101
|
Lu A, Hsieh F, Sharma BR, Vaughn SR, Enrich C, Pfeffer SR. CRISPR screens for lipid regulators reveal a role for ER-bound SNX13 in lysosomal cholesterol export. J Cell Biol 2022; 221:212937. [PMID: 34936700 PMCID: PMC8704955 DOI: 10.1083/jcb.202105060] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/02/2021] [Accepted: 11/29/2021] [Indexed: 11/22/2022] Open
Abstract
We report here two genome-wide CRISPR screens performed to identify genes that, when knocked out, alter levels of lysosomal cholesterol or bis(monoacylglycero)phosphate. In addition, these screens were also performed under conditions of NPC1 inhibition to identify modifiers of NPC1 function in lysosomal cholesterol export. The screens confirm tight coregulation of cholesterol and bis(monoacylglycero)phosphate in cells and reveal an unexpected role for the ER-localized SNX13 protein as a negative regulator of lysosomal cholesterol export and contributor to ER–lysosome membrane contact sites. In the absence of NPC1 function, SNX13 knockdown redistributes lysosomal cholesterol and is accompanied by triacylglycerol-rich lipid droplet accumulation and increased lysosomal bis(monoacylglycero)phosphate. These experiments provide unexpected insight into the regulation of lysosomal lipids and modification of these processes by novel gene products.
Collapse
Affiliation(s)
- Albert Lu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA.,Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | | | - Bikal R Sharma
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA
| | - Sydney R Vaughn
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Suzanne R Pfeffer
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
102
|
Zhong C, Wang B. Regulation of Cholesterol Binding to the Receptor Patched1 by its interactions With the Ligand Sonic Hedgehog (Shh). Front Mol Biosci 2022; 9:831891. [PMID: 35187087 PMCID: PMC8847689 DOI: 10.3389/fmolb.2022.831891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/11/2022] [Indexed: 01/17/2023] Open
Abstract
The Hedgehog (Hh) signaling pathway is essential in cell development and regeneration, which is activated by the ligand Sonic hedgehog (Shh). The binding of Shh to its receptor Patched1 (PTCH1) releases the inhibitory effect on the downstream protein Smoothened (SMO), a G-protein-coupled-receptor (GPCR) protein. Cholesterol was supposed to function as a secondary messenger between PTCH1 and SMO. However, the molecular mechanism of this regulation process is still unclear. Therefore, microsecond coarse-grained molecular dynamics simulations were performed to investigate the protein-lipid interactions of the PTCH1 monomer and dimer-Shh complex. It was observed that the binding of cholesterols to the monomer is more stable than that to the dimer-Shh complex. It is regulated by the enrichment of Ganglioside lipids around proteins and the conformation of Y446, a residue in the sterol-sensing domain (SSD). The regulation of Shh on the dynamics of PTCH1 was further analyzed to explore the allosteric communication pathways between the Shh and the SSD. Our study provides structural and dynamic details of an additional perspective on the regulation of Hh signaling pathway through the lipid micro-environments of PTCH1.
Collapse
Affiliation(s)
- Changqing Zhong
- Centre for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Beibei Wang
- Centre for Advanced Materials Research, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, China
- *Correspondence: Beibei Wang,
| |
Collapse
|
103
|
Griffiths WJ, Wang Y. Cholesterol metabolism: from lipidomics to immunology. J Lipid Res 2022; 63:100165. [PMID: 34953867 PMCID: PMC8953665 DOI: 10.1016/j.jlr.2021.100165] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022] Open
Abstract
Oxysterols, the oxidized forms of cholesterol or of its precursors, are formed in the first steps of cholesterol metabolism. Oxysterols have interested chemists, biologists, and physicians for many decades, but their exact biological relevance in vivo, other than as intermediates in bile acid biosynthesis, has long been debated. However, in the first quarter of this century, a role for side-chain oxysterols and their C-7 oxidized metabolites has been convincingly established in the immune system. 25-Hydroxycholesterol has been shown to be synthesized by macrophages in response to the activation of Toll-like receptors and to offer protection against microbial pathogens, whereas 7α,25-dihydroxycholesterol has been shown to act as a chemoattractant to lymphocytes expressing the G protein-coupled receptor Epstein-Barr virus-induced gene 2 and to be important in coordinating the action of B cells, T cells, and dendritic cells in secondary lymphoid tissue. There is a growing body of evidence that not only these two oxysterols but also many of their isomers are of importance to the proper function of the immune system. Here, we review recent findings related to the roles of oxysterols in immunology.
Collapse
Affiliation(s)
| | - Yuqin Wang
- Swansea University Medical School, Swansea, Wales, United Kingdom.
| |
Collapse
|
104
|
Wall CTJ, Lefebvre G, Metairon S, Descombes P, Wiederkehr A, Santo-Domingo J. Mitochondrial respiratory chain dysfunction alters ER sterol sensing and mevalonate pathway activity. J Biol Chem 2022; 298:101652. [PMID: 35101444 PMCID: PMC8892029 DOI: 10.1016/j.jbc.2022.101652] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/13/2022] [Accepted: 01/25/2022] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial dysfunction induces a strong adaptive retrograde signaling response; however, many of the downstream effectors of this response remain to be discovered. Here, we studied the shared transcriptional responses to three different mitochondrial respiratory chain inhibitors in human primary skin fibroblasts using QuantSeq 3′-RNA-sequencing. We found that genes involved in the mevalonate pathway were concurrently downregulated, irrespective of the respiratory chain complex affected. Targeted metabolomics demonstrated that impaired mitochondrial respiration at any of the three affected complexes also had functional consequences on the mevalonate pathway, reducing levels of cholesterol precursor metabolites. A deeper study of complex I inhibition showed a reduced activity of endoplasmic reticulum–bound sterol-sensing enzymes through impaired processing of the transcription factor Sterol Regulatory Element-Binding Protein 2 and accelerated degradation of the endoplasmic reticulum cholesterol-sensors squalene epoxidase and HMG-CoA reductase. These adaptations of mevalonate pathway activity affected neither total intracellular cholesterol levels nor the cellular free (nonesterified) cholesterol pool. Finally, measurement of intracellular cholesterol using the fluorescent cholesterol binding dye filipin revealed that complex I inhibition elevated cholesterol on intracellular compartments. Taken together, our study shows that mitochondrial respiratory chain dysfunction elevates intracellular free cholesterol levels and therefore attenuates the expression of mevalonate pathway enzymes, which lowers endogenous cholesterol biosynthesis, disrupting the metabolic output of the mevalonate pathway. We conclude that intracellular disturbances in cholesterol homeostasis may alter systemic cholesterol management in diseases associated with declining mitochondrial function.
Collapse
Affiliation(s)
- Christopher Tadhg James Wall
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland; Institute of Bioengineering, Life Science Faculty, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gregory Lefebvre
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sylviane Metairon
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Patrick Descombes
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Andreas Wiederkehr
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland; Institute of Bioengineering, Life Science Faculty, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Jaime Santo-Domingo
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), University of Valladolid and CSIC, Valladolid, Spain.
| |
Collapse
|
105
|
Sphingomyelin-Sequestered Cholesterol Domain Recruits Formin-Binding Protein 17 for Constricting Clathrin-Coated Pits in Influenza Virus Entry. J Virol 2022; 96:e0181321. [PMID: 35020471 DOI: 10.1128/jvi.01813-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Influenza A virus (IAV) is a global health threat. The cellular endocytic machineries harnessed by IAV remain elusive. Here, by tracking single IAV particles and quantifying the internalized IAV, we found that the sphingomyelin (SM)-sequestered cholesterol, but not the accessible cholesterol, is essential for the clathrin-mediated endocytosis (CME) of IAV. The clathrin-independent endocytosis of IAV is cholesterol-independent. Whereas, the CME of transferrin depends on SM-sequestered cholesterol and accessible cholesterol. Furthermore, three-color single-virus tracking and electron microscopy showed that the SM-cholesterol complex nanodomain is recruited to the IAV-containing clathrin-coated structure (CCS) and facilitates neck constriction of the IAV-containing CCS. Meanwhile, formin-binding protein 17 (FBP17), a membrane-bending protein which activates actin nucleation, is recruited to IAV-CCS complex in a manner dependent on the SM-cholesterol complex. We propose that the SM-cholesterol nanodomain at the neck of CCS recruits FBP17 to induce neck constriction by activating actin assembly. These results unequivocally show the physiological importance of the SM-cholesterol complex in IAV entry. Importance: IAV infects the cells by harnessing cellular endocytic machineries. Better understanding of the cellular machineries used for its entry might lead to the development of antiviral strategies, and would also provide important insights into physiological endocytic processes. This work demonstrated that a special pool of cholesterol in plasma membrane, SM-sequestered cholesterol, recruits FBP17 for the constriction of clathrin-coated pits in IAV entry. Meanwhile, the clathrin-independent cell entry of IAV is cholesterol-independent. The internalization of transferrin, the gold-standard cargo endocytosed solely via CME, is much less dependent on the SM-cholesterol complex. These results would provide new insights into IAV infection and pathway/cargo-specific involvement of cholesterol pool(s).
