1
|
Villalaín J. Membrane fusion by dengue virus: The first step. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184400. [PMID: 39522596 DOI: 10.1016/j.bbamem.2024.184400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Flaviviruses include important human pathogens such as Dengue, Zika, West Nile, Yellow fever, Japanese encephalitis, and Tick-borne encephalitis viruses as well as some emerging viruses that affect millions of people worldwide. They fuse their membrane with the late endosomal one in a pH-dependent way and therefore the merging of the membranes is one of the main goals for obtaining new antivirals. The envelope E protein, a membrane fusion protein, is accountable for fusion and encompasses different domains involved in the fusion mechanism, including the fusion peptide segment. In this work we have used molecular dynamics to study the interaction of the distal end of domain II of the DENV envelope E protein with a membrane like the late endosomal membrane in order to observe the initiation of membrane fusion carried out by a number of trimers of the DENV envelope E protein interacting with a complex biomembrane and demonstrate its feasibility. Our results demonstrate the likelihood of membrane disorganization and pore formation by trimer complex organization, the amino acids responsible for such condition and the secondary structure arrangements needed for such fundamental process. At the same time, we define new targets of the envelope E protein sequence which could permit designing potent antiviral bioactive molecules.
Collapse
Affiliation(s)
- José Villalaín
- Institute of Research, Development, and Innovation in Healthcare Biotechnology (IDiBE), Universitas "Miguel Hernández", E-03202 Elche-Alicante, Spain.
| |
Collapse
|
2
|
D'Souza-Schorey C, Stahl PD. Resolving the two-body problem: A postulated role for the V0 sector of the V0V1-ATPase in exosome biogenesis and multivesicular body fate. Mol Biol Cell 2025; 36:pe1. [PMID: 39705591 DOI: 10.1091/mbc.e24-09-0412] [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: 12/22/2024] Open
Abstract
Because the discovery of the multivesicular body (MVB) as the origin of secreted vesicles or exosomes, the question arose and still looms-what distinguishes an MVB destined for fusion with the plasma membrane (EXO-MVB) facilitating exosome release from an MVB involved in transport of content to the lysosome (LYSO-MVB). Do they have independent origins? Hence, the two-body problem. We hypothesize that a key to this conundrum is the membrane spanning V0 sector of the proton pump, V0V1-ATPase. The V0V1-ATPase participates in the acidification of intracellular compartments, although V0 can function separately from V1 and different V0 isoforms are endowed with membrane binding capabilities that allow the V0V1-ATPase to selectively localize to different endocytic compartments including early and late endosomes and lysosomes. We propose that V0, in collaboration with cholesterol and phosphoinositides, plays a central role in the early endosome as a nucleation center to direct the de novo assembly of an EXO-MVB scaffold. The EXO-MVB scaffold may play multiple roles-operating as an assembly platform, participating in membrane fission as well as providing downstream navigational queues necessary for exosome secretion. Thus, V0 may represent an influential nexus, a starting point, in exosome biogenesis.
Collapse
Affiliation(s)
| | - Philip D Stahl
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
3
|
Dinkel L, Hummel S, Zenatti V, Malara M, Tillmann Y, Colombo A, Monasor LS, Suh JH, Logan T, Roth S, Paeger L, Hoffelner P, Bludau O, Schmidt A, Müller SA, Schifferer M, Nuscher B, Njavro JR, Prestel M, Bartos LM, Wind-Mark K, Slemann L, Hoermann L, Kunte ST, Gnörich J, Lindner S, Simons M, Herms J, Paquet D, Lichtenthaler SF, Bartenstein P, Franzmeier N, Liesz A, Grosche A, Bremova-Ertl T, Catarino C, Beblo S, Bergner C, Schneider SA, Strupp M, Di Paolo G, Brendel M, Tahirovic S. Myeloid cell-specific loss of NPC1 in mice recapitulates microgliosis and neurodegeneration in patients with Niemann-Pick type C disease. Sci Transl Med 2024; 16:eadl4616. [PMID: 39630885 DOI: 10.1126/scitranslmed.adl4616] [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: 10/19/2023] [Revised: 07/12/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Niemann-Pick type C (NPC) disease is an inherited lysosomal storage disorder mainly driven by mutations in the NPC1 gene, causing lipid accumulation within late endosomes/lysosomes and resulting in progressive neurodegeneration. Although microglial activation precedes neuronal loss, it remains elusive whether loss of the membrane protein NPC1 in microglia actively contributes to NPC pathology. In a mouse model with depletion of NPC1 in myeloid cells, we report severe alterations in microglial lipidomic profiles, including the enrichment of bis(monoacylglycero)phosphate, increased cholesterol, and a decrease in cholesteryl esters. Lipid dyshomeostasis was associated with microglial hyperactivity, marked by an increase in translocator protein 18 kDa (TSPO). These hyperactive microglia initiated a pathological cascade resembling NPC-like phenotypes, including a shortened life span, motor impairments, astrogliosis, neuroaxonal pathology, and increased neurofilament light chain (NF-L), a neuronal injury biomarker. As observed in the mouse model, patients with NPC showed increased NF-L in the blood and microglial hyperactivity, as visualized by TSPO-PET imaging. Reduced TSPO expression in blood-derived macrophages of patients with NPC was measured after N-acetyl-l-leucine treatment, which has been recently shown to have beneficial effects in patients with NPC, suggesting that TSPO is a potential marker to monitor therapeutic interventions for NPC. Conclusively, these results demonstrate that myeloid dysfunction, driven by the loss of NPC1, contributes to NPC disease and should be further investigated for therapeutic targeting and disease monitoring.
Collapse
Affiliation(s)
- Lina Dinkel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Selina Hummel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Valerio Zenatti
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Mariagiovanna Malara
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Yannik Tillmann
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Alessio Colombo
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | | | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Todd Logan
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Stefan Roth
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Lars Paeger
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Patricia Hoffelner
- Department of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, Ludwig Maximilian University, 82152 Planegg-Martinsried, Germany
| | - Oliver Bludau
- Department of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Andree Schmidt
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig Maximilian University, 82152 Planegg-Martinsried, Germany
- Neuroproteomics School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Neuroproteomics School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Brigitte Nuscher
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Jasenka Rudan Njavro
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Matthias Prestel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Karin Wind-Mark
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Luna Slemann
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Leonie Hoermann
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Sebastian T Kunte
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Institute of Neuronal Cell Biology (TUM-NZB), Technical University of Munich, 80802 Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University München, 81377 Munich, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Neuroproteomics School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Sahlgrenska Academy, Institute of Neuroscience and Physiology, SE-413 90 Mölndal and Gothenburg, Sweden
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Tatiana Bremova-Ertl
- Department of Neurology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Neurology, University Hospital Bern, 3010 Bern, Switzerland
| | - Claudia Catarino
- Friedrich Baur Institute, Department of Neurology, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Skadi Beblo
- Center for Pediatric Research Leipzig, Department of Women and Child Health, Hospital for Children and Adolescents, University Hospital Leipzig; Leipzig University Center for Rare Diseases, 04103 Leipzig, Germany
| | - Caroline Bergner
- Department of Neurology, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Susanne A Schneider
- Department of Neurology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Michael Strupp
- Department of Neurology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | | | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| |
Collapse
|
4
|
Scrima S, Lambrughi M, Favaro L, Maeda K, Jäättelä M, Papaleo E. Acidic sphingomyelinase interactions with lysosomal membranes and cation amphiphilic drugs: A molecular dynamics investigation. Comput Struct Biotechnol J 2024; 23:2516-2533. [PMID: 38974886 PMCID: PMC11226985 DOI: 10.1016/j.csbj.2024.05.049] [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: 01/12/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 07/09/2024] Open
Abstract
Lysosomes are pivotal in cellular functions and disease, influencing cancer progression and therapy resistance with Acid Sphingomyelinase (ASM) governing their membrane integrity. Moreover, cation amphiphilic drugs (CADs) are known as ASM inhibitors and have anti-cancer activity, but the structural mechanisms of their interactions with the lysosomal membrane and ASM are poorly explored. Our study, leveraging all-atom explicit solvent molecular dynamics simulations, delves into the interaction of glycosylated ASM with the lysosomal membrane and the effects of CAD representatives, i.e., ebastine, hydroxyebastine and loratadine, on the membrane and ASM. Our results confirm the ASM association to the membrane through the saposin domain, previously only shown with coarse-grained models. Furthermore, we elucidated the role of specific residues and ASM-induced membrane curvature in lipid recruitment and orientation. CADs also interfere with the association of ASM with the membrane at the level of a loop in the catalytic domain engaging in membrane interactions. Our computational approach, applicable to various CADs or membrane compositions, provides insights into ASM and CAD interaction with the membrane, offering a valuable tool for future studies.
Collapse
Affiliation(s)
- Simone Scrima
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen 2100, Denmark
- Cancer System Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby 2800, Denmark
| | - Matteo Lambrughi
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen 2100, Denmark
| | - Lorenzo Favaro
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen 2100, Denmark
| | - Kenji Maeda
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen 2100, Denmark
| | - Marja Jäättelä
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen 2100, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Elena Papaleo
- Cancer Structural Biology, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen 2100, Denmark
- Cancer System Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby 2800, Denmark
| |
Collapse
|
5
|
Singh S, Dransfeld UE, Ambaw YA, Lopez-Scarim J, Farese RV, Walther TC. PLD3 and PLD4 synthesize S,S-BMP, a key phospholipid enabling lipid degradation in lysosomes. Cell 2024; 187:6820-6834.e24. [PMID: 39423811 DOI: 10.1016/j.cell.2024.09.036] [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: 03/22/2024] [Revised: 07/07/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
Bis(monoacylglycero)phosphate (BMP) is an abundant lysosomal phospholipid required for degradation of lipids, particularly gangliosides. Alterations in BMP levels are associated with neurodegenerative diseases. Unlike typical glycerophospholipids, lysosomal BMP has two chiral glycerol carbons in the S (rather than the R) stereo-conformation, protecting it from lysosomal degradation. How this unusual and yet crucial S,S-stereochemistry is achieved is unknown. Here, we report that phospholipases D3 and D4 (PLD3 and PLD4) synthesize lysosomal S,S-BMP, with either enzyme catalyzing the critical glycerol stereo-inversion reaction in vitro. Deletion of PLD3 or PLD4 markedly reduced BMP levels in cells or in murine tissues where either enzyme is highly expressed (brain for PLD3; spleen for PLD4), leading to gangliosidosis and lysosomal abnormalities. PLD3 mutants associated with neurodegenerative diseases, including risk of Alzheimer's disease, diminished PLD3 catalytic activity. We conclude that PLD3/4 enzymes synthesize lysosomal S,S-BMP, a crucial lipid for maintaining brain health.
Collapse
Affiliation(s)
- Shubham Singh
- Cell Biology Program, Sloan Kettering Institute, MSKCC, New York, NY, USA
| | - Ulrich E Dransfeld
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yohannes A Ambaw
- Cell Biology Program, Sloan Kettering Institute, MSKCC, New York, NY, USA
| | - Joshua Lopez-Scarim
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Robert V Farese
- Cell Biology Program, Sloan Kettering Institute, MSKCC, New York, NY, USA.
| | - Tobias C Walther
- Cell Biology Program, Sloan Kettering Institute, MSKCC, New York, NY, USA; Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
6
|
Bulfon D, Breithofer J, Grabner GF, Fawzy N, Pirchheim A, Wolinski H, Kolb D, Hartig L, Tischitz M, Zitta C, Bramerdorfer G, Lass A, Taschler U, Kratky D, Greimel P, Zimmermann R. Functionally overlapping intra- and extralysosomal pathways promote bis(monoacylglycero)phosphate synthesis in mammalian cells. Nat Commun 2024; 15:9937. [PMID: 39548099 PMCID: PMC11568333 DOI: 10.1038/s41467-024-54213-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
Bis(monoacylglycero)phosphate (BMP) is a major phospholipid constituent of intralumenal membranes in late endosomes/lysosomes, where it regulates the degradation and sorting of lipid cargo. Recent observations suggest that the Batten disease-associated protein CLN5 functions as lysosomal BMP synthase. Here, we show that transacylation reactions catalyzed by cytosolic and secreted enzymes enhance BMP synthesis independently of CLN5. The transacylases identified in this study are capable of acylating the precursor lipid phosphatidylglycerol (PG), generating acyl-PG, which is subsequently hydrolyzed to BMP. Extracellularly, acyl-PG and BMP are generated by endothelial lipase in cooperation with other serum enzymes of the pancreatic lipase family. The intracellular acylation of PG is catalyzed by several members of the cytosolic phospholipase A2 group IV (PLA2G4) family. Overexpression of secreted or cytosolic transacylases was sufficient to correct BMP deficiency in HEK293 cells lacking CLN5. Collectively, our observations suggest that functionally overlapping pathways promote BMP synthesis in mammalian cells.
