1
|
Longo A, Manganelli V, Misasi R, Riitano G, Caglar TR, Fasciolo E, Recalchi S, Sorice M, Garofalo T. Extracellular Vesicles in the Crosstalk of Autophagy and Apoptosis: A Role for Lipid Rafts. Cells 2025; 14:749. [PMID: 40422252 DOI: 10.3390/cells14100749] [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: 04/14/2025] [Revised: 05/13/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
Autophagy and apoptosis are two essential mechanisms regulating cell fate. Although distinct, their signaling pathways are closely interconnected through various crosstalk mechanisms. Lipid rafts are described to act as both physical and functional platforms during the early stages of autophagic and apoptotic processes. Only recently has a role for lipid raft-associated molecules in regulating EV biogenesis and release begun to emerge. In particular, lipids of EV membranes are essential components in conferring stability to these vesicles in different extracellular environments and/or to facilitate binding or uptake into recipient cells. In this review we highlight these aspects, focusing on the role of lipid molecules during apoptosis and secretory autophagy pathways. We describe the molecular machinery that connects autophagy and apoptosis with vesicular trafficking and lipid metabolism during the release of EVs, and how their alterations contribute to the development of various diseases, including autoimmune disorders and cancer. Overall, these findings emphasize the complexity of autophagy/apoptosis crosstalk and its key role in cellular dynamics, supporting the role of lipid rafts as new therapeutic targets.
Collapse
Affiliation(s)
- Agostina Longo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Valeria Manganelli
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Gloria Riitano
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Tuba Rana Caglar
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Elena Fasciolo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Serena Recalchi
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| |
Collapse
|
2
|
Goldmann O, Medina E. Revisiting Pathogen Exploitation of Clathrin-Independent Endocytosis: Mechanisms and Implications. Cells 2025; 14:731. [PMID: 40422234 DOI: 10.3390/cells14100731] [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: 04/03/2025] [Revised: 05/04/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
Endocytosis is a specialized transport mechanism in which the cell membrane folds inward to enclose large molecules, fluids, or particles, forming vesicles that are transported within the cell. It plays a crucial role in nutrient uptake, immune responses, and cellular communication. However, many pathogens exploit the endocytic pathway to invade and survive within host cells, allowing them to evade the immune system and establish infection. Endocytosis can be classified as clathrin-mediated (CME) or clathrin-independent (CIE), based on the mechanism of vesicle formation. Unlike CME, which involves the formation of clathrin-coated vesicles that bud from the plasma membrane, CIE does not rely on clathrin-coated vesicles. Instead, other mechanisms facilitate membrane invagination and vesicle formation. CIE encompasses a variety of pathways, including caveolin-mediated, Arf6-dependent, and flotillin-dependent pathways. In this review, we discuss key features of CIE pathways, including cargo selection, vesicle formation, routes taken by internalized cargo, and the regulatory mechanisms governing CIE. Many viruses and bacteria hijack host cell CIE mechanisms to facilitate intracellular trafficking and persistence. We also revisit the exploitation of CIE by bacterial and viral pathogens, highlighting recent discoveries in entry mechanisms, intracellular fate, and host-pathogen interactions. Understanding how pathogens manipulate CIE in host cells can inform the development of novel antimicrobial and immunomodulatory interventions, offering new avenues for disease prevention and treatment.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| |
Collapse
|
3
|
Chiricozzi E, Lunghi G, Valsecchi M, Carsana EV, Bassi R, Di Biase E, Dobi D, Ciampa MG, Mauri L, Aureli M, Inamori KI, Inokuchi JI, Sonnino S, Fazzari M. Metabolic and Structural Consequences of GM3 Synthase Deficiency: Insights from an HEK293-T Knockout Model. Biomedicines 2025; 13:843. [PMID: 40299395 PMCID: PMC12024672 DOI: 10.3390/biomedicines13040843] [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: 02/20/2025] [Revised: 03/22/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background: GM3 Synthase Deficiency (GM3SD) is a rare autosomal recessive neurodevelopmental disease characterized by recurrent seizures and neurological deficits. The disorder stems from mutations in the ST3GAL5 gene, encoding GM3 synthase (GM3S), a key enzyme in ganglioside biosynthesis. While enzyme deficiencies affecting ganglioside catabolism are well-documented, the consequences of impaired ganglioside biosynthesis remain less explored. Methods: To investigate GM3SD, we used a Human Embryonic Kidney 293-T (HEK293-T) knockout (KO) cell model generated via CRISPR/Cas9 technology. Lipid composition was assessed via high-performance thin-layer chromatography (HPTLC); glycohydrolase activity in lysosomal and plasma membrane (PM) fractions was enzymatically analyzed. Lysosomal homeostasis was evaluated through protein content analysis and immunofluorescence, and cellular bioenergetics was measured using a luminescence-based assay. Results: Lipidome profiling revealed a significant accumulation of lactosylceramide (LacCer), the substrate of GM3S, along with increased levels of monosialyl-globoside Gb5 (MSGb5), indicating a metabolic shift in glycosphingolipid biosynthesis. Lipid raft analysis revealed elevated cholesterol levels, which may impair microdomain fluidity and signal transduction. Furthermore, altered activity of lysosomal and plasma membrane (PM)-associated glycohydrolases suggests secondary deregulation of glycosphingolipid metabolism, potentially contributing to abnormal lipid patterns. In addition, we observed increased lysosomal mass, indicating potential lysosomal homeostasis dysregulation. Finally, decreased adenosine triphosphate (ATP) levels point to impaired cellular bioenergetics, emphasizing the metabolic consequences of GM3SD. Conclusions: Together, these findings provide novel insights into the molecular alterations associated with GM3SD and establish the HEK293-T KO model as a promising platform for evaluating potential therapeutic strategies.
Collapse
Affiliation(s)
- Elena Chiricozzi
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Manuela Valsecchi
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Emma Veronica Carsana
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Rosaria Bassi
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Erika Di Biase
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA;
| | - Dorina Dobi
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Maria Grazia Ciampa
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Kei-ichiro Inamori
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan;
| | - Jin-ichi Inokuchi
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka 565-0871, Japan;
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| | - Maria Fazzari
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (M.V.); (E.V.C.); (R.B.); (D.D.); (M.G.C.); (L.M.); (M.A.); (S.S.)
| |
Collapse
|
4
|
Stea DM, D’Alessio A. Caveolae: Metabolic Platforms at the Crossroads of Health and Disease. Int J Mol Sci 2025; 26:2918. [PMID: 40243482 PMCID: PMC11988808 DOI: 10.3390/ijms26072918] [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: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Caveolae are small flask-shaped invaginations of the plasma membrane enriched in cholesterol and sphingolipids. They play a critical role in various cellular processes, including signal transduction, endocytosis, and mechanotransduction. Caveolin proteins, specifically Cav-1, Cav-2, and Cav-3, in addition to their role as structural components of caveolae, have been found to regulate the activity of signaling molecules. A growing body of research has highlighted the pivotal role of caveolae and caveolins in maintaining cellular metabolic homeostasis. Indeed, studies have demonstrated that caveolins interact with the key components of insulin signaling, glucose uptake, and lipid metabolism, thereby influencing energy production and storage. The dysfunction of caveolae or the altered expression of caveolins has been associated with metabolic disorders, including obesity, type 2 diabetes, and ocular diseases. Remarkably, mutations in caveolin genes can disrupt cellular energy balance, promote oxidative stress, and exacerbate metabolic dysregulation. This review examines current research on the molecular mechanisms through which caveolae and caveolins regulate cellular metabolism, explores their involvement in the pathogenesis of metabolic disorders, and discusses potential therapeutic strategies targeting caveolin function and the stabilization of caveolae to restore metabolic homeostasis.
Collapse
Affiliation(s)
- Dante Maria Stea
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Alessio D’Alessio
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Rome, Italy
| |
Collapse
|
5
|
Tanaka J, Haga K, Urakami N, Imai M, Sakuma Y. Temperature dependence of membrane viscosity of ternary lipid GUV with L o domains. Biophys J 2025; 124:818-828. [PMID: 39905732 PMCID: PMC11897551 DOI: 10.1016/j.bpj.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 11/05/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025] Open
Abstract
In the cell membrane, it is considered that saturated lipids and cholesterol organize liquid-ordered (Lo) domains in a sea of liquid-disordered (Ld) phases and proteins relevant to cellular functions are localized in the Lo domains. Since the diffusion of transmembrane proteins is regulated by the membrane viscosity, we investigate the temperature dependence of the membrane viscosity of the ternary giant unilamellar vesicles (GUVs) composed of the saturated lipid 1,2-dipalmitoyl-sn-glycero-3-phosphocholine, the unsaturated lipid 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), and cholesterol to understand the effect of the phase separation on the membrane viscosity using a microinjection technique. In the microinjection method, membrane viscosity is estimated by comparing the flow pattern induced on a spherical membrane with a hydrodynamic model. For phase-separated GUVs, the flow pattern is visualized by the motion of the domains. In this study, we developed a method to visualize the flow patterns of homogeneous GUVs above the phase separation temperature by using beads attached to the GUVs. We succeeded in measuring the membrane viscosity of ternary GUVs both above phase separation temperature and in the phase-separated region and found that the membrane viscosity decreases dramatically by phase separation. In the phase-separated region, i.e., GUVs with Lo domains, the membrane viscosity is determined by that of the Ld phase, ηLd, and shows weak temperature dependence compared to that of the DOPC single-component GUV, which is a main component of the Ld phase. We revealed that the Moelwyn-Hughest model, which takes into account the effects of the membrane composition, viscosity of the pure component, and interaction between components, well describes the obtained membrane viscosity of the ternary GUV both above the phase separation temperature and in the phase-separated region. The drastic decrease of the membrane viscosity by the phase separation plays an important role in regulating the mobility of constituents in multi-component membranes.
Collapse
Affiliation(s)
- Julia Tanaka
- Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan
| | - Kenya Haga
- Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan
| | - Naohito Urakami
- Department of Physics and Informatics, Graduate School of Science, Yamaguchi University, Yamaguchi, Japan
| | - Masayuki Imai
- Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan
| | - Yuka Sakuma
- Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan.
| |
Collapse
|
6
|
Butikova EA, Basov NV, Rogachev AD, Gaisler EV, Ivanisenko VA, Demenkov PS, Makarova ALA, Ivanisenko TV, Razumov IA, Kolomeyets DA, Cheresiz SV, Solovieva OI, Larionov KP, Sotnikova YS, Patrushev YV, Kolchanov NA, Pokrovsky AG, Vinokurov NA, Kanygin VV, Popik VM, Shevchenko OA. Metabolomic and gene networks approaches reveal the role of mitochondrial membrane proteins in response of human melanoma cells to THz radiation. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159595. [PMID: 39842507 DOI: 10.1016/j.bbalip.2025.159595] [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: 06/02/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Terahertz (THz) radiation has gained attention due to technological advancements, but its biological effects remain unclear. We investigated the impact of 2.3 THz radiation on SK-MEL-28 cells using metabolomic and gene network analysis. Forty metabolites, primarily related to purine, pyrimidine synthesis and breakdown pathways, were significantly altered post-irradiation. Lipids, such as ceramides and phosphatidylcholines, were also affected. Gene network reconstruction and analysis identified key regulators of the enzymes involved in biosynthesis and degradation of significantly altered metabolites. Mitochondrial membrane components, such as the respiratory chain complex, the proton-transporting ATP synthase complex, and components of lipid rafts reacted to THz radiation. We propose that THz radiation induces reversible disruption of the lipid raft macromolecular structure, thereby altering mitochondrial molecule transport while maintaining protein integrity, which explains the high cell survival rate. Our findings enhance the understanding of THz biological effects and emphasize the role of membrane components in the cellular response to THz radiation.
