]
Abstract
Staphylococcus aureus readily adapts to various environments and quickly develops antibiotic resistance, which has led to an increase in multidrug-resistant infections. Hence, S. aureus presents a significant global health issue and its adaptations to the host environment are crucial for understanding pathogenesis and antibiotic susceptibility. When S. aureus is grown conventionally, its membrane lipids contain a mix of branched-chain and straight-chain saturated fatty acids. However, when unsaturated fatty acids are present in the growth medium, they become a major part of the total fatty acid composition. This study explores the biophysical effects of incorporating straight-chain unsaturated fatty acids into S. aureus membrane lipids. Membrane preparations from cultures supplemented with oleic acid showed more complex differential scanning calorimetry scans than those grown in tryptic soy broth alone. When grown in the presence of oleic acid, the cultures exhibited a transition significantly above the growth temperature, attributed to the presence of glycolipids with long-chain fatty acids causing acyl chain packing frustration within the bilayer. Functional aspects of the membrane were assessed by studying the kinetics of dye release from unilamellar vesicles induced by the antimicrobial peptide mastoparan X. Dye release was slower from liposomes prepared from cells grown in oleic acid-supplemented cultures, suggesting that changes in membrane lipid composition and biophysics protect the cell membrane against peptide-induced lysis. These findings underscore the intricate relationship between the growth environment, membrane lipid composition, and the physical properties of the bacterial membrane, which should be considered when developing new strategies against S. aureus infections.
Collapse
Affiliation(s)
- Djuro Raskovic
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, NC, United States of America
| | - Gloria Alvarado
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States of America; School of Biological Sciences, Illinois State University, Normal, IL, United States of America
| | - Kelly M Hines
- Department of Chemistry, University of Georgia, Athens, GA, United States of America
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, United States of America
| | - Craig Gatto
- School of Biological Sciences, Illinois State University, Normal, IL, United States of America
| | - Brian J Wilkinson
- School of Biological Sciences, Illinois State University, Normal, IL, United States of America
| | - Antje Pokorny
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, NC, United States of America.
| |
Collapse
|
16
|
Mengistu BA, Tsegaw T, Demessie Y, Getnet K, Bitew AB, Kinde MZ, Beirhun AM, Mebratu AS, Mekasha YT, Feleke MG, Fenta MD. Comprehensive review of drug resistance in mammalian cancer stem cells: implications for cancer therapy. Cancer Cell Int 2024; 24:406. [PMID: 39695669 DOI: 10.1186/s12935-024-03558-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer remains a significant global challenge, and despite the numerous strategies developed to advance cancer therapy, an effective cure for metastatic cancer remains elusive. A major hurdle in treatment success is the ability of cancer cells, particularly cancer stem cells (CSCs), to resist therapy. These CSCs possess unique abilities, including self-renewal, differentiation, and repair, which drive tumor progression and chemotherapy resistance. The resilience of CSCs is linked to certain signaling pathways. Tumors with pathway-dependent CSCs often develop genetic resistance, whereas those with pathway-independent CSCs undergo epigenetic changes that affect gene regulation. CSCs can evade cytotoxic drugs, radiation, and apoptosis by increasing drug efflux transporter activity and activating survival mechanisms. Future research should prioritize the identification of new biomarkers and signaling molecules to better understand drug resistance. The use of cutting-edge approaches, such as bioinformatics, genomics, proteomics, and nanotechnology, offers potential solutions to this challenge. Key strategies include developing targeted therapies, employing nanocarriers for precise drug delivery, and focusing on CSC-targeted pathways such as the Wnt, Notch, and Hedgehog pathways. Additionally, investigating multitarget inhibitors, immunotherapy, and nanodrug delivery systems is critical for overcoming drug resistance in cancer cells.
Collapse
Affiliation(s)
- Bemrew Admassu Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tirunesh Tsegaw
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayew Demessie
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Kalkidan Getnet
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebe Belete Bitew
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Mebrie Zemene Kinde
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Asnakew Mulaw Beirhun
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yesuneh Tefera Mekasha
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melaku Getahun Feleke
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melkie Dagnaw Fenta
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
17
|
Doktorova M, Symons JL, Zhang X, Wang HY, Schlegel J, Lorent JH, Heberle FA, Sezgin E, Lyman E, Levental KR, Levental I. Cell membranes sustain phospholipid imbalance via cholesterol asymmetry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.30.551157. [PMID: 39713443 PMCID: PMC11661119 DOI: 10.1101/2023.07.30.551157] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Membranes are molecular interfaces that compartmentalize cells to control the flow of nutrients and information. These functions are facilitated by diverse collections of lipids, nearly all of which are distributed asymmetrically between the two bilayer leaflets. Most models of biomembrane structure and function often include the implicit assumption that these leaflets have similar abundances of phospholipids. Here, we show that this assumption is generally invalid and investigate the consequences of lipid abundance imbalances in mammalian plasma membranes (PM). Using quantitative lipidomics, we discovered that cytoplasmic leaflets of human erythrocyte membranes have >50% overabundance of phospholipids compared to exoplasmic leaflets. This imbalance is enabled by an asymmetric interleaflet distribution of cholesterol, which regulates cellular cholesterol homeostasis. These features produce unique functional characteristics, including low PM permeability and resting tension in the cytoplasmic leaflet that regulates protein localization. These largely overlooked aspects of membrane asymmetry represent an evolution of classic paradigms of biomembrane structure and physiology.
Collapse
Affiliation(s)
- Milka Doktorova
- Department of Biochemistry and Biophysics, Stockholm University; Science for Life Laboratory, Solna 171 65, Sweden
| | - Jessica L. Symons
- Department of Integrative Biology & Pharmacology, University of Texas Health Science Center at Houston; Houston, TX 77030, USA
| | - Xiaoxuan Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia; Charlottesville, VA 22903, USA
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia; Charlottesville, VA 22903, USA
| | - Jan Schlegel
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet; 17165 Solna, Sweden
| | - Joseph H. Lorent
- Department of Cellular and Molecular Pharmacology, TFAR, LDRI, UCLouvain; Avenue Mounier 73, B1.73.05, B-1200 Brussels
| | - Frederick A. Heberle
- Department of Chemistry, University of Tennessee Knoxville; Knoxville, TN 37916, USA
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet; 17165 Solna, Sweden
| | - Edward Lyman
- Department of Physics and Astronomy; Department of Chemistry and Biochemistry, University of Delaware; Newark, DE 19716, USA
| | - Kandice R. Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia; Charlottesville, VA 22903, USA
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia; Charlottesville, VA 22903, USA
| |
Collapse
|
18
|
Iacono G, Abay A, Murillo JSG, Aglialoro F, Yagci N, Varga E, Bijlsma T, Sohler J, Fu K, Reisz JA, Argabright A, D'Alessandro A, Svendsen AF, von Lindern M, van den Akker E. Differentiating erythroblasts adapt to mechanical stimulation by upregulation of cholesterol biosynthesis via S1P/SREBP-induced HMGCR expression. Sci Rep 2024; 14:30157. [PMID: 39627481 PMCID: PMC11615233 DOI: 10.1038/s41598-024-81746-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
Understanding how mechanical stress affects erythropoiesis is crucial to produce transfusable erythrocytes in fluid-turbulent bioreactors. We investigated the effects of shear-stress on differentiating CD49d+CD235a+ primary human erythroblasts (EBL) at molecular, cellular, and functional level. Shear-stress, at differentiation onset, enhanced EBL maturation and induced upregulation of genes regulating cholesterol/lipids biosynthesis, causing changes in cell lipid composition. Of note, the osmotic resistance, and the expression of 3-Hydroxy-3-methylglutaryl-CoA reductase (HMGCR), the rate-limiting enzyme of the cholesterol biosynthesis pathway, were higher in dynamic cultures. Inhibition of the S1P-induced proteolytic cleavage, activating SREBPs, led to abrogation of HMCGR expression, and loss of EBL in dynamic cultures, similar to lovastatin administration. This data reveals a role for the S1P-SREBP-HMGCR-axis in the regulation of shear-stress induced adaptation during erythropoiesis, shedding light into mechanisms that will assist the upscaling of erythroid differentiation into bioreactors. Moreover, as shear-stress on hematopoietic cells occurs within the bone-marrow, these results introduce a novel signalling axis in the transduction pathways controlling erythropoiesis.
Collapse
Affiliation(s)
- Giulia Iacono
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands.
| | - Asena Abay
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Joan S Gallego Murillo
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
- Department of Biotechnology Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Francesca Aglialoro
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Nurcan Yagci
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Eszter Varga
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Tieme Bijlsma
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Justine Sohler
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Kerly Fu
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Amy Argabright
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Arthur F Svendsen
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Marieke von Lindern
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Emile van den Akker
- Department of Hematopoiesis Sanquin Research Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Ušaj M, Pavlin M, Kandušer M. Feasibility Study for the Use of Gene Electrotransfer and Cell Electrofusion as a Single-Step Technique for the Generation of Activated Cancer Cell Vaccines. J Membr Biol 2024; 257:377-389. [PMID: 39133276 PMCID: PMC11584437 DOI: 10.1007/s00232-024-00320-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/20/2024] [Indexed: 08/13/2024]
Abstract
Cell-based therapies hold great potential for cancer immunotherapy. This approach is based on manipulation of dendritic cells to activate immune system against specific cancer antigens. For the development of an effective cell vaccine platform, gene transfer, and cell fusion have been used for modification of dendritic or tumor cells to express immune (co)stimulatory signals and to load dendritic cells with tumor antigens. Both, gene transfer and cell fusion can be achieved by single technique, a cell membrane electroporation. The cell membrane exposed to external electric field becomes temporarily permeable, enabling introduction of genetic material, and also fusogenic, enabling the fusion of cells in the close contact. We tested the feasability of combining gene electrotransfer and electrofusion into a single-step technique and evaluated the effects of electroporation buffer, pulse parameters, and cell membrane fluidity for single or combined method of gene delivery or cell fusdion. We determined the percentage of fused cells expressing green fluorescence protein (GFP) in a murine cell model of melanoma B16F1, cell line used in our previous studies. Our results suggest that gene electrotransfer and cell electrofusion can be applied in a single step. The percentage of viable hybrid cells expressing GFP depends on electric pulse parameters and the composition of the electroporation buffer. Furthermore, our results suggest that cell membrane fluidity is not related to the efficiency of the gene electrotransfer and electrofusion. The protocol is compatible with microfluidic devices, however further optimization of electric pulse parameters and buffers is still needed.