Collapse
|
106
|
Interplay between Asters/GRAMD1s and phosphatidylserine in intermembrane transport of LDL cholesterol. Proc Natl Acad Sci U S A 2022; 119:2120411119. [PMID: 34992143 PMCID: PMC8764668 DOI: 10.1073/pnas.2120411119] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2021] [Indexed: 12/23/2022] Open
Abstract
Cholesterol constitutes 50% of lipids in the plasma membrane (PM) of animal cells. Sensors in the endoplasmic reticulum (ER) maintain this level by adjusting cholesterol uptake, synthesis, and storage. Uptake is mediated by LDL receptors, which deliver cholesterol-carrying LDL to lysosomes from which cholesterol moves to the PM and then to the ER. We report PM-to-ER transport of LDL cholesterol requires cholesterol-binding Aster proteins anchored to the ER and phosphatidylserine embedded in the PM. Asters are known to bind phosphatidylserine, and this accounts for part of the phosphatidylserine requirement. However, the current data suggest an additional requirement for phosphatidylserine independent of Asters. These data advance our knowledge of PM cholesterol homeostasis, a control mechanism essential for cell growth and survival. Low-density lipoprotein (LDL) delivers cholesterol to mammalian cells through receptor-mediated endocytosis. The LDL cholesterol is liberated in lysosomes and transported to the plasma membrane (PM) and from there to the endoplasmic reticulum (ER). Excess ER cholesterol is esterified with a fatty acid for storage as cholesteryl esters. Recently, we showed that PM-to-ER transport of LDL cholesterol requires phosphatidylserine (PS). Others showed that PM-to-ER transport of cholesterol derived from other sources requires Asters (also called GRAMD1s), a family of three ER proteins that bridge between the ER and PM by binding to PS. Here, we use a cholesterol esterification assay and other measures of ER cholesterol delivery to demonstrate that Asters participate in PM-to-ER transport of LDL cholesterol in Chinese hamster ovary cells. Knockout of the gene encoding PTDSS1, the major PS-synthesizing enzyme, lowered LDL-stimulated cholesterol esterification by 85%, whereas knockout of all three Aster genes lowered esterification by 65%. The reduction was even greater (94%) when the genes encoding PTDSS1 and the three Asters were knocked out simultaneously. We conclude that Asters participate in LDL cholesterol delivery from PM to ER, and their action depends in large part, but not exclusively, on PS. The data also indicate that PS participates in another delivery pathway, so far undefined, that is independent of Asters.
Collapse
|
107
|
Lu A. Sorting (Nexin-13) out Novel Insights into Endolysosomal Cholesterol Export. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2022; 5:25152564221114513. [PMID: 37366510 PMCID: PMC10243570 DOI: 10.1177/25152564221114513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 07/21/2022] [Indexed: 06/28/2023]
Abstract
Transport in and out of the endolysosomal compartment represents a key step in the regulation of cellular cholesterol homeostasis. Despite important recent advances, how LDL-derived, free cholesterol is exported from the lumen of endolysosomes to other organelles is still a matter of debate. We recently devised a CRISPR/Cas9 genome-scale strategy to uncover genes involved in the regulation of endolysosomal cholesterol homeostasis and the functionally linked phospholipid, bis(monoacylglycerol)-phosphate. This approach confirmed known genes and pathways involved in this process, and more importantly revealed previously unrecognized roles for new players, such as Sorting Nexin-13 (SNX13). Here we discuss the unexpected regulatory role of SNX13 in endolysosomal cholesterol export.
Collapse
Affiliation(s)
- Albert Lu
- Departament de Biomedicina, Unitat de Biologia Cel·lular,
Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut
d’Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
108
|
Korade Z, Heffer M, Mirnics K. Medication effects on developmental sterol biosynthesis. Mol Psychiatry 2022; 27:490-501. [PMID: 33820938 PMCID: PMC8490477 DOI: 10.1038/s41380-021-01074-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/01/2021] [Accepted: 03/19/2021] [Indexed: 02/01/2023]
Abstract
Cholesterol is essential for normal brain function and development. Genetic disruptions of sterol biosynthesis result in intellectual and developmental disabilities. Developing neurons synthesize their own cholesterol, and disruption of this process can occur by both genetic and chemical mechanisms. Many commonly prescribed medications interfere with sterol biosynthesis, including haloperidol, aripiprazole, cariprazine, fluoxetine, trazodone and amiodarone. When used during pregnancy, these compounds might have detrimental effects on the developing brain of the offspring. In particular, inhibition of dehydrocholesterol-reductase 7 (DHCR7), the last enzyme in the biosynthesis pathway, results in accumulation of the immediate cholesterol precursor, 7-dehydrocholesterol (7-DHC). 7-DHC is highly unstable, giving rise to toxic oxysterols; this is particularly pronounced in a mouse model when both the mother and the offspring carry the Dhcr7+/- genotype. Studies of human dermal fibroblasts from individuals who carry DCHR7+/- single allele mutations suggest that the same gene*medication interaction also occurs in humans. The public health relevance of these findings is high, as DHCR7-inhibitors can be considered teratogens, and are commonly used by pregnant women. In addition, sterol biosynthesis inhibiting medications should be used with caution in individuals with mutations in sterol biosynthesis genes. In an age of precision medicine, further research in this area could open opportunities to improve patient and fetal/infant safety by tailoring medication prescriptions according to patient genotype and life stage.
Collapse
Affiliation(s)
- Zeljka Korade
- Department of Pediatrics, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA, 68198.,Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA, 68198
| | - Marija Heffer
- J. J. Strossmayer University of Osijek, Faculty of Medicine Osijek, Department of Medical Biology and Genetics, Josipa Huttlera 4, 31000 Osijek, Croatia
| | - Károly Mirnics
- Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, 68105, USA.
| |
Collapse
|
109
|
Kinnebrew M, Johnson KA, Radhakrishnan A, Rohatgi R. Measuring and Manipulating Membrane Cholesterol for the Study of Hedgehog Signaling. Methods Mol Biol 2022; 2374:73-87. [PMID: 34562244 PMCID: PMC8819901 DOI: 10.1007/978-1-0716-1701-4_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cholesterol is an abundant lipid in mammalian plasma membranes that regulates the reception of the Hedgehog (Hh) signal in target cells. In vertebrates, cell-surface organelles called primary cilia function as compartments for the propagation of Hh signals. Recent structural, biochemical, and cell-biological studies have led to the model that Patched-1 (PTCH1), the receptor for Hh ligands, uses its transporter-like activity to lower cholesterol accessibility in the membrane surrounding primary cilia. Cholesterol restriction at cilia may represent the long-sought-after mechanism by which PTCH1 inhibits Smoothened (SMO), a cholesterol-responsive transmembrane protein of the G protein-coupled receptor superfamily that transmits the Hh signal across the membrane.Protein probes based on microbial cholesterol-binding proteins revealed that PTCH1 controls only a subset of the total cholesterol molecules, a biochemically defined fraction called accessible cholesterol. The accessible cholesterol pool coexists (and exchanges) with a pool of sequestered cholesterol, which is bound to phospholipids like sphingomyelin. In this chapter, we describe how to measure the accessible and sequestered cholesterol pools in live cells with protein-based probes. We discuss how to purify and fluorescently label these probes for use in flow cytometry and microscopy-based measurements of the cholesterol pools. Additionally, we describe how to modulate accessible cholesterol levels to determine if this pool regulates Hh signaling (or any other cellular process of interest).
Collapse
Affiliation(s)
- Maia Kinnebrew
- Department of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristen A Johnson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rajat Rohatgi
- Department of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
110
|
Chang TY, Chang CCY, Harned TC, De La Torre AL, Lee J, Huynh TN, Gow JG. Blocking cholesterol storage to treat Alzheimer's disease. EXPLORATION OF NEUROPROTECTIVE THERAPY 2021; 1:173-184. [PMID: 35199105 PMCID: PMC8863366 DOI: 10.37349/ent.2021.00014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022]
Abstract
Cholesterol serves as an essential lipid molecule in various membrane organelles of mammalian cells. The metabolites of cholesterol also play important functions. Acyl-coenzyme A: cholesterol acyltransferase 1 (ACAT1), also named as sterol O-acyltransferase 1, is a membrane-bound enzyme residing at the endoplasmic reticulum (ER). It converts cholesterol to cholesteryl esters (CEs) for storage, and is expressed in all cells. CEs cannot partition in membranes; they can only coalesce as cytosolic lipid droplets. Excess CEs are found in the vulnerable region of the brains of patients with late-onset Alzheimer's disease (AD), and in cell and mouse models for AD. Reducing CE contents by genetic inactivation of ACAT1, or by pharmacological inhibition of ACAT is shown to reduce amyloidopathy and other hallmarks for AD. To account for the various beneficial actions of the ACAT1 blockade (A1B), a working hypothesis is proposed here: the increase in CE contents observed in the AD brain is caused by damages of cholesterol-rich lipid rafts that are known to occur in neurons affected by AD. These damages cause cholesterol to release from lipid rafts and move to the ER where it will be converted to CEs by ACAT1. In addition, the increase in CE contents may also be caused by overloading with cholesterol-rich substances, or through activation of ACAT1 gene expression by various proinflammatory agents. Both scenarios may occur in microglia of the chronically inflamed brain. A1B ameliorates AD by diverting the cholesterol pool destined for CE biosynthesis such that it can be utilized more efficiently to repair membrane damage in various organelles, and to exert regulatory actions more effectively to defend against AD. To test the validity of the A1B hypothesis in cell culture and in vivo, the current status of various anti-ACAT1 agents that could be further developed is briefly discussed.