Collapse
Affiliation(s)
- Ekaterina A Butikova
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; Research Institute of Clinical and Experimental Lymрhology - Branch of the Institute of Cytology and Genetics SB RAS, Timakova str.,2, 630060 Novosibirsk, Russia.
| | - Nikita V Basov
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, Acad. Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Artem D Rogachev
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, Acad. Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Evgeniy V Gaisler
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia
| | - Vladimir A Ivanisenko
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; Institute of Cytology and Genetics SB RAS, Acad. Lavrentiev Ave.,10, 630090 Novosibirsk, Russia
| | - Pavel S Demenkov
- Institute of Cytology and Genetics SB RAS, Acad. Lavrentiev Ave.,10, 630090 Novosibirsk, Russia
| | - Aelita-Luiza A Makarova
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; Institute of Cytology and Genetics SB RAS, Acad. Lavrentiev Ave.,10, 630090 Novosibirsk, Russia
| | - Timofey V Ivanisenko
- Institute of Cytology and Genetics SB RAS, Acad. Lavrentiev Ave.,10, 630090 Novosibirsk, Russia
| | - Ivan A Razumov
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; Institute of Cytology and Genetics SB RAS, Acad. Lavrentiev Ave.,10, 630090 Novosibirsk, Russia; Budker Institute of Nuclear Physics SB RAS, Acad. Lavrentiev Ave.,11, 630090 Novosibirsk, Russia
| | - Daria A Kolomeyets
- Budker Institute of Nuclear Physics SB RAS, Acad. Lavrentiev Ave.,11, 630090 Novosibirsk, Russia
| | - Sergey V Cheresiz
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia
| | - Olga I Solovieva
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; Institute of Cytology and Genetics SB RAS, Acad. Lavrentiev Ave.,10, 630090 Novosibirsk, Russia; Budker Institute of Nuclear Physics SB RAS, Acad. Lavrentiev Ave.,11, 630090 Novosibirsk, Russia
| | - Kirill P Larionov
- Boreskov Institute of Catalysis, Acad. Lavrentiev Ave., 5, 630090 Novosibirsk, Russia
| | - Yulia S Sotnikova
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, Acad. Lavrentiev Ave., 9, 630090 Novosibirsk, Russia; Boreskov Institute of Catalysis, Acad. Lavrentiev Ave., 5, 630090 Novosibirsk, Russia
| | - Yuri V Patrushev
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; Boreskov Institute of Catalysis, Acad. Lavrentiev Ave., 5, 630090 Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Institute of Cytology and Genetics SB RAS, Acad. Lavrentiev Ave.,10, 630090 Novosibirsk, Russia
| | - Andrey G Pokrovsky
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia
| | - Nikolay A Vinokurov
- Budker Institute of Nuclear Physics SB RAS, Acad. Lavrentiev Ave.,11, 630090 Novosibirsk, Russia
| | - Vladimir V Kanygin
- Novosibirsk State University, Pirogova Str., 2, 630090 Novosibirsk, Russia; Budker Institute of Nuclear Physics SB RAS, Acad. Lavrentiev Ave.,11, 630090 Novosibirsk, Russia
| | - Vasiliy M Popik
- Budker Institute of Nuclear Physics SB RAS, Acad. Lavrentiev Ave.,11, 630090 Novosibirsk, Russia
| | - Oleg A Shevchenko
- Budker Institute of Nuclear Physics SB RAS, Acad. Lavrentiev Ave.,11, 630090 Novosibirsk, Russia
| |
Collapse
|
7
|
Stober K, Schwerdtfeger F, Aigal S, Mely Y, Römer W. The Bacterium P. aeruginosa Disperses Ordered Membrane Domains by Targeting Phase Boundaries. Biomolecules 2025; 15:341. [PMID: 40149877 PMCID: PMC11940534 DOI: 10.3390/biom15030341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Various pathogens use receptors on the host's plasma membrane for their cellular uptake. For the bacterium Pseudomonas aeruginosa, interactions between its lectin LecA and the host cell glycosphingolipid globotriaosylceramide (also known as Gb3) are crucial for its internalization via the so-called lipid zipper mechanism. In this study, we investigated the interactions of the P. aeruginosa strain PAO1 with phase-separated lipid bilayers containing Gb3. Surprisingly, bacteria are mostly bound to the interphase of liquid-ordered (Lo) and liquid-disordered (Ld) membrane domains. Simultaneously with the formation of bacterial aggregates and the accumulation of membrane lipids, the lipid bilayers were drastically reorganized and Lo domains were dissolved. Surprisingly, Gb3 was found to play a role in the localization of the bacterium at the interface, less so LecA. When microspheres were used as a minimal mimic of the bacterium, these beads also localized preferentially at the Lo-Ld phase boundaries, but in contrast to living bacteria, beads were unable to cause membrane reorganization and dissolution of the Lo domain, even when coated with LecA. Targeting phase boundaries as "weak points" in membranes and thereby reorganizing and destabilizing the host cell plasma membrane could be an attractive entry strategy for P. aeruginosa and many other bacteria and viruses.
Collapse
Affiliation(s)
- Kai Stober
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany; (K.S.)
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Fabian Schwerdtfeger
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany; (K.S.)
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Sahaja Aigal
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany; (K.S.)
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Yves Mely
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch CEDEX, France
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany; (K.S.)
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| |
Collapse
|
8
|
Lee SI, Lim H, Kim NY, Yu J, Cho J, Lee H, Moon DW, Seo J. Imaging lipid rafts reveals the principle of ApoE4-induced Aβ upregulation in human neurons. iScience 2025; 28:111893. [PMID: 39995873 PMCID: PMC11848483 DOI: 10.1016/j.isci.2025.111893] [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: 09/11/2024] [Revised: 12/01/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Lipid rafts in plasma membranes are thought to provide a platform for regulating signaling pathways by increasing the expression or proximity of proteins in the same pathway. Despite this understanding, the absence of direct, simultaneous observations of lipid rafts and their affiliated proteins has hindered a comprehensive assessment of their roles across various biological contexts. Amyloid-β (Aβ), a hallmark of Alzheimer's disease (AD), is generated from the sequential cleavage of amyloid precursor proteins (APPs) by β- and γ-secretases, primarily within endosomes after APP endocytosis by canonical clathrin-mediated endocytosis in neurons. In this study, we developed a protocol for imaging APP on lipid rafts using time-of-flight secondary ion mass spectrometry (ToF-SIMS) and found that astrocyte ApoE4 contributes to an increase in APP localization on lipid rafts, subsequently elevating Aβ42 synthesis in a clathrin-independent manner in neurons.
Collapse
Affiliation(s)
- Se-In Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
| | - Heejin Lim
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28199 South Korea
| | - Na Yeon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
| | - Jichang Yu
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
| | - Joonho Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
| | - Hyein Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
| | - Dae Won Moon
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
| | - Jinsoo Seo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988 South Korea
| |
Collapse
|
9
|
Savulescu-Fiedler I, Dorobantu-Lungu LR, Dragosloveanu S, Benea SN, Dragosloveanu CDM, Caruntu A, Scheau AE, Caruntu C, Scheau C. The Cross-Talk Between the Peripheral and Brain Cholesterol Metabolisms. Curr Issues Mol Biol 2025; 47:115. [PMID: 39996836 PMCID: PMC11853762 DOI: 10.3390/cimb47020115] [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: 12/21/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Cholesterol is an essential element for the development and normal function of the central nervous system. While peripheral cholesterol is influenced by liver metabolism and diet, brain cholesterol metabolism takes place in an isolated system due to the impermeability of the blood-brain barrier (BBB). However, cross-talk occurs between the brain and periphery, specifically through metabolites such as oxysterols that play key roles in regulating cholesterol balance. Several neurodegenerative conditions such as Alzheimer's disease or Parkinson's disease are considered to be affected by the loss of this balance. Also, the treatment of hypercholesterolemia needs to consider these discrete interferences between brain and peripheral cholesterol and the possible implications of each therapeutic approach. This is particularly important because of 27-hydroxycholesterol and 24-hydroxycholesterol, which can cross the BBB and are involved in cholesterol metabolism. This paper examines the metabolic pathways of cholesterol metabolism in the brain and periphery and focuses on the complex cross-talk between these metabolisms. Also, we emphasize the regulatory role of the BBB and the need for an integrated approach to cholesterol management.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Luiza-Roxana Dorobantu-Lungu
- Department of Cardiology, Emergency Institute for Cardiovascular Diseases “C.C. Iliescu”, 022328 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Departament of Infectious Diseases, National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania
| | - Christiana Diana Maria Dragosloveanu
- Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
10
|
Li Y, Uhelski ML, North RY, Farson LB, Bankston CB, Roland GH, Fan DH, Sheffield KN, Jia A, Orlando D, Heles M, Yaksh TL, Miller YI, Kosten TA, Dougherty PM. ApoA-I binding protein (AIBP) regulates transient receptor potential vanilloid 1 (TRPV1) activity in rat dorsal root ganglion neurons by selective disruption of toll-like receptor 4 (TLR4)-lipid rafts. Brain Behav Immun 2025; 123:644-655. [PMID: 39414176 DOI: 10.1016/j.bbi.2024.10.017] [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: 05/06/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Toll-like receptor 4 (TLR4) and the transient receptor potential vanilloid subtype 1 (TRPV1) are both upregulated and play key roles in the induction and expression of paclitaxel-related chemotherapy-induced peripheral neuropathy (CIPN). Using Apolipoprotein A-I binding protein, non-specific cholesterol depletion, TLR4 mis-sense rats and a TLR4 inhibitor, we demonstrate that co-localization of TRPV1 with TLR4 to cholesterol-rich lipid membrane rafts in nociceptors is essential for its normal activation as well as for its exaggerated activation that underlies the development and expression of CIPN. The findings suggest that TLR4-lipid rafts may have an essential role in numerous neuroinflammatory and neuropathic pain conditions. This mechanism is also generalized to female rats for the first time.
Collapse
Affiliation(s)
- Yan Li
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Megan L Uhelski
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Robert Y North
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, the United States of America
| | - Luke B Farson
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Christopher B Bankston
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Gavin H Roland
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Dwight H Fan
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | | | - Amy Jia
- Northwestern University, Evanston, IL 60208, the United States of America
| | - Dana Orlando
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Mario Heles
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Tony L Yaksh
- The Department of Anesthesiology, the University of California San Diego, La Jolla, CA, 92093, the United States of America
| | - Yury I Miller
- Department of Medicine, the University of California San Diego, La Jolla, CA, 92093, the United States of America
| | - Therese A Kosten
- Department of Psychology, Health Building 1, 4349 Martin Luther King Blvd, Houston, TX 77204, the United States of America
| | - Patrick M Dougherty
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America.
| |
Collapse
|
11
|
Cuenca VE, Pedroni VI, Morini MA. Role of DHA in a Physicochemical Study of a Model Membrane of Grey Matter. MEMBRANES 2024; 14:256. [PMID: 39728706 DOI: 10.3390/membranes14120256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024]
Abstract
The present study investigates a multicomponent lipid system that simulates the neuronal grey matter membrane, employing molecular acoustics as a precise, straightforward, and cost-effective methodology. Given the significance of omega-3 polyunsaturated fatty acids in the functionality of cellular membranes, this research examines the effects of reducing 1-palmitoyl-2-docosahexaenoylphosphatylcholine (PDPC) content on the compressibility and elasticity of the proposed membrane under physiological conditions. Our results align with bibliographic data obtained through other techniques, showing that as the proportion of PDPC increases in the grey matter membrane model, the system's compressibility decreases, and the membrane's elasticity increases, as evidenced by the reduction in the bulk modulus. These results could be interpreted in light of the emerging model of lipid rafts, in which esterified DHA infiltrates and remodels their architecture. We contend that the results obtained may serve as a bridge between biophysics and cellular biology.
Collapse
Affiliation(s)
- Victor E Cuenca
- Laboratory of Physical-Chemistry, Department of Chemistry, Universidad Nacional del Sur (UNS), Bahía Blanca 8000, Argentina
| | - Viviana I Pedroni
- Laboratory of Physical-Chemistry, Department of Chemistry, Universidad Nacional del Sur (UNS), Bahía Blanca 8000, Argentina
- INQUISUR-CONICET, Bahía Blanca 8000, Argentina
| | - Marcela A Morini
- Laboratory of Physical-Chemistry, Department of Chemistry, Universidad Nacional del Sur (UNS), Bahía Blanca 8000, Argentina
- INQUISUR-CONICET, Bahía Blanca 8000, Argentina
| |
Collapse
|
12
|
de Souza Cardoso R, Ono A. The Effects of Viral Structural Proteins on Acidic Phospholipids in Host Membranes. Viruses 2024; 16:1714. [PMID: 39599829 PMCID: PMC11599007 DOI: 10.3390/v16111714] [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: 09/13/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Enveloped viruses rely on host membranes for trafficking and assembly. A substantial body of literature published over the years supports the involvement of cellular membrane lipids in the enveloped virus assembly processes. In particular, the knowledge regarding the relationship between viral structural proteins and acidic phospholipids has been steadily increasing in recent years. In this review, we will briefly review the cellular functions of plasma membrane-associated acidic phospholipids and the mechanisms that regulate their local distribution within this membrane. We will then explore the interplay between viruses and the plasma membrane acidic phospholipids in the context of the assembly process for two enveloped viruses, the influenza A virus (IAV) and the human immunodeficiency virus type 1 (HIV-1). Among the proteins encoded by these viruses, three viral structural proteins, IAV hemagglutinin (HA), IAV matrix protein-1 (M1), and HIV-1 Gag protein, are known to interact with acidic phospholipids, phosphatidylserine and/or phosphatidylinositol (4,5)-bisphosphate. These interactions regulate the localization of the viral proteins to and/or within the plasma membrane and likely facilitate the clustering of the proteins. On the other hand, these viral proteins, via their ability to multimerize, can also alter the distribution of the lipids and may induce acidic-lipid-enriched membrane domains. We will discuss the potential significance of these interactions in the virus assembly process and the property of the progeny virions. Finally, we will outline key outstanding questions that need to be answered for a better understanding of the relationships between enveloped virus assembly and acidic phospholipids.