Collapse
Affiliation(s)
- Marko Ušaj
- Faculty of Health and Life Sciences, Department of Chemistry and Biomedical Sciences, Linnaeus University, 391 82, Kalmar, Sweden
| | - Mojca Pavlin
- Faculty of Medicine, Institute of Biophysics, University of Ljubljana, Vrazov Trg 2, 1000, Ljubljana, Slovenia
- Faculty of Electrical Engineering, Group for Nano and Biotechnological Applications, University of Ljubljana, Tržaška 25, 1000, Ljubljana, Slovenia
| | - Maša Kandušer
- Institute for Pharmacy, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
20
|
Wessendorf-Rodriguez K, Ruchhoeft ML, Ashley EL, Galvez HM, Murray CW, Huang Y, McGregor GH, Kambhampati S, Shaw RJ, Metallo CM. Modeling compound lipid homeostasis using stable isotope tracing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618599. [PMID: 39463985 PMCID: PMC11507872 DOI: 10.1101/2024.10.16.618599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Lipids represent the most diverse pool of metabolites found in cells, facilitating compartmentation, signaling, and other functions. Dysregulation of lipid metabolism is linked to disease states such as cancer and neurodegeneration. However, limited tools are available for quantifying metabolic fluxes across the lipidome. To directly measure reaction fluxes encompassing compound lipid homeostasis, we applied stable isotope tracing, liquid chromatography-high-resolution mass spectrometry, and network-based isotopologue modeling to non-small cell lung cancer (NSCLC) models. Compound lipid metabolic flux analysis (CL-MFA) enables the concurrent quantitation of fatty acid synthesis, elongation, headgroup assembly, and salvage reactions within virtually any biological system. Here, we resolve liver kinase B1 (LKB1)-mediated regulation of sphingolipid recycling in NSCLC cells and precision-cut lung slice cultures. We also demonstrate that widely used tissue culture conditions drive cells to upregulate fatty acid synthase flux to supraphysiological levels. Finally, we identify previously uncharacterized isozyme specificity of ceramide synthase inhibitors. These results highlight the ability of CL-MFA to quantify lipid cycling in biological systems to discover biological function and elucidate molecular mechanisms in membrane lipid metabolism.
Collapse
|
21
|
Justice I, Kiesel P, Safronova N, von Appen A, Saenz JP. A tuneable minimal cell membrane reveals that two lipid species suffice for life. Nat Commun 2024; 15:9679. [PMID: 39516463 PMCID: PMC11549477 DOI: 10.1038/s41467-024-53975-y] [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: 12/17/2023] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
All cells are encapsulated by a lipid membrane that facilitates their interactions with the environment. How cells manage diverse mixtures of lipids, which dictate membrane property and function, is experimentally challenging to address. Here, we present an approach to tune and minimize membrane lipid composition in the bacterium Mycoplasma mycoides and its derived 'minimal cell' (JCVI-Syn3A), revealing that a two-component lipidome can support life. Systematic reintroduction of phospholipids with different features demonstrates that acyl chain diversity is more important for growth than head group diversity. By tuning lipid chirality, we explore the lipid divide between Archaea and the rest of life, showing that ancestral lipidomes could have been heterochiral. However, in these simple organisms, heterochirality leads to impaired cellular fitness. Thus, our approach offers a tunable minimal membrane system to explore the fundamental lipidomic requirements for life, thereby extending the concept of minimal life from the genome to the lipidome.
Collapse
Affiliation(s)
- Isaac Justice
- Technische Universität Dresden, B CUBE Center for Molecular Bioengineering, Dresden, Germany
| | - Petra Kiesel
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 107, Dresden, Germany
| | - Nataliya Safronova
- Technische Universität Dresden, B CUBE Center for Molecular Bioengineering, Dresden, Germany
| | - Alexander von Appen
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 107, Dresden, Germany
| | - James P Saenz
- Technische Universität Dresden, B CUBE Center for Molecular Bioengineering, Dresden, Germany.
- Technische Universität Dresden, Faculty of Medicine, Dresden, Germany.
| |
Collapse
|
22
|
Bendi A, Vashisth C, Yadav S, Pundeer R, Raghav N. Recent advances in the synthesis of cholesterol-based triazoles and their biological applications. Steroids 2024; 211:109499. [PMID: 39155033 DOI: 10.1016/j.steroids.2024.109499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Double-headed warheads focusing on the pharmacological aspects as well as membrane permeability can contribute a lot to medicinal chemistry. Over the past few decades, a lot of research has been conducted on steroid-heterocycle conjugates as possible therapeutic agents against a variety of disorders. In the second half of the 20th century, successful research was conducted on cholesterol-based heterocyclic moieties. Keeping in view the biological significance of various triazoles, research on fusion with cholesterol has emerged. This review has been designed to explore the chemistry of cholesterol-based triazoles for the duration from 2010 to 2023 and their significance in medicinal chemistry.
Collapse
Affiliation(s)
- Anjaneyulu Bendi
- Department of Chemistry, Presidency University, Rajanukunte, Itgalpura, Bangalore 560064, Karnataka, India
| | - Chanchal Vashisth
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana 136119 India.
| | - Sidhant Yadav
- Department of Chemistry, Indira Gandhi University, Meerpur, Rewari 122502, Haryana, India
| | - Rashmi Pundeer
- Department of Chemistry, Indira Gandhi University, Meerpur, Rewari 122502, Haryana, India
| | - Neera Raghav
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana 136119 India.
| |
Collapse
|
23
|
He J, Yuan Q, Gao S, Wang Y, Lai H, Wang K, Zhou X, Zhang Z. Lipidome analyses reveal radiation induced remodeling of glycerophospholipid unsaturation in lung tumor. Front Immunol 2024; 15:1470269. [PMID: 39512339 PMCID: PMC11540637 DOI: 10.3389/fimmu.2024.1470269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Radiotherapy is a pivotal treatment for lung cancer, significantly impacting tumor control and patient quality of life. Despite its benefits, the molecular mechanisms underlying radiotherapy-induced biological alterations in lung cancer cells remain inadequately understood. In this study, we employed a mass spectrometry-based lipidomics approach to investigate lipid profile changes in a lung cancer mouse model post-radiation. Lewis lung carcinoma (LLC) cells were injected into C57BL/6J mice, followed by radiation treatment with varying split doses. Our results showed an increase in sterol lipids and a decrease in glycerolipids, specifically triacylglycerides, indicating disrupted lipid storage. Additionally, we observed significant changes in glycerophospholipid unsaturation, suggesting a remodeling of membrane properties that may influence cell survival. Linear regression analysis demonstrated a significant negative correlation between glycerophospholipid unsaturation index and tumor weight, indicating a potential role in radiation-induced tumor cell death. These findings provide new insights into the lipid metabolic pathways affected by radiotherapy and could inform the development of improved therapeutic strategies for lung cancer treatment.
Collapse
Affiliation(s)
- Jingquan He
- Department of Radiotherapy, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College, Guangzhou Traditional Chinese Medicine University, Shenzhen, China
| | - Qingqing Yuan
- Department of Radiation Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Song Gao
- Department of Radiotherapy, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College, Guangzhou Traditional Chinese Medicine University, Shenzhen, China
| | - Yue Wang
- Department of Radiotherapy, Shenzhen Luohu People’s Hospital, Shenzhen, China
| | - Haigen Lai
- Department of Radiotherapy, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College, Guangzhou Traditional Chinese Medicine University, Shenzhen, China
| | - Kaiting Wang
- Department of Radiotherapy, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College, Guangzhou Traditional Chinese Medicine University, Shenzhen, China
| | - Xiaoman Zhou
- Department of Radiotherapy, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College, Guangzhou Traditional Chinese Medicine University, Shenzhen, China
| | - Zicheng Zhang
- Department of Radiotherapy, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College, Guangzhou Traditional Chinese Medicine University, Shenzhen, China
| |
Collapse
|
24
|
Justice I, Kiesel P, Safronova N, von Appen A, Saenz JP. A tuneable minimal cell membrane reveals that two lipid species suffice for life. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.24.563757. [PMID: 39464110 PMCID: PMC11507672 DOI: 10.1101/2023.10.24.563757] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
All cells are encapsulated by a lipid membrane which facilitates the interaction between life and its environment. How life exploits the diverse mixtures of lipids that dictate membrane property and function has been experimentally challenging to address. We introduce an approach to tune and minimize lipidomes in Mycoplasma mycoides and the Minimal Cell (JCVI-Syn3A) revealing that a 2-component lipidome can support life. Systematically reintroducing phospholipid features demonstrated that acyl chain diversity is more critical for growth than head group diversity. By tuning lipid chirality, we explored the lipid divide between Archaea and the rest of life, showing that ancestral lipidomes could have been heterochiral. Our approach offers a tunable minimal membrane system to explore the fundamental lipidomic requirements for life, thereby extending the concept of minimal life from the genome to the lipidome.
Collapse
Affiliation(s)
- Isaac Justice
- Technische Universität Dresden, B CUBE Center for Molecular Bioengineering, 01307 Dresden, Germany
| | - Petra Kiesel
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 107, 01307 Dresden
| | - Nataliya Safronova
- Technische Universität Dresden, B CUBE Center for Molecular Bioengineering, 01307 Dresden, Germany
| | - Alexander von Appen
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 107, 01307 Dresden
| | - James P. Saenz
- Technische Universität Dresden, B CUBE Center for Molecular Bioengineering, 01307 Dresden, Germany
- Technische Universität Dresden, Faculty of Medicine, Dresden 01307, Germany
| |
Collapse
|
25
|
Cruz-Martínez Y, Cantú K, Ojeda G, Gálvez-Susano V, Arias-Santiago S, Ibarra-García AP, Borlongan CV, Carrasco-Vargas H, Antonio Vargas-Hernández M, Ibarra A. Two-phase therapy for improving neuroprotection and neurogenesis: Preventive use of omega fatty acids plus Copolymer-1 immunization after stroke. Brain Res 2024; 1846:149277. [PMID: 39406314 DOI: 10.1016/j.brainres.2024.149277] [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: 08/14/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
Stroke is a major global health issue, ranking as the second leading cause of death and the primary cause of disability worldwide. However, current therapeutic options remain limited. Nutritional supplementation as a form of primary prevention stands as a potential stroke therapeutic. In particular, the intake of omega-3 fatty acids (omega-3FA) exerts anti-inflammatory and neuroprotective effects that help reduce the risk of stroke. In parallel, treatment with Copolymer-1 (COP-1), a peptide with immunomodulatory properties through Th1/Th2/Th3 phenotype switching, similarly affords neuroprotective and neurorestorative effects in stroke models. To investigate the combined effects of these treatments, we designed a two-phase therapy: the first phase involved preventive supplementation with omega-3FA, while the second phase included COP-1 immunization following stroke injury. Sprague-Dawley rats were randomly assigned to one of the four groups: 1) control, 2) omega-3FA, 3) COP-1, and 4) omega-3FA + COP-1. Omega-3FAs were administered for 28 days before inducing stroke. Thirty minutes after reperfusion, the respective groups were immunized with COP-1. Seven days post-stroke, neurological deficits were assessed using the Zea-Longa scale, infarct volumes with 2,3,5-triphenyltetrazolium chloride (TTC) staining, and levels of neurogenesis via immunofluorescence imaging. The results showed that the two-phase therapy produced significant synergistic effects, markedly reducing neurological deficits, and infarct volumes, while enhancing neurogenic activities in neurogenic niches. This combined approach underscores the potential of integrating nutritional and pharmacological strategies to enhance stroke recovery.
