51
|
Berg AL, Rowson-Hodel A, Wheeler MR, Hu M, Free SR, Carraway KL. Engaging the Lysosome and Lysosome-Dependent Cell Death in Cancer. Breast Cancer 2022. [DOI: 10.36255/exon-publications-breast-cancer-lysosome] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
52
|
Sagi A, Mukthavaram R, Recatto R, Hong H, Davis M, Trelles RD, El-Mecharrafie N, Acharya G, Gomez A, Leu A, Tachikawa K, Sacchetti C, Soontornniyomkij B, Rajappan K, Karmali P, Chivakula P. Efficacy increase of lipid nanoparticles in vivo by inclusion of bis(monoacylglycerol)phosphate. Nanomedicine (Lond) 2022; 17:1399-1410. [PMID: 36255044 DOI: 10.2217/nnm-2022-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: To investigate the effect of incorporating bis(monoacylglycerol)phosphate (BMP) lipid into a lipid nanoparticle and the functional transport of mRNA by the formulated nanoparticles in vivo. Materials & methods: The nanoparticles were prepared from ionizable lipid, 1,2-distearoyl-sn-glycerol-3-phosphocholine, cholesterol, 1,2-dimyristoyl-sn-glycerol PEG 2000, BMP and formulated mRNA encoding human erythropoietin. We measured the effect of BMP on physicochemical properties and impact on functional efficacy to transport mRNA to its target cells/tissue as measured by protein expression both in vitro and in vivo. Results: Lipid nanoparticles composed of BMP displayed increased endosomal membrane fusion and improved mRNA delivery to the cytosol. Conclusion: The results establish the foundation for future development of these nanoparticulated entities by designing new BMP derivatives and correlating structures to enhanced pharmacokinetic profiles.
Collapse
Affiliation(s)
- Amit Sagi
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Rajesh Mukthavaram
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - River Recatto
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Hyojung Hong
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Michael Davis
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Ramon Diaz Trelles
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Nadja El-Mecharrafie
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Grishma Acharya
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Abraham Gomez
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Angel Leu
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Kiyoshi Tachikawa
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Cristiano Sacchetti
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | | | - Kumar Rajappan
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Priya Karmali
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Padmanabh Chivakula
- Arcturus Therapeutics, 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| |
Collapse
|
53
|
Chinthapally K, Blagg BSJ, Ashfeld BL. Syntheses of Symmetrical and Unsymmetrical Lysobisphosphatidic Acid Derivatives. J Org Chem 2022; 87:10523-10530. [PMID: 35895907 DOI: 10.1021/acs.joc.2c01176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent years have witnessed significant achievements in the field of organic chemistry, which have led to new drugs and the discovery of new and biologically interesting molecules. Herein, we describe a practical and efficient approach to the synthesis of enantiomerically pure and diverse lysobisphosphatidic acid analogues. The key feature of the synthesis is a one-pot, sequential phosphorylation of a protected sn-2-O-oleoyl glycerol or sn-3-O-oleoyl glycerol with 2-cyanoethyl N,N-diisopropylchlorophosphoramidite, followed by oxidation.
Collapse
Affiliation(s)
- Kiran Chinthapally
- Warren Family Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Brian S J Blagg
- Warren Family Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana 46556, United States.,Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Brandon L Ashfeld
- Warren Family Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana 46556, United States.,Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
54
|
Spadea A, Jackman M, Cui L, Pereira S, Lawrence MJ, Campbell RA, Ashford M. Nucleic Acid-Loaded Lipid Nanoparticle Interactions with Model Endosomal Membranes. ACS APPLIED MATERIALS & INTERFACES 2022; 14:30371-30384. [PMID: 35758331 PMCID: PMC9264317 DOI: 10.1021/acsami.2c06065] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Lipid nanoparticles (LNPs) are important delivery systems for RNA-based therapeutics, yet the mechanism of their interaction with endosomal membranes remains unclear. Here, the interactions of nucleic acid-loaded LNPs that contain an ionizable lipid with models of the early and late endosomal membranes are studied, for the first time, using different reflectometry techniques. Novel insight is provided with respect to the subphase pH, the stage of the endosome, and the nature of the nucleic acid cargo. It is found that the insertion of lipids from the LNPs into the model membrane is greatest at pH 6.5 and 5.5, whereas at higher pH, lipid insertion is suppressed with evidence instead for the binding of intact LNPs, demonstrating the importance of the pH in the fusion of LNPs undergoing the endosomal pathway. Furthermore, and independently of the pH, the effect of the early- versus late-stage endosomal models is minimal, suggesting that the increased fluidity and anionic nature of the late endosome has little effect on the extent of LNP interaction. Last, there is greater nucleic acid delivery from LNPs containing mRNA than Poly(A), indicating that the extent of interaction can be tuned according to the nature of the nucleic acid cargo. Such new information on the relative impact of factors influencing nucleic acid delivery by LNP interactions with endosomal membranes is important in the design and tuning of vehicles with improved nucleic acid delivery capacities.
Collapse
Affiliation(s)
- Alice Spadea
- NorthWest
Centre for Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.
- Division
of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.
| | - Mark Jackman
- Advanced
Drug Delivery, Pharmaceutical Sciences, AstraZeneca R&D, Cambridge CB2 0AA, U.K.
| | - Lili Cui
- Advanced
Drug Delivery, Pharmaceutical Sciences, AstraZeneca R&D, Cambridge CB2 0AA, U.K.
| | - Sara Pereira
- Advanced
Drug Delivery, Pharmaceutical Sciences, AstraZeneca R&D, Cambridge CB2 0AA, U.K.
| | - M. Jayne Lawrence
- NorthWest
Centre for Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.
- Division
of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.
| | - Richard A. Campbell
- Division
of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.
| | - Marianne Ashford
- Advanced
Drug Delivery, Pharmaceutical Sciences, AstraZeneca R&D, Macclesfield SK10 2NA, U.K.
| |
Collapse
|
55
|
Villalaín J. Procyanidin C1 Location, Interaction, and Aggregation in Two Complex Biomembranes. MEMBRANES 2022; 12:membranes12070692. [PMID: 35877895 PMCID: PMC9319219 DOI: 10.3390/membranes12070692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 01/25/2023]
Abstract
Procyanidins are known for their many benefits to human health and show a plethora of biological effects. One of the most important procyanidin is the procyanidin trimer C1 (PC1). Due to its relatively high lipid–water partition coefficient, the properties of PC1 could be attributed to its capability to interact with the biomembrane, to modulate its structure and dynamics, and to interact with lipids and proteins, however, its biological mechanism is not known. We have used all-atom molecular dynamics in order to determine the position of PC1 in complex membranes and the presence of its specific interactions with membrane lipids, having simulated a membrane mimicking the plasma membrane and another mimicking the mitochondrial membrane. PC1 has a tendency to be located at the membrane interphase, with part of the molecule exposed to the water solvent and part of it reaching the first carbons of the hydrocarbon chains. It has no preferred orientation, and it completely excludes the CHOL molecule. Remarkably, PC1 has a tendency to spontaneously aggregate, forming high-order oligomers. These data suggest that its bioactive properties could be attributed to its membranotropic effects, which therefore supports the development of these molecules as therapeutic molecules, which would open new opportunities for future medical advances.
Collapse
Affiliation(s)
- José Villalaín
- Institute of Research, Development, and Innovation in Healthcare Biotechnology (IDiBE), Universidad Miguel Hernández, E-03202 Elche, Spain
| |
Collapse
|
56
|
Yamaji-Hasegawa A, Murate M, Inaba T, Dohmae N, Sato M, Fujimori F, Sako Y, Greimel P, Kobayashi T. A novel sterol-binding protein reveals heterogeneous cholesterol distribution in neurite outgrowth and in late endosomes/lysosomes. Cell Mol Life Sci 2022; 79:324. [PMID: 35644822 PMCID: PMC11072113 DOI: 10.1007/s00018-022-04339-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
Abstract
We identified a mushroom-derived protein, maistero-2 that specifically binds 3-hydroxy sterol including cholesterol (Chol). Maistero-2 bound lipid mixture in Chol-dependent manner with a binding threshold of around 30%. Changing lipid composition did not significantly affect the threshold concentration. EGFP-maistero-2 labeled cell surface and intracellular organelle Chol with higher sensitivity than that of well-established Chol probe, D4 fragment of perfringolysin O. EGFP-maistero-2 revealed increase of cell surface Chol during neurite outgrowth and heterogeneous Chol distribution between CD63-positive and LAMP1-positive late endosomes/lysosomes. The absence of strictly conserved Thr-Leu pair present in Chol-dependent cytolysins suggests a distinct Chol-binding mechanism for maistero-2.
Collapse
Affiliation(s)
| | - Motohide Murate
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- UMR 7021, CNRS, Université de Strasbourg, 67401, Illkirch, France
| | - Takehiko Inaba
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN CSRS, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Masayuki Sato
- Yukiguni Maitake Co, Ltd. Yokawa 89, Minamiuonuma, Niigata, 949-6695, Japan
| | - Fumihiro Fujimori
- Laboratory of Biological Science and Technology, Tokyo Kasei University, 1-18-1 Kaga, Itabashi, Tokyo, 173-8062, Japan
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Peter Greimel
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- UMR 7021, CNRS, Université de Strasbourg, 67401, Illkirch, France.
| |
Collapse
|
57
|
Villalaín J. Envelope E protein of dengue virus and phospholipid binding to the late endosomal membrane. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183889. [PMID: 35167815 DOI: 10.1016/j.bbamem.2022.183889] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/11/2022] [Accepted: 02/07/2022] [Indexed: 02/06/2023]
Abstract
Flaviviruses include many significant human pathogens, comprising dengue, West Nile, Yellow fever, Japanese encephalitis, Zika and tick-borne encephalitis viruses and many others, affecting millions of people in the world. These viruses have produced important epidemics in the past, they continue to do it and they will undoubtedly continue to do so in the future. Flaviviruses enter into the cells via receptor-mediated endocytosis by fusing its membrane with the endosomal membrane in a pH-dependent manner with the help of the envelope E protein, a prototypical class II membrane fusion protein. The envelope E protein has a conserved fusion peptide at its distal end, which is responsible in the first instance of inserting the protein into the host membrane. Since the participation of other segments of the E protein in the fusion process should not be ruled out, we have used atomistic molecular dynamics to study the binding of the distal end of domain II of the envelope E protein from Dengue virus (DENV) with a complex membrane similar to the late-endosome one. Our work shows that not only the fusion peptide participates directly in the fusion, but also two other sequences of the protein, next to the fusion peptide it in the three-dimensional structure, are jointly wrapped in the fusion process. Overall, these three sequences represent a new target that would make it possible to obtain effective antivirals against DENV 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
|
58
|
Record M, Attia M, Carayon K, Pucheu L, Bunay J, Soulès R, Ayadi S, Payré B, Perrin‐Cocon L, Bourgailh F, Lamazière A, Lotteau V, Poirot M, Silvente‐Poirot S, de Medina P. Targeting the liver X receptor with dendrogenin A differentiates tumour cells to secrete immunogenic exosome-enriched vesicles. J Extracell Vesicles 2022; 11:e12211. [PMID: 35411723 PMCID: PMC9001168 DOI: 10.1002/jev2.12211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/07/2022] [Accepted: 03/21/2022] [Indexed: 01/02/2023] Open
Abstract
Tumour cells are characterized by having lost their differentiation state. They constitutively secrete small extracellular vesicles (sEV) called exosomes when they come from late endosomes. Dendrogenin A (DDA) is an endogenous tumour suppressor cholesterol‐derived metabolite. It is a new class of ligand of the nuclear Liver X receptors (LXR) which regulate cholesterol homeostasis and immunity. We hypothesized that DDA, which induces tumour cell differentiation, inhibition of tumour growth and immune cell infiltration into tumours, could functionally modify sEV secreted by tumour cells. Here, we have shown that DDA differentiates tumour cells by acting on the LXRβ. This results in an increased production of sEV (DDA‐sEV) which includes exosomes. The DDA‐sEV secreted from DDA‐treated cells were characterized for their content and activity in comparison to sEV secreted from control cells (C‐sEV). DDA‐sEV were enriched, relatively to C‐sEV, in several proteins and lipids such as differentiation antigens, “eat‐me” signals, lipidated LC3 and the endosomal phospholipid bis(monoacylglycero)phosphate, which stimulates dendritic cell maturation and a Th1 T lymphocyte polarization. Moreover, DDA‐sEV inhibited the growth of tumours implanted into immunocompetent mice compared to control conditions. This study reveals a pharmacological control through a nuclear receptor of exosome‐enriched tumour sEV secretion, composition and immune function. Targeting the LXR may be a novel way to reprogram tumour cells and sEV to stimulate immunity against cancer.