Collapse
Affiliation(s)
| | - Akira Ono
- Department of Microbiology and Immunology, The University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
13
|
Hanashima S, Yamanaka A, Ibata Y, Yasuda T, Umegawa Y, Murata M. Lipid Compositions of Liquid-Ordered and Liquid-Disordered Phases in Ternary Membranes of Sphingomyelin, Cholesterol, and Dioleoylphosphatidylcholine Determined by 2H NMR: Stearoyl-Sphingomyelin Compared with Its Palmitoyl Counterpart. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:22973-22981. [PMID: 39429033 DOI: 10.1021/acs.langmuir.4c03104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Sphingomyelin (SM) and cholesterol are the major lipids in the signaling platforms of cell membranes, known as lipid rafts. In particular, SM with a stearoyl chain (C18-SM) is abundant in specific tissues such as the brain, the most cholesterol-rich organ, whereas the distribution of palmitoyl (C16)-SM is ubiquitous. Here, we reveal the differences between palmitoyl- and stearoyl-SM in lipid-lipid interactions based on the tie lines obtained from the 2H solid-state NMR spectra of bilayer systems composed of SM/dioleoylphosphatidylcholine/cholesterol 33:33:33 and 40:40:20. Lipid probes carrying position-selective deuterations, 10',10'-d2-SM, 24-d1-cholesterol, and 6″,6″-d2-dioleoyl-phosphatidylcholine, were incorporated into the membranes. 2H NMR peaks from these probes in the membranes directly provide the lipid compositions of the liquid-ordered (Lo) and liquid-disordered (Ld) regions. Without using bulky fluorescent groups, these probes allow us to obtain the end points of the tie lines in a ternary phase diagram based on the lever rule. Consequently, the tie lines of the stearoyl-SM membranes were steeper than those of the palmitoyl-SM membranes, indicating that cholesterol content was higher in the Lo domains of stearoyl-SM, regardless of the total concentration of unsaturated phospholipids. When comparing the content of unsaturated lipids in the Lo domain, the stearoyl-SM membranes had a higher content than palmitoyl-SM membranes. These results revealed that stearoyl-SM is suitable for stabilizing biologically functional microdomains in cholesterol-rich organs, whereas palmitoyl-SM may be better suited for stabilizing domains in tissue membranes with normal cholesterol content. The small but significant differences in the lipid interactions between stearoyl-SM and palmitoyl-SM may be related to the spatiotemporal formation of functional domains in biological environments.
Collapse
Affiliation(s)
- Shinya Hanashima
- Graduate School of Science, Osaka University, Machikaneyama 1-1, Toyonaka, Osaka 560-0043, Japan
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Koyamacho-minami 4-101, Tottori 680-8550, Japan
- Center for Research on Green Sustainable Chemistry, Tottori University, Koyamacho-minami 4-101, Tottori 680-8550, Japan
| | - Ayana Yamanaka
- Graduate School of Science, Osaka University, Machikaneyama 1-1, Toyonaka, Osaka 560-0043, Japan
| | - Yuki Ibata
- Graduate School of Science, Osaka University, Machikaneyama 1-1, Toyonaka, Osaka 560-0043, Japan
| | - Tomokazu Yasuda
- Graduate School of Science, Osaka University, Machikaneyama 1-1, Toyonaka, Osaka 560-0043, Japan
| | - Yuichi Umegawa
- Graduate School of Science, Osaka University, Machikaneyama 1-1, Toyonaka, Osaka 560-0043, Japan
| | - Michio Murata
- Graduate School of Science, Osaka University, Machikaneyama 1-1, Toyonaka, Osaka 560-0043, Japan
- Forefront Research Centre for Fundamental Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
14
|
Fuchs RM, Reed JR, Connick JP, Paloncýová M, Šrejber M, Čechová P, Otyepka M, Eyer MK, Backes WL. Identification of the N-terminal residues responsible for the differential microdomain localization of CYP1A1 and CYP1A2. J Biol Chem 2024; 300:107891. [PMID: 39447873 PMCID: PMC11603000 DOI: 10.1016/j.jbc.2024.107891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
The endoplasmic reticulum is organized into ordered regions enriched in cholesterol and sphingomyelin, and disordered microdomains characterized by more fluidity. Rabbit CYP1A1 and CYP1A2 localize into disordered and ordered microdomains, respectively. Previously, a CYP1A2 chimera containing the first 109 amino acids of CYP1A1 showed altered microdomain localization. The goal of this study was to identify specific residues responsible for CYP1A microdomain localization. Thus, CYP1A2 chimeras containing substitutions from homologous regions of CYP1A1 were expressed in HEK 293T/17 cells, and the localization was examined after solubilization with Brij 98. A CYP1A2 mutant with the three amino acids from CYP1A1 (VAG) at positions 27 to 29 of CYP1A2 was generated that showed a distribution pattern similar to those of CYP1A1/1A2 chimeras containing both the first 109 amino acids and the first 31 amino acids of CYP1A1 followed by remaining amino acids of CYP1A2. Similarly, the reciprocal substitution of three amino acids from CYP1A2 (AVR) into CYP1A1 resulted in a partial redistribution of the chimera into ordered microdomains. Molecular dynamic simulations indicate that the positive charges of the CYP1A1 and CYP1A2 linker regions between the N termini and catalytic domains resulted in different depths of immersion of the N termini in the membrane. The overlap of the distribution of positively charged residues in CYP1A2 (AVR) and negatively charged phospholipids was higher in the ordered than in the disordered microdomain. These findings identify three residues in the CYP1AN terminus as a novel microdomain-targeting motif of the P450s and provide a mechanistic explanation for the differential microdomain localization of CYP1A.
Collapse
Affiliation(s)
- Robert M Fuchs
- Department of Pharmacology and Experimental Therapeutics, and the Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center - New Orleans, New Orleans, Louisiana, USA
| | - James R Reed
- Department of Pharmacology and Experimental Therapeutics, and the Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center - New Orleans, New Orleans, Louisiana, USA
| | - J Patrick Connick
- Department of Pharmacology and Experimental Therapeutics, and the Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center - New Orleans, New Orleans, Louisiana, USA
| | - Markéta Paloncýová
- Regional Center of Advanced Technologies and Materials, The Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czech Republic
| | - Martin Šrejber
- Regional Center of Advanced Technologies and Materials, The Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czech Republic
| | - Petra Čechová
- Regional Center of Advanced Technologies and Materials, The Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czech Republic
| | - Michal Otyepka
- Regional Center of Advanced Technologies and Materials, The Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czech Republic; IT4Innovations, VŠB - Technical University of Ostrava, Ostrava, Czech Republic
| | - Marilyn K Eyer
- Department of Pharmacology and Experimental Therapeutics, and the Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center - New Orleans, New Orleans, Louisiana, USA
| | - Wayne L Backes
- Department of Pharmacology and Experimental Therapeutics, and the Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center - New Orleans, New Orleans, Louisiana, USA.
| |
Collapse
|
15
|
Skubic C, Trček H, Nassib P, Kreft T, Walakira A, Pohar K, Petek S, Režen T, Ihan A, Rozman D. Knockouts of CYP51A1, DHCR24, or SC5D from cholesterol synthesis reveal pathways modulated by sterol intermediates. iScience 2024; 27:110651. [PMID: 39262789 PMCID: PMC11387598 DOI: 10.1016/j.isci.2024.110651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/20/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024] Open
Abstract
Sterols from cholesterol synthesis are crucial for cholesterol production, but also have individual roles difficult to assess in vivo due to essentiality of cholesterol. We developed HepG2 cell models with knockouts (KOs) for three enzymes of cholesterol synthesis, each accumulating specific sterols. Surprisingly, KOs of CYP51, DHCR24, and SC5D shared only 9% of differentially expressed genes. The most striking was the phenotype of CYP51 KO with highly elevated lanosterol and 24,25-dihydrolanosterol, significant increase in G2+M phase and enhanced cancer and cell cycle pathways. Comparisons with mouse liver Cyp51 KO data suggest 24,25-dihydrolanosterol activates similar cell proliferation pathways, possibly via elevated LEF1 and WNT/NFKB signaling. In contrast, SC5D and DHCR24 KO cells with elevated lathosterol or desmosterol proliferated slowly, with downregulated E2F, mitosis, and enriched HNF1A. These findings demonstrate that increase of lanosterol and 24,25-dihydrolanosterol, but not other sterols, promotes cell proliferation in hepatocytes.
Collapse
Affiliation(s)
- Cene Skubic
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Hana Trček
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Petra Nassib
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Tinkara Kreft
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Andrew Walakira
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Katka Pohar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Sara Petek
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Tadeja Režen
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Damjana Rozman
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| |
Collapse
|
16
|
Lin Z, Hua G, Hu X. Lipid metabolism associated crosstalk: the bidirectional interaction between cancer cells and immune/stromal cells within the tumor microenvironment for prognostic insight. Cancer Cell Int 2024; 24:295. [PMID: 39174964 PMCID: PMC11342506 DOI: 10.1186/s12935-024-03481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Cancer is closely related to lipid metabolism, with the tumor microenvironment (TME) containing numerous lipid metabolic interactions. Cancer cells can bidirectionally interact with immune and stromal cells, the major components of the TME. This interaction is primarily mediated by fatty acids (FAs), cholesterol, and phospholipids. These interactions can lead to various physiological changes, including immune suppression, cancer cell proliferation, dissemination, and anti-apoptotic effects on cancer cells. The physiological modulation resulting from this lipid metabolism-associated crosstalk between cancer cells and immune/stromal cells provides valuable insights into cancer prognosis. A comprehensive literature review was conducted to examine the function of the bidirectional lipid metabolism interactions between cancer cells and immune/stromal cells within the TME, particularly how these interactions influence cancer prognosis. A novel autophagy-extracellular vesicle (EV) pathway has been proposed as a mediator of lipid metabolism interactions between cancer cells and immune cells/stromal cells, impacting cancer prognosis. As a result, different forms of lipid metabolism interactions have been described as being linked to cancer prognosis, including those mediated by the autophagy-EV pathway. In conclusion, understanding the bidirectional lipid metabolism interactions between cancer cells and stromal/immune cells in the TME can help develop more advanced prognostic approaches for cancer patients.
Collapse
Affiliation(s)
- Zhongshu Lin
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Guanxiang Hua
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Xiaojuan Hu
- Queen Mary College, Nanchang University, Nanchang, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
17
|
Santos-Miranda A, Joviano-Santos JV, Marques ILS, Cau S, Carvalho FA, Fraga JR, Alvarez-Leite JI, Roman-Campos D, Cruz JS. Electrocontractile remodeling of isolated cardiomyocytes induced during early-stage hypercholesterolemia. J Bioenerg Biomembr 2024; 56:373-387. [PMID: 38869808 DOI: 10.1007/s10863-024-10026-x] [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: 06/28/2023] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Hypercholesterolemia is one of the most important risk factors for cardiovascular diseases. However, it is mostly associated with vascular dysfunction and atherosclerotic lesions, while evidence of direct effects of hypercholesterolemia on cardiomyocytes and heart function is still incomplete and controversial. In this study, we assessed the direct effects of hypercholesterolemia on heart function and the electro-contractile properties of isolated cardiomyocytes. After 5 weeks, male Swiss mice fed with AIN-93 diet added with 1.25% cholesterol (CHO), developed an increase in total serum cholesterol levels and cardiomyocytes cholesterol content. These changes led to altered electrocardiographic records, with a shortening of the QT interval. Isolated cardiomyocytes displayed a shortening of the action potential duration with increased rate of depolarization, which was explained by increased IK, reduced ICa.L and altered INa voltage-dependent inactivation. Also, reduced diastolic [Ca2+]i was found with preserved adrenergic response and cellular contraction function. However, contraction of isolated hearts is impaired in isolated CHO hearts, before and after ischemia/reperfusion, although CHO heart was less susceptible to arrhythmic contractions. Overall, our results demonstrate that early hypercholesterolemia-driven increase in cellular cholesterol content is associated with direct modulation of the heart and cardiomyocytes' excitability, Ca2+ handling, and contraction.
Collapse
Affiliation(s)
- Artur Santos-Miranda
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, Brazil.
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil.
| | - Julliane V Joviano-Santos
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Laboratório de Investigações NeuroCardíacas, Ciências Médicas de Minas Gerais (LINC CMMG), Minas Gerais, Brazil
| | - Ivan Lobo Sousa Marques
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Stefany Cau
- Department of Pharmacology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Fabrício A Carvalho
- Department of Pharmacology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Júlia R Fraga
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | | - Danilo Roman-Campos
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jader S Cruz
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| |
Collapse
|
18
|
Shouman S, El-Kholy N, Hussien AE, El-Derby AM, Magdy S, Abou-Shanab AM, Elmehrath AO, Abdelwaly A, Helal M, El-Badri N. SARS-CoV-2-associated lymphopenia: possible mechanisms and the role of CD147. Cell Commun Signal 2024; 22:349. [PMID: 38965547 PMCID: PMC11223399 DOI: 10.1186/s12964-024-01718-3] [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: 02/24/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024] Open
Abstract
T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.