Collapse
Affiliation(s)
- Yolanda Cruz-Martínez
- Centro de investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilican CP52786, Edo. de México, México.
| | - Karla Cantú
- Centro de investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilican CP52786, Edo. de México, México.
| | - Gerardo Ojeda
- Centro de investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilican CP52786, Edo. de México, México
| | - Vanessa Gálvez-Susano
- Centro de investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilican CP52786, Edo. de México, México
| | - Stella Arias-Santiago
- Centro de investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilican CP52786, Edo. de México, México
| | - Andrea P Ibarra-García
- Centro de investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilican CP52786, Edo. de México, México
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| | - Humberto Carrasco-Vargas
- Escuela Militar de Graduados en Sanidad, Ciudad de México, México; Secretaria de la Defensa Nacional, Ciudad de México, México
| | - Marco Antonio Vargas-Hernández
- Escuela Militar de Graduados en Sanidad, Ciudad de México, México; Secretaria de la Defensa Nacional, Ciudad de México, México
| | - Antonio Ibarra
- Centro de investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilican CP52786, Edo. de México, México; Escuela Militar de Graduados en Sanidad, Ciudad de México, México; Secretaria de la Defensa Nacional, Ciudad de México, México.
| |
Collapse
|
26
|
Janež Š, Guzelj S, Kocbek P, de Vlieger EA, Slütter B, Jakopin Ž. Distinctive Immune Signatures Driven by Structural Alterations in Desmuramylpeptide NOD2 Agonists. J Med Chem 2024; 67:17585-17607. [PMID: 39344184 PMCID: PMC11472310 DOI: 10.1021/acs.jmedchem.4c01577] [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: 07/10/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Herein we report on the design, synthesis and biological evaluation of a series of nucleotide-binding oligomerization-domain-containing protein 2 (NOD2) desmuramylpeptide agonists. The structural prerequisites that shape both physicochemical and immunomodulatory profiles of desmuramylpeptide NOD2 agonists have been delineated. Within this context, we identified 3, a butyrylated desmuramylpeptide, as a potent in vitro NOD2 agonist (EC50 = 4.6 nM), exhibiting an almost 17-fold enhancement in potency compared to its unsubstituted counterpart 1 (EC50 = 77.0 nM). The novel set of desmuramylpeptides demonstrate unique in vitro immunomodulatory activities. They elicited cytokine production in peripheral blood mononuclear cells (PBMCs), both alone and in conjunction with lipopolysaccharide (LPS). The spermine-decorated 32 also stimulated the LPS-induced cytotoxic activity (2.95-fold) of PBMCs against K562 cancer cells. Notably, the cholesterol-conjugate 26 displayed anti-inflammatory actions, highlighted by its capacity to convert the inflammatory monocyte subset into an anti-inflammatory phenotype. Finally, the eicosapentaenoylated derivative 23 augmented antigen presentation by mouse bone marrow-derived dendritic cells (BMDCs), thus highlighting its potential as a vaccine adjuvant.
Collapse
Affiliation(s)
- Špela Janež
- Faculty
of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Samo Guzelj
- Faculty
of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Petra Kocbek
- Faculty
of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Eveline A. de Vlieger
- Div.
BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Bram Slütter
- Div.
BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Žiga Jakopin
- Faculty
of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
27
|
Fayezi S, Oehms S, Wolff von Gudenberg H, Ponnaiah M, Lhomme M, Strowitzki T, Germeyer A. De novo synthesis of monounsaturated fatty acids modulates exosome-mediated lipid export from human granulosa cells. Mol Cell Endocrinol 2024; 592:112317. [PMID: 38901632 DOI: 10.1016/j.mce.2024.112317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/21/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Ovarian somatic cells support the maturation and fertility of oocytes. Metabolic desaturation of fatty acids in these cells has a positive paracrine impact on the maturation of oocytes. We hypothesized that the enzyme stearoyl-CoA desaturase 1 (SCD1) in granulosa cells regulates the lipid cargo of exosomes secreted from these cells by maintaining the balance between saturated and unsaturated lipids. We investigated the effect of SCD1 on exosome lipid content in a cumulus-granulosa cell model under physiologically relevant in vitro conditions. METHODS Non-luteinized human COV434 granulosa cells were subjected to treatment with an inhibitor of SCD1 (SCDinhib) alone, in combination with oleic acid, or under control conditions. Subsequently, the exosomes were isolated and characterized via nanoparticle tracking analysis, transmission electron microscopy, and Western blotting. We used liquid chromatography mass spectrometry to investigate the lipidomic profiles. We used quantitative PCR with TaqMan primers to assess the expression of genes involved in lipogenesis and control of cell cycle progression. RESULTS A trend toward exosome production was observed with a shift toward smaller exosome sizes in cells treated with SCD1inhib. This trend reached statistical significance when SCDinhib was combined with oleic acid supplementation. SCD1 inhibition led to the accumulation of saturated omega-6 lipids in exosomes. The latter effect was reversed by oleic acid supplementation, which also improved exosome production and suppressed the expression of fatty acid synthase and Cyclin D2. CONCLUSION These findings underscore the critical role of de novo fatty acid desaturation in the regulation of the export of specific lipids through exosomes, with potential implications for controlling intercellular communication within the ovary.
Collapse
Affiliation(s)
- Shabnam Fayezi
- Department of Gynecological Endocrinology and Fertility Disorders, Women's Hospital, University of Heidelberg, 69120 Heidelberg, Germany.
| | - Sophie Oehms
- Department of Gynecological Endocrinology and Fertility Disorders, Women's Hospital, University of Heidelberg, 69120 Heidelberg, Germany
| | - Helena Wolff von Gudenberg
- Department of Gynecological Endocrinology and Fertility Disorders, Women's Hospital, University of Heidelberg, 69120 Heidelberg, Germany
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), ICAN I/O - Data Sciences (MP), ICAN Omics (ML), 75013 Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), ICAN I/O - Data Sciences (MP), ICAN Omics (ML), 75013 Paris, France
| | - Thomas Strowitzki
- Department of Gynecological Endocrinology and Fertility Disorders, Women's Hospital, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ariane Germeyer
- Department of Gynecological Endocrinology and Fertility Disorders, Women's Hospital, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
28
|
Amoakon JP, Lee J, Liyanage P, Arora K, Karlstaedt A, Mylavarapu G, Amin R, Naren AP. Defective CFTR modulates mechanosensitive channels TRPV4 and PIEZO1 and drives endothelial barrier failure. iScience 2024; 27:110703. [PMID: 39252977 PMCID: PMC11382128 DOI: 10.1016/j.isci.2024.110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/25/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Despite reports of CFTR expression on endothelial cells, pulmonary vascular perturbations, and perfusion deficits in CF patients, the mechanism of pulmonary vascular disease in CF remains unclear. Here, our pilot study of 40 CF patients reveals a loss of small pulmonary blood vessels in patients with severe lung disease. Using a vessel-on-a-chip model, we establish a shear-stress-dependent mechanism of endothelial barrier failure in CF involving TRPV4, a mechanosensitive channel. Furthermore, we demonstrate that CFTR deficiency downregulates the function of PIEZO1, another mechanosensitive channel involved in angiogenesis and wound repair, and exacerbates loss of small pulmonary blood vessel. We also show that CFTR directly interacts with PIEZO1 and enhances its function. Our study identifies key cellular targets to mitigate loss of small pulmonary blood vessels in CF.
Collapse
Affiliation(s)
- Jean-Pierre Amoakon
- Department of Systems Biology and Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jesun Lee
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Pramodha Liyanage
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kavisha Arora
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Goutham Mylavarapu
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Raouf Amin
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anjaparavanda P Naren
- Department of Systems Biology and Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
29
|
Zhang H, Liu D, Zhang J, Adams E, Gong J, Li W, Wang B, Liu X, Yang R, Wei F, Allen HC. GMP affected assembly behaviors of phosphatidylethanolamine monolayers elucidated by multi-resolved SFG-VS and BAM. Colloids Surf B Biointerfaces 2024; 241:113995. [PMID: 38870647 DOI: 10.1016/j.colsurfb.2024.113995] [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: 12/18/2023] [Revised: 05/18/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024]
Abstract
The interaction between nucleotide molecules and lipid molecules plays important roles in cell activities, but the molecular mechanism is very elusive. In the present study, a small but noticeable interaction between the negatively charged phosphatidylethanolamine (PE) and Guanosine monophosphate (GMP) molecules was observed from the PE monolayer at the air/water interface. As shown by the sum frequency generation (SFG) spectra and Pi-A isotherm of the PE monolayer, the interaction between the PE and GMP molecules imposes very small changes to the PE molecules. However, the Brewster angle microscopy (BAM) technique revealed that the assembly conformations of PE molecules are significantly changed by the adsorption of GMP molecules. By comparing the SFG spectra of PE monolayers after the adsorption of GMP, guanosine and guanine, it is also shown that the hydrogen bonding effect plays an important role in the nucleotide-PE interactions. These results provide fundamental insight into the structure changes during the nucleotide-lipid interaction, which may shed light on the molecular mechanism of viral infection, DNA drug delivery, and cell membrane curvature control in the brain or neurons.