Collapse
Grants
- Funding: this work was supported by SFB Lipid hydrolysis (10.55776/F73, D.K., R.Z.), 10.55776/P28533 (R.Z.), 10.55776/P35532 (R.Z.), the doctoral program doc-fund “Molecular Metabolism” 10.55776/DOC50 funded by the Austrian Science Fund FWF, Field of Excellence BioHealth – University of Graz, Graz, Austria, Province of Styria, City of Graz, BioTechMed-Graz, and NAWI Graz, and the Glycolipidologue Program of RIKEN (P.G.). For open access purposes, the authors have applied a CC BY public copyright license to any author accepted manuscript version arising from this submission
Collapse
Affiliation(s)
- Dominik Bulfon
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Gernot F Grabner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Nermeen Fawzy
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- Core Facility Ultrastructure Analysis, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Lennart Hartig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martin Tischitz
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Clara Zitta
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Peter Greimel
- Laboratory for Cell Function Dynamics, Center for Brain Science, RIKEN, Wako, Saitama, Japan
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- Field of Excellence BioHealth, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
7
|
Tavakoli A, Hu S, Ebrahim S, Kachar B. Hemifusomes and Interacting Proteolipid Nanodroplets Mediate Multi-Vesicular Body Formation. RESEARCH SQUARE 2024:rs.3.rs-5200876. [PMID: 39502775 PMCID: PMC11537336 DOI: 10.21203/rs.3.rs-5200876/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
The complex, pleiomorphic membrane structure of the vesicular components within the endolysosomal system has been appreciated through decades of classical electron microscopy. However, due to the heavy fixation and staining required in these approaches, in situ visualization of fragile intermediates between early endosomes, late endosomes and ultimately multivesicular bodies (MVBs), remains elusive, raising the likelihood that other structures may have also been overlooked. Here, using in situ cryo-electron tomography in four mammalian cell lines, we discover heterotypic hemifused vesicles that share an extended hemifusion diaphragm, associated with a 42nm proteolipid nanodroplet (PND). We term this previously undescribed vesicular organelle-complex, "hemifusome". Hemifusomes make up approximately 10% of the organelle pool of the endolysosomal system, but do not participate directly in transferrin-mediated endocytosis. Hemifusomes exist in compound conformations and also contain intraluminal vesicles. Based on their range of morphologies, and the consistent presence of the PND at sites of compound hemifused vesicles, we propose that hemifusomes function as platforms for vesicular biogenesis mediated by the PND. These findings offer direct in situ evidence for a long-lived hemifusion diaphragm, and a new, ESCRT-independent model for the formation of late endosomes containing intraluminal vesicles and ultimately MVBs.
Collapse
Affiliation(s)
- Amirrasoul Tavakoli
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlotteville, VA 22903, USA
| | - Shiqiong Hu
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlotteville, VA 22903, USA
| | - Seham Ebrahim
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlotteville, VA 22903, USA
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlotteville, VA 22903, USA
| |
Collapse
|
8
|
Deng S, Liu TA, Ilnytska O, Allada T, Fomina A, Lin N, Petukhova VZ, Pathmasiri KC, Chinthapally K, Blagg BSJ, Ashfeld BL, Cologna SM, Storch J. Molecular determinants of phospholipid treatment to reduce intracellular cholesterol accumulation in NPC1 deficiency. J Biol Chem 2024; 300:107889. [PMID: 39395801 DOI: 10.1016/j.jbc.2024.107889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024] Open
Abstract
Niemann-Pick type C (NPC) disease, caused by mutations in the NPC1 or NPC2 genes, leads to abnormal intracellular cholesterol accumulation in late endosomes/lysosomes. Exogenous enrichment with lysobisphosphatidic acid (LBPA), also known as bis-monoacylglycerol phosphate, either directly or via the LBPA precursor phosphatidylglycerol (PG), has been investigated as a therapeutic intervention to reduce cholesterol accumulation in NPC disease. Here, we report the effects of stereoisomer configuration and acyl chain composition of LBPA on cholesterol clearance in NPC1-deficient cells. We find that S,R, S,S, and S,R LBPA stereoisomers behaved similarly, with all 3 compounds leading to comparable reductions in filipin staining in two NPC1-deficient human fibroblast cell lines. Examination of several LBPA molecular species containing one or two monounsaturated or polyunsaturated acyl chains showed that all LBPA species containing one 18:1 chain significantly reduced cholesterol accumulation, whereas the shorter chain species di-14:0 LBPA had little effect on cholesterol clearance in NPC1-deficient cells. Since cholesterol accumulation in NPC1-deficient cells can also be cleared by PG incubation, we used nonhydrolyzable PG analogs to determine whether conversion to LBPA is required for sterol clearance, or whether PG itself is effective. The results showed that nonhydrolyzable PG species were not appreciably converted to LBPA and showed virtually no cholesterol clearance efficacy in NPC1-deficient cells, supporting the notion that LBPA is the active agent promoting late endosome/lysosome cholesterol clearance. Overall these studies are helping to define the molecular requirements for potential therapeutic use of LBPA as an option for addressing NPC disease.
Collapse
Affiliation(s)
- Shikun Deng
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Ting-Ann Liu
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Olga Ilnytska
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA; Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA
| | - Tamara Allada
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Angelina Fomina
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Nancy Lin
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA; Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA
| | | | | | - Kiran Chinthapally
- Warren Center for Drug Discovery and Development, and Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Brian S J Blagg
- Warren Center for Drug Discovery and Development, and Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Brandon L Ashfeld
- Warren Center for Drug Discovery and Development, and Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Judith Storch
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA; Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA.
| |
Collapse
|
9
|
Komori T, Fukuda M. Two roads diverged in a cell: insights from differential exosome regulation in polarized cells. Front Cell Dev Biol 2024; 12:1451988. [PMID: 39286483 PMCID: PMC11402822 DOI: 10.3389/fcell.2024.1451988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Exosomes are extracellular vesicles involved in intercellular signaling, carrying various cargo from microRNAs to metabolites and proteins. They are released by practically all cells and are highly heterogenous due to their origin and content. Several groups of exosomes are known to be involved in various pathological conditions including autoimmune, neurodegenerative, and infectious diseases as well as cancer, and therefore a substantial understanding of their biogenesis and release is crucial. Polarized cells display an array of specific functions originated from differentiated membrane trafficking systems and could lead to hints in untangling the complex process of exosomes. Indeed, recent advances have successfully revealed specific regulation pathways for releasing different subsets of exosomes from different sides of polarized epithelial cells, underscoring the importance of polarized cells in the field. Here we review current evidence on exosome biogenesis and release, especially in polarized cells, highlight the challenges that need to be combatted, and discuss potential applications related to exosomes of polarized-cell origin.
Collapse
Affiliation(s)
- Tadayuki Komori
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
10
|
Dresselhaus EC, Harris KP, Blanchette CR, Koles K, Del Signore SJ, Pescosolido MF, Ermanoska B, Rozencwaig M, Soslowsky RC, Parisi MJ, Stewart BA, Mosca TJ, Rodal AA. ESCRT disruption provides evidence against trans-synaptic signaling via extracellular vesicles. J Cell Biol 2024; 223:e202405025. [PMID: 38842573 PMCID: PMC11157088 DOI: 10.1083/jcb.202405025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/20/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Extracellular vesicles (EVs) are released by many cell types, including neurons, carrying cargoes involved in signaling and disease. It is unclear whether EVs promote intercellular signaling or serve primarily to dispose of unwanted materials. We show that loss of multivesicular endosome-generating endosomal sorting complex required for transport (ESCRT) machinery disrupts release of EV cargoes from Drosophila motor neurons. Surprisingly, ESCRT depletion does not affect the signaling activities of the EV cargo Synaptotagmin-4 (Syt4) and disrupts only some signaling activities of the EV cargo evenness interrupted (Evi). Thus, these cargoes may not require intercellular transfer via EVs, and instead may be conventionally secreted or function cell-autonomously in the neuron. We find that EVs are phagocytosed by glia and muscles, and that ESCRT disruption causes compensatory autophagy in presynaptic neurons, suggesting that EVs are one of several redundant mechanisms to remove cargoes from synapses. Our results suggest that synaptic EV release serves primarily as a proteostatic mechanism for certain cargoes.
Collapse
Affiliation(s)
| | - Kathryn P. Harris
- Office of the Vice-Principal, Research and Innovation, University of Toronto Mississauga, Mississauga, Canada
| | | | - Kate Koles
- Department of Biology, Brandeis University, Waltham, MA, USA
| | | | | | | | - Mark Rozencwaig
- Department of Biology, Brandeis University, Waltham, MA, USA
| | | | - Michael J. Parisi
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bryan A. Stewart
- Department of Biology, University of Toronto Mississauga, Mississauga, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Timothy J. Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Avital A. Rodal
- Department of Biology, Brandeis University, Waltham, MA, USA
| |
Collapse
|
11
|
Filippini F, Galli T. Unveiling defects of secretion mechanisms in Parkinson's disease. J Biol Chem 2024; 300:107603. [PMID: 39059489 PMCID: PMC11378209 DOI: 10.1016/j.jbc.2024.107603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegenerative diseases are characterized by progressive dysfunction and loss of specific sets of neurons. While extensive research has focused on elucidating the genetic and epigenetic factors and molecular mechanisms underlying these disorders, emerging evidence highlights the critical role of secretion in the pathogenesis, possibly even onset, and progression of neurodegenerative diseases, suggesting the occurrence of non-cell-autonomous mechanisms. Secretion is a fundamental process that regulates intercellular communication, supports cellular homeostasis, and orchestrates various physiological functions in the body. Defective secretion can impair the release of neurotransmitters and other signaling molecules, disrupting synaptic transmission and compromising neuronal survival. It can also contribute to the accumulation, misfolding, and aggregation of disease-associated proteins, leading to neurotoxicity and neuronal dysfunction. In this review, we discuss the implications of defective secretion in the context of Parkinson's disease, emphasizing its role in protein aggregation, synaptic dysfunction, extracellular vesicle secretion, and neuroinflammation. We propose a multiple-hit model whereby protein accumulation and secretory defects must be combined for the onset and progression of the disease.
Collapse
Affiliation(s)
- Francesca Filippini
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thierry Galli
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, Paris, France; Groupe Hospitalier Universitaire Paris Psychiatrie & Neurosciences, Paris, France.
| |
Collapse
|
12
|
Tavakoli A, Hu S, Ebrahim S, Kachar B. Hemifusomes and Interacting Proteolipid Nanodroplets: Formation of a Novel Cellular Organelle Complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610112. [PMID: 39253452 PMCID: PMC11383319 DOI: 10.1101/2024.08.28.610112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Within cells, vesicle fusion, scission, and the formation of intraluminal vesicles are critical processes that facilitate traffic, degradation, and recycling of cellular components, and maintenance of cellular homeostasis. Despite significant advancements in elucidating the molecular mechanisms that drive these dynamic processes, the direct in situ visualization of membrane remodeling intermediates remains challenging. Here, through the application of cryo-electron tomography in mammalian cells, we have identified a previously undescribed vesicular organelle complex with unique membrane topology: heterotypic hemifused vesicles that share extended hemifusion diaphragms (HDs) with a 42 nm proteolipid nanodroplet (PND) at their rim. We have termed these organelle complexes "hemifusomes". The HDs of hemifusomes exhibit a range of sizes and curvatures, including the formation of lens-shaped compartments encapsulated within the membrane bilayer. The morphological diversity of the lens-shaped vesicle aligns with a step-wise process of their intraluminal budding, ultimately leading to their scission and the generation of intraluminal vesicles. We propose that hemifusomes function as versatile platforms for protein and lipid sorting and as central hubs for the biogenesis of intraluminal vesicles and the formation of multivesicular bodies.
Collapse
|
13
|
Jin H, Liu J, Wang D. Antioxidant Potential of Exosomes in Animal Nutrition. Antioxidants (Basel) 2024; 13:964. [PMID: 39199210 PMCID: PMC11351667 DOI: 10.3390/antiox13080964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
This review delves into the advantages of exosomes as novel antioxidants in animal nutrition and their potential for regulating oxidative stress. Although traditional nutritional approaches promote oxidative stress defense systems in mammalian animals, several issues remain to be solved, such as low bioavailability, targeted tissue efficiency, and high-dose by-effect. As an important candidate offering regulation opportunities concerned with cellular communication, disease prevention, and physiology regulation in multiple biological systems, the potential of exosomes in mediating redox status in biological systems has not been well described. A previously reported relationship between redox system regulation and circulating exosomes suggested exosomes as a fundamental candidate for both a regulator and biomarker for a redox system. Herein, we review the effects of oxidative stress on exosomes in animals and the potential application of exosomes as antioxidants in animal nutrition. Then, we highlight the advantages of exosomes as redox regulators due to their higher bioavailability and physiological heterogeneity-targeted properties, providing a theoretical foundation and feed industry application. Therefore, exosomes have shown great potential as novel antioxidants in the field of animal nutrition. They can overcome the limitations of traditional antioxidants in terms of dosage and side effects, which will provide unprecedented opportunities in nutritional management and disease prevention, and may become a major breakthrough in the field of animal nutrition.