Collapse
Affiliation(s)
- Michel Record
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Mehdi Attia
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Kevin Carayon
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Laly Pucheu
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Julio Bunay
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Régis Soulès
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Silia Ayadi
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Bruno Payré
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Laure Perrin‐Cocon
- Team “ VIRal InfectionMetabolism and ImmunityCIRICentre International de Recherche en InfectiologieUniv LyonInsermU1111Université Claude Bernard Lyon 1CNRSUMR5308ENS de LyonLyonFrance
| | - Florence Bourgailh
- Centre de Microscopie Electronique Appliquée à la BiologieFaculté de Médecine RangueilToulouseFrance
| | - Antonin Lamazière
- Sorbonne UniversitéINSERMCentre de Recherche Saint‐AntoineCRSAAP‐HP.SUHôpital Saint AntoineDépartement de métabobolomique cliniqueParisFrance
| | - Vincent Lotteau
- Team “ VIRal InfectionMetabolism and ImmunityCIRICentre International de Recherche en InfectiologieUniv LyonInsermU1111Université Claude Bernard Lyon 1CNRSUMR5308ENS de LyonLyonFrance
| | - Marc Poirot
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Sandrine Silvente‐Poirot
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| | - Philippe de Medina
- Team “Cholesterol Metabolism and Therapeutic Innovations” Cancer Research Centre of Toulouse (CRCT)UMR 1037 INSERMUMR 5071 CNRSUniversité de Toulouse IIIEquipe labellisée par la Ligue Nationale Contre le CancerFrench network for Nutrition And Cancer Research (NACRe network)France
| |
Collapse
|
59
|
Low-dose metformin targets the lysosomal AMPK pathway through PEN2. Nature 2022; 603:159-165. [PMID: 35197629 PMCID: PMC8891018 DOI: 10.1038/s41586-022-04431-8] [Citation(s) in RCA: 321] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
Metformin, the most prescribed antidiabetic medicine, has shown other benefits such as anti-ageing and anticancer effects1-4. For clinical doses of metformin, AMP-activated protein kinase (AMPK) has a major role in its mechanism of action4,5; however, the direct molecular target of metformin remains unknown. Here we show that clinically relevant concentrations of metformin inhibit the lysosomal proton pump v-ATPase, which is a central node for AMPK activation following glucose starvation6. We synthesize a photoactive metformin probe and identify PEN2, a subunit of γ-secretase7, as a binding partner of metformin with a dissociation constant at micromolar levels. Metformin-bound PEN2 forms a complex with ATP6AP1, a subunit of the v-ATPase8, which leads to the inhibition of v-ATPase and the activation of AMPK without effects on cellular AMP levels. Knockout of PEN2 or re-introduction of a PEN2 mutant that does not bind ATP6AP1 blunts AMPK activation. In vivo, liver-specific knockout of Pen2 abolishes metformin-mediated reduction of hepatic fat content, whereas intestine-specific knockout of Pen2 impairs its glucose-lowering effects. Furthermore, knockdown of pen-2 in Caenorhabditis elegans abrogates metformin-induced extension of lifespan. Together, these findings reveal that metformin binds PEN2 and initiates a signalling route that intersects, through ATP6AP1, the lysosomal glucose-sensing pathway for AMPK activation. This ensures that metformin exerts its therapeutic benefits in patients without substantial adverse effects.
Collapse
|
60
|
Calvo V, Izquierdo M. T Lymphocyte and CAR-T Cell-Derived Extracellular Vesicles and Their Applications in Cancer Therapy. Cells 2022; 11:790. [PMID: 35269412 PMCID: PMC8909086 DOI: 10.3390/cells11050790] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Extracellular vesicles (EV) are a very diverse group of cell-derived vesicles released by almost all kind of living cells. EV are involved in intercellular exchange, both nearby and systemically, since they induce signals and transmit their cargo (proteins, lipids, miRNAs) to other cells, which subsequently trigger a wide variety of biological responses in the target cells. However, cell surface receptor-induced EV release is limited to cells from the immune system, including T lymphocytes. T cell receptor activation of T lymphocytes induces secretion of EV containing T cell receptors for antigen and several bioactive molecules, including proapoptotic proteins. These EV are specific for antigen-bearing cells, which make them ideal candidates for a cell-free, EV-dependent cancer therapy. In this review we examine the generation of EV by T lymphocytes and CAR-T cells and some potential therapeutic approaches of these EV.
Collapse
Affiliation(s)
- Victor Calvo
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| | - Manuel Izquierdo
- Departamento de Metabolismo y Señalización Celular, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, 28029 Madrid, Spain
| |
Collapse
|
61
|
Van de Vyver T, De Smedt SC, Raemdonck K. Modulating intracellular pathways to improve non-viral delivery of RNA therapeutics. Adv Drug Deliv Rev 2022; 181:114041. [PMID: 34763002 DOI: 10.1016/j.addr.2021.114041] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022]
Abstract
RNA therapeutics (e.g. siRNA, oligonucleotides, mRNA, etc.) show great potential for the treatment of a myriad of diseases. However, to reach their site of action in the cytosol or nucleus of target cells, multiple intra- and extracellular barriers have to be surmounted. Several non-viral delivery systems, such as nanoparticles and conjugates, have been successfully developed to meet this requirement. Unfortunately, despite these clear advances, state-of-the-art delivery agents still suffer from relatively low intracellular delivery efficiencies. Notably, our current understanding of the intracellular delivery process is largely oversimplified. Gaining mechanistic insight into how RNA formulations are processed by cells will fuel rational design of the next generation of delivery carriers. In addition, identifying which intracellular pathways contribute to productive RNA delivery could provide opportunities to boost the delivery performance of existing nanoformulations. In this review, we discuss both established as well as emerging techniques that can be used to assess the impact of different intracellular barriers on RNA transfection performance. Next, we highlight how several modulators, including small molecules but also genetic perturbation technologies, can boost RNA delivery by intervening at differing stages of the intracellular delivery process, such as cellular uptake, intracellular trafficking, endosomal escape, autophagy and exocytosis.
Collapse
Affiliation(s)
- Thijs Van de Vyver
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
62
|
Vial T, Marti G, Missé D, Pompon J. Lipid Interactions Between Flaviviruses and Mosquito Vectors. Front Physiol 2021; 12:763195. [PMID: 34899388 PMCID: PMC8660100 DOI: 10.3389/fphys.2021.763195] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/20/2021] [Indexed: 11/18/2022] Open
Abstract
Mosquito-borne flaviviruses, such as dengue (DENV), Zika (ZIKV), yellow fever (YFV), West Nile (WNV), and Japanese encephalitis (JEV) viruses, threaten a large part of the human populations. In absence of therapeutics and effective vaccines against each flaviviruses, targeting viral metabolic requirements in mosquitoes may hold the key to new intervention strategies. Development of metabolomics in the last decade opened a new field of research: mosquito metabolomics. It is now clear that flaviviruses rely on mosquito lipids, especially phospholipids, for their cellular cycle and propagation. Here, we review the biosyntheses of, biochemical properties of and flaviviral interactions with mosquito phospholipids. Phospholipids are structural lipids with a polar headgroup and apolar acyl chains, enabling the formation of lipid bilayer that form plasma- and endomembranes. Phospholipids are mostly synthesized through the de novo pathway and remodeling cycle. Variations in headgroup and acyl chains influence phospholipid physicochemical properties and consequently the membrane behavior. Flaviviruses interact with cellular membranes at every step of their cellular cycle. Recent evidence demonstrates that flaviviruses reconfigure the phospholipidome in mosquitoes by regulating phospholipid syntheses to increase virus multiplication. Identifying the phospholipids involved and understanding how flaviviruses regulate these in mosquitoes is required to design new interventions.
Collapse
Affiliation(s)
- Thomas Vial
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.,UMR 152 PHARMADEV-IRD, Université Paul Sabatier, Toulouse, France
| | - Guillaume Marti
- LRSV (UMR 5546), CNRS, Université de Toulouse, Toulouse, France.,MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Dorothée Missé
- MIVEGEC, Université Montpellier, IRD, CNRS, Montpellier, France
| | - Julien Pompon
- MIVEGEC, Université Montpellier, IRD, CNRS, Montpellier, France
| |
Collapse
|
63
|
Glitscher M, Hildt E. Endosomal Cholesterol in Viral Infections - A Common Denominator? Front Physiol 2021; 12:750544. [PMID: 34858206 PMCID: PMC8632007 DOI: 10.3389/fphys.2021.750544] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cholesterol has gained tremendous attention as an essential lipid in the life cycle of virtually all viruses. These seem to have developed manifold strategies to modulate the cholesterol metabolism to the side of lipid uptake and de novo synthesis. In turn, affecting the cholesterol homeostasis has emerged as novel broad-spectrum antiviral strategy. On the other hand, the innate immune system is similarly regulated by the lipid and stimulated by its derivatives. This certainly requires attention in the design of antiviral strategies aiming to decrease cellular cholesterol, as evidence accumulates that withdrawal of cholesterol hampers innate immunity. Secondly, there are exceptions to the rule of the abovementioned virus-induced metabolic shift toward cholesterol anabolism. It therefore is of interest to dissect underlying regulatory mechanisms, which we aimed for in this minireview. We further collected evidence for intracellular cholesterol concentrations being less important in viral life cycles as compared to the spatial distribution of the lipid. Various routes of cholesterol trafficking were found to be hijacked in viral infections with respect to organelle-endosome contact sites mediating cholesterol shuttling. Thus, re-distribution of cellular cholesterol in the context of viral infections requires more attention in ongoing research. As a final aim, a pan-antiviral treatment could be found just within the transport and re-adjustment of local cholesterol concentrations. Thus, we aimed to emphasize the importance of the regulatory roles the endosomal system fulfils herein and hope to stimulate research in this field.