Collapse
Affiliation(s)
- Hongjuan Zhang
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Dongqi Liu
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Jiawei Zhang
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Ellen Adams
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, OH 43210, USA
| | - Jingjing Gong
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Wenhui Li
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Bing Wang
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Xueqing Liu
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Renqiang Yang
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Feng Wei
- School of Optoelectronic Materials and Technology, & Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, Jianghan University, Wuhan 430056, China.
| | - Heather C Allen
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
30
|
Wang Y, Becker S, Finkelstein S, Dyka FM, Liu H, Eminhizer M, Hao Y, Brush RS, Spencer WJ, Arshavsky VY, Ash JD, Du J, Agbaga MP, Vinberg F, Ellis JM, Lobanova ES. Acyl-CoA synthetase 6 controls rod photoreceptor function and survival by shaping the phospholipid composition of retinal membranes. Commun Biol 2024; 7:1027. [PMID: 39169121 PMCID: PMC11339274 DOI: 10.1038/s42003-024-06691-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
The retina is light-sensitive neuronal tissue in the back of the eye. The phospholipid composition of the retina is unique and highly enriched in polyunsaturated fatty acids, including docosahexaenoic fatty acid (DHA). While it is generally accepted that a high DHA content is important for vision, surprisingly little is known about the mechanisms of DHA enrichment in the retina. Furthermore, the biological processes controlled by DHA in the eye remain poorly defined as well. Here, we combined genetic manipulations with lipidomic analysis in mice to demonstrate that acyl-CoA synthetase 6 (Acsl6) serves as a regulator of the unique composition of retinal membranes. Inactivation of Acsl6 reduced the levels of DHA-containing phospholipids, led to progressive loss of light-sensitive rod photoreceptor neurons, attenuated the light responses of these cells, and evoked distinct transcriptional response in the retina involving the Srebf1/2 (sterol regulatory element binding transcription factors 1/2) pathway. This study identifies one of the major enzymes responsible for DHA enrichment in the retinal membranes and introduces a model allowing an evaluation of rod functioning and pathology caused by impaired DHA incorporation/retention in the retina.
Collapse
Affiliation(s)
- Yixiao Wang
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Silke Becker
- Department of Ophthalmology, University of Utah, Salt Lake City, UT, USA
| | | | - Frank M Dyka
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haitao Liu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Eminhizer
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
| | - Ying Hao
- Department of Ophthalmology, Duke University, Durham, NC, USA
| | - Richard S Brush
- Department of Ophthalmology, University of Oklahoma Health Sciences Center and Dean McGee Eye Institute, Oklahoma City, OK, USA
| | - William J Spencer
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - John D Ash
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianhai Du
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
| | - Martin-Paul Agbaga
- Department of Ophthalmology, University of Oklahoma Health Sciences Center and Dean McGee Eye Institute, Oklahoma City, OK, USA
| | - Frans Vinberg
- Department of Ophthalmology, University of Utah, Salt Lake City, UT, USA
| | | | | |
Collapse
|
31
|
Brunner S, Höring M, Liebisch G, Schweizer S, Scheiber J, Giansanti P, Hidrobo M, Hermeling S, Oeckl J, Prudente de Mello N, Perocchi F, Seeliger C, Strohmeyer A, Klingenspor M, Plagge J, Küster B, Burkhardt R, Janssen KP, Ecker J. Mitochondrial lipidomes are tissue specific - low cholesterol contents relate to UCP1 activity. Life Sci Alliance 2024; 7:e202402828. [PMID: 38843936 PMCID: PMC11157264 DOI: 10.26508/lsa.202402828] [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: 05/17/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
Lipid composition is conserved within sub-cellular compartments to maintain cell function. Lipidomic analyses of liver, muscle, white and brown adipose tissue (BAT) mitochondria revealed substantial differences in their glycerophospholipid (GPL) and free cholesterol (FC) contents. The GPL to FC ratio was 50-fold higher in brown than white adipose tissue mitochondria. Their purity was verified by comparison of proteomes with ER and mitochondria-associated membranes. A lipid signature containing PC and FC, calculated from the lipidomic profiles, allowed differentiation of mitochondria from BAT of mice housed at different temperatures. Elevating FC in BAT mitochondria prevented uncoupling protein (UCP) 1 function, whereas increasing GPL boosted it. Similarly, STARD3 overexpression facilitating mitochondrial FC import inhibited UCP1 function in primary brown adipocytes, whereas a knockdown promoted it. We conclude that the mitochondrial GPL/FC ratio is key for BAT function and propose that targeting it might be a promising strategy to promote UCP1 activity.
Collapse
Affiliation(s)
- Sarah Brunner
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Sabine Schweizer
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| | | | - Piero Giansanti
- Bavarian Center for Biomolecular Mass Spectrometry at the University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Maria Hidrobo
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| | - Sven Hermeling
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| | - Josef Oeckl
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Natalia Prudente de Mello
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München and German National Diabetes Center (DZD), Neuherberg, Germany
- Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians University, Munich, Germany
| | - Fabiana Perocchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München and German National Diabetes Center (DZD), Neuherberg, Germany
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- Munich Cluster of Systems Neurology, Munich, Germany
| | - Claudine Seeliger
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| | - Akim Strohmeyer
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Johannes Plagge
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| | - Bernhard Küster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
- Bavarian Biomolecular Mass Spectrometry Center, Technical University of Munich, Freising, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich Germany
| | - Josef Ecker
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| |
Collapse
|
32
|
Ruiz M, Devkota R, Bergh PO, Nik AM, Blid Sköldheden S, Mondejar-Duran J, Tufvesson-Alm M, Bohlooly-Y M, Sanchez D, Carlsson P, Henricsson M, Jerlhag E, Borén J, Pilon M. Aging AdipoR2-deficient mice are hyperactive with enlarged brains excessively rich in saturated fatty acids. FASEB J 2024; 38:e23815. [PMID: 38989587 DOI: 10.1096/fj.202400293rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/04/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
To investigate how the fatty acid composition of brain phospholipids influences brain-specific processes, we leveraged the AdipoR2 (adiponectin receptor 2) knockout mouse model in which the brain is enlarged, and cellular membranes are excessively rich in saturated fatty acids. Lipidomics analysis of brains at 2, 7, and 18 months of age showed that phosphatidylcholines, which make up about two-thirds of all cerebrum membrane lipids, contain a gross excess of saturated fatty acids in AdipoR2 knockout mice, and that this is mostly attributed to an excess palmitic acid (C16:0) at the expense of oleic acid (C18:1), consistent with a defect in fatty acid desaturation and elongation in the mutant. Specifically, there was a ~12% increase in the overall saturated fatty acid content within phosphatidylcholines and a ~30% increase in phosphatidylcholines containing two palmitic acids. Phosphatidylethanolamines, sphingomyelins, ceramides, lactosylceramides, and dihydroceramides also showed an excess of saturated fatty acids in the AdipoR2 knockout mice while nervonic acid (C24:1) was enriched at the expense of shorter saturated fatty acids in glyceroceramides. Similar defects were found in the cerebellum and myelin sheaths. Histology showed that cell density is lower in the cerebrum of AdipoR2 knockout mice, but electron microscopy did not detect reproducible defects in the ultrastructure of cerebrum neurons, though proteomics analysis showed an enrichment of electron transport chain proteins in the cerebellum. Behavioral tests showed that older (33 weeks old) AdipoR2 knockout mice are hyperactive and anxious compared to control mice of a similar age. Also, in contrast to control mice, the AdipoR2 knockout mice do not gain weight in old age but do have normal lifespans. We conclude that an excess fatty acid saturation in brain phospholipids is accompanied by hyperactivity but seems otherwise well tolerated.
Collapse
Affiliation(s)
- Mario Ruiz
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Ranjan Devkota
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Per-Olof Bergh
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ali Moussavi Nik
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Blid Sköldheden
- Department of Pharmacology, Institute of Neuroscience and physiology, University of Gothenburg, Gothenburg, Sweden
| | - Jorge Mondejar-Duran
- Instituto de Biomedicina y Genética Molecular, Excellence Unit, University of Valladolid-CSIC, Valladolid, Spain
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and physiology, University of Gothenburg, Gothenburg, Sweden
| | | | - Diego Sanchez
- Instituto de Biomedicina y Genética Molecular, Excellence Unit, University of Valladolid-CSIC, Valladolid, Spain
| | - Peter Carlsson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and physiology, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
33
|
Longarzo ML, Vázquez RF, Bellini MJ, Zamora RA, Redondo-Morata L, Giannotti MI, Oliveira Jr ON, Fanani ML, Maté SM. Understanding the effects of omega-3 fatty acid supplementation on the physical properties of brain lipid membranes. iScience 2024; 27:110362. [PMID: 39071883 PMCID: PMC11277689 DOI: 10.1016/j.isci.2024.110362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/24/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
A deficiency in omega-3 fatty acids (ω3 FAs) in the brain has been correlated with cognitive impairment, learning deficiencies, and behavioral changes. In this study, we provided ω3 FAs as a supplement to spontaneously hypertensive rats (SHR+ ω3). Our focus was on examining the impact of dietary supplementation on the physicochemical properties of the brain-cell membranes. Significant increases in ω3 levels in the cerebral cortex of SHR+ ω3 were observed, leading to alterations in brain lipid membranes molecular packing, elasticity, and lipid miscibility, resulting in an augmented phase disparity. Results from synthetic lipid mixtures confirmed the disordering effect introduced by ω3 lipids, showing its consequences on the hydration levels of the monolayers and the organization of the membrane domains. These findings suggest that dietary ω3 FAs influence the organization of brain membranes, providing insight into a potential mechanism for the broad effects of dietary fat on brain health and disease.