Collapse
Affiliation(s)
| | | | - Diming Wang
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (H.J.); (J.L.)
| |
Collapse
|
14
|
Robinson BP, Bass NR, Bhakt P, Spiliotis ET. Septin-coated microtubules promote maturation of multivesicular bodies by inhibiting their motility. J Cell Biol 2024; 223:e202308049. [PMID: 38668767 PMCID: PMC11046855 DOI: 10.1083/jcb.202308049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 02/06/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024] Open
Abstract
The microtubule cytoskeleton consists of microtubule subsets with distinct compositions of microtubule-associated proteins, which instruct the position and traffic of subcellular organelles. In the endocytic pathway, these microtubule-associated cues are poorly understood. Here, we report that in MDCK cells, endosomes with multivesicular body (MVB) and late endosome (LE) markers localize preferentially to microtubules coated with septin GTPases. Compared with early endosomes, CD63-containing MVBs/LEs are largely immotile on septin-coated microtubules. In vitro reconstitution assays revealed that the motility of isolated GFP-CD63 endosomes is directly inhibited by microtubule-associated septins. Quantification of CD63-positive endosomes containing the early endosome antigen (EEA1), the Rab7 effector and dynein adaptor RILP or Rab27a, showed that intermediary EEA1- and RILP-positive GFP-CD63 preferentially associate with septin-coated microtubules. Septin knockdown enhanced GFP-CD63 motility and decreased the percentage of CD63-positive MVBs/LEs with lysobiphosphatidic acid without impacting the fraction of EEA1-positive CD63. These results suggest that MVB maturation involves immobilization on septin-coated microtubules, which may facilitate multivesiculation and/or organelle-organelle contacts.
Collapse
Affiliation(s)
| | - Naomi R. Bass
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Priyanka Bhakt
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Elias T. Spiliotis
- Department of Biology, Drexel University, Philadelphia, PA, USA
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
15
|
Stocks CJ, Li X, Stow JL. New advances in innate immune endosomal trafficking. Curr Opin Cell Biol 2024; 89:102395. [PMID: 38970837 DOI: 10.1016/j.ceb.2024.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 07/08/2024]
Abstract
The exocytic and endocytic intracellular trafficking pathways in innate immune cells are known for mediating the secretion of key inflammatory mediators or the internalization of growth factors, nutrients, antigens, cell debris, pathogens and even therapeutics, respectively. Inside cells, these pathways are intertwined as an elaborate network that supports the regulation of immune functions. Endosomal membranes host dynamic platforms for molecular complexes that control signaling and inflammatory responses. High content screens, coupled with elegant microscopy across the scale of resolving molecular complexes to tracking live cellular organelles, have been employed to generate the studies highlighted here. With a focus on deactivation of STING, scaffolding by SLC15A4/TASL complexes and macropinosome shrinkage via the chloride channel protein TMEM206, new studies are identifying molecules, molecular interactions and mechanisms for immune regulation throughout endosomal pathways.
Collapse
Affiliation(s)
- Claudia J Stocks
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xichun Li
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jennifer L Stow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
16
|
Wünkhaus D, Tang R, Nyame K, Laqtom NN, Schweizer M, Scotto Rosato A, Krogsæter EK, Wollnik C, Abu-Remaileh M, Grimm C, Hermey G, Kuhn R, Gruber-Schoffnegger D, Markmann S. TRPML1 activation ameliorates lysosomal phenotypes in CLN3 deficient retinal pigment epithelial cells. Sci Rep 2024; 14:17469. [PMID: 39080379 PMCID: PMC11289453 DOI: 10.1038/s41598-024-67479-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
Mutations in the lysosomal membrane protein CLN3 cause Juvenile Neuronal Ceroid Lipofuscinosis (JNCL). Activation of the lysosomal ion channel TRPML1 has previously been shown to be beneficial in several neurodegenerative disease models. Here, we tested whether TRPML1 activation rescues disease-associated phenotypes in CLN3-deficient retinal pigment epithelial (ARPE-19 CLN3-KO) cells. ARPE-19 CLN3-KO cells accumulate LAMP1 positive organelles and show lysosomal storage of mitochondrial ATPase subunit C (SubC), globotriaosylceramide (Gb3), and glycerophosphodiesters (GPDs), whereas lysosomal bis(monoacylglycero)phosphate (BMP/LBPA) lipid levels were significantly decreased. Activation of TRPML1 reduced lysosomal storage of Gb3 and SubC but failed to restore BMP levels in CLN3-KO cells. TRPML1-mediated decrease of storage was TFEB-independent, and we identified TRPML1-mediated enhanced lysosomal exocytosis as a likely mechanism for clearing storage including GPDs. Therefore, ARPE-19 CLN3-KO cells represent a human cell model for CLN3 disease showing many of the described core lysosomal deficits, some of which can be improved using TRPML1 agonists.
Collapse
Affiliation(s)
| | - R Tang
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
- Charles River Laboratory, Chesterford Research Park, Saffron Walden, UK
| | - K Nyame
- Department of Chemical Engineering and of Genetics and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - N N Laqtom
- Department of Chemical Engineering and of Genetics and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Biological and Environmental Science & Engineering Division, King Abdullah University Of Science And Technology, Thuwal, Saudi Arabia
| | - M Schweizer
- Core Facility Morphology and Electronmicroscopy, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A Scotto Rosato
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - E K Krogsæter
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
- Gladstone Institutes, San Francisco, CA, USA
| | | | - M Abu-Remaileh
- Department of Chemical Engineering and of Genetics and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - C Grimm
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research IIP, Munich/Frankfurt, Germany
| | - G Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - R Kuhn
- Evotec SE, Hamburg, Germany
| | | | | |
Collapse
|
17
|
Benchimol M, de Souza W. Endocytosis in anaerobic parasitic protists. Mem Inst Oswaldo Cruz 2024; 119:e240058. [PMID: 39082582 PMCID: PMC11285859 DOI: 10.1590/0074-02760240058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/05/2024] [Indexed: 08/03/2024] Open
Abstract
The incorporation of different molecules by eukaryotic cells occurs through endocytosis, which is critical to the cell's survival and ability to reproduce. Although this process has been studied in greater detail in mammalian and yeast cells, several groups working with pathogenic protists have made relevant contributions. This review analysed the most relevant data on the endocytic process in anaerobic protists (Entamoeba histolytica, Giardia intestinalis, Trichomonas vaginalis, and Tritrichomonas foetus). Many protozoa can exert endocytic activity across their entire surface and do so with great intensity, as with E. histolytica. The available data on the endocytic pathway and the participation of PI-3 kinase, Rab, and Rho molecular complexes is reviewed from a historical perspective.
Collapse
Affiliation(s)
- Marlene Benchimol
- Universidade Federal do Rio de Janeiro, Centro Nacional de Biologia
Estrutural e Bioimagens, Rio de Janeiro, RJ, Brasil
- Universidade da Grande Rio, Duque de Caxias, RJ, Brasil
| | - Wanderley de Souza
- Universidade Federal do Rio de Janeiro, Centro Nacional de Biologia
Estrutural e Bioimagens, Rio de Janeiro, RJ, Brasil
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica
Carlos Chagas Filho, Laboratório de Ultraestrutura Celular Hertha Meyer, Rio de
Janeiro, RJ, Brasil
| |
Collapse
|
18
|
Abe A, Hinkovska-Galcheva V, Bouchev P, Bouley R, Shayman JA. The role of lysosomal phospholipase A2 in the catabolism of bis(monoacylglycerol)phosphate and association with phospholipidosis. J Lipid Res 2024; 65:100574. [PMID: 38857781 PMCID: PMC11277439 DOI: 10.1016/j.jlr.2024.100574] [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: 03/19/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
Bis(monoacylglycerol)phosphate (BMP) is an acidic glycerophospholipid localized to late endosomes and lysosomes. However, the metabolism of BMP is poorly understood. Because many drugs that cause phospholipidosis inhibit lysosomal phospholipase A2 (LPLA2, PLA2G15, LYPLA3) activity, we investigated whether this enzyme has a role in BMPcatabolism. The incubation of recombinant human LPLA2 (hLPLA2) and liposomes containing the naturally occurring BMP (sn-(2-oleoyl-3-hydroxy)-glycerol-1-phospho-sn-1'-(2'-oleoyl-3'-hydroxy)-glycerol (S,S-(2,2',C18:1)-BMP) resulted in the deacylation of this BMP isomer. The deacylation rate was 70 times lower than that of dioleoyl phosphatidylglycerol (DOPG), an isomer and precursor of BMP. The release rates of oleic acid from DOPG and four BMP stereoisomers by LPLA2 differed. The rank order of the rates of hydrolysis were DOPG>S,S-(3,3',C18:1)-BMP>R,S-(3,1',C18:1)-BMP>R,R-(1,1',C18:1)>S,S-(2,2')-BMP. The cationic amphiphilic drug amiodarone (AMD) inhibited the deacylation of DOPG and BMP isomers by hLPLA2 in a concentration-dependent manner. Under these experimental conditions, the IC50s of amiodarone-induced inhibition of the four BMP isomers and DOPG were less than 20 μM and approximately 30 μM, respectively. BMP accumulation was observed in AMD-treated RAW 264.7 cells. The accumulated BMP was significantly reduced by exogenous treatment of cells with active recombinant hLPLA2 but not with diisopropylfluorophosphate-inactivated recombinant hLPLA2. Finally, a series of cationic amphiphilic drugs known to cause phospholipidosis were screened for inhibition of LPLA2 activity as measured by either the transacylation or fatty acid hydrolysis of BMP or phosphatidylcholine as substrates. Fifteen compounds demonstrated significant inhibition with IC50s ranging from 6.8 to 63.3 μM. These results indicate that LPLA2 degrades BMP isomers with different substrate specificities under acidic conditions and may be the key enzyme associated with BMP accumulation in drug-induced phospholipidosis.
Collapse
Affiliation(s)
- Akira Abe
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Philip Bouchev
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Renee Bouley
- Department of Chemistry and Biochemistry, The Ohio State University at Marion, Marion, OH, USA
| | - James A Shayman
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
19
|
Van Es LJC, Possee RD, King LA. Characterisation of extracellular vesicles in baculovirus infection of Spodoptera frugiperda cells. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e163. [PMID: 38947876 PMCID: PMC11212295 DOI: 10.1002/jex2.163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024]
Abstract
Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is an enveloped DNA virus of the Baculoviridae family. This baculovirus is widely exploited for the biological control of insect pest species and as an expression platform to produce recombinant proteins in insect cells. Extracellular vesicles (EVs) are secreted by all cells and are involved in key roles in many biological processes through their cargo consisting of proteins, RNA or DNA. In viral infections, EVs have been found to transfer both viral and cellular cargo that can elicit either a pro- or antiviral response in recipient cells. Here, small EVs (sEVs) released by Spodoptera frugiperda (Sf) insect cells were characterised for the first time. Using S. frugiperda (SfC1B5) cells stably expressing the baculovirus gp64, the viral envelope protein GP64 was shown to be incorporated into sEVs. Sf9 cells were also transfected with a bacmid AcMNPV genome lacking p6.9 (AcΔP6.9) to prevent budded virus production. The protein content of sEVs from both mock- and AcΔP6.9-transfected cells were analysed by mass spectrometry. In addition to GP64, viral proteins Ac-F, ME-53 and viral ubiquitin were identified, as well as many host proteins including TSG101-which may be useful as a protein marker for sEVs.
Collapse
Affiliation(s)
- Lex J. C. Van Es
- Department of Biological and Medical SciencesOxford Brookes UniversityOxfordUK
- Oxford Expression Technologies LtdOxfordUK
| | | | - Linda A. King
- Department of Biological and Medical SciencesOxford Brookes UniversityOxfordUK
| |
Collapse
|
20
|
Kuchitsu Y, Taguchi T. Lysosomal microautophagy: an emerging dimension in mammalian autophagy. Trends Cell Biol 2024; 34:606-616. [PMID: 38104013 DOI: 10.1016/j.tcb.2023.11.005] [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] [Received: 08/23/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023]
Abstract
Autophagy is a self-catabolic process through which cellular components are delivered to lysosomes for degradation. There are three types of autophagy, i.e., macroautophagy, chaperone-mediated autophagy (CMA), and microautophagy. In macroautophagy, a portion of the cytoplasm is wrapped by the autophagosome, which then fuses with lysosomes and delivers the engulfed cytoplasm for degradation. In CMA, the translocation of cytosolic substrates to the lysosomal lumen is directly across the limiting membrane of lysosomes. In microautophagy, lytic organelles, including endosomes or lysosomes, take up a portion of the cytoplasm directly. Although macroautophagy has been investigated extensively, microautophagy has received much less attention. Nonetheless, it has become evident that microautophagy plays a variety of cellular roles from yeast to mammals. Here we review the very recent updates of microautophagy. In particular, we focus on the feature of the degradative substrates and the molecular machinery that mediates microautophagy.