Collapse
Affiliation(s)
- Shaimaa Shouman
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Nada El-Kholy
- Department of Drug Discovery, H. Lee Moffit Cancer Center& Research Institute, Tampa, FL, 33612, USA
- Cancer Chemical Biology Ph.D. Program, University of South Florida, Tampa, FL, 33620, USA
| | - Alaa E Hussien
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Shireen Magdy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | | | - Ahmad Abdelwaly
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Institute for Computational Molecular Science, Department of Chemistry, Temple University, Philadelphia, PA, 19122, USA
| | - Mohamed Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt.
| |
Collapse
|
19
|
Yu C, Richly M, Hoang TT, El Beheiry M, Türkcan S, Masson JB, Alexandrou A, Bouzigues CI. Confinement energy landscape classification reveals membrane receptor nano-organization mechanisms. Biophys J 2024; 123:1882-1895. [PMID: 38845200 PMCID: PMC11267427 DOI: 10.1016/j.bpj.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/01/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
The cell membrane organization has an essential functional role through the control of membrane receptor confinement in micro- or nanodomains. Several mechanisms have been proposed to account for these properties, although some features have remained controversial, notably the nature, size, and stability of cholesterol- and sphingolipid-rich domains or lipid rafts. Here, we probed the effective energy landscape acting on single-nanoparticle-labeled membrane receptors confined in raft nanodomains- epidermal growth factor receptor (EGFR), Clostridium perfringens ε-toxin receptor (CPεTR), and Clostridium septicum α-toxin receptor (CSαTR)-and compared it with hop-diffusing transferrin receptors. By establishing a new analysis pipeline combining Bayesian inference, decision trees, and clustering approaches, we systematically classified single-protein trajectories according to the type of effective confining energy landscape. This revealed the existence of only two distinct organization modalities: confinement in a quadratic energy landscape for EGFR, CPεTR, and CSαTR (A), and free diffusion in confinement domains resulting from the steric hindrance due to F-actin barriers for transferrin receptor (B). The further characterization of effective confinement energy landscapes by Bayesian inference revealed the role of interactions with the domain environment in cholesterol- and sphingolipid-rich domains with (EGFR) or without (CPεTR and CSαTR) interactions with F-actin to regulate the confinement energy depth. These two distinct mechanisms result in the same organization type (A). We revealed that the apparent domain sizes for these receptor trajectories resulted from Brownian exploration of the energy landscape in a steady-state-like regime at a common effective temperature, independently of the underlying molecular mechanisms. These results highlight that confinement domains may be adequately described as interaction hotspots rather than rafts with abrupt domain boundaries. Altogether, these results support a new model for functional receptor confinement in membrane nanodomains and pave the way to the constitution of an atlas of membrane protein organization.
Collapse
Affiliation(s)
- Chao Yu
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Maximilian Richly
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Thi Thuy Hoang
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Mohammed El Beheiry
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Decision and Bayesian Computation, Paris, France; Épiméthée, INRIA, Paris, France
| | - Silvan Türkcan
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Jean-Baptiste Masson
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Decision and Bayesian Computation, Paris, France; Épiméthée, INRIA, Paris, France
| | - Antigoni Alexandrou
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Cedric I Bouzigues
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France.
| |
Collapse
|
20
|
Bepler T, Barrera MD, Rooney MT, Xiong Y, Kuang H, Goodell E, Goodwin MJ, Harbron E, Fu R, Mihailescu M, Narayanan A, Cotten ML. Antiviral activity of the host defense peptide piscidin 1: investigating a membrane-mediated mode of action. Front Chem 2024; 12:1379192. [PMID: 38988727 PMCID: PMC11233706 DOI: 10.3389/fchem.2024.1379192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/08/2024] [Indexed: 07/12/2024] Open
Abstract
Outbreaks of viral diseases are on the rise, fueling the search for antiviral therapeutics that act on a broad range of viruses while remaining safe to human host cells. In this research, we leverage the finding that the plasma membranes of host cells and the lipid bilayers surrounding enveloped viruses differ in lipid composition. We feature Piscidin 1 (P1), a cationic host defense peptide (HDP) that has antimicrobial effects and membrane activity associated with its N-terminal region where a cluster of aromatic residues and copper-binding motif reside. While few HDPs have demonstrated antiviral activity, P1 acts in the micromolar range against several enveloped viruses that vary in envelope lipid composition. Notably, it inhibits HIV-1, a virus that has an envelope enriched in cholesterol, a lipid associated with higher membrane order and stability. Here, we first document through plaque assays that P1 boasts strong activity against SARS-CoV-2, which has an envelope low in cholesterol. Second, we extend previous studies done with homogeneous bilayers and devise cholesterol-containing zwitterionic membranes that contain the liquid disordered (Ld; low in cholesterol) and ordered (Lo, rich in cholesterol) phases. Using dye leakage assays and cryo-electron microscopy on vesicles, we show that P1 has dramatic permeabilizing capability on the Lo/Ld, an effect matched by a strong ability to aggregate, fuse, and thin the membranes. Differential scanning calorimetry and NMR experiments demonstrate that P1 mixes the lipid content of vesicles and alters the stability of the Lo. Structural studies by NMR indicate that P1 interacts with the Lo/Ld by folding into an α-helix that lies parallel to the membrane surface. Altogether, these results show that P1 is more disruptive to phase-separated than homogenous cholesterol-containing bilayers, suggesting an ability to target domain boundaries. Overall, this multi-faceted research highlights how a peptide that interacts strongly with membranes through an aromatic-rich N-terminal motif disrupt viral envelope mimics. This represents an important step towards the development of novel peptides with broad-spectrum antiviral activity.
Collapse
Affiliation(s)
- Tristan Bepler
- New York Structural Biology Center, New York, NY, United States
| | - Michael D. Barrera
- School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Mary T. Rooney
- Department of Applied Science, William & Mary, Williamsburg, VA, United States
- Department of Chemistry, Hofstra University, Hempstead, NY, United States
| | - Yawei Xiong
- Department of Applied Science, William & Mary, Williamsburg, VA, United States
| | - Huihui Kuang
- New York Structural Biology Center, New York, NY, United States
| | - Evan Goodell
- Department of Applied Science, William & Mary, Williamsburg, VA, United States
| | - Matthew J. Goodwin
- Department of Chemistry, William & Mary, Williamsburg, VA, United States
| | - Elizabeth Harbron
- Department of Chemistry, William & Mary, Williamsburg, VA, United States
| | - Riqiang Fu
- National High Magnetic Field Laboratory, Tallahassee, FL, United States
| | - Mihaela Mihailescu
- Institute for Bioscience and Biotechnology Research, Rockville, MD, United States
| | - Aarthi Narayanan
- Department of Biology, George Mason University, Manassas, VA, United States
| | - Myriam L. Cotten
- Department of Applied Science, William & Mary, Williamsburg, VA, United States
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
21
|
Manganelli V, Dini L, Tacconi S, Dinarelli S, Capozzi A, Riitano G, Recalchi S, Caglar TR, Fratini F, Misasi R, Sorice M, Garofalo T. Autophagy Promotes Enrichment of Raft Components within Extracellular Vesicles Secreted by Human 2FTGH Cells. Int J Mol Sci 2024; 25:6175. [PMID: 38892363 PMCID: PMC11172899 DOI: 10.3390/ijms25116175] [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: 03/21/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Autophagy plays a key role in removing protein aggregates and damaged organelles. In addition to its conventional degradative functions, autophagy machinery contributes to the release of cytosolic proteins through an unconventional secretion pathway. In this research, we analyzed autophagy-induced extracellular vesicles (EVs) in HT1080-derived human fibrosarcoma 2FTGH cells using transmission electron microscopy and atomic force microscopy (AFM). We preliminary observed that autophagy induces the formation of a subset of large heterogeneous intracellular vesicular structures. Moreover, AFM showed that autophagy triggering led to a more visible smooth cell surface with a reduced amount of plasma membrane protrusions. Next, we characterized EVs secreted by cells following autophagy induction, demonstrating that cells release both plasma membrane-derived microvesicles and exosomes. A self-forming iodixanol gradient was performed for cell subfractionation. Western blot analysis showed that endogenous LC3-II co-fractionated with CD63 and CD81. Then, we analyzed whether raft components are enriched within EV cargoes following autophagy triggering. We observed that the raft marker GD3 and ER marker ERLIN1 co-fractionated with LC3-II; dual staining by immunogold electron microscopy and coimmunoprecipitation revealed GD3-LC3-II association, indicating that autophagy promotes enrichment of raft components within EVs. Introducing a new brick in the crosstalk between autophagy and the endolysosomal system may have important implications for the knowledge of pathogenic mechanisms, suggesting alternative raft target therapies in diseases in which the generation of EV is active.
Collapse
Affiliation(s)
- Valeria Manganelli
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (V.M.); (A.C.); (G.R.); (S.R.); (T.R.C.); (R.M.); (T.G.)
| | - Luciana Dini
- Department of Biology and Biotechnology C. Darwin, “Sapienza” University of Rome, 00185 Rome, Italy;
| | - Stefano Tacconi
- CarMeN Laboratory, INSERM 1060-INRAE 1397, Department of Human Nutrition, Lyon Sud Hospital, University of Lyon, 69310 Lyon, France;
| | - Simone Dinarelli
- Institute for the Structure of Matter (ISM), National Research Council (CNR), Via del Fosso del Cavaliere 100, 00133 Rome, Italy;
| | - Antonella Capozzi
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (V.M.); (A.C.); (G.R.); (S.R.); (T.R.C.); (R.M.); (T.G.)
| | - Gloria Riitano
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (V.M.); (A.C.); (G.R.); (S.R.); (T.R.C.); (R.M.); (T.G.)
| | - Serena Recalchi
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (V.M.); (A.C.); (G.R.); (S.R.); (T.R.C.); (R.M.); (T.G.)
| | - Tuba Rana Caglar
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (V.M.); (A.C.); (G.R.); (S.R.); (T.R.C.); (R.M.); (T.G.)
| | - Federica Fratini
- Proteomics Core Facility, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy;
| | - Roberta Misasi
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (V.M.); (A.C.); (G.R.); (S.R.); (T.R.C.); (R.M.); (T.G.)
| | - Maurizio Sorice
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (V.M.); (A.C.); (G.R.); (S.R.); (T.R.C.); (R.M.); (T.G.)
| | - Tina Garofalo
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (V.M.); (A.C.); (G.R.); (S.R.); (T.R.C.); (R.M.); (T.G.)
| |
Collapse
|
22
|
Park CHK, Kim D, Kim B, Rhee SJ, Cho SJ, Ahn YM. Serum lipids as predictive markers for death by suicide. Psychiatry Res 2024; 335:115837. [PMID: 38492263 DOI: 10.1016/j.psychres.2024.115837] [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: 11/22/2023] [Revised: 02/18/2024] [Accepted: 03/03/2024] [Indexed: 03/18/2024]
Abstract
Serum lipid levels have been associated with an increased risk of suicidal behaviors. This retrospective cohort study aimed to investigate the association between serum lipid levels and death by suicide among suicide attempters according to sex. Suicide attempters visiting emergency departments between 2007 and 2011 were followed up until the date of all-cause death or December 31, 2012. Sex-stratified Cox proportional hazards regression and competing risk models were constructed to obtain the hazard ratios (HR) of serum lipid measures and suicide. For each significant lipid variable in the final models, Kaplan-Meier survival analysis and cumulative incidence function (CIF) were employed to compare the time to suicide between the high- and low-lipid groups based on the best cutoff point from the receiver operating characteristic curve. In 408 female attempters (65.8 %), the HR in the Cox regression model and subdistribution HR in the competing risk model for increased total cholesterol (TC) were 0.968 and 0.970, respectively. In the Kaplan-Meier survival analysis and CIF, increased death by suicide was demonstrated in the low-TC group (< 165 mg/dL). Lower serum TC levels among female suicide attempters may predict suicide. More careful monitoring is warranted in women with lower TC levels who recently attempted suicide.
Collapse
Affiliation(s)
- C Hyung Keun Park
- Department of Psychiatry, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea.
| | - Daewook Kim
- Haru Psychiatric Clinic, 775-1 Gaya-daero, Busanjin-gu, Busan 47278, Republic of Korea.
| | - Bora Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, 401 Quarry Road, Palo Alto, CA 94305, United States; Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA 94304, United States.
| | - Sang Jin Rhee
- Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea.
| | - Sung Joon Cho
- Department of Psychiatry, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea.
| | - Yong Min Ahn
- Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea.
| |
Collapse
|
23
|
Li L, Gao J, Milewski Ł, Hu J, Różycki B. Lattice-based mesoscale simulations and mean-field theory of cell membrane adhesion. Methods Enzymol 2024; 701:425-455. [PMID: 39025578 DOI: 10.1016/bs.mie.2024.03.005] [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] [Indexed: 07/20/2024]
Abstract
Adhesion of cell membranes involves multi-scale phenomena, ranging from specific molecular binding at Angstrom scale all the way up to membrane deformations and phase separation at micrometer scale. Consequently, theory and simulations of cell membrane adhesion require multi-scale modeling and suitable approximations that capture the essential physics of these phenomena. Here, we present a mesoscale model for membrane adhesion which we have employed in a series of our recent studies. This model quantifies, in particular, how nanoscale lipid clusters physically affect and respond to the intercellular receptor-ligand binding that mediates membrane adhesion. The goal of this Chapter is to present all details and subtleties of the mean-field theory and Monte Carlo simulations of this mesoscale model, which can be used to further explore physical phenomena related to cell membrane adhesion.