Collapse
Affiliation(s)
- Ta Yuan Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Catherine C. Y. Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Taylor C. Harned
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Adrianna L. De La Torre
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Junghoon Lee
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Thao N. Huynh
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - James G. Gow
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
111
|
Abdulla N, Vincent CT, Kaur M. Mechanistic Insights Delineating the Role of Cholesterol in Epithelial Mesenchymal Transition and Drug Resistance in Cancer. Front Cell Dev Biol 2021; 9:728325. [PMID: 34869315 PMCID: PMC8640133 DOI: 10.3389/fcell.2021.728325] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the significant advancements made in targeted anti-cancer therapy, drug resistance constitutes a multifaceted phenomenon leading to therapy failure and ultimately mortality. Emerging experimental evidence highlight a role of cholesterol metabolism in facilitating drug resistance in cancer. This review aims to describe the role of cholesterol in facilitating multi-drug resistance in cancer. We focus on specific signaling pathways that contribute to drug resistance and the link between these pathways and cholesterol. Additionally, we briefly discuss the molecular mechanisms related to the epithelial-mesenchymal transition (EMT), and the documented link between EMT, metastasis and drug resistance. We illustrate this by specifically focusing on hypoxia and the role it plays in influencing cellular cholesterol content following EMT induction. Finally, we provide a proposed model delineating the crucial role of cholesterol in EMT and discuss whether targeting cholesterol could serve as a novel means of combatting drug resistance in cancer progression and metastasis.
Collapse
Affiliation(s)
- Naaziyah Abdulla
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| | - C Theresa Vincent
- Department of Immunology, Genetics and Pathology, Uppsala, Sweden.,Department of Microbiology, New York University School of Medicine, New York, NY, United States
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
112
|
Barrantes FJ. Fluorescence sensors for imaging membrane lipid domains and cholesterol. CURRENT TOPICS IN MEMBRANES 2021; 88:257-314. [PMID: 34862029 DOI: 10.1016/bs.ctm.2021.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lipid membrane domains are supramolecular lateral heterogeneities of biological membranes. Of nanoscopic dimensions, they constitute specialized hubs used by the cell as transient signaling platforms for a great variety of biologically important mechanisms. Their property to form and dissolve in the bulk lipid bilayer endow them with the ability to engage in highly dynamic processes, and temporarily recruit subpopulations of membrane proteins in reduced nanometric compartments that can coalesce to form larger mesoscale assemblies. Cholesterol is an essential component of these lipid domains; its unique molecular structure is suitable for interacting intricately with crevices and cavities of transmembrane protein surfaces through its rough β face while "talking" to fatty acid acyl chains of glycerophospholipids and sphingolipids via its smooth α face. Progress in the field of membrane domains has been closely associated with innovative improvements in fluorescence microscopy and new fluorescence sensors. These advances enabled the exploration of the biophysical properties of lipids and their supramolecular platforms. Here I review the rationale behind the use of biosensors over the last few decades and their contributions towards elucidation of the in-plane and transbilayer topography of cholesterol-enriched lipid domains and their molecular constituents. The challenges introduced by super-resolution optical microscopy are discussed, as well as possible scenarios for future developments in the field, including virtual ("no staining") staining.
Collapse
Affiliation(s)
- Francisco J Barrantes
- Biomedical Research Institute (BIOMED), Catholic University of Argentina (UCA)-National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
113
|
Varghese FS, Meutiawati F, Teppor M, Jacobs S, de Keyzer C, Taşköprü E, van Woudenbergh E, Overheul GJ, Bouma E, Smit JM, Delang L, Merits A, van Rij RP. Posaconazole inhibits multiple steps of the alphavirus replication cycle. Antiviral Res 2021; 197:105223. [PMID: 34856248 DOI: 10.1016/j.antiviral.2021.105223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/22/2021] [Accepted: 11/27/2021] [Indexed: 11/28/2022]
Abstract
Repurposing drugs is a promising strategy to identify therapeutic interventions against novel and re-emerging viruses. Posaconazole is an antifungal drug used to treat invasive aspergillosis and candidiasis. Recently, posaconazole and its structural analog, itraconazole were shown to inhibit replication of multiple viruses by modifying intracellular cholesterol homeostasis. Here, we show that posaconazole inhibits replication of the alphaviruses Semliki Forest virus (SFV), Sindbis virus and chikungunya virus with EC50 values ranging from 1.4 μM to 9.5 μM. Posaconazole treatment led to a significant reduction of virus entry in an assay using a temperature-sensitive SFV mutant, but time-of-addition and RNA transfection assays indicated that posaconazole also inhibits post-entry stages of the viral replication cycle. Virus replication in the presence of posaconazole was partially rescued by the addition of exogenous cholesterol. A transferrin uptake assay revealed that posaconazole considerably slowed down cellular endocytosis. A single point mutation in the SFV E2 glycoprotein, H255R, provided partial resistance to posaconazole as well as to methyl-β-cyclodextrin, corroborating the effect of posaconazole on cholesterol and viral entry. Our results indicate that posaconazole inhibits multiple steps of the alphavirus replication cycle and broaden the spectrum of viruses that can be targeted in vitro by posaconazole, which could be further explored as a therapeutic agent against emerging viruses.
Collapse
Affiliation(s)
- Finny S Varghese
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Febrina Meutiawati
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mona Teppor
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Sofie Jacobs
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Carolien de Keyzer
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Ezgi Taşköprü
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Esther van Woudenbergh
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands; Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Gijs J Overheul
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ellen Bouma
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jolanda M Smit
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Leen Delang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
114
|
Ikhlef S, Lipp NF, Delfosse V, Fuggetta N, Bourguet W, Magdeleine M, Drin G. Functional analyses of phosphatidylserine/PI(4)P exchangers with diverse lipid species and membrane contexts reveal unanticipated rules on lipid transfer. BMC Biol 2021; 19:248. [PMID: 34801011 PMCID: PMC8606082 DOI: 10.1186/s12915-021-01183-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/04/2021] [Indexed: 11/14/2022] Open
Abstract
Background Lipid species are accurately distributed in the eukaryotic cell so that organelle and plasma membranes have an adequate lipid composition to support numerous cellular functions. In the plasma membrane, a precise regulation of the level of lipids such as phosphatidylserine, PI(4)P, and PI(4,5)P2, is critical for maintaining the signaling competence of the cell. Several lipid transfer proteins of the ORP/Osh family contribute to this fine-tuning by delivering PS, synthesized in the endoplasmic reticulum, to the plasma membrane in exchange for PI(4)P. To get insights into the role of these PS/PI(4)P exchangers in regulating plasma membrane features, we question how they selectively recognize and transfer lipid ligands with different acyl chains, whether these proteins exchange PS exclusively for PI(4)P or additionally for PI(4,5)P2, and how sterol abundance in the plasma membrane impacts their activity. Results We measured in vitro how the yeast Osh6p and human ORP8 transported PS and PI(4)P subspecies of diverse length and unsaturation degree between membranes by fluorescence-based assays. We established that the exchange activity of Osh6p and ORP8 strongly depends on whether these ligands are saturated or not, and is high with representative cellular PS and PI(4)P subspecies. Unexpectedly, we found that the speed at which these proteins individually transfer lipid ligands between membranes is inversely related to their affinity for them and that high-affinity ligands must be exchanged to be transferred more rapidly. Next we determined that Osh6p and ORP8 cannot use PI(4,5)P2 for exchange processes, because it is a low-affinity ligand, and do not transfer more PS into sterol-rich membranes. Conclusions Our study provides new insights into PS/PI(4)P exchangers by indicating the degree to which they can regulate the acyl chain composition of the PM, and how they control PM phosphoinositide levels. Moreover, we establish general rules on how the activity of lipid transfer proteins relates to their affinity for ligands. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01183-1.