Collapse
Affiliation(s)
- Yoshihiko Kuchitsu
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan.
| | - Tomohiko Taguchi
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
21
|
Mosesso N, Lerner NS, Bläske T, Groh F, Maguire S, Niedermeier ML, Landwehr E, Vogel K, Meergans K, Nagel MK, Drescher M, Stengel F, Hauser K, Isono E. Arabidopsis CaLB1 undergoes phase separation with the ESCRT protein ALIX and modulates autophagosome maturation. Nat Commun 2024; 15:5188. [PMID: 38898014 PMCID: PMC11187125 DOI: 10.1038/s41467-024-49485-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Autophagy is relevant for diverse processes in eukaryotic cells, making its regulation of fundamental importance. The formation and maturation of autophagosomes require a complex choreography of numerous factors. The endosomal sorting complex required for transport (ESCRT) is implicated in the final step of autophagosomal maturation by sealing of the phagophore membrane. ESCRT-III components were shown to mediate membrane scission by forming filaments that interact with cellular membranes. However, the molecular mechanisms underlying the recruitment of ESCRTs to non-endosomal membranes remain largely unknown. Here we focus on the ESCRT-associated protein ALG2-interacting protein X (ALIX) and identify Ca2+-dependent lipid binding protein 1 (CaLB1) as its interactor. Our findings demonstrate that CaLB1 interacts with AUTOPHAGY8 (ATG8) and PI(3)P, a phospholipid found in autophagosomal membranes. Moreover, CaLB1 and ALIX localize with ATG8 on autophagosomes upon salt treatment and assemble together into condensates. The depletion of CaLB1 impacts the maturation of salt-induced autophagosomes and leads to reduced delivery of autophagosomes to the vacuole. Here, we propose a crucial role of CaLB1 in augmenting phase separation of ALIX, facilitating the recruitment of ESCRT-III to the site of phagophore closure thereby ensuring efficient maturation of autophagosomes.
Collapse
Affiliation(s)
- Niccolò Mosesso
- Plant Physiology and Biochemistry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Niharika Savant Lerner
- Plant Physiology and Biochemistry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Tobias Bläske
- Plant Physiology and Biochemistry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Felix Groh
- Plant Physiology and Biochemistry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Shane Maguire
- Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
- Biophysical Chemistry, Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Marie Laura Niedermeier
- Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
- Biochemistry and Mass Spectrometry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Eliane Landwehr
- Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
- Spectroscopy of Complex Systems, Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Karin Vogel
- Plant Physiology and Biochemistry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Konstanze Meergans
- Plant Physiology and Biochemistry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Marie-Kristin Nagel
- Plant Physiology and Biochemistry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Malte Drescher
- Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
- Spectroscopy of Complex Systems, Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Florian Stengel
- Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
- Biochemistry and Mass Spectrometry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Karin Hauser
- Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
- Biophysical Chemistry, Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Erika Isono
- Plant Physiology and Biochemistry, Department of Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany.
- Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany.
- Division of Molecular Cell Biology, National Institute for Basic Biology, Nishigonaka 38, Myodaiji, Okazaki, 444-8585 Aichi, Japan.
| |
Collapse
|
22
|
Bienvenu A, Burette M, Cantet F, Gourdelier M, Swain J, Cazevieille C, Clemente T, Sadi A, Dupont C, Le Fe M, Bonetto N, Bordignon B, Muriaux D, Gilk S, Bonazzi M, Martinez E. The multifunction Coxiella effector Vice stimulates macropinocytosis and interferes with the ESCRT machinery. Proc Natl Acad Sci U S A 2024; 121:e2315481121. [PMID: 38870060 PMCID: PMC11194487 DOI: 10.1073/pnas.2315481121] [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: 09/06/2023] [Accepted: 04/25/2024] [Indexed: 06/15/2024] Open
Abstract
Intracellular bacterial pathogens divert multiple cellular pathways to establish their niche and persist inside their host. Coxiella burnetii, the causative agent of Q fever, secretes bacterial effector proteins via its Type 4 secretion system to generate a Coxiella-containing vacuole (CCV). Manipulation of lipid and protein trafficking by these effectors is essential for bacterial replication and virulence. Here, we have characterized the lipid composition of CCVs and found that the effector Vice interacts with phosphoinositides and membranes enriched in phosphatidylserine and lysobisphosphatidic acid. Remarkably, eukaryotic cells ectopically expressing Vice present compartments that resemble early CCVs in both morphology and composition. We found that the biogenesis of these compartments relies on the double function of Vice. The effector protein initially localizes at the plasma membrane of eukaryotic cells where it triggers the internalization of large vacuoles by macropinocytosis. Then, Vice stabilizes these compartments by perturbing the ESCRT machinery. Collectively, our results reveal that Vice is an essential C. burnetii effector protein capable of hijacking two major cellular pathways to shape the bacterial replicative niche.
Collapse
Affiliation(s)
- Arthur Bienvenu
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Melanie Burette
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Franck Cantet
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Manon Gourdelier
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Jitendriya Swain
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Chantal Cazevieille
- Institut des Neurosciences de Montpellier (INM), Université de Montpellier, INSERM, Montpellier34090, France
| | - Tatiana Clemente
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE68198-5900
| | - Arif Sadi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE68198-5900
| | - Claire Dupont
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Manon Le Fe
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Nicolas Bonetto
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Benoit Bordignon
- Montpellier Rio Imaging (MRI), BioCampus Montpellier, CNRS, INSERM, Université de Montpellier, Montpellier34090, France
| | - Delphine Muriaux
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Stacey Gilk
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE68198-5900
| | - Matteo Bonazzi
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| | - Eric Martinez
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier34090, France
| |
Collapse
|
23
|
Keszler A, Weihrauch D, Lindemer B, Broeckel G, Lohr NL. Vitamin E Attenuates Red-Light-Mediated Vasodilation: The Benefits of a Mild Oxidative Stress. Antioxidants (Basel) 2024; 13:668. [PMID: 38929107 PMCID: PMC11200653 DOI: 10.3390/antiox13060668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Red light (670 nm) energy controls vasodilation via the formation of a transferable endothelium-derived nitric oxide (NO)-precursor-containing substance, its intracellular traffic, and exocytosis. Here we investigated the underlying mechanistic effect of oxidative stress on light-mediated vasodilation by using pressure myography on dissected murine arteries and immunofluorescence on endothelial cells. Treatment with antioxidants Trolox and catalase decreased vessel dilation. In the presence of catalase, a lower number of exosomes were detected in the vessel bath. Light exposure resulted in increased cellular free radical levels. Mitochondrial reactive oxygen species were also more abundant but did not alter cellular ATP production. Red light enhanced the co-localization of late exosome marker CD63 and cellular S-nitrosoprotein to a greater extent than high glucose, suggesting that a mild oxidative stress favors the localization of NO precursor in late exosomes. Exocytosis regulating protein Rab11 was more abundant after irradiation. Our findings conclude that red-light-induced gentle oxidative stress facilitates the dilation of blood vessels, most likely through empowering the traffic of vasodilatory substances. Application of antioxidants disfavors this mechanism.
Collapse
Affiliation(s)
- Agnes Keszler
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (A.K.); (D.W.); (B.L.); (G.B.)
| | - Dorothee Weihrauch
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (A.K.); (D.W.); (B.L.); (G.B.)
- Department of Plastic Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian Lindemer
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (A.K.); (D.W.); (B.L.); (G.B.)
| | - Grant Broeckel
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (A.K.); (D.W.); (B.L.); (G.B.)
| | - Nicole L. Lohr
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (A.K.); (D.W.); (B.L.); (G.B.)
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Clement J. Zablocki VA Medical Center, Milwaukee, WI 53295, USA
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 53233, USA
- Birmigham VA Medical Center, Birmingham, AL 53233, USA
| |
Collapse
|
24
|
Sheikhhossein HH, Iommelli F, Di Pietro N, Curia MC, Piattelli A, Palumbo R, Roviello GN, De Rosa V. Exosome-like Systems: From Therapies to Vaccination for Cancer Treatment and Prevention-Exploring the State of the Art. Vaccines (Basel) 2024; 12:519. [PMID: 38793770 PMCID: PMC11125800 DOI: 10.3390/vaccines12050519] [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: 03/22/2024] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer remains one of the main causes of death in the world due to its increasing incidence and treatment difficulties. Although significant progress has been made in this field, innovative approaches are needed to reduce tumor incidence, progression, and spread. In particular, the development of cancer vaccines is currently ongoing as both a preventive and therapeutic strategy. This concept is not new, but few vaccines have been approved in oncology. Antigen-based vaccination emerges as a promising strategy, leveraging specific tumor antigens to activate the immune system response. However, challenges persist in finding suitable delivery systems and antigen preparation methods. Exosomes (EXs) are highly heterogeneous bilayer vesicles that carry several molecule types in the extracellular space. The peculiarity is that they may be released from different cells and may be able to induce direct or indirect stimulation of the immune system. In particular, EX-based vaccines may cause an anti-tumor immune attack or produce memory cells recognizing cancer antigens and inhibiting disease development. This review delves into EX composition, biogenesis, and immune-modulating properties, exploring their role as a tool for prevention and therapy in solid tumors. Finally, we describe future research directions to optimize vaccine efficacy and realize the full potential of EX-based cancer immunotherapy.
Collapse
Affiliation(s)
- Hamid Heydari Sheikhhossein
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
- Villa Serena Foundation for Research, 65013 Città Sant'Angelo, Italy
| | - Francesca Iommelli
- Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy
| | - Natalia Di Pietro
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - Adriano Piattelli
- School of Dentistry, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
- Facultad de Medicina, UCAM Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| | - Rosanna Palumbo
- Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy
| | - Giovanni N Roviello
- Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy
| | - Viviana De Rosa
- Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy
| |
Collapse
|
25
|
Dresselhaus EC, Harris KP, Blanchette CR, Koles K, Del Signore SJ, Pescosolido MF, Ermanoska B, Rozencwaig M, Soslowsky RC, Parisi MJ, Stewart BA, Mosca TJ, Rodal AA. ESCRT disruption provides evidence against transsynaptic signaling functions for extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.22.537920. [PMID: 38746182 PMCID: PMC11092503 DOI: 10.1101/2023.04.22.537920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Extracellular vesicles (EVs) are released by many cell types including neurons, carrying cargoes involved in signaling and disease. It is unclear whether EVs promote intercellular signaling or serve primarily to dispose of unwanted materials. We show that loss of multivesicular endosome-generating ESCRT (endosomal sorting complex required for transport) machinery disrupts release of EV cargoes from Drosophila motor neurons. Surprisingly, ESCRT depletion does not affect the signaling activities of the EV cargo Synaptotagmin-4 (Syt4) and disrupts only some signaling activities of the EV cargo Evenness Interrupted (Evi). Thus, these cargoes may not require intercellular transfer via EVs, and instead may be conventionally secreted or function cell autonomously in the neuron. We find that EVs are phagocytosed by glia and muscles, and that ESCRT disruption causes compensatory autophagy in presynaptic neurons, suggesting that EVs are one of several redundant mechanisms to remove cargoes from synapses. Our results suggest that synaptic EV release serves primarily as a proteostatic mechanism for certain cargoes.