Collapse
Affiliation(s)
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institute, Langen, Germany
| |
Collapse
|
64
|
Kapustin AN, Davey P, Longmire D, Matthews C, Linnane E, Rustogi N, Stavrou M, Devine PWA, Bond NJ, Hanson L, Sonzini S, Revenko A, MacLeod AR, Ross S, Chiarparin E, Puri S. Antisense oligonucleotide activity in tumour cells is influenced by intracellular LBPA distribution and extracellular vesicle recycling. Commun Biol 2021; 4:1241. [PMID: 34725463 PMCID: PMC8560811 DOI: 10.1038/s42003-021-02772-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/08/2021] [Indexed: 12/18/2022] Open
Abstract
Next generation modified antisense oligonucleotides (ASOs) are commercially approved new therapeutic modalities, yet poor productive uptake and endosomal entrapment in tumour cells limit their broad application. Here we compare intracellular traffic of anti KRAS antisense oligonucleotide (AZD4785) in tumour cell lines PC9 and LK2, with good and poor productive uptake, respectively. We find that the majority of AZD4785 is rapidly delivered to CD63+late endosomes (LE) in both cell lines. Importantly, lysobisphosphatidic acid (LBPA) that triggers ASO LE escape is presented in CD63+LE in PC9 but not in LK2 cells. Moreover, both cell lines recycle AZD4785 in extracellular vesicles (EVs); however, AZD4785 quantification by advanced mass spectrometry and proteomic analysis reveals that LK2 recycles more AZD4785 and RNA-binding proteins. Finally, stimulating LBPA intracellular production or blocking EV recycling enhances AZD4785 activity in LK2 but not in PC9 cells thus offering a possible strategy to enhance ASO potency in tumour cells with poor productive uptake of ASOs. Kapustin et al. investigate the intracellular trafficking of anti-KRAS antisense oligonucleotides. They show that the oligonucleotide AZD4785 is recycled via late endosomes in extracellular vesicles in both cells with poor and good oligo productive uptake, and that inducing lysobisphosphatidic acid in late endosomes or blocking EV recycling enhance AZD4785 activity in cells with poor productive uptake, potentially offering improved treatment strategies.
Collapse
Affiliation(s)
- Alexander N Kapustin
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| | - Paul Davey
- Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - David Longmire
- Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Carl Matthews
- Antibody Discovery & Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Emily Linnane
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Nitin Rustogi
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Maria Stavrou
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Paul W A Devine
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Nicholas J Bond
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Lyndsey Hanson
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Alderley Park, UK
| | - Silvia Sonzini
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | - Sarah Ross
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | | | - Sanyogitta Puri
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
65
|
Stévenin V, Giai Gianetto Q, Duchateau M, Matondo M, Enninga J, Chang YY. Purification of infection-associated macropinosomes by magnetic isolation for proteomic characterization. Nat Protoc 2021; 16:5220-5249. [PMID: 34697468 DOI: 10.1038/s41596-021-00610-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 08/03/2021] [Indexed: 02/08/2023]
Abstract
Macropinocytosis refers to the nonselective uptake of extracellular molecules into many different types of eukaryotic cells within large fluid-filled vesicles named macropinosomes. Macropinosomes are relevant for a wide variety of cellular processes, such as antigen sampling in immune cells, homeostasis in the kidney, cell migration or pathogen uptake. Understanding the molecular composition of the different macropinosomes formed during these processes has helped to differentiate their regulations from other endocytic events. Here, we present a magnetic purification protocol that segregates scarce macropinosomes from other endocytic vesicles at a high purity and in a low-cost and unbiased manner. Our protocol takes advantage of moderate-sized magnetic beads of 100 nm in diameter coupled to mass-spectrometry-based proteomic analysis. Passing the cell lysate through a table-top magnet allows the quick retention of the bead-containing macropinosomes. Unlike other cell-fractionation-based methodologies, our protocol minimizes sample loss and production cost without prerequisite knowledge of the macropinosomes and with minimal laboratory experience. We describe a detailed procedure for the isolation of infection-associated macropinosomes during bacterial invasion and the optimization steps to readily adapt it to various studies. The protocol can be performed in 3 d to provide highly purified and enriched macropinosomes for qualitative proteomic composition analysis.
Collapse
Affiliation(s)
- Virginie Stévenin
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France.
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands.
- Université de Paris, Ecole Doctorale BioSPC, Paris, France.
| | - Quentin Giai Gianetto
- Proteomics Platform, Mass Spectrometry for Biology Unit, Institut Pasteur, USR 2000 CNRS, Paris, France
- Hub Bioinformatics et Biostatistics, Computational Biology Department, USR CNRS, Institut Pasteur, Paris, France
| | - Magalie Duchateau
- Proteomics Platform, Mass Spectrometry for Biology Unit, Institut Pasteur, USR 2000 CNRS, Paris, France
| | - Mariette Matondo
- Proteomics Platform, Mass Spectrometry for Biology Unit, Institut Pasteur, USR 2000 CNRS, Paris, France
| | - Jost Enninga
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France
| | - Yuen-Yan Chang
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France.
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
66
|
Structural basis of human PDZD8-Rab7 interaction for the ER-late endosome tethering. Sci Rep 2021; 11:18859. [PMID: 34552186 PMCID: PMC8458453 DOI: 10.1038/s41598-021-98419-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/31/2021] [Indexed: 02/02/2023] Open
Abstract
The membrane contact sites (MCSs) between the ER and late endosomes (LEs) are essential for the regulation of endosomal protein sorting, dynamics, and motility. PDZD8 is an ER transmembrane protein containing a Synaptotagmin-like Mitochondrial lipid-binding Proteins (SMP) domain. PDZD8 tethers the ER to late endosomes and lysosomes by associating its C-terminal coiled-coil (CC) with the LE Rab7. To identify the structural determinants for the PDZD8–Rab7 interaction, we determined the crystal structure of the human PDZD8 CC domain in complex with the GTP-bound form of Rab7. The PDZD8 CC contains one short helix and the two helices forming an antiparallel coiled-coil. Two Rab7 molecules bind to the opposite sides of the PDZD8 CC in a 2:1 ratio. The switch I/II and interswitch regions of the GTP-loaded Rab7 form the binding interfaces, which correlates with the GTP-dependent interaction of PDZD8 and Rab7. Analysis of the protein interaction by isothermal titration calorimetry confirms that two Rab7 molecules bind the PDZD8 CC in a GTP-dependent manner. The structural model of the PDZD8 CC–Rab7 complex correlates with the recruitment of PDZD8 at the LE–ER interface and its role in lipid transport and regulation.
Collapse
|
67
|
The late endosome-resident lipid bis(monoacylglycero)phosphate is a cofactor for Lassa virus fusion. PLoS Pathog 2021; 17:e1009488. [PMID: 34492091 PMCID: PMC8448326 DOI: 10.1371/journal.ppat.1009488] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 09/17/2021] [Accepted: 08/25/2021] [Indexed: 11/20/2022] Open
Abstract
Arenavirus entry into host cells occurs through a low pH-dependent fusion with late endosomes that is mediated by the viral glycoprotein complex (GPC). The mechanisms of GPC-mediated membrane fusion and of virus targeting to late endosomes are not well understood. To gain insights into arenavirus fusion, we examined cell-cell fusion induced by the Old World Lassa virus (LASV) GPC complex. LASV GPC-mediated cell fusion is more efficient and occurs at higher pH with target cells expressing human LAMP1 compared to cells lacking this cognate receptor. However, human LAMP1 is not absolutely required for cell-cell fusion or LASV entry. We found that GPC-induced fusion progresses through the same lipid intermediates as fusion mediated by other viral glycoproteins–a lipid curvature-sensitive intermediate upstream of hemifusion and a hemifusion intermediate downstream of acid-dependent steps that can be arrested in the cold. Importantly, GPC-mediated fusion and LASV pseudovirus entry are specifically augmented by an anionic lipid, bis(monoacylglycero)phosphate (BMP), which is highly enriched in late endosomes. This lipid also specifically promotes cell fusion mediated by Junin virus GPC, an unrelated New World arenavirus. We show that BMP promotes late steps of LASV fusion downstream of hemifusion–the formation and enlargement of fusion pores. The BMP-dependence of post-hemifusion stages of arenavirus fusion suggests that these viruses evolved to use this lipid as a cofactor to selectively fuse with late endosomes. Pathogenic arenaviruses pose a serious health threat. The viral envelope glycoprotein GPC mediates attachment to host cells and drives virus entry via endocytosis and low pH-dependent fusion within late endosomes. Understanding the host factors and processes that are essential for arenavirus fusion may identify novel therapeutic targets. To delineate the mechanism of arenavirus entry, we examined cell-cell fusion induced by the Old World Lassa virus GPC proteins at low pH. Lassa GPC-mediated fusion was augmented by the human LAMP1 receptor and progressed through lipid curvature-sensitive intermediates, such as hemifusion (merger of contacting leaflets of viral and cell membrane without the formation of a fusion pore). We found that most GPC-mediated fusion events were off-path hemifusion structures and that the transition from hemifusion to full fusion and fusion pore enlargement were specifically promoted by an anionic lipid, bis(monoacylglycero)phosphate, which is highly enriched in late endosomes. This lipid also specifically promotes fusion of unrelated New World Junin arenavirus. Our results imply that arenaviruses evolved to use bis(monoacylglycero)phosphate to enter cells from late endosomes.
Collapse
|
68
|
Breiden B, Sandhoff K. Acid Sphingomyelinase, a Lysosomal and Secretory Phospholipase C, Is Key for Cellular Phospholipid Catabolism. Int J Mol Sci 2021; 22:9001. [PMID: 34445706 PMCID: PMC8396676 DOI: 10.3390/ijms22169001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
Here, we present the main features of human acid sphingomyelinase (ASM), its biosynthesis, processing and intracellular trafficking, its structure, its broad substrate specificity, and the proposed mode of action at the surface of the phospholipid substrate carrying intraendolysosomal luminal vesicles. In addition, we discuss the complex regulation of its phospholipid cleaving activity by membrane lipids and lipid-binding proteins. The majority of the literature implies that ASM hydrolyses solely sphingomyelin to generate ceramide and ignores its ability to degrade further substrates. Indeed, more than twenty different phospholipids are cleaved by ASM in vitro, including some minor but functionally important phospholipids such as the growth factor ceramide-1-phosphate and the unique lysosomal lysolipid bis(monoacylglycero)phosphate. The inherited ASM deficiency, Niemann-Pick disease type A and B, impairs mainly, but not only, cellular sphingomyelin catabolism, causing a progressive sphingomyelin accumulation, which furthermore triggers a secondary accumulation of lipids (cholesterol, glucosylceramide, GM2) by inhibiting their turnover in late endosomes and lysosomes. However, ASM appears to be involved in a variety of major cellular functions with a regulatory significance for an increasing number of metabolic disorders. The biochemical characteristics of ASM, their potential effect on cellular lipid turnover, as well as a potential impact on physiological processes will be discussed.
Collapse
Affiliation(s)
| | - Konrad Sandhoff
- Membrane Biology and Lipid Biochemistry Unit, LIMES Institute, University of Bonn, 53121 Bonn, Germany
| |
Collapse
|
69
|
Donoso‐Quezada J, Ayala‐Mar S, González‐Valdez J. The role of lipids in exosome biology and intercellular communication: Function, analytics and applications. Traffic 2021; 22:204-220. [PMID: 34053166 PMCID: PMC8361711 DOI: 10.1111/tra.12803] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/18/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Exosomes are extracellular vesicles that in recent years have received special attention for their regulatory functions in numerous biological processes. Recent evidence suggests a correlation between the composition of exosomes in body fluids and the progression of some disorders, such as cancer, diabetes and neurodegenerative diseases. In consequence, numerous studies have been performed to evaluate the composition of these vesicles, aiming to develop new biomarkers for diagnosis and to find novel therapeutic targets. On their part, lipids represent one of the most important components of exosomes, with important structural and regulatory functions during exosome biogenesis, release, targeting and cellular uptake. Therefore, exosome lipidomics has emerged as an innovative discipline for the discovery of novel lipid species with biomedical applications. This review summarizes the current knowledge about exosome lipids and their roles in exosome biology and intercellular communication. Furthermore, it presents the state-of-the-art analytical procedures used in exosome lipidomics while emphasizing how this emerging discipline is providing new insights for future applications of exosome lipids in biomedicine.