Collapse
Affiliation(s)
- Souade Ikhlef
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France
| | - Nicolas-Frédéric Lipp
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France.,Current position: Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Vanessa Delfosse
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Nicolas Fuggetta
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France
| | - William Bourguet
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Maud Magdeleine
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France
| | - Guillaume Drin
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France.
| |
Collapse
|
115
|
Axmann M, Plochberger B, Mikula M, Weber F, Strobl WM, Stangl H. Plasma Membrane Lipids: An Important Binding Site for All Lipoprotein Classes. MEMBRANES 2021; 11:membranes11110882. [PMID: 34832111 PMCID: PMC8622984 DOI: 10.3390/membranes11110882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 12/23/2022]
Abstract
Cholesterol is one of the main constituents of plasma membranes; thus, its supply is of utmost importance. This review covers the known mechanisms of cholesterol transfer from circulating lipoprotein particles to the plasma membrane, and vice versa. To achieve homeostasis, the human body utilizes cellular de novo synthesis and extracellular transport particles for supply of cholesterol and other lipids via the blood stream. These lipoprotein particles can be classified according to their density: chylomicrons, very low, low, and high-density lipoprotein (VLDL, LDL, and HDL, respectively). They deliver and receive their lipid loads, most importantly cholesterol, to and from cells by several redundant routes. Defects in one of these pathways (e.g., due to mutations in receptors) usually are not immediately fatal. Several redundant pathways, at least temporarily, compensate for the loss of one or more of them, but the defects trigger systemic diseases, such as atherosclerosis later on. Recently, intracellular membrane–membrane contact sites were shown to be involved in intracellular cholesterol transfer and the plasma membrane itself has been proposed to act as a binding site for lipoprotein-mediated cargo unloading.
Collapse
Affiliation(s)
- Markus Axmann
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Birgit Plochberger
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Mario Mikula
- Center for Pathobiochemistry and Genetics, Institute for Medical Genetics, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
| | - Florian Weber
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Witta Monika Strobl
- Center for Pathobiochemistry and Genetics, Institute for Medical Chemistry, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
| | - Herbert Stangl
- Center for Pathobiochemistry and Genetics, Institute for Medical Chemistry, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
- Correspondence:
| |
Collapse
|
116
|
Bi J, Khan A, Tang J, Armando AM, Wu S, Zhang W, Gimple RC, Reed A, Jing H, Koga T, Wong ITL, Gu Y, Miki S, Yang H, Prager B, Curtis EJ, Wainwright DA, Furnari FB, Rich JN, Cloughesy TF, Kornblum HI, Quehenberger O, Rzhetsky A, Cravatt BF, Mischel PS. Targeting glioblastoma signaling and metabolism with a re-purposed brain-penetrant drug. Cell Rep 2021; 37:109957. [PMID: 34731610 PMCID: PMC8856626 DOI: 10.1016/j.celrep.2021.109957] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/10/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022] Open
Abstract
The highly lethal brain cancer glioblastoma (GBM) poses a daunting challenge because the blood-brain barrier renders potentially druggable amplified or mutated oncoproteins relatively inaccessible. Here, we identify sphingomyelin phosphodiesterase 1 (SMPD1), an enzyme that regulates the conversion of sphingomyelin to ceramide, as an actionable drug target in GBM. We show that the highly brain-penetrant antidepressant fluoxetine potently inhibits SMPD1 activity, killing GBMs, through inhibition of epidermal growth factor receptor (EGFR) signaling and via activation of lysosomal stress. Combining fluoxetine with temozolomide, a standard of care for GBM, causes massive increases in GBM cell death and complete tumor regression in mice. Incorporation of real-world evidence from electronic medical records from insurance databases reveals significantly increased survival in GBM patients treated with fluoxetine, which was not seen in patients treated with other selective serotonin reuptake inhibitor (SSRI) antidepressants. These results nominate the repurposing of fluoxetine as a potentially safe and promising therapy for patients with GBM and suggest prospective randomized clinical trials.
Collapse
Affiliation(s)
- Junfeng Bi
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA.
| | - Atif Khan
- Department of Medicine, Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Jun Tang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA
| | - Aaron M Armando
- Department of Pharmacology, UCSD School of Medicine, La Jolla, CA, USA
| | - Sihan Wu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA; Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wei Zhang
- Department of Medicine, UCSD School of Medicine, La Jolla, CA, USA
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alex Reed
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Hui Jing
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tomoyuki Koga
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA; Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Ivy Tsz-Lo Wong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA
| | - Yuchao Gu
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Shunichiro Miki
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Huijun Yang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA
| | - Briana Prager
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ellis J Curtis
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA; Department of Medicine, UCSD School of Medicine, La Jolla, CA, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Frank B Furnari
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA; Department of Pathology, UCSD School of Medicine, La Jolla, CA, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA, USA
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, CA, USA
| | - Harley I Kornblum
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen UCLA School of Medicine, Los Angeles, CA, USA
| | | | - Andrey Rzhetsky
- Department of Medicine, Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA; Department of Human Genetics, Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Benjamin F Cravatt
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Paul S Mischel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA.
| |
Collapse
|
117
|
The impact of TiO 2 nanoparticle exposure on transmembrane cholesterol transport and enhanced bacterial infectivity in HeLa cells. Acta Biomater 2021; 135:606-616. [PMID: 34400307 DOI: 10.1016/j.actbio.2021.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 02/04/2023]
Abstract
We have previously shown that exposure to TiO2 nanoparticles (NPs) reduces the resistance of HeLa cells to bacterial infection. Here we demonstrate that the increased infectivity is associated with enhanced asymmetry in the cholesterol distribution. We applied a live cell imaging method which uses tunable orthogonal cholesterol sensors to visualize and quantify in-situ cholesterol distribution between the two leaflets of the plasma membrane (PM). In the control culture, we found marked transbilayer asymmetry of cholesterol, with the concentration in the outer plasma membrane (OPM) being 13 ± 2-fold higher than that in the inner plasma membrane (IPM). Exposure of the culture to 0.1 mg/mL of rutile TiO2 NPs increased the asymmetry such that the concentration in the OPM was 51 ± 10 times higher, while the total cholesterol content increased only 21 ± 2%. This change in cholesterol gradient may explain the increase in bacterial infectivity in HeLa cells exposed to TiO2 NPs since many pathogens, including Staphylococcus aureus used in the present study, require cholesterol for proper membrane attachment and virulence. RT-PCR indicated that exposure to TiO2 was responsible for upregulation of the ABCA1 and ABCG1 mRNAs, which are responsible for the production of the cholesterol transporter proteins that facilitate cholesterol transport across cellular membranes. This was confirmed by the observation of an overall decrease in bacterial infection in ABCA1 knockout or methyl-β-cyclodextrin-treated HeLa cells, as regardless of TiO2 NP exposure. Hence rather than preventing bacterial infection, TiO2 nanoparticles upregulate genes associated with membrane cholesterol production and distribution, hence increasing infectivity. STATEMENT OF SIGNIFICANCE: A great deal of work has been done regarding the toxicology of the particles, especially focusing on detrimental outcomes associated with reactive oxygen species (ROS) production. In this paper we show unambiguously a very surprising result, namely the ability of these particles to enhance bacterial infection even at very small exposure levels, where none of the deleterious effects of ROS products can yet be detected. Using a new imaging technique, we are able to demonstrate, in operando, the effect of the particles on cholesterol generation and distribution in live HeLa cells. This paper also represents the first in a series where we explore other consequences of increased membrane cholesterol, due to particle exposure, which are known to have multiple other consequences on human tissue function and development.
Collapse
|
118
|
Jurášek M, Valečka J, Novotný I, Kejík Z, Fähnrich J, Marešová A, Tauchen J, Bartůněk P, Dolenský B, Jakubek M, Drašar PB, Králová J. Synthesis and biological evaluation of cationic TopFluor cholesterol analogues. Bioorg Chem 2021; 117:105410. [PMID: 34700109 DOI: 10.1016/j.bioorg.2021.105410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/25/2021] [Accepted: 10/03/2021] [Indexed: 12/17/2022]
Abstract
Cholesterol is not only a major component of the cell membrane, but also plays an important role in a wide range of biological processes and pathologies. It is therefore crucial to develop appropriate tools for visualizing intracellular cholesterol transport. Here, we describe new cationic analogues of BODIPY-Cholesterol (TopFluor-Cholesterol, TF-Chol), which combine a positive charge on the sterol side chain and a BODIPY group connected via a C-4 linker. In contrast to TF-Chol, the new analogues TF-1 and TF-3 possessing acetyl groups on the A ring (C-3 position on steroid) internalized much faster and displayed slightly different levels of intracellular localization. Their applicability for cholesterol monitoring was indicated by the fact that they strongly label compartments with accumulated cholesterol in cells carrying a mutation of the Niemann-Pick disease-associated cholesterol transporter, NPC1.