Collapse
Affiliation(s)
| | - Kathryn P. Harris
- Office of the Vice-Principal, Research and Innovation, University of Toronto, Mississauga, Mississauga, Canada
| | | | - Kate Koles
- Department of Biology, Brandeis University, Waltham, MA
| | | | | | | | | | | | - Michael J. Parisi
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA
| | - Bryan A. Stewart
- Department of Biology, University of Toronto Mississauga, Mississauga, Canada; Department of Cell and Systems Biology University of Toronto, Toronto, Canada
| | - Timothy J. Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA
| | | |
Collapse
|
26
|
Radulescu D, Mihai FD, Trasca MET, Caluianu EI, Calafeteanu CDM, Radulescu PM, Mercut R, Ciupeanu-Calugaru ED, Marinescu GA, Siloşi CA, Nistor CCE, Danoiu S. Oxidative Stress in Military Missions-Impact and Management Strategies: A Narrative Analysis. Life (Basel) 2024; 14:567. [PMID: 38792589 PMCID: PMC11121804 DOI: 10.3390/life14050567] [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: 03/01/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
This narrative review comprehensively examines the impact of oxidative stress on military personnel, highlighting the crucial role of physical exercise and tailored diets, particularly the ketogenic diet, in minimizing this stress. Through a meticulous analysis of the recent literature, the study emphasizes how regular physical exercise not only enhances cardiovascular, cognitive, and musculoskeletal health but is also essential in neutralizing the effects of oxidative stress, thereby improving endurance and performance during long-term missions. Furthermore, the implementation of the ketogenic diet provides an efficient and consistent energy source through ketone bodies, tailored to the specific energy requirements of military activities, and significantly contributes to the reduction in reactive oxygen species production, thus protecting against cellular deterioration under extreme stress. The study also underlines the importance of integrating advanced technologies, such as wearable devices and smart sensors that allow for the precise and real-time monitoring of oxidative stress and physiological responses, thus facilitating the customization of training and nutritional regimes. Observations from this review emphasize significant variability among individuals in responses to oxidative stress, highlighting the need for a personalized approach in formulating intervention strategies. It is crucial to develop and implement well-monitored, personalized supplementation protocols to ensure that each member of the military personnel receives a regimen tailored to their specific needs, thereby maximizing the effectiveness of measures to combat oxidative stress. This analysis makes a valuable contribution to the specialized literature, proposing a detailed framework for addressing oxidative stress in the armed forces and opening new directions for future research with the aim of optimizing clinical practices and improving the health and performance of military personnel under stress and specific challenges of the military field.
Collapse
Affiliation(s)
- Dumitru Radulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Florina-Diana Mihai
- Doctoral School, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | - Major Emil-Tiberius Trasca
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Elena-Irina Caluianu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Captain Dan Marian Calafeteanu
- Department of Ortopedics, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania;
| | - Patricia-Mihaela Radulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Razvan Mercut
- Department of Plastic and Reconstructive Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | | | - Georgiana-Andreea Marinescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Cristian-Adrian Siloşi
- Doctoral School, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | | | - Suzana Danoiu
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| |
Collapse
|
27
|
Varadharajan V, Ramachandiran I, Massey WJ, Jain R, Banerjee R, Horak AJ, McMullen MR, Huang E, Bellar A, Lorkowski SW, Gulshan K, Helsley RN, James I, Pathak V, Dasarathy J, Welch N, Dasarathy S, Streem D, Reizes O, Allende DS, Smith JD, Simcox J, Nagy LE, Brown JM. Membrane-bound O-acyltransferase 7 (MBOAT7) shapes lysosomal lipid homeostasis and function to control alcohol-associated liver injury. eLife 2024; 12:RP92243. [PMID: 38648183 PMCID: PMC11034944 DOI: 10.7554/elife.92243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Recent genome-wide association studies (GWAS) have identified a link between single-nucleotide polymorphisms (SNPs) near the MBOAT7 gene and advanced liver diseases. Specifically, the common MBOAT7 variant (rs641738) associated with reduced MBOAT7 expression is implicated in non-alcoholic fatty liver disease (NAFLD), alcohol-associated liver disease (ALD), and liver fibrosis. However, the precise mechanism underlying MBOAT7-driven liver disease progression remains elusive. Previously, we identified MBOAT7-driven acylation of lysophosphatidylinositol lipids as key mechanism suppressing the progression of NAFLD (Gwag et al., 2019). Here, we show that MBOAT7 loss of function promotes ALD via reorganization of lysosomal lipid homeostasis. Circulating levels of MBOAT7 metabolic products are significantly reduced in heavy drinkers compared to healthy controls. Hepatocyte- (Mboat7-HSKO), but not myeloid-specific (Mboat7-MSKO), deletion of Mboat7 exacerbates ethanol-induced liver injury. Lipidomic profiling reveals a reorganization of the hepatic lipidome in Mboat7-HSKO mice, characterized by increased endosomal/lysosomal lipids. Ethanol-exposed Mboat7-HSKO mice exhibit dysregulated autophagic flux and lysosomal biogenesis, associated with impaired transcription factor EB-mediated lysosomal biogenesis and autophagosome accumulation. This study provides mechanistic insights into how MBOAT7 influences ALD progression through dysregulation of lysosomal biogenesis and autophagic flux, highlighting hepatocyte-specific MBOAT7 loss as a key driver of ethanol-induced liver injury.
Collapse
Affiliation(s)
- Venkateshwari Varadharajan
- Department of Cancer Biology, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Iyappan Ramachandiran
- Department of Cancer Biology, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - William J Massey
- Department of Cancer Biology, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Raghav Jain
- Department of Biochemistry, University of Wisconsin-MadisonMadisonUnited States
| | - Rakhee Banerjee
- Department of Cancer Biology, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Anthony J Horak
- Department of Cancer Biology, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Megan R McMullen
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Emily Huang
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Annette Bellar
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Shuhui W Lorkowski
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
| | - Kailash Gulshan
- Center for Gene Regulation in Health and Disease (GRHD), Cleveland State UniversityClevelandUnited States
| | - Robert N Helsley
- Department of Cancer Biology, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Department of Pharmacology & Nutritional Sciences, Saha Cardiovascular Research Center, University of Kentucky College of MedicineLexingtonUnited States
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-MadisonMadisonUnited States
| | - Vai Pathak
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Jaividhya Dasarathy
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Family Medicine, Metro Health Medical Center, Case Western Reserve UniversityClevelandUnited States
| | - Nicole Welch
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Srinivasan Dasarathy
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - David Streem
- Lutheran Hospital, Cleveland ClinicClevelandUnited States
| | - Ofer Reizes
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Daniela S Allende
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Anatomical Pathology, Cleveland ClinicClevelandUnited States
| | - Jonathan D Smith
- Department of Cancer Biology, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
| | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-MadisonMadisonUnited States
| | - Laura E Nagy
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - J Mark Brown
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Northern Ohio Alcohol Center (NOAC), Lerner Research Institute, Cleveland ClinicClevelandUnited States
| |
Collapse
|
28
|
Read CB, Ali AN, Stephenson DJ, Macknight HP, Maus KD, Cockburn CL, Kim M, Xie X, Carlyon JA, Chalfant CE. Ceramide-1-phosphate is a regulator of Golgi structure and is co-opted by the obligate intracellular bacterial pathogen Anaplasma phagocytophilum. mBio 2024; 15:e0029924. [PMID: 38415594 PMCID: PMC11005342 DOI: 10.1128/mbio.00299-24] [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: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Many intracellular pathogens structurally disrupt the Golgi apparatus as an evolutionarily conserved promicrobial strategy. Yet, the host factors and signaling processes involved are often poorly understood, particularly for Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis. We found that A. phagocytophilum elevated cellular levels of the bioactive sphingolipid, ceramide-1-phosphate (C1P), to promote Golgi fragmentation that enables bacterial proliferation, conversion from its non-infectious to infectious form, and productive infection. A. phagocytophilum poorly infected mice deficient in ceramide kinase, the Golgi-localized enzyme responsible for C1P biosynthesis. C1P regulated Golgi morphology via activation of a PKCα/Cdc42/JNK signaling axis that culminates in phosphorylation of Golgi structural proteins, GRASP55 and GRASP65. siRNA-mediated depletion of Cdc42 blocked A. phagocytophilum from altering Golgi morphology, which impaired anterograde trafficking of trans-Golgi vesicles into and maturation of the pathogen-occupied vacuole. Cells overexpressing phosphorylation-resistant versions of GRASP55 and GRASP65 presented with suppressed C1P- and A. phagocytophilum-induced Golgi fragmentation and poorly supported infection by the bacterium. By studying A. phagocytophilum, we identify C1P as a regulator of Golgi structure and a host factor that is relevant to disease progression associated with Golgi fragmentation.IMPORTANCECeramide-1-phosphate (C1P), a bioactive sphingolipid that regulates diverse processes vital to mammalian physiology, is linked to disease states such as cancer, inflammation, and wound healing. By studying the obligate intracellular bacterium Anaplasma phagocytophilum, we discovered that C1P is a major regulator of Golgi morphology. A. phagocytophilum elevated C1P levels to induce signaling events that promote Golgi fragmentation and increase vesicular traffic into the pathogen-occupied vacuole that the bacterium parasitizes. As several intracellular microbial pathogens destabilize the Golgi to drive their infection cycles and changes in Golgi morphology is also linked to cancer and neurodegenerative disorder progression, this study identifies C1P as a potential broad-spectrum therapeutic target for infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Curtis B. Read
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Anika N. Ali
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Daniel J. Stephenson
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - H. Patrick Macknight
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Kenneth D. Maus
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Chelsea L. Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Minjung Kim
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Xiujie Xie
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Charles E. Chalfant
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Cancer Biology, University of Virginia Cancer Center, Charlottesville, Virginia, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, Virginia, USA
| |
Collapse
|
29
|
Lee YJ, Shin KJ, Chae YC. Regulation of cargo selection in exosome biogenesis and its biomedical applications in cancer. Exp Mol Med 2024; 56:877-889. [PMID: 38580812 PMCID: PMC11059157 DOI: 10.1038/s12276-024-01209-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 04/07/2024] Open
Abstract
Extracellular vesicles (EVs), including exosomes, are increasingly recognized as potent mediators of intercellular communication due to their capacity to transport a diverse array of bioactive molecules. They assume vital roles in a wide range of physiological and pathological processes and hold significant promise as emerging disease biomarkers, therapeutic agents, and carriers for drug delivery. Exosomes encompass specific groups of membrane proteins, lipids, nucleic acids, cytosolic proteins, and other signaling molecules within their interior. These cargo molecules dictate targeting specificity and functional roles upon reaching recipient cells. Despite our growing understanding of the significance of exosomes in diverse biological processes, the molecular mechanisms governing the selective sorting and packaging of cargo within exosomes have not been fully elucidated. In this review, we summarize current insights into the molecular mechanisms that regulate the sorting of various molecules into exosomes, the resulting biological functions, and potential clinical applications, with a particular emphasis on their relevance in cancer and other diseases. A comprehensive understanding of the loading processes and mechanisms involved in exosome cargo sorting is essential for uncovering the physiological and pathological roles of exosomes, identifying therapeutic targets, and advancing the clinical development of exosome-based therapeutics.
Collapse
Affiliation(s)
- Yu Jin Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| | - Kyeong Jin Shin
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Young Chan Chae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
30
|
Singh S, Dransfeld U, Ambaw Y, Lopez-Scarim J, Farese RV, Walther TC. PLD3 and PLD4 synthesize S,S-BMP, a key phospholipid enabling lipid degradation in lysosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586175. [PMID: 38562702 PMCID: PMC10983895 DOI: 10.1101/2024.03.21.586175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Bis(monoacylglycero)phosphate (BMP) is an abundant lysosomal phospholipid required for degradation of lipids, in particular gangliosides. Alterations in BMP levels are associated with neurodegenerative diseases. Unlike typical glycerophospholipids, lysosomal BMP has two chiral glycerol carbons in the S (rather than the R) stereo-conformation, protecting it from lysosomal degradation. How this unusual and yet crucial S,S-stereochemistry is achieved is unknown. Here we report that phospholipases D3 and D4 (PLD3 and PLD4) synthesize lysosomal S,S-BMP, with either enzyme catalyzing the critical glycerol stereo-inversion reaction in vitro. Deletion of PLD3 or PLD4 markedly reduced BMP levels in cells or in murine tissues where either enzyme is highly expressed (brain for PLD3; spleen for PLD4), leading to gangliosidosis and lysosomal abnormalities. PLD3 mutants associated with neurodegenerative diseases, including Alzheimer's disease risk, diminished PLD3 catalytic activity. We conclude that PLD3/4 enzymes synthesize lysosomal S,S-BMP, a crucial lipid for maintaining brain health.