Collapse
Affiliation(s)
| | - Sergio Ayala‐Mar
- Tecnologico de MonterreySchool of Engineering and ScienceMonterreyNuevo LeónMexico
| | - José González‐Valdez
- Tecnologico de MonterreySchool of Engineering and ScienceMonterreyNuevo LeónMexico
| |
Collapse
|
70
|
Ilnytska O, Lai K, Gorshkov K, Schultz ML, Tran BN, Jeziorek M, Kunkel TJ, Azaria RD, McLoughlin HS, Waghalter M, Xu Y, Schlame M, Altan-Bonnet N, Zheng W, Lieberman AP, Dobrowolski R, Storch J. Enrichment of NPC1-deficient cells with the lipid LBPA stimulates autophagy, improves lysosomal function, and reduces cholesterol storage. J Biol Chem 2021; 297:100813. [PMID: 34023384 PMCID: PMC8294588 DOI: 10.1016/j.jbc.2021.100813] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/29/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
Niemann-Pick C (NPC) is an autosomal recessive disorder characterized by mutations in the NPC1 or NPC2 genes encoding endolysosomal lipid transport proteins, leading to cholesterol accumulation and autophagy dysfunction. We have previously shown that enrichment of NPC1-deficient cells with the anionic lipid lysobisphosphatidic acid (LBPA; also called bis(monoacylglycerol)phosphate) via treatment with its precursor phosphatidylglycerol (PG) results in a dramatic decrease in cholesterol storage. However, the mechanisms underlying this reduction are unknown. In the present study, we showed using biochemical and imaging approaches in both NPC1-deficient cellular models and an NPC1 mouse model that PG incubation/LBPA enrichment significantly improved the compromised autophagic flux associated with NPC1 disease, providing a route for NPC1-independent endolysosomal cholesterol mobilization. PG/LBPA enrichment specifically enhanced the late stages of autophagy, and effects were mediated by activation of the lysosomal enzyme acid sphingomyelinase. PG incubation also led to robust and specific increases in LBPA species with polyunsaturated acyl chains, potentially increasing the propensity for membrane fusion events, which are critical for late-stage autophagy progression. Finally, we demonstrated that PG/LBPA treatment efficiently cleared cholesterol and toxic protein aggregates in Purkinje neurons of the NPC1I1061T mouse model. Collectively, these findings provide a mechanistic basis supporting cellular LBPA as a potential new target for therapeutic intervention in NPC disease.
Collapse
Affiliation(s)
- 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.
| | - Kimberly Lai
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark L Schultz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Bruce Nguyen Tran
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Maciej Jeziorek
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Thaddeus J Kunkel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ruth D Azaria
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hayley S McLoughlin
- Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Miriam Waghalter
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Yang Xu
- Departments of Anesthesiology and Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Michael Schlame
- Departments of Anesthesiology and Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA; Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, 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
|
71
|
Huda MN, Nafiujjaman M, Deaguero IG, Okonkwo J, Hill ML, Kim T, Nurunnabi M. Potential Use of Exosomes as Diagnostic Biomarkers and in Targeted Drug Delivery: Progress in Clinical and Preclinical Applications. ACS Biomater Sci Eng 2021; 7:2106-2149. [PMID: 33988964 PMCID: PMC8147457 DOI: 10.1021/acsbiomaterials.1c00217] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Exosomes are cell-derived vesicles containing heterogeneous active biomolecules such as proteins, lipids, mRNAs, receptors, immune regulatory molecules, and nucleic acids. They typically range in size from 30 to 150 nm in diameter. An exosome's surfaces can be bioengineered with antibodies, fluorescent dye, peptides, and tailored for small molecule and large active biologics. Exosomes have enormous potential as a drug delivery vehicle due to enhanced biocompatibility, excellent payload capability, and reduced immunogenicity compared to alternative polymeric-based carriers. Because of active targeting and specificity, exosomes are capable of delivering their cargo to exosome-recipient cells. Additionally, exosomes can potentially act as early stage disease diagnostic tools as the exosome carries various protein biomarkers associated with a specific disease. In this review, we summarize recent progress on exosome composition, biological characterization, and isolation techniques. Finally, we outline the exosome's clinical applications and preclinical advancement to provide an outlook on the importance of exosomes for use in targeted drug delivery, biomarker study, and vaccine development.
Collapse
Affiliation(s)
- Md Nurul Huda
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX 79968
| | - Md Nafiujjaman
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Isaac G Deaguero
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968
| | - Jude Okonkwo
- John A Paulson School of Engineering, Harvard University, Cambridge, MA 02138
| | - Meghan L. Hill
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Taeho Kim
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Md Nurunnabi
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX 79968
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968
| |
Collapse
|
72
|
Gruenberg J. Life in the lumen: The multivesicular endosome. Traffic 2021; 21:76-93. [PMID: 31854087 PMCID: PMC7004041 DOI: 10.1111/tra.12715] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022]
Abstract
The late endosomes/endo‐lysosomes of vertebrates contain an atypical phospholipid, lysobisphosphatidic acid (LBPA) (also termed bis[monoacylglycero]phosphate [BMP]), which is not detected elsewhere in the cell. LBPA is abundant in the membrane system present in the lumen of this compartment, including intralumenal vesicles (ILVs). In this review, the current knowledge on LBPA and LBPA‐containing membranes will be summarized, and their role in the control of endosomal cholesterol will be outlined. Some speculations will also be made on how this system may be overwhelmed in the cholesterol storage disorder Niemann‐Pick C. Then, the roles of intralumenal membranes in endo‐lysosomal dynamics and functions will be discussed in broader terms. Likewise, the mechanisms that drive the biogenesis of intralumenal membranes, including ESCRTs, will also be discussed, as well as their diverse composition and fate, including degradation in lysosomes and secretion as exosomes. This review will also discuss how intralumenal membranes are hijacked by pathogenic agents during intoxication and infection, and what is the biochemical composition and function of the intra‐endosomal lumenal milieu. Finally, this review will allude to the size limitations imposed on intralumenal vesicle functions and speculate on the possible role of LBPA as calcium chelator in the acidic calcium stores of endo‐lysosomes.
Collapse
Affiliation(s)
- Jean Gruenberg
- Biochemistry Department, University of Geneva, Geneva, Switzerland
| |
Collapse
|
73
|
What Can Mushroom Proteins Teach Us about Lipid Rafts? MEMBRANES 2021; 11:membranes11040264. [PMID: 33917311 PMCID: PMC8067419 DOI: 10.3390/membranes11040264] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 12/25/2022]
Abstract
The lipid raft hypothesis emerged as a need to explain the lateral organization and behavior of lipids in the environment of biological membranes. The idea, that lipids segregate in biological membranes to form liquid-disordered and liquid-ordered states, was faced with a challenge: to show that lipid-ordered domains, enriched in sphingomyelin and cholesterol, actually exist in vivo. A great deal of indirect evidence and the use of lipid-binding probes supported this idea, but there was a lack of tools to demonstrate the existence of such domains in living cells. A whole new toolbox had to be invented to biochemically characterize lipid rafts and to define how they are involved in several cellular functions. A potential solution came from basic biochemical experiments in the late 1970s, showing that some mushroom extracts exert hemolytic activities. These activities were later assigned to aegerolysin-based sphingomyelin/cholesterol-specific cytolytic protein complexes. Recently, six sphingomyelin/cholesterol binding proteins from different mushrooms have been identified and have provided some insight into the nature of sphingomyelin/cholesterol-rich domains in living vertebrate cells. In this review, we dissect the accumulated knowledge and introduce the mushroom lipid raft binding proteins as molecules of choice to study the dynamics and origins of these liquid-ordered domains in mammalian cells.
Collapse
|
74
|
Abstract
Extracellular vesicles (EVs) are increasingly being recognised as players in intercellular communication within the human body. EVs are nano-sized vesicles that are secreted by virtually all cells, primarily arising from either the plasma membrane or the endocytic system. They contain a wide range of proteins and nucleic acids in their lumen, as well as cell surface proteins on their exterior. The proteins and nucleic acids within are the 'cargo' that EVs deliver into the cytosol of recipient cells to elicit a response or phenotypic change. For delivery to occur, the cargo needs to cross two lipid bilayers; one that makes up the vesicle itself, and the other of the recipient cell. Exactly how this process works is a topic that is poorly understood, despite being pivotal for their function. Furthermore, extracellular vesicles have therapeutic potential as drug delivery vehicles. Therefore, understanding their delivery mechanism and harnessing its action for drug delivery is of great importance. This chapter will focus on the proposed mechanisms for cargo delivery and discuss existing evidence for cargo delivery from EVs into the cytosol of recipient cells.
Collapse
|
75
|
Tomishige N, Murate M, Didier P, Richert L, Mély Y, Kobayashi T. The use of pore-forming toxins to image lipids and lipid domains. Methods Enzymol 2021; 649:503-542. [PMID: 33712198 DOI: 10.1016/bs.mie.2021.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Very few proteins are reported to bind specific lipids. Because of the high selectivity and strong binding to specific lipids, lipid-targeting pore forming toxins (PFTs) have been employed to study the distribution of lipids in cell- and model-membranes. Non-toxic and monomeric PFT-derivatives are especially useful to study living cells. In this chapter we highlight sphingomyelin (SM)-binding PFT, lysenin (Lys), its derivatives, and newly identified SM/cholesterol binding protein, nakanori. We describe the preparation of non-toxic mutant of Lys (NT-Lys) and its application in optical and super resolution microscopy. We also discuss the observation of nanometer scale lipid domains labeled with nakanori and maltose-binding protein (MBP)-Lys in electron microscopy.
Collapse
Affiliation(s)
| | | | - Pascal Didier
- UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | | | - Yves Mély
- UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | | |
Collapse
|
76
|
Schmitt MV, Reichel A, Liu X, Fricker G, Lienau P. Extension of the Mechanistic Tissue Distribution Model of Rodgers and Rowland by Systematic Incorporation of Lysosomal Trapping: Impact on Unbound Partition Coefficient and Volume of Distribution Predictions in the Rat. Drug Metab Dispos 2021; 49:53-61. [PMID: 33148688 DOI: 10.1124/dmd.120.000161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/28/2020] [Indexed: 11/22/2022] Open
Abstract
Physiologically based pharmacokinetic modeling has become a standard tool to predict drug distribution in early stages of drug discovery; however, this does not currently encompass lysosomal trapping. For basic lipophilic compounds, lysosomal sequestration is known to potentially influence intracellular as well as tissue distribution. The aim of our research was to reliably predict the lysosomal drug content and ultimately integrate this mechanism into pharmacokinetic prediction models. First, we further validated our previously presented method to predict the lysosomal drug content (Schmitt et al., 2019) for a larger set of compounds (n = 41) showing a very good predictivity. Using the lysosomal marker lipid bis(monoacylglycero)phosphate, we estimated the lysosomal volume fraction for all major tissues in the rat, ranging from 0.03% for adipose up to 5.3% for spleen. The pH-driven lysosomal trapping was then estimated and fully integrated into the mechanistic distribution model published by Rodgers et al. (2005) Predictions of Kpu improved for all lysosome-rich tissues. For instance, Kpu increased for nicotine 4-fold (spleen) and 2-fold (lung and kidney) and for quinidine 1.8-fold (brain), although for most other drugs the effects were much less (≤7%). Overall, the effect was strongest for basic compounds with a lower lipophilicity, such as nicotine, for which the unbound volume of distribution at steady-state prediction changed from 1.34 to 1.58 l/kg. For more lipophilic (basic) compounds or those that already show strong interactions with acidic phospholipids, the additional contribution of lysosomal trapping was less pronounced. Nevertheless, lysosomal trapping will also affect intracellular distribution of such compounds. SIGNIFICANCE STATEMENT: The estimation of the lysosomal content in all body tissues facilitated the incorporation of lysosomal sequestration into a general physiologically based pharmacokinetic model, leading to improved predictions as well as elucidating its influence on tissue and subcellular distribution in the rat.