Collapse
Affiliation(s)
- María L. Longarzo
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, (1900), La Plata, Argentina
| | - Romina F. Vázquez
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, (1900), La Plata, Argentina
| | - María J. Bellini
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, (1900), La Plata, Argentina
| | - Ricardo A. Zamora
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Instituto de Investigación Interdisciplinaria (I³), Vicerrectoría Académica, and Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Campus Lircay, Talca 3460000, Chile
| | - Lorena Redondo-Morata
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL—Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Marina I. Giannotti
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- CIBER-BBN, ISCIII, 08028 Barcelona, Spain
- Department of Materials Science and Physical Chemistry, University of Barcelona, 08028 Barcelona, Spain
| | - Osvaldo N. Oliveira Jr
- São Carlos Institute of Physics (IFSC-USP), University of São Paulo, 13566-590 São Carlos, São Paulo, Brazil
| | - María L. Fanani
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Cordoba, Argentina
- Departamento de Química Biológica Raquel Caputto, Facultad de Cs. Químicas, Universidad Nacional de Córdoba. Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| | - Sabina M. Maté
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, (1900), La Plata, Argentina
| |
Collapse
|
34
|
Liu CH, Shen PC, Lin WJ, Liu HC, Tsai MH, Huang TY, Chen IC, Lai YL, Wang YD, Hung MC, Cheng WC. LipidSig 2.0: integrating lipid characteristic insights into advanced lipidomics data analysis. Nucleic Acids Res 2024; 52:W390-W397. [PMID: 38709887 PMCID: PMC11223864 DOI: 10.1093/nar/gkae335] [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: 02/08/2024] [Revised: 03/29/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
In the field of lipidomics, where the complexity of lipid structures and functions presents significant analytical challenges, LipidSig stands out as the first web-based platform providing integrated, comprehensive analysis for efficient data mining of lipidomic datasets. The upgraded LipidSig 2.0 (https://lipidsig.bioinfomics.org/) simplifies the process and empowers researchers to decipher the complex nature of lipids and link lipidomic data to specific characteristics and biological contexts. This tool markedly enhances the efficiency and depth of lipidomic research by autonomously identifying lipid species and assigning 29 comprehensive characteristics upon data entry. LipidSig 2.0 accommodates 24 data processing methods, streamlining diverse lipidomic datasets. The tool's expertise in automating intricate analytical processes, including data preprocessing, lipid ID annotation, differential expression, enrichment analysis, and network analysis, allows researchers to profoundly investigate lipid properties and their biological implications. Additional innovative features, such as the 'Network' function, offer a system biology perspective on lipid interactions, and the 'Multiple Group' analysis aids in examining complex experimental designs. With its comprehensive suite of features for analyzing and visualizing lipid properties, LipidSig 2.0 positions itself as an indispensable tool for advanced lipidomics research, paving the way for new insights into the role of lipids in cellular processes and disease development.
Collapse
Affiliation(s)
- Chia-Hsin Liu
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
| | - Pei-Chun Shen
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
| | - Wen-Jen Lin
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
- School of Medicine, China Medical University, Taichung 404328, Taiwan
| | - Hsiu-Cheng Liu
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
| | - Meng-Hsin Tsai
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
| | - Tzu-Ya Huang
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
| | - I-Chieh Chen
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
| | - Yo-Liang Lai
- Department of Radiation Oncology, China Medical University, Taichung 404328, Taiwan
| | - Yu-De Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404328, Taiwan
- Department of Urology, China Medical University, Taichung 404328, Taiwan
| | - Mien-Chie Hung
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 404328, Taiwan
- Molecular Medicine Center, China Medical University Hospital, China Medical University, Taichung 404328, Taiwan
- Department of Biotechnology, Asia University, Taichung 413305, Taiwan
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 404328, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404328, Taiwan
- The Ph.D. program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 404328, Taiwan
| |
Collapse
|
35
|
Hart NR. Paradoxes: Cholesterol and Hypoxia in Preeclampsia. Biomolecules 2024; 14:691. [PMID: 38927094 PMCID: PMC11201883 DOI: 10.3390/biom14060691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Preeclampsia, a hypertensive disease of pregnancy of unknown etiology, is intensely studied as a model of cardiovascular disease (CVD) not only due to multiple shared pathologic elements but also because changes that develop over decades in CVD appear and resolve within days in preeclampsia. Those affected by preeclampsia and their offspring experience increased lifetime risks of CVD. At the systemic level, preeclampsia is characterized by increased cellular, membrane, and blood levels of cholesterol; however, cholesterol-dependent signaling, such as canonical Wnt/βcatenin, Hedgehog, and endothelial nitric oxide synthase, is downregulated indicating a cholesterol deficit with the upregulation of cholesterol synthesis and efflux. Hypoxia-related signaling in preeclampsia also appears to be paradoxical with increased Hypoxia-Inducible Factors in the placenta but measurably increased oxygen in maternal blood in placental villous spaces. This review addresses the molecular mechanisms by which excessive systemic cholesterol and deficient cholesterol-dependent signaling may arise from the effects of dietary lipid variance and environmental membrane modifiers causing the cellular hypoxia that characterizes preeclampsia.
Collapse
Affiliation(s)
- Nancy R Hart
- PeaceHealth St. Joseph Medical Center, Bellingham, WA 98225, USA
| |
Collapse
|
36
|
Ding S, Grossi V, Hopmans EC, Bale NJ, Cravo-Laureau C, Sinninghe Damsté JS. Nitrogen and sulfur for phosphorus: Lipidome adaptation of anaerobic sulfate-reducing bacteria in phosphorus-deprived conditions. Proc Natl Acad Sci U S A 2024; 121:e2400711121. [PMID: 38833476 PMCID: PMC11181052 DOI: 10.1073/pnas.2400711121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024] Open
Abstract
Understanding how microbial lipidomes adapt to environmental and nutrient stress is crucial for comprehending microbial survival and functionality. Certain anaerobic bacteria can synthesize glycerolipids with ether/ester bonds, yet the complexities of their lipidome remodeling under varying physicochemical and nutritional conditions remain largely unexplored. In this study, we thoroughly examined the lipidome adaptations of Desulfatibacillum alkenivorans strain PF2803T, a mesophilic anaerobic sulfate-reducing bacterium known for its high proportions of alkylglycerol ether lipids in its membrane, under various cultivation conditions including temperature, pH, salinity, and ammonium and phosphorous concentrations. Employing an extensive analytical and computational lipidomic methodology, we identified an assemblage of nearly 400 distinct lipids, including a range of glycerol ether/ester lipids with various polar head groups. Information theory-based analysis revealed that temperature fluctuations and phosphate scarcity profoundly influenced the lipidome's composition, leading to an enhanced diversity and specificity of novel lipids. Notably, phosphorous limitation led to the biosynthesis of novel glucuronosylglycerols and sulfur-containing aminolipids, termed butyramide cysteine glycerols, featuring various ether/ester bonds. This suggests a novel adaptive strategy for anaerobic heterotrophs to thrive under phosphorus-depleted conditions, characterized by a diverse array of nitrogen- and sulfur-containing polar head groups, moving beyond a reliance on conventional nonphospholipid types.
Collapse
Affiliation(s)
- Su Ding
- Department of Marine Microbiology and Biogeochemistry, NIOZ Royal Netherlands Institute for Sea Research, TexelSZ 1797, The Netherlands
| | - Vincent Grossi
- Laboratoire de Géologie de Lyon: Terre, Planètes, Environnement, CNRS, Université Claude Bernard Lyon 1, Villeurbanne69622, France
| | - Ellen C. Hopmans
- Department of Marine Microbiology and Biogeochemistry, NIOZ Royal Netherlands Institute for Sea Research, TexelSZ 1797, The Netherlands
| | - Nicole J. Bale
- Department of Marine Microbiology and Biogeochemistry, NIOZ Royal Netherlands Institute for Sea Research, TexelSZ 1797, The Netherlands
| | - Cristiana Cravo-Laureau
- Institut des Sciences Analytiques et de Physico-chimie pour l’environnement et les Matériaux, Universite de Pau et des Pays de l’Adour, CNRS, Pau64000, France
| | - Jaap S. Sinninghe Damsté
- Department of Marine Microbiology and Biogeochemistry, NIOZ Royal Netherlands Institute for Sea Research, TexelSZ 1797, The Netherlands
- Department of Earth Sciences, Faculty of Geosciences, Utrecht University, Utrecht, CB3584, The Netherlands
| |
Collapse
|
37
|
Pennington ER, Virk R, Bridges MD, Bathon BE, Beatty N, Gray RS, Kelley P, Wassall SR, Manke J, Armstrong M, Reisdorph N, Vanduinen R, Fenton JI, Gowdy KM, Shaikh SR. Docosahexaenoic Acid Controls Pulmonary Macrophage Lipid Raft Size and Inflammation. J Nutr 2024; 154:1945-1958. [PMID: 38582385 PMCID: PMC11217028 DOI: 10.1016/j.tjnut.2024.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Docosahexaenoic acid (DHA) controls the biophysical organization of plasma membrane sphingolipid/cholesterol-enriched lipid rafts to exert anti-inflammatory effects, particularly in lymphocytes. However, the impact of DHA on the spatial arrangement of alveolar macrophage lipid rafts and inflammation is unknown. OBJECTIVES The primary objective was to determine how DHA controls lipid raft organization and function of alveolar macrophages. As proof-of-concept, we also investigated DHA's anti-inflammatory effects on select pulmonary inflammatory markers with a murine influenza model. METHODS MH-S cells, an alveolar macrophage line, were treated with 50 μM DHA or vehicle control and were used to study plasma membrane molecular organization with fluorescence-based methods. Biomimetic membranes and coarse grain molecular dynamic (MD) simulations were employed to investigate how DHA mechanistically controls lipid raft size. qRT-PCR, mass spectrometry, and ELISAs were used to quantify downstream inflammatory signaling transcripts, oxylipins, and cytokines, respectively. Lungs from DHA-fed influenza-infected mice were analyzed for specific inflammatory markers. RESULTS DHA increased the size of lipid rafts while decreasing the molecular packing of the MH-S plasma membrane. Adding a DHA-containing phospholipid to a biomimetic lipid raft-containing membrane led to condensing, which was reversed with the removal of cholesterol. MD simulations revealed DHA nucleated lipid rafts by driving cholesterol and sphingomyelin into rafts. Downstream of the plasma membrane, DHA lowered the concentration of select inflammatory transcripts, oxylipins, and IL-6 secretion. DHA lowered pulmonary Il6 and Tnf-α mRNA expression and increased anti-inflammatory oxylipins of influenza-infected mice. CONCLUSIONS The data suggest a model in which the localization of DHA acyl chains to nonrafts is driving sphingomyelin and cholesterol molecules into larger lipid rafts, which may serve as a trigger to impede signaling and lower inflammation. These findings also identify alveolar macrophages as a target of DHA and underscore the anti-inflammatory properties of DHA for lung inflammation.