Collapse
Affiliation(s)
- Shubham Singh
- Cell Biology Program, Sloan Kettering Institute, MSKCC, New York, NY, USA
| | - Ulrich Dransfeld
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yohannes Ambaw
- Cell Biology Program, Sloan Kettering Institute, MSKCC, New York, NY, USA
| | - Joshua Lopez-Scarim
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Robert V. Farese
- Cell Biology Program, Sloan Kettering Institute, MSKCC, New York, NY, USA
| | - Tobias C. Walther
- Cell Biology Program, Sloan Kettering Institute, MSKCC, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| |
Collapse
|
31
|
Heins-Marroquin U, Singh RR, Perathoner S, Gavotto F, Merino Ruiz C, Patraskaki M, Gomez-Giro G, Kleine Borgmann F, Meyer M, Carpentier A, Warmoes MO, Jäger C, Mittelbronn M, Schwamborn JC, Cordero-Maldonado ML, Crawford AD, Schymanski EL, Linster CL. CLN3 deficiency leads to neurological and metabolic perturbations during early development. Life Sci Alliance 2024; 7:e202302057. [PMID: 38195117 PMCID: PMC10776888 DOI: 10.26508/lsa.202302057] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Juvenile neuronal ceroid lipofuscinosis (or Batten disease) is an autosomal recessive, rare neurodegenerative disorder that affects mainly children above the age of 5 yr and is most commonly caused by mutations in the highly conserved CLN3 gene. Here, we generated cln3 morphants and stable mutant lines in zebrafish. Although neither morphant nor mutant cln3 larvae showed any obvious developmental or morphological defects, behavioral phenotyping of the mutant larvae revealed hyposensitivity to abrupt light changes and hypersensitivity to pro-convulsive drugs. Importantly, in-depth metabolomics and lipidomics analyses revealed significant accumulation of several glycerophosphodiesters (GPDs) and cholesteryl esters, and a global decrease in bis(monoacylglycero)phosphate species, two of which (GPDs and bis(monoacylglycero)phosphates) were previously proposed as potential biomarkers for CLN3 disease based on independent studies in other organisms. We could also demonstrate GPD accumulation in human-induced pluripotent stem cell-derived cerebral organoids carrying a pathogenic variant for CLN3 Our models revealed that GPDs accumulate at very early stages of life in the absence of functional CLN3 and highlight glycerophosphoinositol and BMP as promising biomarker candidates for pre-symptomatic CLN3 disease.
Collapse
Affiliation(s)
- Ursula Heins-Marroquin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Randolph R Singh
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- https://ror.org/00hj8s172 Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Simon Perathoner
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Floriane Gavotto
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Carla Merino Ruiz
- Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain
- Biosfer Teslab SL, Reus, Spain
| | - Myrto Patraskaki
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Gemma Gomez-Giro
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Felix Kleine Borgmann
- National Center of Pathology (NCP), Laboratoire national de santé (LNS), Dudelange, Luxembourg
- Department of Oncology (DONC), Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Melanie Meyer
- National Center of Pathology (NCP), Laboratoire national de santé (LNS), Dudelange, Luxembourg
| | - Anaïs Carpentier
- National Center of Pathology (NCP), Laboratoire national de santé (LNS), Dudelange, Luxembourg
| | - Marc O Warmoes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Christian Jäger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Michel Mittelbronn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- National Center of Pathology (NCP), Laboratoire national de santé (LNS), Dudelange, Luxembourg
- Department of Oncology (DONC), Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Life Science and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | | | - Alexander D Crawford
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences (NMBU), Ås, Norway
- Institute for Orphan Drug Discovery, Bremerhaven, Germany
| | - Emma L Schymanski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Carole L Linster
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
32
|
Sambre P, Ho JCS, Parikh AN. Intravesicular Solute Delivery and Surface Area Regulation in Giant Unilamellar Vesicles Driven by Cycles of Osmotic Stresses. J Am Chem Soc 2024; 146:3250-3261. [PMID: 38266489 PMCID: PMC10859933 DOI: 10.1021/jacs.3c11679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024]
Abstract
Phospholipid bilayers are dynamic cellular components that undergo constant changes in their topology, facilitating a broad diversity of physiological functions including endo- and exocytosis, cell division, and intracellular trafficking. These shape transformations consume energy, supplied invariably by the activity of proteins. Here, we show that cycles of oppositely directed osmotic stresses─unassisted by any protein activity─can induce well-defined remodeling of giant unilamellar vesicles, minimally recapitulating the phenomenologies of surface area homeostasis and macropinocytosis. We find that a stress cycle consisting of deflationary hypertonic stress followed by an inflationary hypotonic one prompts an elaborate sequence of membrane shape changes ultimately transporting molecular cargo from the outside into the intravesicular milieu. The initial osmotic deflation produces microscopic spherical invaginations. During the subsequent inflation, the first subpopulation contributes area to the swelling membrane, thereby providing a means for surface area regulation and tensional homeostasis. The second subpopulation vesiculates into the lumens of the mother vesicles, producing pinocytic vesicles. Remarkably, the gradients of solute concentrations between the GUV and the daughter pinocytic vesicles create cascades of water current, inducing pulsatory transient poration that enable solute exchange between the buds and the GUV interior. This results in an efficient water-flux-mediated delivery of molecular cargo across the membrane boundary. Our findings suggest a primitive physical mechanism for communication and transport across protocellular compartments driven only by osmotic stresses. They also suggest plausible physical routes for intravesicular, and possibly intracellular, delivery of ions, solutes, and molecular cargo stimulated simply by cycles of osmotic currents of water.
Collapse
Affiliation(s)
- Pallavi
D. Sambre
- Department
of Materials Science and Engineering, University
of California, Davis, One Shields Avenue, Davis, California 95616, United States
| | - James C. S. Ho
- Singapore
Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 59 Nanyang Drive, 636921 Singapore
- Institute
for Digital Molecular Analytics and Science, Nanyang Technological University, 60 Nanyang Drive, 637551Singapore
| | - Atul N. Parikh
- Department
of Materials Science and Engineering, University
of California, Davis, One Shields Avenue, Davis, California 95616, United States
- Singapore
Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 59 Nanyang Drive, 636921 Singapore
- Institute
for Digital Molecular Analytics and Science, Nanyang Technological University, 60 Nanyang Drive, 637551Singapore
- Department
of Biomedical Engineering, University of
California, Davis, One Shields Avenue, Davis, California 95616, United States
| |
Collapse
|
33
|
Almeida FC, Patra K, Giannisis A, Niesnerova A, Nandakumar R, Ellis E, Oliveira TG, Nielsen HM. APOE genotype dictates lipidomic signatures in primary human hepatocytes. J Lipid Res 2024; 65:100498. [PMID: 38216055 PMCID: PMC10875595 DOI: 10.1016/j.jlr.2024.100498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/27/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024] Open
Abstract
Apolipoprotein E (APOE) genetic variants are most notably known for their divergent impact on the risk of developing Alzheimer's disease. While APOE genotype has been consistently shown to modulate lipid metabolism in a variety of cellular contexts, the effect of APOE alleles on the lipidome in hepatocytes is unknown. In this study, we investigated the contribution of APOE alleles to lipidomic profiles of donor-derived primary human hepatocytes from 77 subjects. Lipidomic data obtained by liquid chromatography-mass spectrometry were analyzed across ε2/ε3, ε3/ε3, and ε3/ε4 genotypes to reveal how APOE modulates lipid relative levels over age and between groups. Hepatic APOE concentration, measured by ELISA, was assessed for correlation with lipid abundance in subjects grouped as per APOE genotype and sex. APOE genotype-specific differential lipidomic signatures associated with age for multiple lipid classes but did not differ between sexes. Compared to ε2/ε3, ε3/ε4 hepatocytes had higher abundance of acylcarnitines (AC) and acylphosphatidylglycerol (AcylPG) as a class, as well as higher medium and long-chain ACs, AcylPG, phosphatidylglycerol (PG), bis(monoacylglycerol)phosphate (BMP), monoacylglycerol (MG) and diacylglycerol (DG) species. The ε3/ε4 hepatocytes also exhibited a higher abundance of medium and long-chain ACs compared to the ε3/ε3 hepatocytes. Only in the ε3/ε4 hepatocytes, APOE concentration was lower and showed a negative correlation with BMP levels, specifically in females. APOE genotype dictates a differential lipidome in primary human hepatocytes. The lipids involved suggest mitochondrial dysfunction with accompanying alterations in neutral lipid storage, reflective of a general disturbance of free fatty acid metabolism in human hepatocytes with the ε4 allele.
Collapse
Affiliation(s)
- Francisco C Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Neuroradiology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Kalicharan Patra
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Andreas Giannisis
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Anezka Niesnerova
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Renu Nandakumar
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, USA
| | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology, (CLINTEC), Division of Transplantation surgery, Karolinska Institutet and ME Transplantation, Karolinska University Hospital, Huddinge, Sweden
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Neuroradiology, Hospital de Braga, Braga, Portugal.
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
34
|
Weihrauch D, Keszler A, Broeckel G, Aranda E, Lindemer B, Lohr NL. Red light mediates the exocytosis of vasodilatory vesicles from cultured endothelial cells: a cellular, and ex vivo murine model. Photochem Photobiol Sci 2024; 23:355-364. [PMID: 38277065 PMCID: PMC10917865 DOI: 10.1007/s43630-023-00522-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024]
Abstract
We have previously established that 670 nm energy induces relaxation of blood vessels via an endothelium derived S-nitrosothiol (RSNO) suggested to be embedded in vesicles. Here, we confirm that red light facilitates the exocytosis of this vasodilator from cultured endothelial cells and increases ex vivo blood vessel diameter. Ex vivo pressurized and pre-constricted facial arteries from C57Bl6/J mice relaxed 14.7% of maximum diameter when immersed in the medium removed from red-light exposed Bovine Aortic Endothelial Cells. In parallel experiments, 0.49 nM RSNO equivalent species was measured in the medium over the irradiated cells vs dark control. Electron microscopy of light exposed endothelium revealed significant increases in the size of the Multi Vesicular Body (MVB), a regulator of exosome trafficking, while RSNO accumulated in the MVBs as detected with immunogold labeling electron microscopy (1.8-fold of control). Moreover, red light enhanced the presence of F-actin related stress fibers (necessary for exocytosis), and the endothelial specific marker VE-cadherin levels suggesting an endothelial origin of the extracellular vesicles. Flow cytometry coupled with DAF staining, an indirect sensor of nitric oxide (NO), indicated significant amounts of NO within the extracellular vesicles (1.4-fold increase relative to dark control). Therefore, we further define the mechanism on the 670 nm light mediated traffic of endothelial vasodilatory vesicles and plan to leverage this insight into the delivery of red-light therapies.
Collapse
Affiliation(s)
- Dorothee Weihrauch
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Anesthesiology, Milwaukee, WI, USA
- Department of Plastic Surgery, Milwaukee, WI, USA
| | - Agnes Keszler
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Grant Broeckel
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eva Aranda
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Anesthesiology, Milwaukee, WI, USA
| | - Brian Lindemer
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nicole L Lohr
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.
- Clement J Zablocki VA Medical Center, Milwaukee, WI, USA.
- Cardiovascular Institute, University of Birmingham, Alabama, USA.
| |
Collapse
|
35
|
Varadharajan V, Ramachandiran L, Massey WJ, Jain R, Banerjee R, Horak AJ, McMullen MR, Huang E, Bellar A, Lorkowski SW, Guilshan K, Helsley RN, James I, Pathak V, Dasarathy J, Welch N, Dasarathy S, Streem D, Reizes O, Allende DS, Smith JD, Simcox J, Nagy LE, Brown JM. Membrane Bound O-Acyltransferase 7 (MBOAT7) Shapes Lysosomal Lipid Homeostasis and Function to Control Alcohol-Associated Liver Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.26.559533. [PMID: 37808828 PMCID: PMC10557709 DOI: 10.1101/2023.09.26.559533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Several recent genome-wide association studies (GWAS) have identified single nucleotide polymorphism (SNPs) near the gene encoding membrane-bound O -acyltransferase 7 ( MBOAT7 ) that is associated with advanced liver diseases. In fact, a common MBOAT7 variant (rs641738), which is associated with reduced MBOAT7 expression, confers increased susceptibility to non-alcoholic fatty liver disease (NAFLD), alcohol-associated liver disease (ALD), and liver fibrosis in those chronically infected with hepatitis viruses B and C. The MBOAT7 gene encodes a lysophosphatidylinositol (LPI) acyltransferase enzyme that produces the most abundant form of phosphatidylinositol 38:4 (PI 18:0/20:4). Although these recent genetic studies clearly implicate MBOAT7 function in liver disease progression, the mechanism(s) by which MBOAT7-driven LPI acylation regulates liver disease is currently unknown. Previously we showed that antisense oligonucleotide (ASO)-mediated knockdown of Mboat7 promoted non-alcoholic fatty liver disease (NAFLD) in mice (Helsley et al., 2019). Here, we provide mechanistic insights into how MBOAT7 loss of function promotes alcohol-associated liver disease (ALD). In agreement with GWAS studies, we find that circulating levels of metabolic product of MBOAT7 (PI 38:4) are significantly reduced in heavy drinkers compared to age-matched healthy controls. Hepatocyte specific genetic deletion ( Mboat7 HSKO ), but not myeloid-specific deletion ( Mboat7 MSKO ), of Mboat7 in mice results in enhanced ethanol-induced hepatic steatosis and high concentrations of plasma alanine aminotransferase (ALT). Given MBOAT7 is a lipid metabolic enzyme, we performed comprehensive lipidomic profiling of the liver and identified a striking reorganization of the hepatic lipidome upon ethanol feeding in Mboat7 HSKO mice. Specifically, we observed large increases in the levels of endosomal/lysosomal lipids including bis(monoacylglycero)phosphates (BMP) and phosphatidylglycerols (PGs) in ethanol-exposed Mboat7 HSKO mice. In parallel, ethanol-fed Mboat7 HSKO mice exhibited marked dysregulation of autophagic flux and lysosomal biogenesis when exposed to ethanol. This was associated with impaired transcription factor EB (TFEB)-mediated lysosomal biogenesis and accumulation of autophagosomes. Collectively, this works provides new molecular insights into how genetic variation in MBOAT7 impacts ALD progression in humans and mice. This work is the first to causally link MBOAT7 loss of function in hepatocytes, but not myeloid cells, to ethanol-induced liver injury via dysregulation of lysosomal biogenesis and autophagic flux.