Collapse
Affiliation(s)
- Maximilian V Schmitt
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Andreas Reichel
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Xiaohui Liu
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Gert Fricker
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Philip Lienau
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| |
Collapse
|
77
|
Vazquez Reyes S, Ray S, Aguilera J, Sun J. Development of an In Vitro Membrane Model to Study the Function of EsxAB Heterodimer and Establish the Role of EsxB in Membrane Permeabilizing Activity of Mycobacterium tuberculosis. Pathogens 2020; 9:pathogens9121015. [PMID: 33276541 PMCID: PMC7761419 DOI: 10.3390/pathogens9121015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 11/17/2022] Open
Abstract
EsxA and EsxB are secreted as a heterodimer and have been shown to play critical roles in phagosome rupture and translocation of Mycobacterium tuberculosis into the cytosol. Recent in vitro studies have suggested that the EsxAB heterodimer is dissociated upon acidification, which might allow EsxA insertion into lipid membranes. While the membrane permeabilizing activity (MPA) of EsxA has been well characterized in liposomes composed of di-oleoyl-phosphatidylcholine (DOPC), the MPA of EsxAB heterodimer has not been detected through in vitro assays due to its negligible activity with DOPC liposomes. In this study, we established a new in vitro membrane assay to test the MPA activity of N-terminal acetylated EsxA (N-EsxA). We established that a dose-dependent increase in anionic charged lipids enhances the MPA of N-EsxA. The MPA of both N-EsxA and EsxAB were significantly increased with this new liposome system and made it possible to characterize the MPA of EsxAB in more physiologically-relevant conditions. We tested, for the first time, the effect of temperature on the MPA of N-EsxA and EsxAB in this new system. Interestingly, the MPA of N-EsxA was lower at 37 °C than at RT, and on the contrary, the MPA of EsxAB was higher at 37 °C than at RT. Surprisingly, after incubation at 37 °C, the MPA of N-EsxA continuously decreased over time, while MPA of EsxAB remained stable, suggesting EsxB plays a key role in stabilizing N-EsxA to preserve its MPA at 37 °C. In summary, this study established a new in vitro model system that characterizes the MPA of EsxAB and the role of EsxB at physiological-relevant conditions.
Collapse
Affiliation(s)
- Salvador Vazquez Reyes
- Department of Biological Sciences, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA; (S.V.R.); (J.A.)
- Border Biomedical Research Center at University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
| | - Supriyo Ray
- Department of Chemistry & Biochemistry, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
- Department of Natural Sciences, Bowie State University, 14000 Jericho Park Rd, Bowie, MD 20715, USA
- Correspondence: (S.R.); (J.S.)
| | - Javier Aguilera
- Department of Biological Sciences, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA; (S.V.R.); (J.A.)
- Border Biomedical Research Center at University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
| | - Jianjun Sun
- Department of Biological Sciences, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA; (S.V.R.); (J.A.)
- Border Biomedical Research Center at University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
- Correspondence: (S.R.); (J.S.)
| |
Collapse
|
78
|
Carrière F, Longhi S, Record M. The endosomal lipid bis(monoacylglycero) phosphate as a potential key player in the mechanism of action of chloroquine against SARS-COV-2 and other enveloped viruses hijacking the endocytic pathway. Biochimie 2020; 179:237-246. [PMID: 32485205 PMCID: PMC7261073 DOI: 10.1016/j.biochi.2020.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
The anti-malarial drug Chloroquine (CQ) and its derivative hydroxychloroquine have shown antiviral activities in vitro against many viruses, including coronaviruses, dengue virus and the biosafety level 4 Nipah and Hendra paramyxoviruses. The in vivo efficacy of CQ in the treatment of COVID-19 is currently a matter of debate. CQ is a lysosomotrophic compound that accumulates in lysosomes, as well as in food vacuoles of Plasmodium falciparum. In the treatment of malaria, CQ impairs the digestion and growth of the parasite by increasing the pH of the food vacuole. Similarly, it is assumed that the antiviral effects of CQ results from the increase of lysosome pH and the inhibition of acidic proteases involved in the maturation of virus fusion protein. CQ has however other effects, among which phospholipidosis, characterized by the accumulation of multivesicular bodies within the cell. The increase in phospholipid species particularly concerns bis(monoacylglycero)phosphate (BMP), a specific lipid of late endosomes involved in vesicular trafficking and pH-dependent vesicle budding. It was shown previously that drugs like progesterone, the cationic amphiphile U18666A and the phospholipase inhibitor methyl arachidonyl fluoro phosphonate (MAFP) induce the accumulation of BMP in THP-1 cells and decrease cell infection by human immunodeficiency virus. HIV viral particles were found to be retained into large endosomal-type vesicles, preventing virus spreading. Since BMP was also reported to favour virus entry through hijacking of the endocytic pathway, we propose here that BMP could play a dual role in viral infection, with its antiviral effects triggered by lysosomotropic drugs like CQ.
Collapse
Affiliation(s)
- Frédéric Carrière
- Aix Marseille Univ, CNRS, BIP, UMR7281, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 9, France.
| | - Sonia Longhi
- Aix Marseille Univ, CNRS, AFMB, UMR7257, 163 Avenue de Luminy, 13288, Marseille cedex 09, France
| | - Michel Record
- UMR INSERM 1037-CRCT (Cancer Research Center of Toulouse), University of Toulouse III Paul Sabatier, Team "Cholesterol Metabolism and Therapeutic Innovations,", 2 Avenue Hubert Curien, 31037, Toulouse cedex 1, France.
| |
Collapse
|
79
|
Luquain-Costaz C, Rabia M, Hullin-Matsuda F, Delton I. Bis(monoacylglycero)phosphate, an important actor in the host endocytic machinery hijacked by SARS-CoV-2 and related viruses. Biochimie 2020; 179:247-256. [PMID: 33159981 PMCID: PMC7642752 DOI: 10.1016/j.biochi.2020.10.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
Viruses, including the novel coronavirus SARS-CoV-2, redirect infected cell metabolism to their own purposes. After binding to its receptor angiotensin-converting enzyme 2 (ACE2) on the cell surface, the SARS-CoV-2 is taken up by receptor-mediated endocytosis ending in the acidic endolysosomal compartment. The virus hijacks the endosomal machinery leading to fusion of viral and endosomal membranes and release of the viral RNA into the cytosol. This mini-review specifically highlights the membrane lipid organization of the endosomal system focusing on the unconventional and late endosome/lysosome-specific phospholipid, bis(monoacylglycero)phosphate (BMP). BMP is enriched in alveolar macrophages of lung, one of the target tissue of SARS-CoV-2. This review details the BMP structure, its unsaturated fatty acid composition and fusogenic properties that are essential for the highly dynamic formation of the intraluminal vesicles inside the endosomes. Interestingly, BMP is necessary for infection and replication of enveloped RNA virus such as SARS-CoV-1 and Dengue virus. We also emphasize the role of BMP in lipid sorting and degradation, especially cholesterol transport in cooperation with Niemann Pick type C proteins (NPC 1 and 2) and with some oxysterol-binding protein (OSBP)-related proteins (ORPs) as well as in sphingolipid degradation. Interestingly, numerous virus infection required NPC1 as well as ORPs along the endocytic pathway. Furthermore, BMP content is increased during pathological endosomal lipid accumulation in various lysosomal storage disorders. This is particularly important knowing the high percentage of patients with metabolic disorders among the SARS-CoV-2 infected patients presenting severe forms of COVID-19.
Collapse
Affiliation(s)
- Céline Luquain-Costaz
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Maxence Rabia
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | | | - Isabelle Delton
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France.
| |
Collapse
|
80
|
Tsubone TM, Martins WK, Franco MSF, Silva MN, Itri R, Baptista MS. Cellular compartments challenged by membrane photo-oxidation. Arch Biochem Biophys 2020; 697:108665. [PMID: 33159891 DOI: 10.1016/j.abb.2020.108665] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/20/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022]
Abstract
The lipid composition impacts directly on the structure and function of the cytoplasmic as well as organelle membranes. Depending on the type of membrane, specific lipids are required to accommodate, intercalate, or pack membrane proteins to the proper functioning of the cells/organelles. Rather than being only a physical barrier that separates the inner from the outer spaces, membranes are responsible for many biochemical events such as cell-to-cell communication, protein-lipid interaction, intracellular signaling, and energy storage. Photochemical reactions occur naturally in many biological membranes and are responsible for diverse processes such as photosynthesis and vision/phototaxis. However, excessive exposure to light in the presence of absorbing molecules produces excited states and other oxidant species that may cause cell aging/death, mutations and innumerable diseases including cancer. At the same time, targeting key compartments of diseased cells with light can be a promising strategy to treat many diseases in a clinical procedure called Photodynamic Therapy. Here we analyze the relationships between membrane alterations induced by photo-oxidation and the biochemical responses in mammalian cells. We specifically address the impact of photosensitization reactions in membranes of different organelles such as mitochondria, lysosome, endoplasmic reticulum, and plasma membrane, and the subsequent responses of eukaryotic cells.
Collapse
Affiliation(s)
| | | | - Marcia S F Franco
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, Brazil
| | | | - Rosangela Itri
- Department of Applied Physics, Institute of Physics, University of São Paulo, SP, Brazil
| | - Mauricio S Baptista
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, Brazil.
| |
Collapse
|
81
|
Showalter MR, Berg AL, Nagourney A, Heil H, Carraway KL, Fiehn O. The Emerging and Diverse Roles of Bis(monoacylglycero) Phosphate Lipids in Cellular Physiology and Disease. Int J Mol Sci 2020; 21:ijms21218067. [PMID: 33137979 PMCID: PMC7663174 DOI: 10.3390/ijms21218067] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
Although understudied relative to many phospholipids, accumulating evidence suggests that bis(monoacylglycero)phosphate (BMP) is an important class of regulatory lipid that plays key roles in lysosomal integrity and function. BMPs are rare in most mammalian tissues, comprising only a few percent of total cellular lipid content, but are elevated in cell types such as macrophages that rely heavily on lysosomal function. BMPs are markedly enriched in endosomal and lysosomal vesicles compared to other organelles and membranous structures, and their unique sn-1:sn-1′ stereoconfiguration may confer stability within the hydrolytic lysosomal environment. BMP-enriched vesicles serve in endosomal-lysosomal trafficking and function as docking structures for the activation of lysosomal hydrolytic enzymes, notably those involved in the catabolic breakdown of sphingolipids. BMP levels are dysregulated in lysosomal storage disorders, phospholipidosis, metabolic diseases, liver and kidney diseases and neurodegenerative disorders. However, whether BMP alteration is a mediator or simply a marker of pathological states is unclear. Likewise, although BMP acyl chain composition may be altered with disease states, the functional significance of specific BMP species remains to be resolved. Newly developed tools for untargeted lipidomic analysis, together with a deeper understanding of enzymes mediating BMP synthesis and degradation, will help shed further light on the functional significance of BMPs in cellular physiology and pathology.