Collapse
Affiliation(s)
- Edward Ross Pennington
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, NC, United States
| | - Rafia Virk
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, NC, United States
| | - Meagan D Bridges
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, NC, United States
| | - Brooke E Bathon
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, NC, United States
| | - Nari Beatty
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, NC, United States
| | - Rosemary S Gray
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, NC, United States
| | - Patrick Kelley
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Stephen R Wassall
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Jonathan Manke
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rachel Vanduinen
- Department of Food Science and Human Nutrition, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, United States
| | - Jenifer I Fenton
- Department of Food Science and Human Nutrition, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, United States
| | - Kymberly M Gowdy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, the Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, NC, United States.
| |
Collapse
|
38
|
Mak KM, Shekhar AC. Soybean polyenylphosphatidylcholine (PPC) is beneficial in liver and extrahepatic tissue injury: An update in experimental research. Anat Rec (Hoboken) 2024; 307:2162-2186. [PMID: 37814787 DOI: 10.1002/ar.25333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/11/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023]
Abstract
Polyenylphosphatidylcholine (PPC) is a purified polyunsaturated phosphatidylcholine extract of soybeans. This article updates PPC's beneficial effects on various forms of liver cell injury and other tissues in experimental research. PPC downregulates hepatocyte CYP2E1 expression and associated hepatotoxicity, as well as attenuates oxidative stress, apoptosis, lipoprotein oxidation and steatosis in alcoholic and nonalcoholic liver injury. PPC inhibits pro-inflammatory cytokine production, while stimulating anti-inflammatory cytokine secretion in ethanol or lipopolysaccharide-stimulated Kupffer cells/macrophages. It promotes M2-type macrophage polarization and metabolic reprogramming of glucose and lipid metabolism. PPC mitigates steatosis in NAFLD through inhibiting polarization of pro-inflammatory M1-type Kupffer cells, alleviating metabolic inflammation, remodeling hepatic lipid metabolism, correcting imbalances between lipogenesis and lipolysis and enhancing lipoprotein secretion from hepatocytes. PPC is antifibrotic by preventing progression of alcoholic hepatic fibrosis in baboons and also prevents CCl4-induced fibrosis in rats. PPC supplementation replenishes the phosphatidylcholine content of damaged cell membranes, resulting in increased membrane fluidity and functioning. Phosphatidylcholine repletion prevents increased membrane curvature of the endoplasmic reticulum and Golgi and decreases sterol regulatory element binding protein-1-mediated lipogenesis, reducing steatosis. PPC remodels gut microbiota and affects hepatic lipid metabolism via the gut-hepatic-axis and also alleviates brain inflammatory responses and cognitive impairment via the gut-brain-axis. Additionally, PPC protects extrahepatic tissues from injury caused by various toxic compounds by reducing oxidative stress, inflammation, and membrane damage. It also stimulates liver regeneration, enhances sensitivity of cancer cells to radiotherapy/chemotherapy, and inhibits experimental hepatocarcinogenesis. PPC's beneficial effects justify it as a supportive treatment of liver disease.
Collapse
Affiliation(s)
- Ki M Mak
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aditya C Shekhar
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
39
|
Lin WJ, Chiang AWT, Zhou EH, Liang C, Liu CH, Ma WL, Cheng WC, Lewis NE. iLipidome: enhancing statistical power and interpretability using hidden biosynthetic interdependencies in the lipidome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594607. [PMID: 38826229 PMCID: PMC11142111 DOI: 10.1101/2024.05.16.594607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Numerous biological processes and diseases are influenced by lipid composition. Advances in lipidomics are elucidating their roles, but analyzing and interpreting lipidomics data at the systems level remain challenging. To address this, we present iLipidome, a method for analyzing lipidomics data in the context of the lipid biosynthetic network, thus accounting for the interdependence of measured lipids. iLipidome enhances statistical power, enables reliable clustering and lipid enrichment analysis, and links lipidomic changes to their genetic origins. We applied iLipidome to investigate mechanisms driving changes in cellular lipidomes following supplementation of docosahexaenoic acid (DHA) and successfully identified the genetic causes of alterations. We further demonstrated how iLipidome can disclose enzyme-substrate specificity and pinpoint prospective glioblastoma therapeutic targets. Finally, iLipidome enabled us to explore underlying mechanisms of cardiovascular disease and could guide the discovery of early lipid biomarkers. Thus, iLipidome can assist researchers studying the essence of lipidomic data and advance the field of lipid biology.
Collapse
|
40
|
Raskovic D, Alvarado G, Hines KM, Xu L, Gatto C, Wilkinson BJ, Pokorny A. Growth of Staphylococcus aureus in the presence of oleic acid shifts the glycolipid fatty acid profile and increases resistance to antimicrobial peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592415. [PMID: 38746422 PMCID: PMC11092785 DOI: 10.1101/2024.05.03.592415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Staphylococcus aureus readily adapts to various environments and quickly develops antibiotic resistance, which has led to an increase in multidrug-resistant infections. Hence, S. aureus presents a significant global health issue and its adaptations to the host environment are crucial for understanding pathogenesis and antibiotic susceptibility. When S. aureus is grown conventionally, its membrane lipids contain a mix of branched-chain and straight-chain saturated fatty acids. However, when unsaturated fatty acids are present in the growth medium, they become a major part of the total fatty acid composition. This study explores the biophysical effects of incorporating straight-chain unsaturated fatty acids into S. aureus membrane lipids. Membrane preparations from cultures supplemented with oleic acid showed more complex differential scanning calorimetry scans than those grown in tryptic soy broth alone. When grown in the presence of oleic acid, the cultures exhibited a transition significantly above the growth temperature, attributed to the presence of glycolipids with long-chain fatty acids causing acyl chain packing frustration within the bilayer. Functional aspects of the membrane were assessed by studying the kinetics of dye release from unilamellar vesicles induced by the antimicrobial peptide mastoparan X. Dye release was slower from liposomes prepared from cells grown in oleic acid-supplemented cultures, suggesting that changes in membrane lipid composition and biophysics protect the cell membrane against peptide-induced lysis. These findings underscore the intricate relationship between the growth environment, membrane lipid composition, and the physical properties of the bacterial membrane, which should be considered when developing new strategies against S. aureus infections.
Collapse
Affiliation(s)
- Djuro Raskovic
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, United States of America
| | - Gloria Alvarado
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Kelly M Hines
- Department of Chemistry, University of Georgia, Athens, Georgia, United States of America
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, United States of America
| | - Craig Gatto
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Brian J Wilkinson
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Antje Pokorny
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina, United States of America
| |
Collapse
|
41
|
Santinho A, Carpentier M, Lopes Sampaio J, Omrane M, Thiam AR. Giant organelle vesicles to uncover intracellular membrane mechanics and plasticity. Nat Commun 2024; 15:3767. [PMID: 38704407 PMCID: PMC11069511 DOI: 10.1038/s41467-024-48086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
Tools for accessing and studying organelles remain underdeveloped. Here, we present a method by which giant organelle vesicles (GOVs) are generated by submitting cells to a hypotonic medium followed by plasma membrane breakage. By this means, GOVs ranging from 3 to over 10 µm become available for micromanipulation. GOVs are made from organelles such as the endoplasmic reticulum, endosomes, lysosomes and mitochondria, or in contact with one another such as giant mitochondria-associated ER membrane vesicles. We measure the mechanical properties of each organelle-derived GOV and find that they have distinct properties. In GOVs procured from Cos7 cells, for example, bending rigidities tend to increase from the endoplasmic reticulum to the plasma membrane. We also found that the mechanical properties of giant endoplasmic reticulum vesicles (GERVs) vary depending on their interactions with other organelles or the metabolic state of the cell. Lastly, we demonstrate GERVs' biochemical activity through their capacity to synthesize triglycerides and assemble lipid droplets. These findings underscore the potential of GOVs as valuable tools for studying the biophysics and biology of organelles.
Collapse
Affiliation(s)
- Alexandre Santinho
- Laboratoire de Physique de l'École normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, F-75005, Paris, France
| | - Maxime Carpentier
- Laboratoire de Physique de l'École normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, F-75005, Paris, France
| | - Julio Lopes Sampaio
- Institut Curie, PSL Research University, Plateforme de Métabolomique et Lipidomique, 26 rue d'Ulm, Paris, France
| | - Mohyeddine Omrane
- Laboratoire de Physique de l'École normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, F-75005, Paris, France
| | - Abdou Rachid Thiam
- Laboratoire de Physique de l'École normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, F-75005, Paris, France.
| |
Collapse
|
42
|
Tanaka T, Matsumoto A, Klymchenko AS, Tsurumaki E, Ikenouchi J, Konishi G. Fluorescent Solvatochromic Probes for Long-Term Imaging of Lipid Order in Living Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309721. [PMID: 38468355 PMCID: PMC11077641 DOI: 10.1002/advs.202309721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/28/2024] [Indexed: 03/13/2024]
Abstract
High-resolution spatio-temporal monitoring of the cell membrane lipid order provides visual insights into the complex and sophisticated systems that control cellular physiological functions. Solvatochromic fluorescent probes are highly promising noninvasive visualization tools for identifying the ordering of the microenvironment of plasma membrane microdomains. However, conventional probes, although capable of structural analysis, lack the necessary long-term photostability required for live imaging at the cellular level. Here, an ultra-high-light-resistant solvatochromic fluorescence probe, 2-N,N-diethylamino-7-(4-methoxycarbonylphenyl)-9,9-dimethylfluorene (FπCM) is reported, which enables live lipid order imaging of cell division. This probe and its derivatives exhibit sufficient fluorescence wavelengths, brightness, polarity responsiveness, low phototoxicity, and remarkable photostability under physiological conditions compared to conventional solvatochromic probes. Therefore, these probes have the potential to overcome the limitations of fluorescence microscopy, particularly those associated with photobleaching. FπCM probes can serve as valuable tools for elucidating mechanisms of cellular processes at the bio-membrane level.