Collapse
Affiliation(s)
- Long Li
- Kuang Yaming Honors School, Nanjing University, Nanjing, P.R. China; State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Jie Gao
- Kuang Yaming Honors School, Nanjing University, Nanjing, P.R. China
| | - Łukasz Milewski
- Institute of Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing, P.R. China.
| | - Bartosz Różycki
- Institute of Physics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
24
|
Erazo-Oliveras A, Muñoz-Vega M, Salinas ML, Wang X, Chapkin RS. Dysregulation of cellular membrane homeostasis as a crucial modulator of cancer risk. FEBS J 2024; 291:1299-1352. [PMID: 36282100 PMCID: PMC10126207 DOI: 10.1111/febs.16665] [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: 06/18/2022] [Revised: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Cellular membranes serve as an epicentre combining extracellular and cytosolic components with membranous effectors, which together support numerous fundamental cellular signalling pathways that mediate biological responses. To execute their functions, membrane proteins, lipids and carbohydrates arrange, in a highly coordinated manner, into well-defined assemblies displaying diverse biological and biophysical characteristics that modulate several signalling events. The loss of membrane homeostasis can trigger oncogenic signalling. More recently, it has been documented that select membrane active dietaries (MADs) can reshape biological membranes and subsequently decrease cancer risk. In this review, we emphasize the significance of membrane domain structure, organization and their signalling functionalities as well as how loss of membrane homeostasis can steer aberrant signalling. Moreover, we describe in detail the complexities associated with the examination of these membrane domains and their association with cancer. Finally, we summarize the current literature on MADs and their effects on cellular membranes, including various mechanisms of dietary chemoprevention/interception and the functional links between nutritional bioactives, membrane homeostasis and cancer biology.
Collapse
Affiliation(s)
- Alfredo Erazo-Oliveras
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Mónica Muñoz-Vega
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Michael L. Salinas
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Xiaoli Wang
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
- Center for Environmental Health Research; Texas A&M University; College Station, Texas, 77843; USA
| |
Collapse
|
25
|
Polita A, Žvirblis R, Dodonova-Vaitkūnienė J, Shivabalan AP, Maleckaitė K, Valinčius G. Bimodal effects on lipid droplets induced in cancer and non-cancer cells by chemotherapy drugs as revealed with a green-emitting BODIPY fluorescent probe. J Mater Chem B 2024; 12:3022-3030. [PMID: 38426244 DOI: 10.1039/d3tb02979d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Lipid droplets (LDs) are cytoplasmic lipid-rich organelles with important roles in lipid storage and metabolism, cell signaling and membrane biosynthesis. Additionally, multiple diseases, such as obesity, fatty liver, cardiovascular diseases and cancer, are related to the metabolic disorders of LDs. In various cancer cells, LD accumulation is associated with resistance to cell death, reduced effectiveness of chemotherapeutic drugs, and increased proliferation and aggressiveness. In this work, we present a new viscosity-sensitive, green-emitting BODIPY probe capable of distinguishing between ordered and disordered lipid phases and selectively internalising into LDs of live cells. Through the use of fluorescence lifetime imaging microscopy (FLIM), we demonstrate that LDs in live cancer (A549) and non-cancer (HEK 293T) cells have vastly different microviscosities. Additionally, we quantify the microviscosity changes in LDs under the influence of DNA-damaging chemotherapy drugs doxorubicin and etoposide. Finally, we show that doxorubicin and etoposide have different effects on the microviscosities of LDs in chemotherapy-resistant A549 cancer cells.
Collapse
Affiliation(s)
- Artūras Polita
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, LT-10257, Lithuania.
| | - Rokas Žvirblis
- Life Sciences Center, Institute of Biotechnology, Vilnius University, Saulėtekio av. 7, Vilnius, LT-10257, Lithuania
| | - Jelena Dodonova-Vaitkūnienė
- Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko st. 24, Vilnius, LT-03225, Lithuania
| | - Arun Prabha Shivabalan
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, LT-10257, Lithuania.
| | - Karolina Maleckaitė
- Center of Physical Sciences and Technology, Saulėtekio av. 3, Vilnius, LT-10257, Lithuania
| | - Gintaras Valinčius
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, LT-10257, Lithuania.
| |
Collapse
|
26
|
Ruiz GCM, do Carmo Morato LF, Pazin WM, Oliveira ON, Constantino CJL. In situ interaction between the hormone 17α-ethynylestradiol and the liquid-ordered phase composed of the lipid rafts sphingomyelin and cholesterol. Bioorg Chem 2024; 143:107002. [PMID: 38006790 DOI: 10.1016/j.bioorg.2023.107002] [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: 09/26/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023]
Abstract
Hormone treatments are frequently associated with cardiovascular diseases and cancers in women. Additionally, the detrimental effects of their presence as contaminants in water remain a concern. The transport of hormones through cell membranes is essential for their biological action, but investigating cell permeability is challenging owing to the experimental difficulty in dealing with whole cells. In this paper, we study the interaction of the synthetic hormone 17α-ethynylestradiol (EE2) with membrane models containing the key raft components sphingomyelin (SM) and cholesterol (Chol). The models consisted of Langmuir monolayers and giant unilamellar vesicles (GUVs) that represent bilayers. EE2 induced expansion of SM monolayers upon interacting with the non-hydrated amide group of SM head, but it had practically no effect on SM GUVs because these group are not available for interaction in bilayers. In contrast, EE2 interacted with hydrated phosphate group (PO2-) and amide group of SM/Chol mixture monolayer, which could explain the loss in phase contrast of liquid-ordered GUVs suggesting pore formation. A comparison with reported EE2 effects on GUVs in the fluid phase, for which no loss in phase contrast was observed, indicates that the liquid-ordered phase consisting of lipid rafts is relevant to be associated with the changes on cell permeability caused by the hormones.
Collapse
Affiliation(s)
- Gilia Cristine Marques Ruiz
- Department of Physics, School of Technology and Applied Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil.
| | - Luis Fernando do Carmo Morato
- Department of Physics, School of Technology and Applied Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | | | - Osvaldo N Oliveira
- Sao Carlos Institute of Physics, University of Sao Paulo (USP), Sao Carlos, SP, Brazil
| | - Carlos José Leopoldo Constantino
- Department of Physics, School of Technology and Applied Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| |
Collapse
|
27
|
Mlinac-Jerkovic K, Kalanj-Bognar S, Heffer M, Blažetić S. Methodological Pitfalls of Investigating Lipid Rafts in the Brain: What Are We Still Missing? Biomolecules 2024; 14:156. [PMID: 38397393 PMCID: PMC10886647 DOI: 10.3390/biom14020156] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The purpose of this review is to succinctly examine the methodologies used in lipid raft research in the brain and to highlight the drawbacks of some investigative approaches. Lipid rafts are biochemically and biophysically different from the bulk membrane. A specific lipid environment within membrane domains provides a harbor for distinct raftophilic proteins, all of which in concert create a specialized platform orchestrating various cellular processes. Studying lipid rafts has proved to be arduous due to their elusive nature, mobility, and constant dynamic reorganization to meet the cellular needs. Studying neuronal lipid rafts is particularly cumbersome due to the immensely complex regional molecular architecture of the central nervous system. Biochemical fractionation, performed with or without detergents, is still the most widely used method to isolate lipid rafts. However, the differences in solubilization when various detergents are used has exposed a dire need to find more reliable methods to study particular rafts. Biochemical methods need to be complemented with other approaches such as live-cell microscopy, imaging mass spectrometry, and the development of specific non-invasive fluorescent probes to obtain a more complete image of raft dynamics and to study the spatio-temporal expression of rafts in live cells.
Collapse
Affiliation(s)
| | | | - Marija Heffer
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Senka Blažetić
- Department of Biology, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
28
|
Chiribao ML, Díaz-Viraqué F, Libisch MG, Batthyány C, Cunha N, De Souza W, Parodi-Talice A, Robello C. Paracrine Signaling Mediated by the Cytosolic Tryparedoxin Peroxidase of Trypanosoma cruzi. Pathogens 2024; 13:67. [PMID: 38251374 PMCID: PMC10818299 DOI: 10.3390/pathogens13010067] [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: 11/30/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Peroxiredoxins are abundant and ubiquitous proteins that participate in different cellular functions, such as oxidant detoxification, protein folding, and intracellular signaling. Under different cellular conditions, peroxiredoxins can be secreted by different parasites, promoting the induction of immune responses in hosts. In this work, we demonstrated that the cytosolic tryparedoxin peroxidase of Trypanosoma cruzi (cTXNPx) is secreted by epimastigotes and trypomastigotes associated with extracellular vesicles and also as a vesicle-free protein. By confocal microscopy, we show that cTXNPx can enter host cells by an active mechanism both through vesicles and as a recombinant protein. Transcriptomic analysis revealed that cTXNPx induces endoplasmic reticulum stress and interleukin-8 expression in epithelial cells. This analysis also suggested alterations in cholesterol metabolism in cTXNPx-treated cells, which was confirmed by immunofluorescence showing the accumulation of LDL and the induction of LDL receptors in both epithelial cells and macrophages. BrdU incorporation assays and qPCR showed that cTXNPx has a mitogenic, proliferative, and proinflammatory effect on these cells in a dose-dependent manner. Importantly, we also demonstrated that cTXNPx acts as a paracrine virulence factor, increasing the susceptibility to infection in cTXNPx-pretreated epithelial cells by approximately 40%. Although the results presented in this work are from in vitro studies and likely underestimate the complexity of parasite-host interactions, our work suggests a relevant role for this protein in establishing infection.
Collapse
Affiliation(s)
- María Laura Chiribao
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11000, Uruguay;
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - Florencia Díaz-Viraqué
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - María Gabriela Libisch
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Montevideo 11000, Uruguay;
| | - Narcisa Cunha
- Instituto de Biofísica Carlos Chagas Filho, Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (N.C.); (W.D.S.)
| | - Wanderley De Souza
- Instituto de Biofísica Carlos Chagas Filho, Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (N.C.); (W.D.S.)
| | - Adriana Parodi-Talice
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
- Sección Genética Evolutiva, Instituto de Biología, Facultad de Ciencias, Universidad de la República, Montevideo 11000, Uruguay
| | - Carlos Robello
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11000, Uruguay;
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| |
Collapse
|
29
|
Coradini D. Impact of De Novo Cholesterol Biosynthesis on the Initiation and Progression of Breast Cancer. Biomolecules 2024; 14:64. [PMID: 38254664 PMCID: PMC10813427 DOI: 10.3390/biom14010064] [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: 11/28/2023] [Revised: 12/26/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024] Open
Abstract
Cholesterol (CHOL) is a multifaceted lipid molecule. It is an essential structural component of cell membranes, where it cooperates in regulating the intracellular trafficking and signaling pathways. Additionally, it serves as a precursor for vital biomolecules, including steroid hormones, isoprenoids, vitamin D, and bile acids. Although CHOL is normally uptaken from the bloodstream, cells can synthesize it de novo in response to an increased requirement due to physiological tissue remodeling or abnormal proliferation, such as in cancer. Cumulating evidence indicated that increased CHOL biosynthesis is a common feature of breast cancer and is associated with the neoplastic transformation of normal mammary epithelial cells. After an overview of the multiple biological activities of CHOL and its derivatives, this review will address the impact of de novo CHOL production on the promotion of breast cancer with a focus on mammary stem cells. The review will also discuss the effect of de novo CHOL production on in situ and invasive carcinoma and its impact on the response to adjuvant treatment. Finally, the review will discuss the present and future therapeutic strategies to normalize CHOL biosynthesis.
Collapse
Affiliation(s)
- Danila Coradini
- Laboratory of Medical Statistics and Biometry, "Giulio A. Maccacaro", Department of Clinical Sciences and Community Health, University of Milan, Campus Cascina Rosa, 20133 Milan, Italy
| |
Collapse
|
30
|
Ding C, Chen Y, Miao G, Qi Z. Research Advances on the Role of Lipids in the Life Cycle of Human Coronaviruses. Microorganisms 2023; 12:63. [PMID: 38257890 PMCID: PMC10820681 DOI: 10.3390/microorganisms12010063] [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: 11/13/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Coronaviruses (CoVs) are emerging pathogens with a significant potential to cause life-threatening harm to human health. Since the beginning of the 21st century, three highly pathogenic and transmissible human CoVs have emerged, triggering epidemics and posing major threats to global public health. CoVs are enveloped viruses encased in a lipid bilayer. As fundamental components of cells, lipids can play an integral role in many physiological processes, which have been reported to play important roles in the life cycle of CoVs, including viral entry, uncoating, replication, assembly, and release. Therefore, research on the role of lipids in the CoV life cycle can provide a basis for a better understanding of the infection mechanism of CoVs and provide lipid targets for the development of new antiviral strategies. In this review, research advances on the role of lipids in different stages of viral infection and the possible targets of lipids that interfere with the viral life cycle are discussed.