Collapse
Affiliation(s)
- Michal Jurášek
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jan Valečka
- Light microscopy core facility, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Ivan Novotný
- Light microscopy core facility, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Zdeněk Kejík
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jan Fähnrich
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Anna Marešová
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jan Tauchen
- Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague 6, Czech Republic
| | - Petr Bartůněk
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Bohumil Dolenský
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Milan Jakubek
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Pavel B Drašar
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jarmila Králová
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vídeňská 1083, 142 20 Prague 4, Czech Republic.
| |
Collapse
|
119
|
Griffiths WJ, Wang Y. Sterols, Oxysterols, and Accessible Cholesterol: Signalling for Homeostasis, in Immunity and During Development. Front Physiol 2021; 12:723224. [PMID: 34690800 PMCID: PMC8531217 DOI: 10.3389/fphys.2021.723224] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/10/2021] [Indexed: 12/14/2022] Open
Abstract
In this article we discuss the concept of accessible plasma membrane cholesterol and its involvement as a signalling molecule. Changes in plasma membrane accessible cholesterol, although only being minor in the context of total cholesterol plasma membrane cholesterol and total cell cholesterol, are a key regulator of overall cellular cholesterol homeostasis by the SREBP pathway. Accessible cholesterol also provides the second messenger between patched 1 and smoothened in the hedgehog signalling pathway important during development, and its depletion may provide a mechanism of resistance to microbial pathogens including SARS-CoV-2. We revise the hypothesis that oxysterols are a signalling form of cholesterol, in this instance as a rapidly acting and paracrine version of accessible cholesterol.
Collapse
Affiliation(s)
| | - Yuqin Wang
- Swansea University Medical School, Swansea, United Kingdom
| |
Collapse
|
120
|
Kinnebrew M, Luchetti G, Sircar R, Frigui S, Viti LV, Naito T, Beckert F, Saheki Y, Siebold C, Radhakrishnan A, Rohatgi R. Patched 1 reduces the accessibility of cholesterol in the outer leaflet of membranes. eLife 2021; 10:e70504. [PMID: 34698632 PMCID: PMC8654371 DOI: 10.7554/elife.70504] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
A long-standing mystery in vertebrate Hedgehog signaling is how Patched 1 (PTCH1), the receptor for Hedgehog ligands, inhibits the activity of Smoothened, the protein that transmits the signal across the membrane. We previously proposed (Kinnebrew et al., 2019) that PTCH1 inhibits Smoothened by depleting accessible cholesterol from the ciliary membrane. Using a new imaging-based assay to directly measure the transport activity of PTCH1, we find that PTCH1 depletes accessible cholesterol from the outer leaflet of the plasma membrane. This transport activity is terminated by binding of Hedgehog ligands to PTCH1 or by dissipation of the transmembrane potassium gradient. These results point to the unexpected model that PTCH1 moves cholesterol from the outer to the inner leaflet of the membrane in exchange for potassium ion export in the opposite direction. Our study provides a plausible solution for how PTCH1 inhibits SMO by changing the organization of cholesterol in membranes and establishes a general framework for studying how proteins change cholesterol accessibility to regulate membrane-dependent processes in cells.
Collapse
Affiliation(s)
- Maia Kinnebrew
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| | - Giovanni Luchetti
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
- Department of Physiological Chemistry, GenentechSouth San FranciscoUnited States
| | - Ria Sircar
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| | - Sara Frigui
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| | - Lucrezia Vittoria Viti
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Tomoki Naito
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
| | - Francis Beckert
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Rajat Rohatgi
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
121
|
Ormsby TJR, Owens SE, Horlock AD, Davies D, Griffiths WJ, Wang Y, Cronin JG, Bromfield JJ, Sheldon IM. Oxysterols protect bovine endometrial cells against pore-forming toxins from pathogenic bacteria. FASEB J 2021; 35:e21889. [PMID: 34569656 PMCID: PMC9272411 DOI: 10.1096/fj.202100036r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/16/2021] [Accepted: 08/16/2021] [Indexed: 11/11/2022]
Abstract
Many species of pathogenic bacteria secrete toxins that form pores in mammalian cell membranes. These membrane pores enable the delivery of virulence factors into cells, result in the leakage of molecules that bacteria can use as nutrients, and facilitate pathogen invasion. Inflammatory responses to bacteria are regulated by the side-chain-hydroxycholesterols 27-hydroxycholesterol and 25-hydroxycholesterol, but their effect on the intrinsic protection of cells against pore-forming toxins is unclear. Here, we tested the hypothesis that 27-hydroxycholesterol and 25-hydroxycholesterol help protect cells against pore-forming toxins. We treated bovine endometrial epithelial and stromal cells with 27-hydroxycholesterol or 25-hydroxycholesterol, and then challenged the cells with pyolysin, which is a cholesterol-dependent cytolysin from Trueperella pyogenes that targets these endometrial cells. We found that treatment with 27-hydroxycholesterol or 25-hydroxycholesterol protected both epithelial and stomal cells against pore formation and the damage caused by pyolysin. The oxysterols limited pyolysin-induced leakage of potassium and lactate dehydrogenase from cells, and reduced cytoskeletal changes and cytolysis. This oxysterol cytoprotection against pyolysin was partially dependent on reducing cytolysin-accessible cholesterol in the cell membrane and on activating liver X receptors. Treatment with 27-hydroxycholesterol also protected the endometrial cells against Staphylococcus aureus α-hemolysin. Using mass spectrometry, we found 27-hydroxycholesterol and 25-hydroxycholesterol in uterine and follicular fluid. Furthermore, epithelial cells released additional 25-hydroxycholesterol in response to pyolysin. In conclusion, both 27-hydroxycholesterol and 25-hydroxycholesterol increased the intrinsic protection of bovine endometrial cells against pore-forming toxins. Our findings imply that side-chain-hydroxycholesterols may help defend the endometrium against pathogenic bacteria.
Collapse
Affiliation(s)
| | - Sian E Owens
- Swansea University Medical School, Swansea University, Swansea, UK
| | | | - Daphne Davies
- Swansea University Medical School, Swansea University, Swansea, UK
| | | | - Yuqin Wang
- Swansea University Medical School, Swansea University, Swansea, UK
| | - James G Cronin
- Swansea University Medical School, Swansea University, Swansea, UK
| | - John J Bromfield
- Department of Animal Sciences, University of Florida, Gainesville, Florida, USA
| | - Iain M Sheldon
- Swansea University Medical School, Swansea University, Swansea, UK
| |
Collapse
|
122
|
Steck TL, Tabei SMA, Lange Y. A basic model for cell cholesterol homeostasis. Traffic 2021; 22:471-481. [PMID: 34528339 DOI: 10.1111/tra.12816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/26/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022]
Abstract
Cells manage their cholesterol by negative feedback using a battery of sterol-responsive proteins. How these activities are coordinated so as to specify the abundance and distribution of the sterol is unclear. We present a simple mathematical model that addresses this question. It assumes that almost all of the cholesterol is associated with phospholipids in stoichiometric complexes. A small fraction of the sterol is uncomplexed and thermodynamically active. It equilibrates among the organelles, setting their sterol level according to the affinity of their phospholipids. The activity of the homeostatic proteins in the cytoplasmic membranes is then set by their fractional saturation with uncomplexed cholesterol in competition with the phospholipids. The high-affinity phospholipids in the plasma membrane (PM) are filled to near stoichiometric equivalence, giving it most of the cell sterol. Notably, the affinity of the phospholipids in the endomembranes (EMs) is lower by orders of magnitude than that of the phospholipids in the PM. Thus, the small amount of sterol in the EMs rests far below stoichiometric capacity. Simulations match a variety of experimental data. The model captures the essence of cell cholesterol homeostasis, makes coherent a diverse set of experimental findings, provides a surprising prediction and suggests new experiments.
Collapse
Affiliation(s)
- Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| | - S M Ali Tabei
- Department of Physics, University of Northern Iowa, Cedar Falls, Iowa, USA
| | - Yvonne Lange
- Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
123
|
Segawa K, Kikuchi A, Noji T, Sugiura Y, Hiraga K, Suzuki C, Haginoya K, Kobayashi Y, Matsunaga M, Ochiai Y, Yamada K, Nishimura T, Iwasawa S, Shoji W, Sugihara F, Nishino K, Kosako H, Ikawa M, Uchiyama Y, Suematsu M, Ishikita H, Kure S, Nagata S. A sublethal ATP11A mutation associated with neurological deterioration causes aberrant phosphatidylcholine flipping in plasma membranes. J Clin Invest 2021; 131:e148005. [PMID: 34403372 DOI: 10.1172/jci148005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 08/05/2021] [Indexed: 01/01/2023] Open
Abstract
ATP11A translocates phosphatidylserine (PtdSer), but not phosphatidylcholine (PtdCho), from the outer to the inner leaflet of plasma membranes, thereby maintaining the asymmetric distribution of PtdSer. Here, we detected a de novo heterozygous point mutation of ATP11A in a patient with developmental delays and neurological deterioration. Mice carrying the corresponding mutation died perinatally of neurological disorders. This mutation caused an amino acid substitution (Q84E) in the first transmembrane segment of ATP11A, and mutant ATP11A flipped PtdCho. Molecular dynamics simulations revealed that the mutation allowed PtdCho binding at the substrate entry site. Aberrant PtdCho flipping markedly decreased the concentration of PtdCho in the outer leaflet of plasma membranes, whereas sphingomyelin (SM) concentrations in the outer leaflet increased. This change in the distribution of phospholipids altered cell characteristics, including cell growth, cholesterol homeostasis, and sensitivity to sphingomyelinase. Matrix-assisted laser desorption ionization-imaging mass spectrometry (MALDI-IMS) showed a marked increase of SM levels in the brains of Q84E-knockin mouse embryos. These results provide insights into the physiological importance of the substrate specificity of plasma membrane flippases for the proper distribution of PtdCho and SM.