Collapse
Affiliation(s)
- Takuya Tanaka
- Department of Chemical Science and EngineeringTokyo Institute of TechnologyTokyo152‐8552Japan
| | - Atsushi Matsumoto
- Department of BiologyFaculty of SciencesKyushu UniversityFukuoka819‐0395Japan
| | - Andery S. Klymchenko
- Laboratoire de Bioimagerie et PathologiesUMR 7021 CNRSUniversité de Strasbourg74 route du RhinIllkirch67401France
| | - Eiji Tsurumaki
- Department of ChemistryTokyo Institute of TechnologyTokyo152‐8552Japan
| | - Junichi Ikenouchi
- Department of BiologyFaculty of SciencesKyushu UniversityFukuoka819‐0395Japan
| | - Gen‐ichi Konishi
- Department of Chemical Science and EngineeringTokyo Institute of TechnologyTokyo152‐8552Japan
| |
Collapse
|
43
|
Gopalan AB, van Uden L, Sprenger RR, Fernandez-Novel Marx N, Bogetofte H, Neveu PA, Meyer M, Noh KM, Diz-Muñoz A, Ejsing CS. Lipotype acquisition during neural development is not recapitulated in stem cell-derived neurons. Life Sci Alliance 2024; 7:e202402622. [PMID: 38418090 PMCID: PMC10902711 DOI: 10.26508/lsa.202402622] [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: 01/26/2024] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 03/01/2024] Open
Abstract
During development, different tissues acquire distinct lipotypes that are coupled to tissue function and homeostasis. In the brain, where complex membrane trafficking systems are required for neural function, specific glycerophospholipids, sphingolipids, and cholesterol are highly abundant, and defective lipid metabolism is associated with abnormal neural development and neurodegenerative disease. Notably, the production of specific lipotypes requires appropriate programming of the underlying lipid metabolic machinery during development, but when and how this occurs is unclear. To address this, we used high-resolution MSALL lipidomics to generate an extensive time-resolved resource of mouse brain development covering early embryonic and postnatal stages. This revealed a distinct bifurcation in the establishment of the neural lipotype, whereby the canonical lipid biomarkers 22:6-glycerophospholipids and 18:0-sphingolipids begin to be produced in utero, whereas cholesterol attains its characteristic high levels after birth. Using the resource as a reference, we next examined to which extent this can be recapitulated by commonly used protocols for in vitro neuronal differentiation of stem cells. Here, we found that the programming of the lipid metabolic machinery is incomplete and that stem cell-derived cells can only partially acquire a neural lipotype when the cell culture media is supplemented with brain-specific lipid precursors. Altogether, our work provides an extensive lipidomic resource for early mouse brain development and highlights a potential caveat when using stem cell-derived neuronal progenitors for mechanistic studies of lipid biochemistry, membrane biology and biophysics, which nonetheless can be mitigated by further optimizing in vitro differentiation protocols.
Collapse
Affiliation(s)
- Anusha B Gopalan
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Faculty of Biosciences, Candidate for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
| | - Lisa van Uden
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Richard R Sprenger
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | | | - Helle Bogetofte
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Pierre A Neveu
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Odense University Hospital, Odense, Denmark
- BRIDGE, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kyung-Min Noh
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christer S Ejsing
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
44
|
Pang SJ, Liu TT, Pan JC, Man QQ, Song S, Zhang J. The Association between the Plasma Phospholipid Profile and Insulin Resistance: A Population-Based Cross-Section Study from the China Adult Chronic Disease and Nutrition Surveillance. Nutrients 2024; 16:1205. [PMID: 38674894 PMCID: PMC11054597 DOI: 10.3390/nu16081205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The dysfunction of phospholipid metabolism enzymes and the change in membrane phospholipid composition are associated with insulin resistance, indicating that phospholipids play an important role in the regulation of insulin sensitivity. The reflection of phospholipid changes in blood might provide clues for both mechanism understanding and intervention. Using a targeted phospholipidomic approach, 199 phospholipid molecular species were identified and quantified in the plasma of 1053 middle-aged participants from a national investigation. The associations of the phospholipid matrix, clusters, and molecular species with insulin resistance were investigated. A significant association was confirmed between the phospholipid matrix and the homeostatic-model assessment of insulin resistance (HOMA-IR) by a distance-based linear model. Furthermore, three clustered phospholipid modules and 32 phospholipid molecular species were associated with HOMA-IR with the strict control of demographic and lifestyle parameters, family history of diabetes, BMI, WC, and blood lipid parameters. The overall decline in lysophosphatidylcholines (LPCs), the decrease in saturated lysophosphatidylethanolamines (LPEs), the decrease in polyunsaturated/plasmenyl phosphatidylcholines (PCs), and the increase in polyunsaturated phatidylethanolamines (PEs) were the prominent characters of plasma phospholipid perturbation associated with insulin resistance. This suggested that PC- and PE-related metabolic pathways were widely involved in the process of insulin resistance, especially the disorder of LPC acylation to diacyl-PC.
Collapse
Affiliation(s)
- Shao-Jie Pang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
- Key Laboratory of Public Nutrition and Health, National Health Commission of the People’s Republic of China, Beijing 100050, China
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Beijing 100015, China;
| | - Ting-Ting Liu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
| | - Jian-Cun Pan
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Beijing 100015, China;
| | - Qing-Qing Man
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
| | - Shuang Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
| | - Jian Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
- Key Laboratory of Public Nutrition and Health, National Health Commission of the People’s Republic of China, Beijing 100050, China
| |
Collapse
|
45
|
Reinhard J, Starke L, Klose C, Haberkant P, Hammarén H, Stein F, Klein O, Berhorst C, Stumpf H, Sáenz JP, Hub J, Schuldiner M, Ernst R. MemPrep, a new technology for isolating organellar membranes provides fingerprints of lipid bilayer stress. EMBO J 2024; 43:1653-1685. [PMID: 38491296 PMCID: PMC11021466 DOI: 10.1038/s44318-024-00063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/16/2024] [Accepted: 02/26/2024] [Indexed: 03/18/2024] Open
Abstract
Biological membranes have a stunning ability to adapt their composition in response to physiological stress and metabolic challenges. Little is known how such perturbations affect individual organelles in eukaryotic cells. Pioneering work has provided insights into the subcellular distribution of lipids in the yeast Saccharomyces cerevisiae, but the composition of the endoplasmic reticulum (ER) membrane, which also crucially regulates lipid metabolism and the unfolded protein response, remains insufficiently characterized. Here, we describe a method for purifying organelle membranes from yeast, MemPrep. We demonstrate the purity of our ER membrane preparations by proteomics, and document the general utility of MemPrep by isolating vacuolar membranes. Quantitative lipidomics establishes the lipid composition of the ER and the vacuolar membrane. Our findings provide a baseline for studying membrane protein biogenesis and have important implications for understanding the role of lipids in regulating the unfolded protein response (UPR). The combined preparative and analytical MemPrep approach uncovers dynamic remodeling of ER membranes in stressed cells and establishes distinct molecular fingerprints of lipid bilayer stress.
Collapse
Affiliation(s)
- John Reinhard
- Saarland University, Medical Biochemistry and Molecular Biology, Homburg, Germany
- Saarland University, Preclinical Center for Molecular Signaling (PZMS), Homburg, Germany
| | - Leonhard Starke
- Saarland University, Theoretical Physics and Center for Biophysics, Saarbrücken, Germany
| | | | - Per Haberkant
- EMBL Heidelberg, Proteomics Core Facility, Heidelberg, Germany
| | | | - Frank Stein
- EMBL Heidelberg, Proteomics Core Facility, Heidelberg, Germany
| | - Ofir Klein
- Weizmann Institute of Science, Department of Molecular Genetics, Rehovot, Israel
| | - Charlotte Berhorst
- Saarland University, Medical Biochemistry and Molecular Biology, Homburg, Germany
- Saarland University, Preclinical Center for Molecular Signaling (PZMS), Homburg, Germany
| | - Heike Stumpf
- Saarland University, Medical Biochemistry and Molecular Biology, Homburg, Germany
- Saarland University, Preclinical Center for Molecular Signaling (PZMS), Homburg, Germany
| | - James P Sáenz
- Technische Universität Dresden, B CUBE, Dresden, Germany
| | - Jochen Hub
- Saarland University, Theoretical Physics and Center for Biophysics, Saarbrücken, Germany
| | - Maya Schuldiner
- Weizmann Institute of Science, Department of Molecular Genetics, Rehovot, Israel
| | - Robert Ernst
- Saarland University, Medical Biochemistry and Molecular Biology, Homburg, Germany.
- Saarland University, Preclinical Center for Molecular Signaling (PZMS), Homburg, Germany.
| |
Collapse
|
46
|
Henry WS, Müller S, Yang JS, Innes-Gold S, Das S, Reinhardt F, Sigmund K, Phadnis VV, Wan Z, Eaton E, Sampaio JL, Bell GW, Viravalli A, Hammond PT, Kamm RD, Cohen AE, Boehnke N, Hsu VW, Levental KR, Rodriguez R, Weinberg RA. Ether lipids influence cancer cell fate by modulating iron uptake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585922. [PMID: 38562716 PMCID: PMC10983928 DOI: 10.1101/2024.03.20.585922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cancer cell fate has been widely ascribed to mutational changes within protein-coding genes associated with tumor suppressors and oncogenes. In contrast, the mechanisms through which the biophysical properties of membrane lipids influence cancer cell survival, dedifferentiation and metastasis have received little scrutiny. Here, we report that cancer cells endowed with a high metastatic ability and cancer stem cell-like traits employ ether lipids to maintain low membrane tension and high membrane fluidity. Using genetic approaches and lipid reconstitution assays, we show that these ether lipid-regulated biophysical properties permit non-clathrin-mediated iron endocytosis via CD44, leading directly to significant increases in intracellular redox-active iron and enhanced ferroptosis susceptibility. Using a combination of in vitro three-dimensional microvascular network systems and in vivo animal models, we show that loss of ether lipids also strongly attenuates extravasation, metastatic burden and cancer stemness. These findings illuminate a mechanism whereby ether lipids in carcinoma cells serve as key regulators of malignant progression while conferring a unique vulnerability that can be exploited for therapeutic intervention.