Collapse
|
36
|
Staheli JP, Neal ML, Navare A, Mast FD, Aitchison JD. Predicting host-based, synthetic lethal antiviral targets from omics data. NAR MOLECULAR MEDICINE 2024; 1:ugad001. [PMID: 38994440 PMCID: PMC11233254 DOI: 10.1093/narmme/ugad001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/08/2023] [Accepted: 01/03/2024] [Indexed: 07/13/2024]
Abstract
Traditional antiviral therapies often have limited effectiveness due to toxicity and the emergence of drug resistance. Host-based antivirals are an alternative, but can cause nonspecific effects. Recent evidence shows that virus-infected cells can be selectively eliminated by targeting synthetic lethal (SL) partners of proteins disrupted by viral infection. Thus, we hypothesized that genes depleted in CRISPR knockout (KO) screens of virus-infected cells may be enriched in SL partners of proteins altered by infection. To investigate this, we established a computational pipeline predicting antiviral SL drug targets. First, we identified SARS-CoV-2-induced changes in gene products via a large compendium of omics data. Second, we identified SL partners for each altered gene product. Last, we screened CRISPR KO data for SL partners required for cell viability in infected cells. Despite differences in virus-induced alterations detected by various omics data, they share many predicted SL targets, with significant enrichment in CRISPR KO-depleted datasets. Our comparison of SARS-CoV-2 and influenza infection data revealed potential broad-spectrum, host-based antiviral SL targets. This suggests that CRISPR KO data are replete with common antiviral targets due to their SL relationship with virus-altered states and that such targets can be revealed from analysis of omics datasets and SL predictions.
Collapse
Affiliation(s)
- Jeannette P Staheli
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Maxwell L Neal
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Arti Navare
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Fred D Mast
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| |
Collapse
|
37
|
Barrado-Gil L, García-Dorival I, Galindo I, Alonso C, Cuesta-Geijo MÁ. Insights into the function of ESCRT complex and LBPA in ASFV infection. Front Cell Infect Microbiol 2023; 13:1163569. [PMID: 38125905 PMCID: PMC10731053 DOI: 10.3389/fcimb.2023.1163569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
The African swine fever virus (ASFV) is strongly dependent on an intact endocytic pathway and a certain cellular membrane remodeling for infection, possibly regulated by the endosomal sorting complexes required for transport (ESCRT). The ESCRT machinery is mainly involved in the coordination of membrane dynamics; hence, several viruses exploit this complex and its accessory proteins VPS4 and ALIX for their own benefit. In this work, we found that shRNA-mediated knockdown of VPS4A decreased ASFV replication and viral titers, and this silencing resulted in an enhanced expression of ESCRT-0 component HRS. ASFV infection slightly increased HRS expression but not under VPS4A depletion conditions. Interestingly, VPS4A silencing did not have an impact on ALIX expression, which was significantly overexpressed upon ASFV infection. Further analysis revealed that ALIX silencing impaired ASFV infection at late stages of the viral cycle, including replication and viral production. In addition to ESCRT, the accessory protein ALIX is involved in endosomal membrane dynamics in a lysobisphosphatydic acid (LBPA) and Ca2+-dependent manner, which is relevant for intraluminal vesicle (ILV) biogenesis and endosomal homeostasis. Moreover, LBPA interacts with NPC2 and/or ALIX to regulate cellular cholesterol traffic, and would affect ASFV infection. Thus, we show that LBPA blocking impacted ASFV infection at both early and late infection, suggesting a function for this unconventional phospholipid in the ASFV viral cycle. Here, we found for the first time that silencing of VPS4A and ALIX affects the infection later on, and blocking LBPA function reduces ASFV infectivity at early and later stages of the viral cycle, while ALIX was overexpressed upon infection. These data suggested the relevance of ESCRT-related proteins in ASFV infection.
Collapse
Affiliation(s)
| | | | | | | | - Miguel Ángel Cuesta-Geijo
- Departmento Biotecnología, INIA-CSIC, Centro Nacional Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| |
Collapse
|
38
|
Barreca V, Boussadia Z, Polignano D, Galli L, Tirelli V, Sanchez M, Falchi M, Bertuccini L, Iosi F, Tatti M, Sargiacomo M, Fiani ML. Metabolic labelling of a subpopulation of small extracellular vesicles using a fluorescent palmitic acid analogue. J Extracell Vesicles 2023; 12:e12392. [PMID: 38072803 PMCID: PMC10710952 DOI: 10.1002/jev2.12392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Exosomes are among the most puzzling vehicles of intercellular communication, but several crucial aspects of their biogenesis remain elusive, primarily due to the difficulty in purifying vesicles with similar sizes and densities. Here we report an effective methodology for labelling small extracellular vesicles (sEV) using Bodipy FL C16, a fluorescent palmitic acid analogue. In this study, we present compelling evidence that the fluorescent sEV population derived from Bodipy C16-labelled cells represents a discrete subpopulation of small exosomes following an intracellular pathway. Rapid cellular uptake and metabolism of Bodipy C16 resulted in the incorporation of fluorescent phospholipids into intracellular organelles specifically excluding the plasma membrane and ultimately becoming part of the exosomal membrane. Importantly, our fluorescence labelling method facilitated accurate quantification and characterization of exosomes, overcoming the limitations of nonspecific dye incorporation into heterogeneous vesicle populations. The characterization of Bodipy-labelled exosomes reveals their enrichment in tetraspanin markers, particularly CD63 and CD81, and in minor proportion CD9. Moreover, we employed nanoFACS sorting and electron microscopy to confirm the exosomal nature of Bodipy-labelled vesicles. This innovative metabolic labelling approach, based on the fate of a fatty acid, offers new avenues for investigating exosome biogenesis and functional properties in various physiological and pathological contexts.
Collapse
Affiliation(s)
- Valeria Barreca
- National Center for Global HealthIstituto Superiore di SanitàRomeItaly
| | | | - Deborah Polignano
- National Center for Global HealthIstituto Superiore di SanitàRomeItaly
| | - Lorenzo Galli
- National Center for Global HealthIstituto Superiore di SanitàRomeItaly
| | | | | | - Mario Falchi
- National AIDS CenterIstituto Superiore di SanitàRomeItaly
| | | | | | - Massimo Tatti
- Department of Oncology and Molecular MedicineIstituto Superiore di SanitàRomeItaly
| | | | - Maria Luisa Fiani
- National Center for Global HealthIstituto Superiore di SanitàRomeItaly
| |
Collapse
|
39
|
Komori T, Kuwahara T. An Update on the Interplay between LRRK2, Rab GTPases and Parkinson's Disease. Biomolecules 2023; 13:1645. [PMID: 38002327 PMCID: PMC10669493 DOI: 10.3390/biom13111645] [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: 10/19/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Over the last decades, research on the pathobiology of neurodegenerative diseases has greatly evolved, revealing potential targets and mechanisms linked to their pathogenesis. Parkinson's disease (PD) is no exception, and recent studies point to the involvement of endolysosomal defects in PD. The endolysosomal system, which tightly controls a flow of endocytosed vesicles targeted either for degradation or recycling, is regulated by a number of Rab GTPases. Their associations with leucine-rich repeat kinase 2 (LRRK2), a major causative and risk protein of PD, has also been one of the hot topics in the field. Understanding their interactions and functions is critical for unraveling their contribution to PD pathogenesis. In this review, we summarize recent studies on LRRK2 and Rab GTPases and attempt to provide more insight into the interaction of LRRK2 with each Rab and its relationship to PD.
Collapse
Affiliation(s)
| | - Tomoki Kuwahara
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
40
|
Ma X, Wu F, Peng C, Chen H, Zhang D, Han T. Exploration of mRNA nanoparticles based on DOTAP through optimization of the helper lipids. Biotechnol J 2023; 18:e2300123. [PMID: 37545293 DOI: 10.1002/biot.202300123] [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: 03/14/2023] [Revised: 06/15/2023] [Accepted: 08/02/2023] [Indexed: 08/08/2023]
Abstract
Lipid nanoparticles (LNPs) are one of the most efficient carriers for RNA packaging and delivery, and vaccines based on mRNA-LNPs have received substantial attention since the outbreak of the COVID-19 pandemic. LNPs based on 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) have been widely used in preclinical and clinical settings. A novel non-viral gene delivery system called LNP3 was previously developed, which was composed of DOTAP, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and cholesterol. One of the helper lipids in this carrier was DOPE, which belongs to phospholipids. Given that substituting DOPE with non-phospholipids as helper lipids can increase the delivery efficiency of some LNPs, this study aimed to examine whether non-phospholipids can be formulated with DOTAP as helper lipids. It was found that monoglycerides with C14:0, C16:0, C18:0, C18:1, and C18:2 mediated mRNA transfection, and the transfection efficiency varied between C18:0, C18:1, and C18:2. Furthermore, substituting of the glycerol with other moieties such as the cholesterol or the ethanolamine similarly mediated mRNA transfection. The introduction of cholesterol can further improve the transfection capacity of some DOTAP-based LNPs. One of the best-performing formulations, LNP3-MO, was used to mediate luciferase-mRNA expression in vivo, and the luminescence signal was found to be mainly enriched in the lung and spleen. In addition, the level of SARS-CoV-2 spike antibody in the serum increased after three doses of LNP3-MO mediated SARS-CoV-2 spike mRNA. Altogether, this study demonstrates that non-phospholipids are promising helper lipids that can be formulated with DOTAP to facilitate efficient delivery of mRNAs in vitro and in vivo with organ-specific targeting.
Collapse
Affiliation(s)
- Xueni Ma
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Fanqi Wu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Respiratory, Lanzhou University Second Hospital, Lanzhou, China
| | - Caihong Peng
- Department of Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Huiling Chen
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Tiyun Han
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
41
|
Lisi V, Senesi G, Balbi C. Converging protective pathways: Exploring the linkage between physical exercise, extracellular vesicles and oxidative stress. Free Radic Biol Med 2023; 208:718-727. [PMID: 37739138 DOI: 10.1016/j.freeradbiomed.2023.09.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Physical Exercise (EXR) has been shown to have numerous beneficial effects on various systems in the human body. It leads to a decrease in the risk of mortality from chronic diseases, such as cardiovascular disease, cancer, metabolic and central nervous system disorders. EXR results in improving cardiovascular fitness, cognitive function, immune activity, endocrine action, and musculoskeletal health. These positive effects make EXR a valuable intervention for promoting overall health and well-being in individuals of all ages. These beneficial effects are partially mediated by the role of the regular EXR in the adaptation to redox homeostasis counteracting the sudden increase of ROS, the hallmark of many chronic diseases. EXR can trigger the release of numerous humoral factors, e.g. protein, microRNA (miRs), and DNA, that can be shuttled as cargo of Extracellular vesicles (EVs). EVs show different cargo modification after oxidative stress stimuli as well as after EXR. In this review, we aim to highlight the main studies on the role of EVs released during EXR and oxidative stress conditions in enhancing the antioxidant enzymes pathway and in the decrease of oxidative stress environment mediated by their cargo.
Collapse
Affiliation(s)
- Veronica Lisi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy.
| | - Giorgia Senesi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Carolina Balbi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| |
Collapse
|
42
|
Villalaín J. Phospholipid binding of the dengue virus envelope E protein segment containing the conserved His residue. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184198. [PMID: 37437754 DOI: 10.1016/j.bbamem.2023.184198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/16/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Flaviviruses encompass many important human pathogens, including Dengue, Zika, West Nile, Yellow fever, Japanese encephalitis, and Tick-borne encephalitis viruses as well as several emerging viruses that affect millions of people worldwide. They enter cells by endocytosis, fusing their membrane with the late endosomal one in a pH-dependent manner, so membrane fusion is one of the main targets for obtaining new antiviral inhibitors. The envelope E protein, a class II membrane fusion protein, is responsible for fusion and contains different domains involved in the fusion mechanism, including the fusion peptide. However, other segments, apart from the fusion peptide, have been implicated in the mechanism of membrane fusion, in particular a segment containing a His residue supposed to act as a specific pH sensor. We have used atomistic molecular dynamics to study the binding of the envelope E protein segment containing the conserved His residue in its three different tautomer forms with a complex membrane mimicking the late-endosomal one. We show that this His-containing segment is capable of spontaneous membrane binding, preferentially binds electronegatively charged phospholipids and does not bind cholesterol. Since Flaviviruses have caused epidemics in the past, continue to do so and will undoubtedly continue to do so, this specific segment could characterise a new target that would allow finding effective antiviral molecules against DENV virus in particular and Flaviviruses in general.