Collapse
Affiliation(s)
- Cuiling Ding
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (C.D.); (Y.C.)
| | - Yibo Chen
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (C.D.); (Y.C.)
| | - Gen Miao
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China;
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (C.D.); (Y.C.)
| |
Collapse
|
31
|
Mills JT, Minogue SC, Snowden JS, Arden WKC, Rowlands DJ, Stonehouse NJ, Wobus CE, Herod MR. Amino acid substitutions in norovirus VP1 dictate host dissemination via variations in cellular attachment. J Virol 2023; 97:e0171923. [PMID: 38032199 PMCID: PMC10734460 DOI: 10.1128/jvi.01719-23] [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: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023] Open
Abstract
IMPORTANCE All viruses initiate infection by utilizing receptors to attach to target host cells. These virus-receptor interactions can therefore dictate viral replication and pathogenesis. Understanding the nature of virus-receptor interactions could also be important for the development of novel therapies. Noroviruses are non-enveloped icosahedral viruses of medical importance. They are a common cause of acute gastroenteritis with no approved vaccine or therapy and are a tractable model for studying fundamental virus biology. In this study, we utilized the murine norovirus model system to show that variation in a single amino acid of the major capsid protein alone can affect viral infectivity through improved attachment to suspension cells. Modulating plasma membrane mobility reduced infectivity, suggesting an importance of membrane mobility for receptor recruitment and/or receptor conformation. Furthermore, different substitutions at this site altered viral tissue distribution in a murine model, illustrating how in-host capsid evolution could influence viral infectivity and/or immune evasion.
Collapse
Affiliation(s)
- Jake T. Mills
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Susanna C. Minogue
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Joseph S. Snowden
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Wynter K. C. Arden
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - David J. Rowlands
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nicola J. Stonehouse
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Christiane E. Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Morgan R. Herod
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
32
|
Guo Y, Livelo C, Melkani G. Time-restricted feeding regulates lipid metabolism under metabolic challenges. Bioessays 2023; 45:e2300157. [PMID: 37850554 PMCID: PMC10841423 DOI: 10.1002/bies.202300157] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023]
Abstract
Dysregulation of lipid metabolism is a commonly observed feature associated with metabolic syndrome and leads to the development of negative health outcomes such as obesity, diabetes mellitus, non-alcoholic fatty liver disease, or atherosclerosis. Time-restricted feeding/eating (TRF/TRE), an emerging dietary intervention, has been shown to promote pleiotropic health benefits including the alteration of diurnal expression of genes associated with lipid metabolism, as well as levels of lipid species. Although TRF likely induces a response in multiple organs leading to the modulation of lipid metabolism, a majority of the studies related to TRF effects on lipids have focused only on individual tissues, and furthermore there is a lack of insight into potential underlying mechanisms. In this review, we summarize the current insights regarding TRF effects on lipid metabolism and the potential mechanisms in adipose tissue, liver, skeletal muscle, and heart, and conclude by outlining possible avenues for future exploration.
Collapse
Affiliation(s)
- Yiming Guo
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christopher Livelo
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Girish Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
33
|
Chen R. Cholesterol modulation of interactions between psychostimulants and dopamine transporters. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:35-59. [PMID: 38467486 DOI: 10.1016/bs.apha.2023.09.004] [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] [Indexed: 03/13/2024]
Abstract
The dopamine transporter (DAT) is a key site of action for cocaine and amphetamines. Dysfunctional DAT is associated with aberrant synaptic dopamine transmission and enhanced drug-seeking and taking behavior. Studies in cultured cells and ex vivo suggest that DAT function is sensitive to membrane cholesterol content. Although it is largely unknown whether psychostimulants alter cholesterol metabolism in the brain, emerging evidence indicates that peripheral cholesterol metabolism is altered in patients with psychostimulant use disorder and circulating cholesterol levels are associated with vulnerability to relapse. Cholesterol interacts with sphingolipids forming lipid raft microdomains on the membrane. These cholesterol-rich lipid raft microdomains serve to recruit and assemble other lipids and proteins to initiate signal transduction. There are two spatially and functionally distinct populations of the DAT segregated by cholesterol-rich lipid raft microdomains and cholesterol-scarce non-raft microdomains on the plasma membrane. These two DAT populations are differentially regulated by DAT blockers (e.g. cocaine), substrates (e.g. amphetamine), and protein kinase C providing distinct cholesterol-dependent modulation of dopamine uptake and efflux. In this chapter, we summarize the impact of depletion and addition of membrane cholesterol on DAT conformational changes between the outward-facing and the inward-facing states, lipid raft-associated DAT localization, basal and induced DAT internalization, and DAT function. In particular, we focus on how the interactions of the DAT with cocaine and amphetamine are influenced by membrane cholesterol. Lastly, we discuss the therapeutic potential of cholesterol-modifying drugs as a new avenue to normalize DAT function and dopamine transmission in patients with psychostimulant use disorder.
Collapse
Affiliation(s)
- Rong Chen
- Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Winston Salem, NC, United States.
| |
Collapse
|
34
|
Varela L, van de Lest CHA, Boere J, Libregts SFWM, Lozano-Andrés E, van Weeren PR, Wauben MHM. Acute joint inflammation induces a sharp increase in the number of synovial fluid EVs and modifies their phospholipid profile. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159367. [PMID: 37473834 DOI: 10.1016/j.bbalip.2023.159367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Inflammation is the hallmark of most joint disorders. However, the precise regulation of induction, perpetuation, and resolution of joint inflammation is not entirely understood. Since extracellular vesicles (EVs) are critical for intercellular communication, we aim to unveil their role in these processes. Here, we investigated the EVs' dynamics and phospholipidome profile from synovial fluid (SF) of healthy equine joints and from horses with lipopolysaccharide (LPS)-induced synovitis. LPS injection triggered a sharp increase of SF-EVs at 5-8 h post-injection, which started to decline at 24 h post-injection. Importantly, we identified significant changes in the lipid profile of SF-EVs after synovitis induction. Compared to healthy joint-derived SF-EVs (0 h), SF-EVs collected at 5, 24, and 48 h post-LPS injection were strongly increased in hexosylceramides. At the same time, phosphatidylserine, phosphatidylcholine, and sphingomyelin were decreased in SF-EVs at 5 h and 24 h post-LPS injection. Based on the lipid changes during acute inflammation, we composed specific lipid profiles associated with healthy and inflammatory state-derived SF-EVs. The sharp increase in SF-EVs during acute synovitis and the correlation of specific lipids with either healthy or inflamed states-derived SF-EVs are findings of potential interest for unveiling the role of SF-EVs in joint inflammation, as well as for the identification of EV-biomarkers of joint inflammation.
Collapse
Affiliation(s)
- Laura Varela
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Chris H A van de Lest
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Janneke Boere
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sten F W M Libregts
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Estefanía Lozano-Andrés
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division of Infectious Diseases & Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - P René van Weeren
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Marca H M Wauben
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
35
|
Rani S, Lai A, Nair S, Sharma S, Handberg A, Carrion F, Möller A, Salomon C. Extracellular vesicles as mediators of cell-cell communication in ovarian cancer and beyond - A lipids focus. Cytokine Growth Factor Rev 2023; 73:52-68. [PMID: 37423866 DOI: 10.1016/j.cytogfr.2023.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
Extracellular vesicles (EVs) are messengers that carry information in the form of proteins, lipids, and nucleic acids and are not only essential for intercellular communication but also play a critical role in the progression of various pathologies, including ovarian cancer. There has been recent substantial research characterising EV cargo, specifically, the lipid profile of EVs. Lipids are involved in formation and cargo sorting of EVs, their release and cellular uptake. Numerous lipidomic studies demonstrated the enrichment of specific classes of lipids in EVs derived from cancer cells suggesting that the EV associated lipids can potentially be employed as minimally invasive biomarkers for early diagnosis of various malignancies, including ovarian cancer. In this review, we aim to provide a general overview of the heterogeneity of EV, biogenesis, their lipid content, and function in cancer progression focussing on ovarian cancer.
Collapse
Affiliation(s)
- Shikha Rani
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Andrew Lai
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Soumya Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Shayna Sharma
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Andreas Möller
- Department of Otorhinolaryngology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia; Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
36
|
York JM. Temperature activated transient receptor potential ion channels from Antarctic fishes. Open Biol 2023; 13:230215. [PMID: 37848053 PMCID: PMC10581778 DOI: 10.1098/rsob.230215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/01/2023] [Indexed: 10/19/2023] Open
Abstract
Antarctic notothenioid fishes (cryonotothenioids) live in waters that range between -1.86°C and an extreme maximum +4°C. Evidence suggests these fish sense temperature peripherally, but the molecular mechanism of temperature sensation in unknown. Previous work identified transient receptor potential (TRP) channels TRPA1b, TRPM4 and TRPV1a as the top candidates for temperature sensors. Here, cryonotothenioid TRPA1b and TRPV1a are characterized using Xenopus oocyte electrophysiology. TRPA1b and TRPV1a showed heat-evoked currents with Q10s of 11.1 ± 2.2 and 20.5 ± 2.4, respectively. Unexpectedly, heat activation occurred at a threshold of 22.9 ± 1.3°C for TRPA1b and 32.1 ± 0.6°C for TRPV1a. These fish have not experienced such temperatures for at least 15 Myr. Either (1) another molecular mechanism underlies temperature sensation, (2) these fishes do not sense temperatures below these thresholds despite having lethal limits as low as 5°C, or (3) native cellular conditions modify the TRP channels to function at relevant temperatures. The effects of osmolytes, pH, oxidation, phosphorylation, lipids and accessory proteins were tested. No conditions shifted the activity range of TRPV1a. Oxidation in combination with reduced cholesterol significantly dropped activation threshold of TRPA1b to 11.3 ± 2.3°C, it is hypothesized the effect may be due to lipid raft disruption.
Collapse
Affiliation(s)
- Julia M. York
- Department of Integrative Biology, Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- School of Integrative Biology, University of Illinois Urbana–Champaign, Urbana, Illinois, USA
| |
Collapse
|
37
|
Marques-da-Silva D, Lagoa R. Rafting on the Evidence for Lipid Raft-like Domains as Hubs Triggering Environmental Toxicants' Cellular Effects. Molecules 2023; 28:6598. [PMID: 37764374 PMCID: PMC10536579 DOI: 10.3390/molecules28186598] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The plasma membrane lipid rafts are cholesterol- and sphingolipid-enriched domains that allow regularly distributed, sub-micro-sized structures englobing proteins to compartmentalize cellular processes. These membrane domains can be highly heterogeneous and dynamic, functioning as signal transduction platforms that amplify the local concentrations and signaling of individual components. Moreover, they participate in cell signaling routes that are known to be important targets of environmental toxicants affecting cell redox status and calcium homeostasis, immune regulation, and hormonal functions. In this work, the evidence that plasma membrane raft-like domains operate as hubs for toxicants' cellular actions is discussed, and suggestions for future research are provided. Several studies address the insertion of pesticides and other organic pollutants into membranes, their accumulation in lipid rafts, or lipid rafts' disruption by polychlorinated biphenyls (PCBs), benzo[a]pyrene (B[a]P), and even metals/metalloids. In hepatocytes, macrophages, or neurons, B[a]P, airborne particulate matter, and other toxicants caused rafts' protein and lipid remodeling, oxidative changes, or amyloidogenesis. Different studies investigated the role of the invaginated lipid rafts present in endothelial cells in mediating the vascular inflammatory effects of PCBs. Furthermore, in vitro and in vivo data strongly implicate raft-localized NADPH oxidases, the aryl hydrocarbon receptor, caveolin-1, and protein kinases in the toxic mechanisms of occupational and environmental chemicals.
Collapse
Affiliation(s)
- Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Ricardo Lagoa
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| |
Collapse
|
38
|
Matveeva M, Lefebvre M, Chahinian H, Yahi N, Fantini J. Host Membranes as Drivers of Virus Evolution. Viruses 2023; 15:1854. [PMID: 37766261 PMCID: PMC10535233 DOI: 10.3390/v15091854] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The molecular mechanisms controlling the adaptation of viruses to host cells are generally poorly documented. An essential issue to resolve is whether host membranes, and especially lipid rafts, which are usually considered passive gateways for many enveloped viruses, also encode informational guidelines that could determine virus evolution. Due to their enrichment in gangliosides which confer an electronegative surface potential, lipid rafts impose a first control level favoring the selection of viruses with enhanced cationic areas, as illustrated by SARS-CoV-2 variants. Ganglioside clusters attract viral particles in a dynamic electrostatic funnel, the more cationic viruses of a viral population winning the race. However, electrostatic forces account for only a small part of the energy of raft-virus interaction, which depends mainly on the ability of viruses to form a network of hydrogen bonds with raft gangliosides. This fine tuning of virus-ganglioside interactions, which is essential to stabilize the virus on the host membrane, generates a second level of selection pressure driven by a typical induced-fit mechanism. Gangliosides play an active role in this process, wrapping around the virus spikes through a dynamic quicksand-like mechanism. Viruses are thus in an endless race for access to lipid rafts, and they are bound to evolve perpetually, combining speed (electrostatic potential) and precision (fine tuning of amino acids) under the selective pressure of the immune system. Deciphering the host membrane guidelines controlling virus evolution mechanisms may open new avenues for the design of innovative antivirals.