Collapse
Affiliation(s)
- Megan R. Showalter
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (A.N.); (H.H.)
| | - Anastasia L. Berg
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (A.L.B.); (K.L.C.III)
| | - Alexander Nagourney
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (A.N.); (H.H.)
| | - Hailey Heil
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (A.N.); (H.H.)
| | - Kermit L. Carraway
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (A.L.B.); (K.L.C.III)
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (A.N.); (H.H.)
- Correspondence:
| |
Collapse
|
82
|
Wang G, Wang Y, Liu N, Liu M. The role of exosome lipids in central nervous system diseases. Rev Neurosci 2020; 31:743-756. [PMID: 32681787 DOI: 10.1515/revneuro-2020-0013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022]
Abstract
Central nervous system (CNS) diseases are common diseases that threaten human health. The CNS is highly enriched in lipids, which play important roles in maintaining normal physiological functions of the nervous system. Moreover, many CNS diseases are closely associated with abnormal lipid metabolism. Exosomes are a subtype of extracellular vesicles (EVs) secreted from multivesicular bodies (MVBs) . Through novel forms of intercellular communication, exosomes secreted by brain cells can mediate inter-neuronal signaling and play important roles in the pathogenesis of CNS diseases. Lipids are essential components of exosomes, with cholesterol and sphingolipid as representative constituents of its bilayer membrane. In the CNS, lipids are closely related to the formation and function of exosomes. Their dysregulation causes abnormalities in exosomes, which may, in turn, lead to dysfunctions in inter-neuronal communication and promote diseases. Therefore, the role of lipids in the treatment of neurological diseases through exosomes has received increasing attention. The aim of this review is to discuss the relationship between lipids and exosomes and their roles in CNS diseases.
Collapse
Affiliation(s)
- Ge Wang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
- Xiangya School of MedicineCentral South University, Changsha, 410078, Hunan, China
| | - Yong Wang
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Ningyuan Liu
- Xiangya School of MedicineCentral South University, Changsha, 410078, Hunan, China
| | - Mujun Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| |
Collapse
|
83
|
Umbaugh DS, Jaeschke H. Extracellular vesicles: Roles and applications in drug-induced liver injury. Adv Clin Chem 2020; 102:63-125. [PMID: 34044913 DOI: 10.1016/bs.acc.2020.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EV) are defined as nanosized particles, with a lipid bilayer, that are unable to replicate. There has been an exponential increase of research investigating these particles in a wide array of diseases and deleterious states (inflammation, oxidative stress, drug-induced liver injury) in large part due to increasing recognition of the functional capacity of EVs. Cells can package lipids, proteins, miRNAs, DNA, and RNA into EVs and send these discrete packages of molecular information to distant, recipient cells to alter the physiological state of that cell. EVs are innately heterogeneous as a result of the diverse molecular pathways that are used to generate them. However, this innate heterogeneity of EVs is amplified due to the diversity in isolation techniques and lack of standardized nomenclature in the literature making it unclear if one scientist's "exosome" is another scientist's "microvesicle." One goal of this chapter is to provide the contextual understanding of EV origin so one can discern between divergent nomenclature. Further, the chapter will explore the potential protective and harmful roles that EVs play in DILI, and the potential of EVs and their cargo as a biomarker. The use of EVs as a therapeutic as well as a vector for therapeutic delivery will be discussed.
Collapse
Affiliation(s)
- David S Umbaugh
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
84
|
Nagakannan P, Tabeshmehr P, Eftekharpour E. Oxidative damage of lysosomes in regulated cell death systems: Pathophysiology and pharmacologic interventions. Free Radic Biol Med 2020; 157:94-127. [PMID: 32259579 DOI: 10.1016/j.freeradbiomed.2020.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022]
Abstract
Lysosomes are small specialized organelles containing a variety of different hydrolase enzymes that are responsible for degradation of all macromolecules, entering the cells through the endosomal system or originated from the internal sources. This allows for transport and recycling of nutrients and internalization of surface proteins for antigen presentation as well as maintaining cellular homeostasis. Lysosomes are also important storage compartments for metal ions and nutrients. The integrity of lysosomal membrane is central to maintaining their normal function, but like other cellular membranes, lysosomal membrane is subject to damage mediated by reactive oxygen species. This results in spillage of lysosomal enzymes into the cytoplasm, leading to proteolytic damage to cellular systems and organelles. Several forms of lysosomal dependent cell death have been identified in diseases. Examination of these events are important for finding treatment strategies relevant to cancer or neurodegenerative diseases as well as autoimmune deficiencies. In this review, we have examined the current literature on involvement of lysosomes in induction of programed cell death and have provided an extensive list of therapeutic approaches that can modulate cell death. Exploitation of these mechanisms can lead to novel therapies for cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Pandian Nagakannan
- Regenerative Medicine Program and Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Parisa Tabeshmehr
- Regenerative Medicine Program and Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Eftekhar Eftekharpour
- Regenerative Medicine Program and Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
85
|
Extracellular vesicle signalling in atherosclerosis. Cell Signal 2020; 75:109751. [PMID: 32860954 PMCID: PMC7534042 DOI: 10.1016/j.cellsig.2020.109751] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a major cardiovascular disease and in 2016, the World Health Organisation (WHO) estimated 17.5 million global deaths, corresponding to 31% of all global deaths, were driven by inflammation and deposition of lipids into the arterial wall. This leads to the development of plaques which narrow the vessel lumen, particularly in the coronary and carotid arteries. Atherosclerotic plaques can become unstable and rupture, leading to myocardial infarction or stroke. Extracellular vesicles (EVs) are a heterogeneous population of vesicles secreted from cells with a wide range of biological functions. EVs participate in cell-cell communication and signalling via transport of cargo including enzymes, DNA, RNA and microRNA in both physiological and patholophysiological settings. EVs are present in atherosclerotic plaques and have been implicated in cellular signalling processes in atherosclerosis development, including immune responses, inflammation, cell proliferation and migration, cell death and vascular remodeling during progression of the disease. In this review, we summarise the current knowledge regarding EV signalling in atherosclerosis progression and the potential of utilising EV signatures as biomarkers of disease.
Collapse
|
86
|
Brock DJ, Kondow-McConaghy H, Allen J, Brkljača Z, Kustigian L, Jiang M, Zhang J, Rye H, Vazdar M, Pellois JP. Mechanism of Cell Penetration by Permeabilization of Late Endosomes: Interplay between a Multivalent TAT Peptide and Bis(monoacylglycero)phosphate. Cell Chem Biol 2020; 27:1296-1307.e5. [PMID: 32783962 DOI: 10.1016/j.chembiol.2020.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/06/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023]
Abstract
Many cellular delivery reagents enter the cytosolic space of cells by escaping the lumen of endocytic organelles and, more specifically, late endosomes. The mechanisms involved in endosomal membrane permeation remain largely unresolved, which impedes the improvement of delivery agents. Here, we investigate how 3TAT, a branched analog of the cell-penetrating peptide (CPP) TAT, achieves the permeabilization of bilayers containing bis(monoacylglycero)phosphate (BMP), a lipid found in late endosomes. We establish that the peptide does not induce the leakage of individual lipid bilayers. Instead, leakage requires contact between membranes. Peptide-driven bilayer contacts lead to fusion, lipid mixing, and, critically, peptide encapsulation within proximal bilayers. Notably, this encapsulation is a distinctive property of BMP that explains the specificity of CPP's membrane leakage activity. These results therefore support a model of cell penetration that requires both BMP and the vicinity between bilayers, two features unique to BMP-rich and multivesicular late endosomes.
Collapse
Affiliation(s)
- Dakota J Brock
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Helena Kondow-McConaghy
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Jason Allen
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Zlatko Brkljača
- Division of Organic Chemistry and Biochemistry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Lauren Kustigian
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Mengqiu Jiang
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Junjie Zhang
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Hays Rye
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Mario Vazdar
- Division of Organic Chemistry and Biochemistry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA; Department of Chemistry, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
87
|
Elbaz-Alon Y, Guo Y, Segev N, Harel M, Quinnell DE, Geiger T, Avinoam O, Li D, Nunnari J. PDZD8 interacts with Protrudin and Rab7 at ER-late endosome membrane contact sites associated with mitochondria. Nat Commun 2020; 11:3645. [PMID: 32686675 PMCID: PMC7371716 DOI: 10.1038/s41467-020-17451-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 06/23/2020] [Indexed: 12/02/2022] Open
Abstract
Endosomes are compositionally dynamic organelles that regulate signaling, nutrient status and organelle quality by specifying whether material entering the cells will be shuttled back to the cell surface or degraded by the lysosome. Recently, membrane contact sites (MCSs) between the endoplasmic reticulum (ER) and endosomes have emerged as important players in endosomal protein sorting, dynamics and motility. Here, we show that PDZD8, a Synaptotagmin-like Mitochondrial lipid-binding Proteins (SMP) domain-containing ER transmembrane protein, utilizes distinct domains to interact with Rab7-GTP and the ER transmembrane protein Protrudin and together these components localize to an ER-late endosome MCS. At these ER-late endosome MCSs, mitochondria are also recruited to form a three-way contact. Thus, our data indicate that PDZD8 is a shared component of two distinct MCSs and suggest a role for SMP-mediated lipid transport in the regulation of endosome function. Membrane contact sites between organelles have been shown to play important biological roles. Here, the authors show that at the ER, PDZD8 associates with Protrudin and also with Rab7 endosomes and recruits mitochondria to form three-way contacts.
Collapse
Affiliation(s)
- Yael Elbaz-Alon
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA, USA. .,Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Yuting Guo
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Nadav Segev
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Harel
- Department of Human Molecular Genetics and Biochemistry, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Daniel E Quinnell
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA, USA
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Dong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jodi Nunnari
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA, USA.
| |
Collapse
|
88
|
Rabia M, Leuzy V, Soulage C, Durand A, Fourmaux B, Errazuriz-Cerda E, Köffel R, Draeger A, Colosetti P, Jalabert A, Di Filippo M, Villard-Garon A, Bergerot C, Luquain-Costaz C, Moulin P, Rome S, Delton I, Hullin-Matsuda F. Bis(monoacylglycero)phosphate, a new lipid signature of endosome-derived extracellular vesicles. Biochimie 2020; 178:26-38. [PMID: 32659447 DOI: 10.1016/j.biochi.2020.07.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/29/2020] [Accepted: 07/08/2020] [Indexed: 01/05/2023]
Abstract
Bis(monoacylglycero)phosphate (BMP), also known as lysobisphosphatidic acid (LBPA), is a phospholipid specifically enriched in the late endosome-lysosome compartment playing a crucial role for the fate of endocytosed components. Due to its presence in extracellular fluids during diseases associated with endolysosomal dysfunction, it is considered as a possible biomarker of disorders such as genetic lysosomal storage diseases and cationic amphiphilic drug-induced phospholipidosis. However, there is no true validation of this biomarker in human studies, nor a clear identification of the carrier of this endolysosome-specific lipid in biofluids. The present study demonstrates that in absence of any sign of renal failure, BMP, especially all docosahexaenoyl containing species, are significantly increased in the urine of patients treated with the antiarrhythmic drug amiodarone. Such urinary BMP increase could reflect a generalized drug-induced perturbation of the endolysosome compartment as observed in vitro with amiodarone-treated human macrophages. Noteworthy, BMP was associated with extracellular vesicles (EVs) isolated from human urines and extracellular medium of human embryonic kidney HEK293 cells and co-localizing with classical EV protein markers CD63 and ALIX. In the context of drug-induced endolysosomal dysfunction, increased BMP-rich EV release could be useful to remove excess of undigested material. This first human pilot study not only reveals BMP as a urinary biomarker of amiodarone-induced endolysosomal dysfunction, but also highlights its utility to prove the endosomal origin of EVs, also named as exosomes. This peculiar lipid already known as a canonical late endosome-lysosome marker, may be thus considered as a new lipid marker of urinary exosomes.