Collapse
Affiliation(s)
- José Villalaín
- Institute of Research, Development, and Innovation in Healthcare Biotechnology (IDiBE), Universitas "Miguel Hernández", E-03202 Elche, Alicante, Spain.
| |
Collapse
|
43
|
Abstract
There are at least 21 families of enveloped viruses that infect mammals, and many contain members of high concern for global human health. All enveloped viruses have a dedicated fusion protein or fusion complex that enacts the critical genome-releasing membrane fusion event that is essential before viral replication within the host cell interior can begin. Because all enveloped viruses enter cells by fusion, it behooves us to know how viral fusion proteins function. Viral fusion proteins are also major targets of neutralizing antibodies, and hence they serve as key vaccine immunogens. Here we review current concepts about viral membrane fusion proteins focusing on how they are triggered, structural intermediates between pre- and postfusion forms, and their interplay with the lipid bilayers they engage. We also discuss cellular and therapeutic interventions that thwart virus-cell membrane fusion.
Collapse
Affiliation(s)
- Judith M White
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA;
| | - Amanda E Ward
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Laura Odongo
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Lukas K Tamm
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
44
|
Almeida BR, Barros BCSC, Barros DTL, Orikaza CM, Suzuki E. Paracoccidioides brasiliensis Induces α3 Integrin Lysosomal Degradation in Lung Epithelial Cells. J Fungi (Basel) 2023; 9:912. [PMID: 37755020 PMCID: PMC10532483 DOI: 10.3390/jof9090912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Studies on the pathogen-host interaction are crucial for the understanding of the mechanisms involved in the establishment, maintenance, and spread of infection. In recent years, our research group has observed that the P. brasiliensis species interact with integrin family receptors and increase the expression of α3 integrin in lung epithelial cells within 5 h of infection. Interestingly, α3 integrin levels were reduced by approximately 99% after 24 h of infection with P. brasiliensis compared to non-infected cells. In this work, we show that, during infection with this fungus, α3 integrin is increased in the late endosomes of A549 lung epithelial cells. We also observed that the inhibitor of the lysosomal activity bafilomycin A1 was able to inhibit the decrease in α3 integrin levels. In addition, the silencing of the charged multivesicular body protein 3 (CHMP3) inhibited the reduction in α3 integrin levels induced by P. brasiliensis in A549 cells. Thus, together, these results indicate that this fungus induces the degradation of α3 integrin in A549 lung epithelial cells by hijacking the host cell endolysosomal pathway.
Collapse
Affiliation(s)
| | | | | | | | - Erika Suzuki
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Ed. Antonio C. M. Paiva, São Paulo 04023-062, SP, Brazil; (B.R.A.)
| |
Collapse
|
45
|
Sarmento MJ, Llorente A, Petan T, Khnykin D, Popa I, Nikolac Perkovic M, Konjevod M, Jaganjac M. The expanding organelle lipidomes: current knowledge and challenges. Cell Mol Life Sci 2023; 80:237. [PMID: 37530856 PMCID: PMC10397142 DOI: 10.1007/s00018-023-04889-3] [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: 02/13/2023] [Revised: 06/13/2023] [Accepted: 07/19/2023] [Indexed: 08/03/2023]
Abstract
Lipids in cell membranes and subcellular compartments play essential roles in numerous cellular processes, such as energy production, cell signaling and inflammation. A specific organelle lipidome is characterized by lipid synthesis and metabolism, intracellular trafficking, and lipid homeostasis in the organelle. Over the years, considerable effort has been directed to the identification of the lipid fingerprints of cellular organelles. However, these fingerprints are not fully characterized due to the large variety and structural complexity of lipids and the great variability in the abundance of different lipid species. The process becomes even more challenging when considering that the lipidome differs in health and disease contexts. This review summarizes the information available on the lipid composition of mammalian cell organelles, particularly the lipidome of the nucleus, mitochondrion, endoplasmic reticulum, Golgi apparatus, plasma membrane and organelles in the endocytic pathway. The lipid compositions of extracellular vesicles and lamellar bodies are also described. In addition, several examples of subcellular lipidome dynamics under physiological and pathological conditions are presented. Finally, challenges in mapping organelle lipidomes are discussed.
Collapse
Affiliation(s)
- Maria J Sarmento
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Department for Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, 0167, Oslo, Norway
- Faculty of Medicine, Centre for Cancer Cell Reprogramming, University of Oslo, Montebello, 0379, Oslo, Norway
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Denis Khnykin
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Iuliana Popa
- Pharmacy Department, Bâtiment Henri Moissan, University Paris-Saclay, 17 Avenue des Sciences, 91400, Orsay, France
| | | | - Marcela Konjevod
- Division of Molecular Medicine, Ruder Boskovic Institute, 10000, Zagreb, Croatia
| | - Morana Jaganjac
- Division of Molecular Medicine, Ruder Boskovic Institute, 10000, Zagreb, Croatia.
| |
Collapse
|
46
|
Skotland T, Llorente A, Sandvig K. Lipids in Extracellular Vesicles: What Can Be Learned about Membrane Structure and Function? Cold Spring Harb Perspect Biol 2023; 15:a041415. [PMID: 37277192 PMCID: PMC10411865 DOI: 10.1101/cshperspect.a041415] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Extracellular vesicles, such as exosomes, can be used as interesting models to study the structure and function of biological membranes as these vesicles contain only one membrane (i.e., one lipid bilayer). In addition to lipids, they contain proteins, nucleic acids, and various other molecules. The lipid composition of exosomes is here compared to HIV particles and detergent-resistant membranes, which also have a high content of sphingolipids, cholesterol, and phosphatidylserine (PS). We discuss interactions between the lipids in the two bilayers, and especially those between PS 18:0/18:1 in the inner leaflet and the very-long-chain sphingolipids in the outer leaflet, and the importance of cholesterol for these interactions. We also briefly discuss the involvement of ether-linked phospholipids (PLs) in such lipid raft-like structures, and the possible involvement of these and other lipid classes in the formation of exosomes. The urgent need to improve the quality of quantitative lipidomic studies is highlighted.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
- Department of Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, 0167 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
- Department of Molecular Biosciences, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
47
|
Ribovski L, Joshi B, Gao J, Zuhorn I. Breaking free: endocytosis and endosomal escape of extracellular vesicles. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:283-305. [PMID: 39697985 PMCID: PMC11648447 DOI: 10.20517/evcna.2023.26] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/17/2023] [Accepted: 06/25/2023] [Indexed: 12/20/2024]
Abstract
Extracellular vesicles (EVs) are natural micro-/nanoparticles that play an important role in intercellular communication. They are secreted by producer/donor cells and subsequent uptake by recipient/acceptor cells may result in phenotypic changes in these cells due to the delivery of cargo molecules, including lipids, RNA, and proteins. The process of endocytosis is widely described as the main mechanism responsible for cellular uptake of EVs, with endosomal escape of cargo molecules being a necessity for the functional delivery of EV cargo. Equivalent to synthetic micro-/nanoparticles, the properties of EVs, such as size and composition, together with environmental factors such as temperature, pH, and extracellular fluid composition, codetermine the interactions of EVs with cells, from binding to uptake, intracellular trafficking, and cargo release. Innovative assays for detection and quantification of the different steps in the EV formation and EV-mediated cargo delivery process have provided valuable insight into the biogenesis and cellular processing of EVs and their cargo, revealing the occurrence of EV recycling and degradation, next to functional cargo delivery, with the back fusion of the EV with the endosomal membrane standing out as a common cargo release pathway. In view of the significant potential for developing EVs as drug delivery systems, this review discusses the interaction of EVs with biological membranes en route to cargo delivery, highlighting the reported techniques for studying EV internalization and intracellular trafficking, EV-membrane fusion, endosomal permeabilization, and cargo delivery, including functional delivery of RNA cargo.
Collapse
Affiliation(s)
- Laís Ribovski
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen 9713 AV, the Netherlands
- Authors contributed equally
| | - Bhagyashree Joshi
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft 2629 HZ, the Netherlands
- Authors contributed equally
| | - Jie Gao
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen 9713 AV, the Netherlands
| | - Inge Zuhorn
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen 9713 AV, the Netherlands
| |
Collapse
|
48
|
He Y, Cheng M, Yang R, Li H, Lu Z, Jin Y, Feng J, Tu L. Research Progress on the Mechanism of Nanoparticles Crossing the Intestinal Epithelial Cell Membrane. Pharmaceutics 2023; 15:1816. [PMID: 37514003 PMCID: PMC10384977 DOI: 10.3390/pharmaceutics15071816] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Improving the stability of drugs in the gastrointestinal tract and their penetration ability in the mucosal layer by implementing a nanoparticle delivery strategy is currently a research focus in the pharmaceutical field. However, for most drugs, nanoparticles failed in enhancing their oral absorption on a large scale (4 folds or above), which hinders their clinical application. Recently, several researchers have proved that the intestinal epithelial cell membrane crossing behaviors of nanoparticles deeply influenced their oral absorption, and relevant reviews were rare. In this paper, we systematically review the behaviors of nanoparticles in the intestinal epithelial cell membrane and mainly focus on their intracellular mechanism. The three key complex intracellular processes of nanoparticles are described: uptake by intestinal epithelial cells on the apical side, intracellular transport and basal side exocytosis. We believe that this review will help scientists understand the in vivo performance of nanoparticles in the intestinal epithelial cell membrane and assist in the design of novel strategies for further improving the bioavailability of nanoparticles.
Collapse
Affiliation(s)
- Yunjie He
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Meng Cheng
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Ruyue Yang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Haocheng Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Zhiyang Lu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Jianfang Feng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Liangxing Tu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| |
Collapse
|
49
|
Shelke GV, Williamson CD, Jarnik M, Bonifacino JS. Inhibition of endolysosome fusion increases exosome secretion. J Cell Biol 2023; 222:e202209084. [PMID: 37213076 PMCID: PMC10202829 DOI: 10.1083/jcb.202209084] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/04/2023] [Accepted: 03/17/2023] [Indexed: 05/23/2023] Open
Abstract
Exosomes are small vesicles that are secreted from cells to dispose of undegraded materials and mediate intercellular communication. A major source of exosomes is intraluminal vesicles within multivesicular endosomes that undergo exocytic fusion with the plasma membrane. An alternative fate of multivesicular endosomes is fusion with lysosomes, resulting in degradation of the intraluminal vesicles. The factors that determine whether multivesicular endosomes fuse with the plasma membrane or with lysosomes are unknown. In this study, we show that impairment of endolysosomal fusion by disruption of a pathway involving the BLOC-one-related complex (BORC), the small GTPase ARL8, and the tethering factor HOPS increases exosome secretion by preventing the delivery of intraluminal vesicles to lysosomes. These findings demonstrate that endolysosomal fusion is a critical determinant of the amount of exosome secretion and suggest that suppression of the BORC-ARL8-HOPS pathway could be used to boost exosome yields in biotechnology applications.
Collapse
Affiliation(s)
- Ganesh Vilas Shelke
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Chad D. Williamson
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Michal Jarnik
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Juan S. Bonifacino
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
50
|
Rädler J, Gupta D, Zickler A, Andaloussi SE. Exploiting the biogenesis of extracellular vesicles for bioengineering and therapeutic cargo loading. Mol Ther 2023; 31:1231-1250. [PMID: 36805147 PMCID: PMC10188647 DOI: 10.1016/j.ymthe.2023.02.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Extracellular vesicles (EVs) are gaining increasing attention for diagnostic and therapeutic applications in various diseases. These natural nanoparticles benefit from favorable safety profiles and unique biodistribution capabilities, rendering them attractive drug-delivery modalities over synthetic analogs. However, the widespread use of EVs is limited by technological shortcomings and biological knowledge gaps that fail to unravel their heterogeneity. An in-depth understanding of their biogenesis is crucial to unlocking their full therapeutic potential. Here, we explore how knowledge about EV biogenesis can be exploited for EV bioengineering to load therapeutic protein or nucleic acid cargos into or onto EVs. We summarize more than 75 articles and discuss their findings on the formation and composition of exosomes and microvesicles, revealing multiple pathways that may be stimulation and/or cargo dependent. Our analysis further identifies key regulators of natural EV cargo loading and we discuss how this knowledge is integrated to develop engineered EV biotherapeutics.
Collapse
Affiliation(s)
- Julia Rädler
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Dhanu Gupta
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden; Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK
| | - Antje Zickler
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Samir El Andaloussi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden.
| |
Collapse
|