Collapse
Affiliation(s)
| | | | | | | | - Jacques Fantini
- Department of Biology, Faculty of Medicine, University of Aix-Marseille, INSERM UMR_S 1072, 13015 Marseille, France; (M.M.); (M.L.); (H.C.); (N.Y.)
| |
Collapse
|
39
|
Garvey CJ, Bryant SJ, Elbourne A, Hunt T, Kent B, Kreuzer M, Strobl M, Steitz R, Bryant G. Phase separation in a ternary DPPC/DOPC/POPC system with reducing hydration. J Colloid Interface Sci 2023; 638:719-732. [PMID: 36774881 DOI: 10.1016/j.jcis.2023.01.145] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/10/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The maintenance of plasma membrane structure is vital for the viability of cells. Disruption of this structure can lead to cell death. One important example is the macroscopic phase separation observed during dehydration associated with desiccation and freezing, often leading to loss of permeability and cell death. It has previously been shown that the hybrid lipid 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) can act as a line-active component in ternary lipid systems, inhibiting macroscopic phase separation and stabilising membrane microdomains in lipid vesicles [1]. The domain size is found to decrease with increasing POPC concentration until complete mixing is observed. However, no such studies have been carried out at reduced hydration. To examine if this phase separation is unique to vesicles in excess water, we have conducted studies on several binary and ternary model membrane systems at both reduced hydration ("powder" type samples and oriented membrane stacks) and in excess water (supported lipid bilayers) at 0.2 mol fraction POPC, in the range where microdomain stabilisation is reported. Differential scanning calorimetry (DSC) and Fourier transform infrared spectroscopy (FTIR) are used to map phase transition temperatures, with X-ray and neutron scattering providing details of the changes in lipid packing and phase information within these boundaries. Atomic force microscopy (AFM) is used to image bilayers on a substrate in excess water. In all cases, macroscopic phase separation was observed rather than microdomain formation at this molar ratio. Thus POPC does not stabilise microdomains under these conditions, regardless of the type of model membrane, hydration or temperature. Thus we conclude that the driving force for separation under these conditions overcomes any linactant effects of the hybrid lipid.
Collapse
Affiliation(s)
- Christopher J Garvey
- Heinz Maier-Leibnitz Zentrum (MLZ), Technische Universität München, Lichtenbergstraße 1, 85748 Garching, Germany.
| | | | - Aaron Elbourne
- School of Science, RMIT University, Melbourne, Australia
| | - Taavi Hunt
- School of Science, RMIT University, Melbourne, Australia
| | - Ben Kent
- Centre for Advanced Macromolecular Design, School of Chemistry, The University of New South Wales, Sydney 2052, Australia; Institute for Soft Matter and Functional Materials, Helmholtz Zentrum Berlin, Hahn-Meitner-Platz 1, Berlin, Germany
| | - Martin Kreuzer
- Institute for Soft Matter and Functional Materials, Helmholtz Zentrum Berlin, Hahn-Meitner-Platz 1, Berlin, Germany; ALBA Synchrotron, Barcelona, Spain
| | - Markus Strobl
- Institute for Soft Matter and Functional Materials, Helmholtz Zentrum Berlin, Hahn-Meitner-Platz 1, Berlin, Germany; Laboratory for Neutron Scattering and Imaging, Paul Scherrer Institut, 5232 Villigen, Switzerland
| | - Roland Steitz
- Institute for Soft Matter and Functional Materials, Helmholtz Zentrum Berlin, Hahn-Meitner-Platz 1, Berlin, Germany
| | - Gary Bryant
- School of Science, RMIT University, Melbourne, Australia.
| |
Collapse
|
40
|
Toledo E, Wittenbecher C, Razquin C, Ruiz-Canela M, Clish CB, Liang L, Alonso A, Hernández-Alonso P, Becerra-Tomás N, Arós-Borau F, Corella D, Ros E, Estruch R, García-Rodríguez A, Fitó M, Lapetra J, Fiol M, Alonso-Gomez ÁM, Serra-Majem L, Deik A, Salas-Salvadó J, Hu FB, Martínez-González MA. Plasma lipidome and risk of atrial fibrillation: results from the PREDIMED trial. J Physiol Biochem 2023; 79:355-364. [PMID: 37004634 PMCID: PMC10300169 DOI: 10.1007/s13105-023-00958-0] [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: 11/18/2021] [Accepted: 03/16/2023] [Indexed: 04/04/2023]
Abstract
The potential role of the lipidome in atrial fibrillation (AF) development is still widely unknown. We aimed to assess the association between lipidome profiles of the Prevención con Dieta Mediterránea (PREDIMED) trial participants and incidence of AF. We conducted a nested case-control study (512 incident centrally adjudicated AF cases and 735 controls matched by age, sex, and center). Baseline plasma lipids were profiled using a Nexera X2 U-HPLC system coupled to an Exactive Plus orbitrap mass spectrometer. We estimated the association between 216 individual lipids and AF using multivariable conditional logistic regression and adjusted the p values for multiple testing. We also examined the joint association of lipid clusters with AF incidence. Hitherto, we estimated the lipidomics network, used machine learning to select important network-clusters and AF-predictive lipid patterns, and summarized the joint association of these lipid patterns weighted scores. Finally, we addressed the possible interaction by the randomized dietary intervention.Forty-one individual lipids were associated with AF at the nominal level (p < 0.05), but no longer after adjustment for multiple-testing. However, the network-based score identified with a robust data-driven lipid network showed a multivariable-adjusted ORper+1SD of 1.32 (95% confidence interval: 1.16-1.51; p < 0.001). The score included PC plasmalogens and PE plasmalogens, palmitoyl-EA, cholesterol, CE 16:0, PC 36:4;O, and TG 53:3. No interaction with the dietary intervention was found. A multilipid score, primarily made up of plasmalogens, was associated with an increased risk of AF. Future studies are needed to get further insights into the lipidome role on AF.Current Controlled Trials number, ISRCTN35739639.
Collapse
Affiliation(s)
- Estefania Toledo
- Department of Preventive Medicine and Public Health, Edificio de Investigación, University of Navarra, Planta 2, Calle Irunlarrea 1, 31008, Pamplona, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Clemens Wittenbecher
- SciLifeLab & Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Cristina Razquin
- Department of Preventive Medicine and Public Health, Edificio de Investigación, University of Navarra, Planta 2, Calle Irunlarrea 1, 31008, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, Edificio de Investigación, University of Navarra, Planta 2, Calle Irunlarrea 1, 31008, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Liming Liang
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Pablo Hernández-Alonso
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Bioquímica I Biotecnologia, Universitat Rovira I Virgili, Unitat de Nutrició Humana, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari San Joan de Reus, Reus, Spain
- Unidad de Gestión Clínica de Endocrinología Y Nutrición del Hospital Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Nerea Becerra-Tomás
- Departament de Bioquímica I Biotecnologia, Universitat Rovira I Virgili, Unitat de Nutrició Humana, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari San Joan de Reus, Reus, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain
| | - Fernando Arós-Borau
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Bioaraba Health Research Institute Osakidetza Basque Health Service, Araba University Hospital University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Dolores Corella
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain
| | - Emilio Ros
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital Clinic, Barcelona, Spain
| | - Ramón Estruch
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Internal Medicine, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | | | - Montserrat Fitó
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Cardiovascular Risk and Nutrition Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - José Lapetra
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, 41013, Seville, Spain
| | - Miquel Fiol
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- IdISBa. Health Research Institute of the Balearis Islands, Palma, Spain
| | - Ángel M Alonso-Gomez
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Bioaraba Health Research Institute Osakidetza Basque Health Service, Araba University Hospital University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Luis Serra-Majem
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Research Institute of Biomedical and Health Sciences, University of Las Palmas de Gran Canaria, & Centro Hospitalario Universitario Insular Materno Infantil (CHUIMI) del Servicio Canario de Salud, Gobierno de Canarias, Las Palmas de Gran Canaria, Spain
| | - Amy Deik
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jordi Salas-Salvadó
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Bioquímica I Biotecnologia, Universitat Rovira I Virgili, Unitat de Nutrició Humana, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari San Joan de Reus, Reus, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Miguel A Martínez-González
- Department of Preventive Medicine and Public Health, Edificio de Investigación, University of Navarra, Planta 2, Calle Irunlarrea 1, 31008, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
41
|
Raji AO. Optimization of Roasting Assisted Oil Extraction from Tiger Nut Using Response Surface Methodology. CHEMISTRY AFRICA 2023. [DOI: 10.1007/s42250-023-00639-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
42
|
Edwards-Hicks J, Apostolova P, Buescher JM, Maib H, Stanczak MA, Corrado M, Klein Geltink RI, Maccari ME, Villa M, Carrizo GE, Sanin DE, Baixauli F, Kelly B, Curtis JD, Haessler F, Patterson A, Field CS, Caputa G, Kyle RL, Soballa M, Cha M, Paul H, Martin J, Grzes KM, Flachsmann L, Mitterer M, Zhao L, Winkler F, Rafei-Shamsabadi DA, Meiss F, Bengsch B, Zeiser R, Puleston DJ, O'Sullivan D, Pearce EJ, Pearce EL. Phosphoinositide acyl chain saturation drives CD8 + effector T cell signaling and function. Nat Immunol 2023; 24:516-530. [PMID: 36732424 PMCID: PMC10908374 DOI: 10.1038/s41590-023-01419-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 01/03/2023] [Indexed: 02/04/2023]
Abstract
How lipidome changes support CD8+ effector T (Teff) cell differentiation is not well understood. Here we show that, although naive T cells are rich in polyunsaturated phosphoinositides (PIPn with 3-4 double bonds), Teff cells have unique PIPn marked by saturated fatty acyl chains (0-2 double bonds). PIPn are precursors for second messengers. Polyunsaturated phosphatidylinositol bisphosphate (PIP2) exclusively supported signaling immediately upon T cell antigen receptor activation. In late Teff cells, activity of phospholipase C-γ1, the enzyme that cleaves PIP2 into downstream mediators, waned, and saturated PIPn became essential for sustained signaling. Saturated PIP was more rapidly converted to PIP2 with subsequent recruitment of phospholipase C-γ1, and loss of saturated PIPn impaired Teff cell fitness and function, even in cells with abundant polyunsaturated PIPn. Glucose was the substrate for de novo PIPn synthesis, and was rapidly utilized for saturated PIP2 generation. Thus, separate PIPn pools with distinct acyl chain compositions and metabolic dependencies drive important signaling events to initiate and then sustain effector function during CD8+ T cell differentiation.
Collapse
Affiliation(s)
- Joy Edwards-Hicks
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Petya Apostolova
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Hannes Maib
- Division of Cell & Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Michal A Stanczak
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mauro Corrado
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | | - Maria Elena Maccari
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matteo Villa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Gustavo E Carrizo
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David E Sanin
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Francesc Baixauli
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Beth Kelly
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan D Curtis
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fabian Haessler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Annette Patterson
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Cameron S Field
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - George Caputa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Ryan L Kyle
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Melanie Soballa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Minsun Cha
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Harry Paul
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacob Martin
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katarzyna M Grzes
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lea Flachsmann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Michael Mitterer
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Liang Zhao
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frances Winkler
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - David Ali Rafei-Shamsabadi
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Frank Meiss
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bertram Bengsch
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel J Puleston
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David O'Sullivan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Edward J Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Erika L Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
43
|
Mills JT, Minogue SC, Snowden JS, Arden WKC, Rowlands DJ, Stonehouse NJ, Wobus CE, Herod MR. Amino acid substitutions in norovirus VP1 dictate cell tropism via an attachment process dependent on membrane mobility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.17.528071. [PMID: 36824911 PMCID: PMC9949111 DOI: 10.1101/2023.02.17.528071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Viruses interact with receptors on the cell surface to initiate and co-ordinate infection. The distribution of receptors on host cells can be a key determinant of viral tropism and host infection. Unravelling the complex nature of virus-receptor interactions is, therefore, of fundamental importance to understanding viral pathogenesis. Noroviruses are non-enveloped, icosahedral, positive-sense RNA viruses of global importance to human health, with no approved vaccine or antiviral agent available. Here we use murine norovirus as a model for the study of molecular mechanisms of virus-receptor interactions. We show that variation at a single amino acid residue in the major viral capsid protein had a key impact on the interaction between virus and receptor. This variation did not affect virion production or virus growth kinetics, but a specific amino acid was rapidly selected through evolution experiments, and significantly improved cellular attachment when infecting immune cells in suspension. However, reducing plasma membrane mobility counteracted this phenotype, providing insight into for the role of membrane fluidity and receptor recruitment in norovirus cellular attachment. When the infectivity of a panel of recombinant viruses with single amino acid variations was compared in vivo, there were significant differences in the distribution of viruses in a murine model, demonstrating a role in cellular tropism in vivo. Overall, these results highlight the importance of lipid rafts and virus-induced receptor recruitment in viral infection, as well as how capsid evolution can greatly influence cellular tropism, within-host spread and pathogenicity.