Collapse
Affiliation(s)
- Maxence Rabia
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Valentin Leuzy
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Christophe Soulage
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Annie Durand
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Baptiste Fourmaux
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Functional Lipidomics Platform, CarMeN Laboratory / IMBL-INSA Lyon, 69621, Villeurbanne Cedex, France
| | | | - René Köffel
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Annette Draeger
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Pascal Colosetti
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Audrey Jalabert
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Mathilde Di Filippo
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Department of Biochemistry and Molecular Biology, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Bron, France
| | - Audrey Villard-Garon
- Department of Endocrinology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Cyrille Bergerot
- Department of Cardiology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Céline Luquain-Costaz
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Philippe Moulin
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Department of Endocrinology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Sophie Rome
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Isabelle Delton
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | | |
Collapse
|
89
|
Grabner GF, Fawzy N, Schreiber R, Pusch LM, Bulfon D, Koefeler H, Eichmann TO, Lass A, Schweiger M, Marsche G, Schoiswohl G, Taschler U, Zimmermann R. Metabolic regulation of the lysosomal cofactor bis(monoacylglycero)phosphate in mice. J Lipid Res 2020; 61:995-1003. [PMID: 32350080 PMCID: PMC7328040 DOI: 10.1194/jlr.ra119000516] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/23/2020] [Indexed: 01/02/2023] Open
Abstract
Bis(monoacylglycero)phosphate (BMP), also known as lysobisphosphatidic acid, is a phospholipid that promotes lipid sorting in late endosomes/lysosomes by activating lipid hydrolases and lipid transfer proteins. Changes in the cellular BMP content therefore reflect an altered metabolic activity of the endolysosomal system. Surprisingly, little is known about the physiological regulation of BMP. In this study, we investigated the effects of nutritional and metabolic factors on BMP profiles of whole tissues and parenchymal and nonparenchymal cells. Tissue samples were obtained from fed, fasted, 2 h refed, and insulin-treated mice, as well as from mice housed at 5°C, 22°C, or 30°C. These tissues exhibited distinct BMP profiles that were regulated by the nutritional state in a tissue-specific manner. Insulin treatment was not sufficient to mimic refeeding-induced changes in tissue BMP levels, indicating that BMP metabolism is regulated by other hormonal or nutritional factors. Tissue fractionation experiments revealed that fasting drastically elevates BMP levels in hepatocytes and pancreatic cells. Furthermore, we observed that the BMP content in brown adipose tissue strongly depends on housing temperatures. In conclusion, our observations suggest that BMP concentrations adapt to the metabolic state in a tissue- and cell-type-specific manner in mice. Drastic changes observed in hepatocytes, pancreatic cells, and brown adipocytes suggest that BMP plays a role in the functional adaption to nutrient starvation and ambient temperature.
Collapse
Affiliation(s)
- Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Nermeen Fawzy
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Lisa M Pusch
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Dominik Bulfon
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Harald Koefeler
- Otto Loewi Research Center, and Center for Medical Research, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Center for Explorative Lipidomics, BioTechMed-Graz, Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | | | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria. mailto:
| |
Collapse
|
90
|
Ohashi Y, Tremel S, Masson GR, McGinney L, Boulanger J, Rostislavleva K, Johnson CM, Niewczas I, Clark J, Williams RL. Membrane characteristics tune activities of endosomal and autophagic human VPS34 complexes. eLife 2020; 9:58281. [PMID: 32602837 PMCID: PMC7326497 DOI: 10.7554/elife.58281] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
The lipid kinase VPS34 orchestrates diverse processes, including autophagy, endocytic sorting, phagocytosis, anabolic responses and cell division. VPS34 forms various complexes that help adapt it to specific pathways, with complexes I and II being the most prominent ones. We found that physicochemical properties of membranes strongly modulate VPS34 activity. Greater unsaturation of both substrate and non-substrate lipids, negative charge and curvature activate VPS34 complexes, adapting them to their cellular compartments. Hydrogen/deuterium exchange mass spectrometry (HDX-MS) of complexes I and II on membranes elucidated structural determinants that enable them to bind membranes. Among these are the Barkor/ATG14L autophagosome targeting sequence (BATS), which makes autophagy-specific complex I more active than the endocytic complex II, and the Beclin1 BARA domain. Interestingly, even though Beclin1 BARA is common to both complexes, its membrane-interacting loops are critical for complex II, but have only a minor role for complex I.
Collapse
Affiliation(s)
- Yohei Ohashi
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| | - Shirley Tremel
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| | - Glenn Robert Masson
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| | - Lauren McGinney
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| | - Jerome Boulanger
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| | - Ksenia Rostislavleva
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| | - Christopher M Johnson
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| | | | | | - Roger L Williams
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| |
Collapse
|
91
|
Ghosh U, Weliky DP. 2H nuclear magnetic resonance spectroscopy supports larger amplitude fast motion and interference with lipid chain ordering for membrane that contains β sheet human immunodeficiency virus gp41 fusion peptide or helical hairpin influenza virus hemagglutinin fusion peptide at fusogenic pH. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183404. [PMID: 32585207 DOI: 10.1016/j.bbamem.2020.183404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/27/2020] [Accepted: 06/19/2020] [Indexed: 01/02/2023]
Abstract
Enveloped viruses are surrounded by a membrane which is obtained from an infected host cell during budding. Infection of a new cell requires joining (fusion) of the virus and cell membranes. This process is mediated by a monotopic viral fusion protein with a large ectodomain outside the virus. The ectodomains of class I enveloped viruses have a N-terminal "fusion peptide" (fp) domain that is critical for fusion and binds to the cell membrane. In this study, 2H NMR spectra are analyzed for deuterated membrane with fp from either HIV gp41 (GP) or influenza hemagglutinin (HA) fusion proteins. In addition, the HAfp samples are studied at more fusogenic pH 5 and less fusogenic pH 7. GPfp adopts intermolecular antiparallel β sheet structure whereas HAfp is a monomeric helical hairpin. The data are obtained for a set of temperatures between 35 and 0 °C using DMPC-d54 lipid with perdeuterated acyl chains. The DMPC has liquid-crystalline (Lα) phase with disordered chains at higher temperature and rippled gel (Pβ') or gel phase (Lβ') with ordered chains at lower temperature. At given temperature T, the no peptide and HAfp, pH 7 samples exhibit similar spectral lineshapes. Spectral broadening with reduced temperature correlates with the transition from Lα to Pβ' and then Lβ' phases. At given T, the lineshapes are narrower for HAfp, pH 5 vs. no peptide and HAfp, pH 7 samples, and even narrower for the GPfp sample. These data support larger-amplitude fast (>105 Hz) lipid acyl chain motion for samples with fusogenic peptides, and peptide interference with chain ordering. The NMR data of the present paper correlate with insertion of these peptides into the hydrocarbon core of the membrane and support a significant fusion contribution from the resultant lipid acyl chain disorder, perhaps because of reduced barriers between the different membrane topologies in the fusion pathway. Membrane insertion and lipid perturbation appear common to both β sheet and helical hairpin peptides.
Collapse
Affiliation(s)
- Ujjayini Ghosh
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - David P Weliky
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
92
|
Nadal‐Bufí F, Henriques ST. How to overcome endosomal entrapment of cell‐penetrating peptides to release the therapeutic potential of peptides? Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Ferran Nadal‐Bufí
- School of Biomedical Sciences, Faculty of Health, Institute of Health & Biomedical Innovation, Queensland University of Technology Translational Research Institute Brisbane Queensland Australia
| | - Sónia Troeira Henriques
- School of Biomedical Sciences, Faculty of Health, Institute of Health & Biomedical Innovation, Queensland University of Technology Translational Research Institute Brisbane Queensland Australia
| |
Collapse
|
93
|
Mori A, Imai Y, Hattori N. Lipids: Key Players That Modulate α-Synuclein Toxicity and Neurodegeneration in Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21093301. [PMID: 32392751 PMCID: PMC7247581 DOI: 10.3390/ijms21093301] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease; it is characterized by the loss of dopaminergic neurons in the midbrain and the accumulation of neuronal inclusions, mainly consisting of α-synuclein (α-syn) fibrils in the affected regions. The prion-like property of the pathological forms of α-syn transmitted via neuronal circuits has been considered inherent in the nature of PD. Thus, one of the potential targets in terms of PD prevention is the suppression of α-syn conversion from the functional form to pathological forms. Recent studies suggested that α-syn interacts with synaptic vesicle membranes and modulate the synaptic functions. A series of studies suggest that transient interaction of α-syn as multimers with synaptic vesicle membranes composed of phospholipids and other lipids is required for its physiological function, while an α-syn-lipid interaction imbalance is believed to cause α-syn aggregation and the resultant pathological α-syn conversion. Altered lipid metabolisms have also been implicated in the modulation of PD pathogenesis. This review focuses on the current literature reporting the role of lipids, especially phospholipids, and lipid metabolism in α-syn dynamics and aggregation processes.
Collapse
Affiliation(s)
- Akio Mori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan;
| | - Yuzuru Imai
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan;
- Department of Research for Parkinson’s Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Correspondence: (Y.I.); (N.H.); Tel.: +81-3-6801-8332 (Y.I. & N.H.)
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan;
- Department of Research for Parkinson’s Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Correspondence: (Y.I.); (N.H.); Tel.: +81-3-6801-8332 (Y.I. & N.H.)
| |
Collapse
|
94
|
Joshi B, de Beer MA, Giepmans BNG, Zuhorn IS. Endocytosis of Extracellular Vesicles and Release of Their Cargo from Endosomes. ACS NANO 2020; 14:4444-4455. [PMID: 32282185 PMCID: PMC7199215 DOI: 10.1021/acsnano.9b10033] [Citation(s) in RCA: 342] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/13/2020] [Indexed: 05/22/2023]
Abstract
Extracellular vesicles (EVs), such as exosomes, can mediate long-distance communication between cells by delivering biomolecular cargo. It is speculated that EVs undergo back-fusion at multivesicular bodies (MVBs) in recipient cells to release their functional cargo. However, direct evidence is lacking. Tracing the cellular uptake of EVs with high resolution as well as acquiring direct evidence for the release of EV cargo is challenging mainly because of technical limitations. Here, we developed an analytical methodology, combining state-of-the-art molecular tools and correlative light and electron microscopy, to identify the intracellular site for EV cargo release. GFP was loaded inside EVs through the expression of GFP-CD63, a fusion of GFP to the cytosolic tail of CD63, in EV producer cells. In addition, we genetically engineered a cell line which expresses anti-GFP fluobody that specifically recognizes the EV cargo (GFP). Incubation of anti-GFP fluobody-expressing cells with GFP-CD63 EVs resulted in the formation of fluobody punctae, designating cytosolic exposure of GFP. Endosomal damage was not observed in EV acceptor cells. Ultrastructural analysis of the underlying structures at GFP/fluobody double-positive punctae demonstrated that EV cargo release occurs from endosomes/lysosomes. Finally, we show that neutralization of endosomal pH and cholesterol accumulation in endosomes leads to blockage of EV cargo exposure. In conclusion, we report that a fraction of internalized EVs fuse with the limiting membrane of endosomes/lysosomes in an acidification-dependent manner, which results in EV cargo exposure to the cell cytosol.