Collapse
Affiliation(s)
- Katsumori Segawa
- Laboratory of Biochemistry and Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Tomoyasu Noji
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Keita Hiraga
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Chigure Suzuki
- Department of Cellular and Molecular Pharmacology and.,Department of Cellular and Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhiro Haginoya
- Department of Pediatric Neurology, Takuto Rehabilitation Center for Children, Sendai, Miyagi, Japan.,Department of Pediatric Neurology, Miyagi Children's Hospital, Sendai, Miyagi, Japan
| | - Yasuko Kobayashi
- Department of Pediatric Neurology, Takuto Rehabilitation Center for Children, Sendai, Miyagi, Japan.,Department of Pediatrics, National Hospital Organization Sendai-Nishitaga Hospital, Sendai, Miyagi, Japan
| | - Mitsuhiro Matsunaga
- Laboratory of Biochemistry and Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yuki Ochiai
- Laboratory of Biochemistry and Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Kyoko Yamada
- Laboratory of Biochemistry and Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Takuo Nishimura
- Laboratory of Biochemistry and Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Shinya Iwasawa
- Department of Pediatrics, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Wataru Shoji
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Fuminori Sugihara
- Central Instrumentation Laboratory, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kohei Nishino
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Pharmacology and.,Department of Cellular and Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Ishikita
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shigeo Kure
- Department of Pediatrics, Tohoku University School of Medicine, Sendai, Miyagi, Japan.,Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
124
|
Bassot A, Prip-Buus C, Alves A, Berdeaux O, Perrier J, Lenoir V, Ji-Cao J, Berger MA, Loizon E, Cabaret S, Panthu B, Rieusset J, Morio B. Loss and gain of function of Grp75 or mitofusin 2 distinctly alter cholesterol metabolism, but all promote triglyceride accumulation in hepatocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159030. [PMID: 34419589 DOI: 10.1016/j.bbalip.2021.159030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022]
Abstract
In the liver, contact sites between the endoplasmic reticulum (ER) and mitochondria (named MAMs) may be crucial hubs for the regulation of lipid metabolism, thus contributing to the exacerbation or prevention of fatty liver. We hypothesized that tether proteins located at MAMs could play a key role in preventing triglyceride accumulation in hepatocytes and nonalcoholic fatty liver disease (NAFLD) occurrence. To test this, we explored the role of two key partners in building MAM integrity and functionality, the glucose-regulated protein 75 (Grp75) and mitofusin 2 (Mfn2), which liver contents are altered in obesity and NAFLD. Grp75 or Mfn2 expression was either silenced using siRNA or overexpressed with adenoviruses in Huh7 cells. Silencing of Grp75 and Mfn2 resulted in decreased ER-mitochondria interactions, mitochondrial network fusion state and mitochondrial oxidative capacity, while overexpression of the two proteins induced mirror impacts on these parameters. Furthermore, Grp75 or Mfn2 silencing decreased cellular cholesterol content and enhanced triglyceride secretion in ApoB100 lipoproteins, while their overexpression led to reverse effects. Cellular phosphatidylcholine/phosphatidylethanolamine ratio was decreased only upon overexpression of the proteins, potentially contributing to altered ApoB100 assembly and secretion. Despite the opposite differences, both silencing and overexpression of Grp75 or Mfn2 induced triglyceride storage, although a fatty acid challenge was required to express the alteration upon protein silencing. Among the mechanisms potentially involved in this phenotype, ER stress was closely associated with altered triglyceride metabolism after Grp75 or Mfn2 overexpression, while blunted mitochondrial FA oxidation capacity may be the main defect causing triglyceride accumulation upon Grp75 or Mfn2 silencing. Further studies are required to decipher the link between modulation of Grp75 or Mfn2 expression, change in MAM integrity and alteration of cholesterol content of the cell. In conclusion, Grp75 or Mfn2 silencing and overexpression in Huh7 cells contribute to altering MAM integrity and cholesterol storage in opposite directions, but all promote triglyceride accumulation through distinct cellular pathways. This study also highlights that besides Mfn2, Grp75 could play a central role in hepatic lipid and cholesterol metabolism in obesity and NAFLD.
Collapse
Affiliation(s)
- Arthur Bassot
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Carina Prip-Buus
- Institut Cochin, Département d'Endocrinologie, Métabolisme et Diabète, INSERM U1016/CNRS UMR8104/Université de Paris, 75014 Paris, France.
| | - Anaïs Alves
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Olivier Berdeaux
- ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Université Bourgogne Franche-Comté, Agrosup Dijon, F-21000 Dijon, France.
| | - Johan Perrier
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Véronique Lenoir
- Institut Cochin, Département d'Endocrinologie, Métabolisme et Diabète, INSERM U1016/CNRS UMR8104/Université de Paris, 75014 Paris, France.
| | - Jingwei Ji-Cao
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Marie-Agnès Berger
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Emmanuelle Loizon
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Stephanie Cabaret
- ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Université Bourgogne Franche-Comté, Agrosup Dijon, F-21000 Dijon, France.
| | - Baptiste Panthu
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Jennifer Rieusset
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Béatrice Morio
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| |
Collapse
|
125
|
Henne WM. How NPC1 Loss Twists the TORCque of Lysosomes. Dev Cell 2021; 56:251-252. [PMID: 33561418 DOI: 10.1016/j.devcel.2021.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Niemann-Pick type C is a neurological disorder caused by mutations in the lysosome cholesterol exporter NPC1. In this issue of Developmental Cell, Davis et al. dissect how NPC1 loss elevates mTORC1 signaling, and demonstrate that suppression of mTORC1 signaling can rescue disease pathology in NPC1-deficient cell models.
Collapse
Affiliation(s)
- W Mike Henne
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
126
|
Naito T, Saheki Y. GRAMD1-mediated accessible cholesterol sensing and transport. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158957. [PMID: 33932585 DOI: 10.1016/j.bbalip.2021.158957] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 01/19/2023]
Abstract
Cholesterol, an essential lipid for cell signaling and structural integrity of cellular membranes, is highly enriched in the plasma membrane (PM). However, the regulatory mechanisms that control its biosynthesis and uptake both reside in the endoplasmic reticulum (ER). Thus, the ER needs to constantly monitor the levels of PM cholesterol. This is in part mediated by regulated transport of a biochemically defined pool of cholesterol, termed "accessible" cholesterol, from the PM to the ER via evolutionarily conserved ER-anchored lipid transfer proteins, the GRAMD1s/Asters (GRAMD1a/1b/1c) (Lam/Ltc proteins in yeast). GRAMD1s possess cytosolically exposed GRAM domain and StART-like domain followed by a transmembrane ER anchor. They form homo- and hetero-meric complexes and move to the contacts formed between the ER and the PM by sensing a transient expansion of the accessible pool of cholesterol in the PM via the GRAM domain and facilitate its extraction and transport to the ER via the StART-like domain. The GRAMD1b GRAM domain possesses distinct, but synergistic sites, for recognizing accessible cholesterol and anionic lipids, including phosphatidylserine, within the PM. This property of the GRAM domain contributes to regulated tethering of the PM to ER membrane where GRAMD1s are anchored and fine-tunes StART-like domain-dependent accessible cholesterol transport. Thus, cells use GRAMD1s to sense the levels of cholesterol in the PM and regulate transport of accessible PM cholesterol to the ER in order to maintain cholesterol homeostasis.
Collapse
Affiliation(s)
- Tomoki Naito
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore; Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan.
| |
Collapse
|
127
|
Ristovski M, Farhat D, Bancud SEM, Lee JY. Lipid Transporters Beam Signals from Cell Membranes. MEMBRANES 2021; 11:562. [PMID: 34436325 PMCID: PMC8399137 DOI: 10.3390/membranes11080562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022]
Abstract
Lipid composition in cellular membranes plays an important role in maintaining the structural integrity of cells and in regulating cellular signaling that controls functions of both membrane-anchored and cytoplasmic proteins. ATP-dependent ABC and P4-ATPase lipid transporters, two integral membrane proteins, are known to contribute to lipid translocation across the lipid bilayers on the cellular membranes. In this review, we will highlight current knowledge about the role of cholesterol and phospholipids of cellular membranes in regulating cell signaling and how lipid transporters participate this process.