Collapse
Affiliation(s)
- Jake T Mills
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Susanna C Minogue
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Joseph S Snowden
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Wynter K C Arden
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48130, USA
| | - David J Rowlands
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Nicola J Stonehouse
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48130, USA
| | - Morgan R Herod
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
44
|
Ullo MF, Case LB. How cells sense and integrate information from different sources. WIREs Mech Dis 2023:e1604. [PMID: 36781396 DOI: 10.1002/wsbm.1604] [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: 06/24/2022] [Revised: 01/06/2023] [Accepted: 01/24/2023] [Indexed: 02/15/2023]
Abstract
Cell signaling is a fundamental cellular process that enables cells to sense and respond to information in their surroundings. At the molecular level, signaling is primarily carried out by transmembrane protein receptors that can initiate complex downstream signal transduction cascades to alter cellular behavior. In the human body, different cells can be exposed to a wide variety of environmental conditions, and cells express diverse classes of receptors capable of sensing and integrating different signals. Furthermore, different receptors and signaling pathways can crosstalk with each other to calibrate the cellular response. Crosstalk occurs through multiple mechanisms at different levels of signaling pathways. In this review, we discuss how cells sense and integrate different chemical, mechanical, and spatial signals as well as the mechanisms of crosstalk between pathways. To illustrate these concepts, we use a few well-studied signaling pathways, including receptor tyrosine kinases and integrin receptors. Finally, we discuss the implications of dysregulated cellular sensing on driving diseases such as cancer. This article is categorized under: Cancer > Molecular and Cellular Physiology Metabolic Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Maria F Ullo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Lindsay B Case
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
45
|
Fantini J, Azzaz F, Chahinian H, Yahi N. Electrostatic Surface Potential as a Key Parameter in Virus Transmission and Evolution: How to Manage Future Virus Pandemics in the Post-COVID-19 Era. Viruses 2023; 15:284. [PMID: 36851498 PMCID: PMC9964723 DOI: 10.3390/v15020284] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Virus-cell interactions involve fundamental parameters that need to be considered in strategies implemented to control viral outbreaks. Among these, the surface electrostatic potential can give valuable information to deal with new epidemics. In this article, we describe the role of this key parameter in the hemagglutination of red blood cells and in the co-evolution of synaptic receptors and neurotransmitters. We then establish the functional link between lipid rafts and the electrostatic potential of viruses, with special emphasis on gangliosides, which are sialic-acid-containing, electronegatively charged plasma membrane components. We describe the common features of ganglioside binding domains, which include a wide variety of structures with little sequence homology but that possess key amino acids controlling ganglioside recognition. We analyze the role of the electrostatic potential in the transmission and intra-individual evolution of HIV-1 infections, including gatekeeper and co-receptor switch mechanisms. We show how to organize the epidemic surveillance of influenza viruses by focusing on mutations affecting the hemagglutinin surface potential. We demonstrate that the electrostatic surface potential, by modulating spike-ganglioside interactions, controls the hemagglutination properties of coronaviruses (SARS-CoV-1, MERS-CoV, and SARS-CoV-2) as well as the structural dynamics of SARS-CoV-2 evolution. We relate the broad-spectrum antiviral activity of repositioned molecules to their ability to disrupt virus-raft interactions, challenging the old concept that an antibiotic or anti-parasitic cannot also be an antiviral. We propose a new concept based on the analysis of the electrostatic surface potential to develop, in real time, therapeutic and vaccine strategies adapted to each new viral epidemic.
Collapse
Affiliation(s)
- Jacques Fantini
- Department of Biology, Faculty of Medicine, University of Aix-Marseille, INSERM UMR_S 1072, 13015 Marseille, France
| | | | | | | |
Collapse
|
46
|
Rosenhouse-Dantsker A, Gazgalis D, Logothetis DE. PI(4,5)P 2 and Cholesterol: Synthesis, Regulation, and Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:3-59. [PMID: 36988876 DOI: 10.1007/978-3-031-21547-6_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is the most abundant membrane phosphoinositide and cholesterol is an essential component of the plasma membrane (PM). Both lipids play key roles in a variety of cellular functions including as signaling molecules and major regulators of protein function. This chapter provides an overview of these two important lipids. Starting from a brief description of their structure, synthesis, and regulation, the chapter continues to describe the primary functions and signaling processes in which PI(4,5)P2 and cholesterol are involved. While PI(4,5)P2 and cholesterol can act independently, they often act in concert or affect each other's impact. The chapters in this volume on "Cholesterol and PI(4,5)P2 in Vital Biological Functions: From Coexistence to Crosstalk" focus on the emerging relationship between cholesterol and PI(4,5)P2 in a variety of biological systems and processes. In this chapter, the next section provides examples from the ion channel field demonstrating that PI(4,5)P2 and cholesterol can act via common mechanisms. The chapter ends with a discussion of future directions.
Collapse
Affiliation(s)
| | - Dimitris Gazgalis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Diomedes E Logothetis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| |
Collapse
|
47
|
Zhang C, Kikushima K, Endo M, Kahyo T, Horikawa M, Matsudaira T, Tanaka T, Takanashi Y, Sato T, Takahashi Y, Xu L, Takayama N, Islam A, Mamun MA, Ozawa T, Setou M. Imaging and Manipulation of Plasma Membrane Fatty Acid Clusters Using TOF-SIMS Combined Optogenetics. Cells 2022; 12:cells12010010. [PMID: 36611804 PMCID: PMC9818728 DOI: 10.3390/cells12010010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
The plasma membrane (PM) serves multiple functions to support cell activities with its heterogeneous molecular distribution. Fatty acids (FAs) are hydrophobic components of the PM whose saturation and length determine the membrane's physical properties. The FA distribution contributes to the PM's lateral heterogeneity. However, the distribution of PM FAs is poorly understood. Here, we proposed the FA cluster hypothesis, which suggested that FAs on the PM exist as clusters. By the optogenetic tool translocating the endoplasmic reticulum (ER), we were able to manipulate the distribution of PM FAs. We used time-of-flight combined secondary ion mass spectrometry (TOF-SIMS) to image PM FAs and discovered that PM FAs were presented and distributed as clusters and are also manipulated as clusters. We also found the existence of multi-FA clusters formed by the colocalization of more than one FA. Our optogenetic tool also decreased the clustering degree of FA clusters and the formation probability of multi-FA clusters. This research opens up new avenues and perspectives to study PM heterogeneity from an FA perspective. This research also suggests a possible treatment for diseases caused by PM lipid aggregation and furnished a convenient tool for therapeutic development.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kenji Kikushima
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Mizuki Endo
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Makoto Horikawa
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Hiroshima Research Center for Healthy Aging, Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8530, Japan
| | - Takaomi Matsudaira
- Foundation for Promotion of Material Science and Technology of Japan, 1-18-6 Kitami, Setagaya-ku, Tokyo 157-0067, Japan
| | - Tatsuya Tanaka
- Foundation for Promotion of Material Science and Technology of Japan, 1-18-6 Kitami, Setagaya-ku, Tokyo 157-0067, Japan
| | - Yusuke Takanashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Lili Xu
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Naoki Takayama
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Ariful Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Md. Al Mamun
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Correspondence:
| |
Collapse
|
48
|
Ramasubramanian L, Jyothi H, Goldbloom-Helzner L, Light BM, Kumar P, Carney RP, Farmer DL, Wang A. Development and Characterization of Bioinspired Lipid Raft Nanovesicles for Therapeutic Applications. ACS APPLIED MATERIALS & INTERFACES 2022; 14:54458-54477. [PMID: 36448709 PMCID: PMC9756296 DOI: 10.1021/acsami.2c13868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Lipid rafts are highly ordered regions of the plasma membrane enriched in signaling proteins and lipids. Their biological potential is realized in exosomes, a subclass of extracellular vesicles (EVs) that originate from the lipid raft domains. Previous studies have shown that EVs derived from human placental mesenchymal stromal cells (PMSCs) possess strong neuroprotective and angiogenic properties. However, clinical translation of EVs is challenged by very low, impure, and heterogeneous yields. Therefore, in this study, lipid rafts are validated as a functional biomaterial that can recapitulate the exosomal membrane and then be synthesized into biomimetic nanovesicles. Lipidomic and proteomic analyses show that lipid raft isolates retain functional lipids and proteins comparable to PMSC-EV membranes. PMSC-derived lipid raft nanovesicles (LRNVs) are then synthesized at high yields using a facile, extrusion-based methodology. Evaluation of biological properties reveals that LRNVs can promote neurogenesis and angiogenesis through modulation of lipid raft-dependent signaling pathways. A proof-of-concept methodology further shows that LRNVs could be loaded with proteins or other bioactive cargo for greater disease-specific functionalities, thus presenting a novel type of biomimetic nanovesicles that can be leveraged as targeted therapeutics for regenerative medicine.
Collapse
Affiliation(s)
- Lalithasri Ramasubramanian
- Department
of Surgery, School of Medicine, University of California-Davis, Sacramento, California 95817, United States
- Institute
for Pediatric Regenerative Medicine, Shriners
Hospitals for Children, Sacramento, California 95817, United States
- Department
of Biomedical Engineering, University of
California-Davis, Davis, California 95616, United States
| | - Harsha Jyothi
- Department
of Surgery, School of Medicine, University of California-Davis, Sacramento, California 95817, United States
| | - Leora Goldbloom-Helzner
- Department
of Surgery, School of Medicine, University of California-Davis, Sacramento, California 95817, United States
- Institute
for Pediatric Regenerative Medicine, Shriners
Hospitals for Children, Sacramento, California 95817, United States
- Department
of Biomedical Engineering, University of
California-Davis, Davis, California 95616, United States
| | - Brandon M. Light
- Department
of Surgery, School of Medicine, University of California-Davis, Sacramento, California 95817, United States
| | - Priyadarsini Kumar
- Department
of Surgery, School of Medicine, University of California-Davis, Sacramento, California 95817, United States
- Institute
for Pediatric Regenerative Medicine, Shriners
Hospitals for Children, Sacramento, California 95817, United States
| | - Randy P. Carney
- Department
of Biomedical Engineering, University of
California-Davis, Davis, California 95616, United States
| | - Diana L. Farmer
- Department
of Surgery, School of Medicine, University of California-Davis, Sacramento, California 95817, United States
- Institute
for Pediatric Regenerative Medicine, Shriners
Hospitals for Children, Sacramento, California 95817, United States
| | - Aijun Wang
- Department
of Surgery, School of Medicine, University of California-Davis, Sacramento, California 95817, United States
- Institute
for Pediatric Regenerative Medicine, Shriners
Hospitals for Children, Sacramento, California 95817, United States
- Department
of Biomedical Engineering, University of
California-Davis, Davis, California 95616, United States
| |
Collapse
|
49
|
Kim JH, Lee CH, Baek MC. Dissecting exosome inhibitors: therapeutic insights into small-molecule chemicals against cancer. Exp Mol Med 2022; 54:1833-1843. [PMID: 36446847 PMCID: PMC9707221 DOI: 10.1038/s12276-022-00898-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
Intensive research in the field of cancer biology has revealed unique methods of communication between cells through extracellular vesicles called exosomes. Exosomes are released from a broad spectrum of cell types and serve as functional mediators under physiological or pathological conditions. Hence, blocking the release of exosome bio carriers may prove useful for slowing the progression of certain types of cancers. Therefore, efforts are being made to develop exosome inhibitors to be used both as research tools and as therapies in clinical trials. Thus, studies on exosomes may lead to a breakthrough in cancer research, for which new clinical targets for different types of cancers are urgently needed. In this review, we briefly outline exosome inhibitors and discuss their modes of action and potential for use as therapeutic tools for cancer.
Collapse
Affiliation(s)
- Jong Hyun Kim
- grid.412072.20000 0004 0621 4958Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, 42472 South Korea
| | - Chan-Hyeong Lee
- grid.258803.40000 0001 0661 1556Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu, 41944 South Korea
| | - Moon-Chang Baek
- grid.258803.40000 0001 0661 1556Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu, 41944 South Korea
| |
Collapse
|
50
|
Myconoside Affects the Viability of Polarized Epithelial MDCKII Cell Line by Interacting with the Plasma Membrane and the Apical Junctional Complexes. SEPARATIONS 2022. [DOI: 10.3390/separations9090239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The phenyl glycoside myconoside, extracted from Balkan endemic Haberlea rhodopensis, has a positive effect on human health, but the exact molecular mechanism of its action is still unknown. The cell membrane and its associated junctional complex are the first targets of exogenous compound action. We aimed to study the effect of myconoside on membrane organization and cytoskeleton components involved in the maintenance of cell polarity in the MDCKII cell line. By fluorescent spectroscopy and microscopy, we found that at low concentrations, myconoside increases the cell viability by enhancing membrane lipid order and adherent junctions. The opposite effect is observed in high myconoside doses. We hypothesized that the cell morphological and physicochemical changes of the analyzed cell compartments are directly related to cell viability and cell apical-basal polarity. Our finding contributes to a better understanding of the beneficial application of phytochemical myconoside in pharmacology and medicine.
Collapse
|