Collapse
Affiliation(s)
- Bhagyashree
S. Joshi
- Department
of Biomedical Engineering, University of
Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Marit A. de Beer
- Department
of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Ben N. G. Giepmans
- Department
of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Inge S. Zuhorn
- Department
of Biomedical Engineering, University of
Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
95
|
Le Joncour V, Filppu P, Hyvönen M, Holopainen M, Turunen SP, Sihto H, Burghardt I, Joensuu H, Tynninen O, Jääskeläinen J, Weller M, Lehti K, Käkelä R, Laakkonen P. Vulnerability of invasive glioblastoma cells to lysosomal membrane destabilization. EMBO Mol Med 2020; 11:emmm.201809034. [PMID: 31068339 PMCID: PMC6554674 DOI: 10.15252/emmm.201809034] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The current clinical care of glioblastomas leaves behind invasive, radio‐ and chemo‐resistant cells. We recently identified mammary‐derived growth inhibitor (MDGI/FABP3) as a biomarker for invasive gliomas. Here, we demonstrate a novel function for MDGI in the maintenance of lysosomal membrane integrity, thus rendering invasive glioma cells unexpectedly vulnerable to lysosomal membrane destabilization. MDGI silencing impaired trafficking of polyunsaturated fatty acids into cells resulting in significant alterations in the lipid composition of lysosomal membranes, and subsequent death of the patient‐derived glioma cells via lysosomal membrane permeabilization (LMP). In a preclinical model, treatment of glioma‐bearing mice with an antihistaminergic LMP‐inducing drug efficiently eradicated invasive glioma cells and secondary tumours within the brain. This unexpected fragility of the aggressive infiltrating cells to LMP provides new opportunities for clinical interventions, such as re‐positioning of an established antihistamine drug, to eradicate the inoperable, invasive, and chemo‐resistant glioma cells from sustaining disease progression and recurrence.
Collapse
Affiliation(s)
- Vadim Le Joncour
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pauliina Filppu
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maija Hyvönen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Minna Holopainen
- Helsinki University Lipidomics Unit, Helsinki Institute of Life Science (HiLIFE) and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - S Pauliina Turunen
- Research Programs Unit, Genome-Scale Biology, University of Helsinki, Helsinki, Finland.,Department of Microbiology, Tumour and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Harri Sihto
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Isabel Burghardt
- Department of Neurology and Brain Tumour Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Heikki Joensuu
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Oncology, Helsinki University Hospital, Helsinki, Finland
| | - Olli Tynninen
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB, Helsinki, Finland
| | | | - Michael Weller
- Department of Neurology and Brain Tumour Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Kaisa Lehti
- Research Programs Unit, Genome-Scale Biology, University of Helsinki, Helsinki, Finland.,Department of Microbiology, Tumour and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, Helsinki Institute of Life Science (HiLIFE) and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland .,Laboratory Animal Centre, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
96
|
Richter F, Martin L, Leer K, Moek E, Hausig F, Brendel JC, Traeger A. Tuning of endosomal escape and gene expression by functional groups, molecular weight and transfection medium: a structure-activity relationship study. J Mater Chem B 2020; 8:5026-5041. [PMID: 32319993 DOI: 10.1039/d0tb00340a] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The use of genetic material by non-viral transfer systems is still in its initial stages, but there are high expectations for the development of targeted therapies. However, nucleic acids cannot enter cells without help, they must be well protected to prevent degradation and overcome a variety of biological barriers, the endosomal barrier being one of the greatest cellular challenges. Herein, the structure-property-relationship was investigated in detail, using well-defined polymers. Polyacrylamides were synthesized via RAFT polymerization resulting in a polymer library of (i) different cationic groups as aminoethyl acrylamide (AEAm), dimethylaminoethyl acrylamide (DMAEAm), dimethylaminopropyl acrylamide (DMAPAm) and guanidinopropyl acrylamide (GPAm); (ii) different degree of polymerization; and investigated (iii) in different cell culture settings. The influence of molar mass and cationic moiety on complex formation with pDNA, cytotoxicity and transfection efficiency of the polymers were investigated. The systematic approach identified a pH-independent guanidinium-containing homopolymer (PGPAm89) as the polymer with the highest transfection efficiency and superior endosomal release under optimal conditions. Since PGPAm89 is not further protonated inside endosomes, common escape theories appear unsuitable. Therefore, the interaction with bis(monoacryloylglycerol)phosphate, a lipid specific for endosomal vesicles, was investigated. Our research suggests that the interactions between amines and lipids may be more relevant than anticipated.
Collapse
Affiliation(s)
- Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.
| | | | | | | | | | | | | |
Collapse
|
97
|
Larios J, Mercier V, Roux A, Gruenberg J. ALIX- and ESCRT-III-dependent sorting of tetraspanins to exosomes. J Cell Biol 2020; 219:e201904113. [PMID: 32049272 PMCID: PMC7054990 DOI: 10.1083/jcb.201904113] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/31/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
The intraluminal vesicles (ILVs) of endosomes mediate the delivery of activated signaling receptors and other proteins to lysosomes for degradation, but they also modulate intercellular communication when secreted as exosomes. The formation of ILVs requires four complexes, ESCRT-0, -I, -II, and -III, with ESCRT-0, -I, and -II presumably involved in cargo sorting and ESCRT-III in membrane deformation and fission. Here, we report that an active form of the ESCRT-associated protein ALIX efficiently recruits ESCRT-III proteins to endosomes. This recruitment occurs independently of other ESCRTs but requires lysobisphosphatidic acid (LBPA) in vivo, and can be reconstituted on supported bilayers in vitro. Our data indicate that this ALIX- and ESCRT-III-dependent pathway promotes the sorting and delivery of tetraspanins to exosomes. We conclude that ALIX provides an additional pathway of ILV formation, secondary to the canonical pathway, and that this pathway controls the targeting of exosomal proteins.
Collapse
|
98
|
Nathan L, Lai AL, Millet JK, Straus MR, Freed JH, Whittaker GR, Daniel S. Calcium Ions Directly Interact with the Ebola Virus Fusion Peptide To Promote Structure-Function Changes That Enhance Infection. ACS Infect Dis 2020; 6:250-260. [PMID: 31746195 DOI: 10.1021/acsinfecdis.9b00296] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ebola virus disease is a serious global health concern given its periodic occurrence, high lethality, and the lack of approved therapeutics. Certain drugs that alter intracellular calcium, particularly in endolysosomes, have been shown to inhibit Ebola virus infection; however, the underlying mechanism is unknown. Here, we provide evidence that Zaire ebolavirus (EBOV) infection is promoted in the presence of calcium as a result of the direct interaction of calcium with the EBOV fusion peptide (FP). We identify the glycoprotein residues D522 and E540 in the FP as functionally critical to EBOV's interaction with calcium. We show using spectroscopic and biophysical assays that interactions of the fusion peptide with Ca2+ ions lead to lipid ordering in the host membrane during membrane fusion, and these changes are promoted at low pH and can be correlated with infectivity. We further demonstrate using circular dichroism spectroscopy that calcium interaction with the fusion peptide promotes α-helical structure of the fusion peptide, a conformational change that enhances membrane fusion, as validated using functional assays of membrane fusion. This study shows that calcium directly targets the Ebola virus fusion peptide and influences its conformation. As these residues are highly conserved across the Filoviridae, calcium's impact on fusion, and subsequently infectivity, is a key interaction that can be leveraged for developing strategies to defend against Ebola infection. This mechanistic insight provides a rationale for the use of calcium-interfering drugs already approved by the FDA as therapeutics against Ebola and enables further development of novel drugs to combat the virus.
Collapse
Affiliation(s)
- Lakshmi Nathan
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New York 14853, United States
| | - Alex L. Lai
- Baker Laboratory, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Jean Kaoru Millet
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Marco R. Straus
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Jack H. Freed
- Baker Laboratory, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New York 14853, United States
| |
Collapse
|
99
|
Carpio LE, Villalaín J. Identification of the phospholipid binding regions of the envelope E protein of flaviviruses by molecular dynamics. J Biomol Struct Dyn 2019; 38:5136-5147. [PMID: 31779533 DOI: 10.1080/07391102.2019.1697368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The Flavivirus genus comprise several important human pathogens, including dengue, West Nile, Yellow fever, Japanese encephalitis, Zika, and tick-borne encephalitis viruses. These enveloped viruses affect more than 2 billion people in the world, mainly in less developed countries. Although some vaccines exist for some flaviviruses, these vaccines are not universally available due to many factors and since their infections are a world-wide public health issue, the development of antiviral molecules is fundamental. Flavivirus membranes, through the help of the envelope E glycoprotein, fuse with endosomal compartments in a pH-dependent way to release their genome into the cytoplasm and require specific lipids, such as bis(monoacylglycero)phosphate (BMP), for efficient fusion. The fundamental role the envelope E protein has on viral entry and membrane fusion suggest that it is an essential antiviral target. In this work, we have used atomistic molecular dynamics simulations to study the binding of the head-group of BMP to the tip of the envelope E proteins of ZIKV, DENV, TBEV and JEV viruses whose three-dimensional structures are known. Our results indicate that, apart from the fusion loop, there are different amino acid residues in different regions of the envelope E proteins of flaviviruses capable of binding the head-group of BMP. These regions should work together to accomplish the binding and fusion of the envelope and endosomal membranes and represent a new target to develop and design potent and effective antiviral agents capable of blocking flavivirus-endosome membrane fusion. [Formula: see text].
Collapse
Affiliation(s)
- Laureano E Carpio
- Molecular and Cellular Biology Institute (IBMC) and Institute of Research, Development, and Innovation in Healthcare Biotechnology (IDiBE), Universidad 'Miguel Hernández', Elche-Alicante, Spain
| | - José Villalaín
- Molecular and Cellular Biology Institute (IBMC) and Institute of Research, Development, and Innovation in Healthcare Biotechnology (IDiBE), Universidad 'Miguel Hernández', Elche-Alicante, Spain
| |
Collapse
|
100
|
Botté A, Potier MC. Focusing on cellular biomarkers: The endo-lysosomal pathway in Down syndrome. PROGRESS IN BRAIN RESEARCH 2019; 251:209-243. [PMID: 32057308 DOI: 10.1016/bs.pbr.2019.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Down syndrome (DS) is the most frequent chromosomal disorder. It is caused by the triplication of human chromosome 21, leading to increased dosage of a variety of genes including APP (Amyloid Precursor Protein). Mainly for this reason, individuals with DS are at high risk to develop Alzheimer's disease (AD). Extensive literature identified various morphological and molecular abnormalities in the endo-lysosomal pathway both in DS and AD. Most studies in this field investigated the causative role of APP (Amyloid Precursor Protein) in endo-lysosomal dysfunctions, thus linking phenotypes observed in DS and AD. In DS context, several lines of evidence and emerging hypotheses suggest that other molecular players and pathways may be implicated in these complex phenotypes. In this review, we outline the normal functioning of endosomal trafficking and summarize the research on endo-lysosomal dysfunction in DS in light of AD findings. We emphasize the role of genes of chromosome 21 implicated in endocytosis to explain endosomal abnormalities and set the limitations and perspectives of models used to explore endo-lysosomal dysfunction in DS and find new biomarkers. The review highlights the complexity of endo-lysosomal dysfunction in DS and suggests directions for future research in the field.
Collapse
Affiliation(s)
- Alexandra Botté
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Marie-Claude Potier
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France.
| |
Collapse
|