Collapse
Affiliation(s)
- Miliça Ristovski
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.R.); (D.F.); (S.E.M.B.)
- Translational and Molecular Medicine Program, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Danny Farhat
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.R.); (D.F.); (S.E.M.B.)
- Biomedical Sciences Program, Faculty of Science, University of Ottawa, Ottawa, ON K1H 6N5, Canada
| | - Shelly Ellaine M. Bancud
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.R.); (D.F.); (S.E.M.B.)
- Translational and Molecular Medicine Program, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jyh-Yeuan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.R.); (D.F.); (S.E.M.B.)
| |
Collapse
|
128
|
Pospiech M, Owens SE, Miller DJ, Austin-Muttitt K, Mullins JGL, Cronin JG, Allemann RK, Sheldon IM. Bisphosphonate inhibitors of squalene synthase protect cells against cholesterol-dependent cytolysins. FASEB J 2021; 35:e21640. [PMID: 33991130 DOI: 10.1096/fj.202100164r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 01/29/2023]
Abstract
Certain species of pathogenic bacteria damage tissues by secreting cholesterol-dependent cytolysins, which form pores in the plasma membranes of animal cells. However, reducing cholesterol protects cells against these cytolysins. As the first committed step of cholesterol biosynthesis is catalyzed by squalene synthase, we explored whether inhibiting this enzyme protected cells against cholesterol-dependent cytolysins. We first synthesized 22 different nitrogen-containing bisphosphonate molecules that were designed to inhibit squalene synthase. Squalene synthase inhibition was quantified using a cell-free enzyme assay, and validated by computer modeling of bisphosphonate molecules binding to squalene synthase. The bisphosphonates were then screened for their ability to protect HeLa cells against the damage caused by the cholesterol-dependent cytolysin, pyolysin. The most effective bisphosphonate reduced pyolysin-induced leakage of lactate dehydrogenase into cell supernatants by >80%, and reduced pyolysin-induced cytolysis from >75% to <25%. In addition, this bisphosphonate reduced pyolysin-induced leakage of potassium from cells, limited changes in the cytoskeleton, prevented mitogen-activated protein kinases cell stress responses, and reduced cellular cholesterol. The bisphosphonate also protected cells against another cholesterol-dependent cytolysin, streptolysin O, and protected lung epithelial cells and primary dermal fibroblasts against cytolysis. Our findings imply that treatment with bisphosphonates that inhibit squalene synthase might help protect tissues against pathogenic bacteria that secrete cholesterol-dependent cytolysins.
Collapse
Affiliation(s)
- Mateusz Pospiech
- Swansea University Medical School, Swansea University, Swansea, UK
| | - Siân E Owens
- Swansea University Medical School, Swansea University, Swansea, UK
| | | | | | | | - James G Cronin
- Swansea University Medical School, Swansea University, Swansea, UK
| | | | - I Martin Sheldon
- Swansea University Medical School, Swansea University, Swansea, UK
| |
Collapse
|
129
|
Induction of microRNA hsa-let-7d-5p, and repression of HMGA2, contribute protection against lipid accumulation in macrophage 'foam' cells. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159005. [PMID: 34274506 DOI: 10.1016/j.bbalip.2021.159005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022]
Abstract
Accumulation of excess cholesterol and cholesteryl ester in macrophage 'foam' cells within the arterial intima characterises early 'fatty streak' atherosclerotic lesions, and is accompanied by epigenetic changes, including altered expression of microRNA sequences which determine of gene and protein expression. This study established that exposure to lipoproteins, including acetylated LDL, induced macrophage expression of microRNA hsa-let-7d-5p, a sequence previously linked with tumour suppression, and repressed expression of one of its target genes, high mobility group AT hook 2 (HMGA2). A let-7d-5p mimic repressed expression of HMGA2 (18%; p < 0.05) while a marked increase (2.9-fold; p < 0.05) in expression of HMGA2 was noted in the presence of let-7d-5p inhibitor. Under these conditions, let-7d-5p mimic significantly (p < 0.05) decreased total (10%), free (8%) and cholesteryl ester (21%) mass, while the inhibitor significantly (p < 0.05) increased total (29%) and free cholesterol (29%) mass, compared with the relevant controls. Let-7d-5p inhibition significantly (p < 0.05) increased endogenous biosynthesis of cholesterol (38%) and cholesteryl ester (39%) pools in macrophage 'foam' cells, without altering the cholesterol efflux pathway, or esterification of exogenous radiolabelled oleate. Let-7d-5p inhibition in sterol-loaded cells increased the level of HMGA2 protein (32%; p < 0.05), while SiRNA knockdown of this protein (29%; p < 0.05) resulted in a (21%, p < 0.05) reduction in free cholesterol mass. Thus, induction of let-7d-5p, and repression of its target HMGA2, in macrophages is a protective response to the challenge of increased cholesterol influx into these cells; dysregulation of this response may contribute to atherosclerosis and other disorders such as cancer.
Collapse
|
130
|
Takahashi K, Kanerva K, Vanharanta L, Almeida‐Souza L, Lietha D, Olkkonen VM, Ikonen E. ORP2 couples LDL-cholesterol transport to FAK activation by endosomal cholesterol/PI(4,5)P 2 exchange. EMBO J 2021; 40:e106871. [PMID: 34124795 PMCID: PMC8281050 DOI: 10.15252/embj.2020106871] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
Low-density lipoprotein (LDL)-cholesterol delivery from late endosomes to the plasma membrane regulates focal adhesion dynamics and cell migration, but the mechanisms controlling it are poorly characterized. Here, we employed auxin-inducible rapid degradation of oxysterol-binding protein-related protein 2 (ORP2/OSBPL2) to show that endogenous ORP2 mediates the transfer of LDL-derived cholesterol from late endosomes to focal adhesion kinase (FAK)-/integrin-positive recycling endosomes in human cells. In vitro, cholesterol enhances membrane association of FAK to PI(4,5)P2 -containing lipid bilayers. In cells, ORP2 stimulates FAK activation and PI(4,5)P2 generation in endomembranes, enhancing cell adhesion. Moreover, ORP2 increases PI(4,5)P2 in NPC1-containing late endosomes in a FAK-dependent manner, controlling their tubulovesicular trafficking. Together, these results provide evidence that ORP2 controls FAK activation and LDL-cholesterol plasma membrane delivery by promoting bidirectional cholesterol/PI(4,5)P2 exchange between late and recycling endosomes.
Collapse
Affiliation(s)
- Kohta Takahashi
- Department of Anatomy and Stem Cells and Metabolism Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
- Present address:
Laboratory of Microbiology and ImmunologyGraduate School of Pharmaceutical SciencesChiba UniversityChibaJapan
| | - Kristiina Kanerva
- Department of Anatomy and Stem Cells and Metabolism Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Lauri Vanharanta
- Department of Anatomy and Stem Cells and Metabolism Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Leonardo Almeida‐Souza
- Helsinki Institute of Life Science, HiLIFEUniversity of HelsinkiHelsinkiFinland
- Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| | - Daniel Lietha
- Centro de Investigaciones Biológicas Margarita Salas (CIB)Spanish National Research Council (CSIC)MadridSpain
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| |
Collapse
|
131
|
Mollinedo F, Gajate C. Mitochondrial Targeting Involving Cholesterol-Rich Lipid Rafts in the Mechanism of Action of the Antitumor Ether Lipid and Alkylphospholipid Analog Edelfosine. Pharmaceutics 2021; 13:763. [PMID: 34065546 PMCID: PMC8161315 DOI: 10.3390/pharmaceutics13050763] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
The ether lipid edelfosine induces apoptosis selectively in tumor cells and is the prototypic molecule of a family of synthetic antitumor compounds collectively known as alkylphospholipid analogs. Cumulative evidence shows that edelfosine interacts with cholesterol-rich lipid rafts, endoplasmic reticulum (ER) and mitochondria. Edelfosine induces apoptosis in a number of hematological cancer cells by recruiting death receptors and downstream apoptotic signaling into lipid rafts, whereas it promotes apoptosis in solid tumor cells through an ER stress response. Edelfosine-induced apoptosis, mediated by lipid rafts and/or ER, requires the involvement of a mitochondrial-dependent step to eventually elicit cell death, leading to the loss of mitochondrial membrane potential, cytochrome c release and the triggering of cell death. The overexpression of Bcl-2 or Bcl-xL blocks edelfosine-induced apoptosis. Edelfosine induces the redistribution of lipid rafts from the plasma membrane to the mitochondria. The pro-apoptotic action of edelfosine on cancer cells is associated with the recruitment of F1FO-ATP synthase into cholesterol-rich lipid rafts. Specific inhibition of the FO sector of the F1FO-ATP synthase, which contains the membrane-embedded c-subunit ring that constitutes the mitochondrial permeability transcription pore, hinders edelfosine-induced cell death. Taking together, the evidence shown here suggests that the ether lipid edelfosine could modulate cell death in cancer cells by direct interaction with mitochondria, and the reorganization of raft-located mitochondrial proteins that critically modulate cell death or survival. Here, we summarize and discuss the involvement of mitochondria in the antitumor action of the ether lipid edelfosine, pointing out the mitochondrial targeting of this drug as a major therapeutic approach, which can be extrapolated to other alkylphospholipid analogs. We also discuss the involvement of cholesterol transport and cholesterol-rich lipid rafts in the interactions between the organelles as well as in the role of mitochondria in the regulation of apoptosis in cancer cells and cancer therapy.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, C/Ramiro de Maeztu 9, E-28040 Madrid, Spain;
| | | |
Collapse
|
132
|
Selective Aster inhibitors distinguish vesicular and nonvesicular sterol transport mechanisms. Proc Natl Acad Sci U S A 2021; 118:2024149118. [PMID: 33376205 DOI: 10.1073/pnas.2024149118] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Aster proteins (encoded by the Gramd1a-c genes) contain a ligand-binding fold structurally similar to a START domain and mediate nonvesicular plasma membrane (PM) to endoplasmic reticulum (ER) cholesterol transport. In an effort to develop small molecule modulators of Asters, we identified 20α-hydroxycholesterol (HC) and U18666A as lead compounds. Unfortunately, both 20α-HC and U18666A target other sterol homeostatic proteins, limiting their utility. 20α-HC inhibits sterol regulatory element-binding protein 2 (SREBP2) processing, and U18666A is an inhibitor of the vesicular trafficking protein Niemann-Pick C1 (NPC1). To develop potent and selective Aster inhibitors, we synthesized a series of compounds by modifying 20α-HC and U18666A. Among these, AI (Aster inhibitor)-1l, which has a longer side chain than 20α-HC, selectively bound to Aster-C. The crystal structure of Aster-C in complex with AI-1l suggests that sequence and flexibility differences in the loop that gates the binding cavity may account for the ligand specificity for Aster C. We further identified the U18666A analog AI-3d as a potent inhibitor of all three Aster proteins. AI-3d blocks the ability of Asters to bind and transfer cholesterol in vitro and in cells. Importantly, AI-3d also inhibits the movement of low-density lipoprotein (LDL) cholesterol to the ER, although AI-3d does not block NPC1. This finding positions the nonvesicular Aster pathway downstream of NPC1-dependent vesicular transport in the movement of LDL cholesterol to the ER. Selective Aster inhibitors represent useful chemical tools to distinguish vesicular and nonvesicular sterol transport mechanisms in mammalian cells.