Collapse
Affiliation(s)
- Whitney S Henry
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Sebastian Müller
- Institut Curie, CNRS, INSERM, PSL Research University, Equipe Labellisée Ligue Contre le Cancer, Paris 75005, France
| | - Jia-Shu Yang
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, and Dept. of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah Innes-Gold
- Dept. of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Sunny Das
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Ferenc Reinhardt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Kim Sigmund
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Vaishnavi V Phadnis
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Dept. of Biology, MIT, Cambridge, MA 02139, USA
| | - Zhengpeng Wan
- Dept. of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Elinor Eaton
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Julio L Sampaio
- Institut Curie, INSERM, Mines ParisTech, Paris 75005, France
| | - George W Bell
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Amartya Viravalli
- Dept. of Chemical Engineering and Materials Science, University of Minnesota Minneapolis, MN 55455, USA
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Dept. of Chemical Engineering, MIT, Cambridge, MA 02139, USA
- Senior author
| | - Roger D Kamm
- Dept. of Biological Engineering, MIT, Cambridge, MA 02139, USA
- Dept. of Physics, Harvard University, Cambridge, MA 02138, USA
- Senior author
| | - Adam E Cohen
- Dept. of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Dept. of Physics, Harvard University, Cambridge, MA 02138, USA
- Senior author
| | - Natalie Boehnke
- Dept. of Chemical Engineering and Materials Science, University of Minnesota Minneapolis, MN 55455, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Senior author
| | - Victor W Hsu
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, and Dept. of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Senior author
| | - Kandice R Levental
- Dept. of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
- Senior author
| | - Raphaël Rodriguez
- Institut Curie, CNRS, INSERM, PSL Research University, Equipe Labellisée Ligue Contre le Cancer, Paris 75005, France
- Senior author
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Dept. of Biology, MIT, Cambridge, MA 02139, USA
- Ludwig Center for Molecular Oncology, Cambridge, MA 02139, USA
- Senior author
| |
Collapse
|
47
|
Qiu B, Zandkarimi F, Bezjian CT, Reznik E, Soni RK, Gu W, Jiang X, Stockwell BR. Phospholipids with two polyunsaturated fatty acyl tails promote ferroptosis. Cell 2024; 187:1177-1190.e18. [PMID: 38366593 PMCID: PMC10940216 DOI: 10.1016/j.cell.2024.01.030] [Citation(s) in RCA: 105] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/16/2023] [Accepted: 01/19/2024] [Indexed: 02/18/2024]
Abstract
Phospholipids containing a single polyunsaturated fatty acyl tail (PL-PUFA1s) are considered the driving force behind ferroptosis, whereas phospholipids with diacyl-PUFA tails (PL-PUFA2s) have been rarely characterized. Dietary lipids modulate ferroptosis, but the mechanisms governing lipid metabolism and ferroptosis sensitivity are not well understood. Our research revealed a significant accumulation of diacyl-PUFA phosphatidylcholines (PC-PUFA2s) following fatty acid or phospholipid treatments, correlating with cancer cell sensitivity to ferroptosis. Depletion of PC-PUFA2s occurred in aging and Huntington's disease brain tissue, linking it to ferroptosis. Notably, PC-PUFA2s interacted with the mitochondrial electron transport chain, generating reactive oxygen species (ROS) for initiating lipid peroxidation. Mitochondria-targeted antioxidants protected cells from PC-PUFA2-induced mitochondrial ROS (mtROS), lipid peroxidation, and cell death. These findings reveal a critical role for PC-PUFA2s in controlling mitochondria homeostasis and ferroptosis in various contexts and explain the ferroptosis-modulating mechanisms of free fatty acids. PC-PUFA2s may serve as diagnostic and therapeutic targets for modulating ferroptosis.
Collapse
Affiliation(s)
- Baiyu Qiu
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Fereshteh Zandkarimi
- Department of Chemistry, Columbia University, New York, NY 10027, USA; Mass Spectrometry Core Facility, Columbia University, New York, NY 10027, USA
| | - Carla T Bezjian
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Eduard Reznik
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY 10027, USA; Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Pathology and Cell Biology and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
48
|
Ashrafuzzaman M, Koeppe RE, Andersen OS. Intrinsic Lipid Curvature and Bilayer Elasticity as Regulators of Channel Function: A Comparative Single-Molecule Study. Int J Mol Sci 2024; 25:2758. [PMID: 38474005 PMCID: PMC10931550 DOI: 10.3390/ijms25052758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Perturbations in bilayer material properties (thickness, lipid intrinsic curvature and elastic moduli) modulate the free energy difference between different membrane protein conformations, thereby leading to changes in the conformational preferences of bilayer-spanning proteins. To further explore the relative importance of curvature and elasticity in determining the changes in bilayer properties that underlie the modulation of channel function, we investigated how the micelle-forming amphiphiles Triton X-100, reduced Triton X-100 and the HII lipid phase promoter capsaicin modulate the function of alamethicin and gramicidin channels. Whether the amphiphile-induced changes in intrinsic curvature were negative or positive, amphiphile addition increased gramicidin channel appearance rates and lifetimes and stabilized the higher conductance states in alamethicin channels. When the intrinsic curvature was modulated by altering phospholipid head group interactions, however, maneuvers that promote a negative-going curvature stabilized the higher conductance states in alamethicin channels but destabilized gramicidin channels. Using gramicidin channels of different lengths to probe for changes in bilayer elasticity, we found that amphiphile adsorption increases bilayer elasticity, whereas altering head group interactions does not. We draw the following conclusions: first, confirming previous studies, both alamethicin and gramicidin channels are modulated by changes in lipid bilayer material properties, the changes occurring in parallel yet differing dependent on the property that is being changed; second, isolated, negative-going changes in curvature stabilize the higher current levels in alamethicin channels and destabilize gramicidin channels; third, increases in bilayer elasticity stabilize the higher current levels in alamethicin channels and stabilize gramicidin channels; and fourth, the energetic consequences of changes in elasticity tend to dominate over changes in curvature.
Collapse
Affiliation(s)
- Mohammad Ashrafuzzaman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Roger E. Koeppe
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Olaf S. Andersen
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA;
| |
Collapse
|
49
|
Sublette ME, Daray FM, Ganança L, Shaikh SR. The role of polyunsaturated fatty acids in the neurobiology of major depressive disorder and suicide risk. Mol Psychiatry 2024; 29:269-286. [PMID: 37993501 DOI: 10.1038/s41380-023-02322-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023]
Abstract
Long-chain polyunsaturated fatty acids (LC-PUFAs) are obtained from diet or derived from essential shorter-chain fatty acids, and are crucial for brain development and functioning. Fundamentally, LC-PUFAs' neurobiological effects derive from their physicochemical characteristics, including length and double bond configuration, which differentiate LC-PUFA species and give rise to functional differences between n(omega)-3 and n-6 LC-PUFAs. LC-PUFA imbalances are implicated in psychiatric disorders, including major depression and suicide risk. Dietary intake and genetic variants in enzymes involved in biosynthesis of LC-PUFAs from shorter chain fatty acids influence LC-PUFA status. Domains impacted by LC-PUFAs include 1) cell signaling, 2) inflammation, and 3) bioenergetics. 1) As major constituents of lipid bilayers, LC-PUFAs are determinants of cell membrane properties of viscosity and order, affecting lipid rafts, which play a role in regulation of membrane-bound proteins involved in cell-cell signaling, including monoaminergic receptors and transporters. 2) The n-3:n-6 LC-PUFA balance profoundly influences inflammation. Generally, metabolic products of n-6 LC-PUFAs (eicosanoids) are pro-inflammatory, while those of n-3 LC-PUFAs (docosanoids) participate in the resolution of inflammation. Additionally, n-3 LC-PUFAs suppress microglial activation and the ensuing proinflammatory cascade. 3) N-3 LC-PUFAs in the inner mitochondrial membrane affect oxidative stress, suppressing production of and scavenging reactive oxygen species (ROS), with neuroprotective benefits. Until now, this wealth of knowledge about LC-PUFA biomechanisms has not been adequately tapped to develop translational studies of LC-PUFA clinical effects in humans. Future studies integrating neurobiological mechanisms with clinical outcomes may suggest ways to identify depressed individuals most likely to respond to n-3 LC-PUFA supplementation, and mechanistic research may generate new treatment strategies.
Collapse
Affiliation(s)
- M Elizabeth Sublette
- Department of Psychiatry, Columbia University, New York, NY, USA.
- Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA.
| | - Federico Manuel Daray
- University of Buenos Aires, School of Medicine, Institute of Pharmacology, Buenos Aires, Argentina
- National Scientific and Technical Research Council, Buenos Aires, Argentina
| | - Licínia Ganança
- Clínica Universitária de Psiquiatria e Psicologia Médica, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Departamento de Psiquiatria e Saúde Mental, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Saame Raza Shaikh
- Nutritional Obesity Research Center, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
50
|
Tan Y, Li Y, Ren L, Fu H, Li Q, Liu S. Integrative proteome and metabolome analyses reveal molecular basis underlying growth and nutrient composition in the Pacific oyster, Crassostrea gigas. J Proteomics 2024; 290:105021. [PMID: 37838097 DOI: 10.1016/j.jprot.2023.105021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
In order to comprehend the molecular basis of growth, nutrient composition, and color pigmentation in oysters, comparative proteome and metabolome analyses of two selectively bred oyster strains with contrasting growth rate and shell color were used in this study. A total of 289 proteins and 224 metabolites were identified differentially expressed between the two strains. We identified a series of specifically enriched functional clusters implicated in protein biosynthesis (RPL4, MRPS7, and CARS), fatty acid metabolism (ACSL5, PEX3, ACOXI, CPTIA, FABP6, and HSD17B12), energy metabolism (FH, PPP1R7, CLAM2, and RGN), cell proliferation (MYB, NFYC, DOHH, TOP2a, SMARCA5, and SMARCC2), material transport (ABCB1, ABCB8, VPS16, and VPS33a), and pigmentation (RDH7, RDH13, Retsat, COX15, and Cyp3a9). Integrated proteome and metabolome analyses indicate that fast-growing strain utilize energy-efficient mechanisms of ATP generation while promoting protein and polyunsaturated fatty acid synthesis, activating the cell cycle to increase cell proliferation and thus promoting their biomass increase. These results uncovered molecular mechanisms underlying growth regulation, nutrition quality, and pigmentation and provided candidate biomarkers for molecular breeding in oysters. SIGNIFICANCE: Rapid growth has always been the primary breeding objective to increase the production profits of Pacific oyster (Crassostrea gigas), while favorable nutritional quality and beautiful color add commercial value. In recent years, proteomic and metabolomic techniques have been widely used in marine organisms, although these techniques are seldom utilized to study oyster growth and development. In this study, two C. gigas strains with contrasted phenotypes in growth and shell color provided an ideal model for unraveling the molecular basis of growth and nutrient composition through a comparison of the proteome and metabolome. Since proteins and metabolites are the critical undertakers and the end products of cellular regulatory processes, identifying the differentially expressed proteins and metabolites would allow for discovering biomarkers and pathways that were implicated in cell growth, proliferation, and other critical functions. This work provides valuable resources in assistance with molecular breeding of oyster strains with superior production traits of fast-growth and high-quality nutrient value.
Collapse
Affiliation(s)
- Ying Tan
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Yongjing Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Liting Ren
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Huiru Fu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Qi Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Shikai Liu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|