Collapse
|
133
|
Buwaneka P, Ralko A, Liu SL, Cho W. Evaluation of the available cholesterol concentration in the inner leaflet of the plasma membrane of mammalian cells. J Lipid Res 2021; 62:100084. [PMID: 33964305 PMCID: PMC8178126 DOI: 10.1016/j.jlr.2021.100084] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
Cholesterol is an essential component of the mammalian plasma membrane involved in diverse cellular processes. Our recent quantitative imaging analysis using ratiometric cholesterol sensors showed that the available cholesterol concentration in the inner leaflet of the plasma membrane (IPM) is low in unstimulated cells and increased in a stimulus-specific manner to trigger cell signaling events. However, the transbilayer distribution of cholesterol in the plasma membrane of mammalian cells remains controversial. Here we report a systematic and rigorous evaluation of basal IPM cholesterol levels in a wide range of mammalian cells with different properties employing cholesterol sensors derived from the D4 domain of the Perfringolysin O toxin and a sterol-transfer protein, Osh4. Results consistently showed that, although basal IPM cholesterol levels vary significantly among cells, they remain significantly lower than cholesterol levels in the outer leaflets. We found that IPM cholesterol levels were particularly low in all tested primary cells. These results support the universality of the low basal IPM cholesterol concentration under physiological conditions. We also report here the presence of sequestered IPM cholesterol pools, which may become available to cytosolic proteins under certain physiological conditions. We hypothesize that these pools may partly account for the low basal level of available IPM cholesterol. In conclusion, we provide new experimental data that confirm the asymmetric transbilayer distribution of the plasma membrane cholesterol, which may contribute to regulation of various cellular signaling processes at the plasma membrane.
Collapse
Affiliation(s)
- Pawanthi Buwaneka
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Arthur Ralko
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Shu-Lin Liu
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Wonhwa Cho
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
134
|
Ikonen E, Zhou X. Cholesterol transport between cellular membranes: A balancing act between interconnected lipid fluxes. Dev Cell 2021; 56:1430-1436. [PMID: 34004151 DOI: 10.1016/j.devcel.2021.04.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022]
Abstract
Cholesterol represents the most abundant single lipid in mammalian cells. How its asymmetric distribution between subcellular membranes is achieved and maintained attracts considerable interest. One of the challenges is that cholesterol rarely is transported alone, but rather is coupled with heterotypic transport and metabolism of other lipids, in particular phosphoinositides, phosphatidylserine, and sphingolipids. This perspective summarizes the major exo- and endocytic cholesterol transport routes and how lipid transfer proteins at membrane contacts and membrane transport intersect along these routes. It discusses the co-transport of cholesterol with other lipids in mammalian cells and reviews emerging evidence related to the physiological relevance of this process.
Collapse
Affiliation(s)
- Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Xin Zhou
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| |
Collapse
|
135
|
Li YE, Wang Y, Du X, Zhang T, Mak HY, Hancock SE, McEwen H, Pandzic E, Whan RM, Aw YC, Lukmantara IE, Yuan Y, Dong X, Don A, Turner N, Qi S, Yang H. TMEM41B and VMP1 are scramblases and regulate the distribution of cholesterol and phosphatidylserine. J Cell Biol 2021; 220:212020. [PMID: 33929485 PMCID: PMC8077175 DOI: 10.1083/jcb.202103105] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023] Open
Abstract
TMEM41B and VMP1 are integral membrane proteins of the endoplasmic reticulum (ER) and regulate the formation of autophagosomes, lipid droplets (LDs), and lipoproteins. Recently, TMEM41B was identified as a crucial host factor for infection by all coronaviruses and flaviviruses. The molecular function of TMEM41B and VMP1, which belong to a large evolutionarily conserved family, remains elusive. Here, we show that TMEM41B and VMP1 are phospholipid scramblases whose deficiency impairs the normal cellular distribution of cholesterol and phosphatidylserine. Their mechanism of action on LD formation is likely to be different from that of seipin. Their role in maintaining cellular phosphatidylserine and cholesterol homeostasis may partially explain their requirement for viral infection. Our results suggest that the proper sorting and distribution of cellular lipids are essential for organelle biogenesis and viral infection.
Collapse
Affiliation(s)
- Yang Emma Li
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Yichang Wang
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China
| | - Ximing Du
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Tizhong Zhang
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China
| | - Hoi Yin Mak
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Sarah E Hancock
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Holly McEwen
- Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia.,National Health and Medical Research Council Clinical Trials Center, The University of Sydney, Camperdown, New South Wales, Australia
| | - Elvis Pandzic
- Biomedical Imaging Facility, Mark Wainwright Analytical Center, New South Wales, Australia
| | - Renee M Whan
- Biomedical Imaging Facility, Mark Wainwright Analytical Center, New South Wales, Australia
| | - Yvette Celine Aw
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Ivan E Lukmantara
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Yiqiong Yuan
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xiuju Dong
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China
| | - Anthony Don
- Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia.,National Health and Medical Research Council Clinical Trials Center, The University of Sydney, Camperdown, New South Wales, Australia
| | - Nigel Turner
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Shiqian Qi
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
136
|
Sanders DW, Jumper CC, Ackerman PJ, Bracha D, Donlic A, Kim H, Kenney D, Castello-Serrano I, Suzuki S, Tamura T, Tavares AH, Saeed M, Holehouse AS, Ploss A, Levental I, Douam F, Padera RF, Levy BD, Brangwynne CP. SARS-CoV-2 requires cholesterol for viral entry and pathological syncytia formation. eLife 2021; 10:e65962. [PMID: 33890572 PMCID: PMC8104966 DOI: 10.7554/elife.65962] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/01/2021] [Indexed: 12/27/2022] Open
Abstract
Many enveloped viruses induce multinucleated cells (syncytia), reflective of membrane fusion events caused by the same machinery that underlies viral entry. These syncytia are thought to facilitate replication and evasion of the host immune response. Here, we report that co-culture of human cells expressing the receptor ACE2 with cells expressing SARS-CoV-2 spike, results in synapse-like intercellular contacts that initiate cell-cell fusion, producing syncytia resembling those we identify in lungs of COVID-19 patients. To assess the mechanism of spike/ACE2-driven membrane fusion, we developed a microscopy-based, cell-cell fusion assay to screen ~6000 drugs and >30 spike variants. Together with quantitative cell biology approaches, the screen reveals an essential role for biophysical aspects of the membrane, particularly cholesterol-rich regions, in spike-mediated fusion, which extends to replication-competent SARS-CoV-2 isolates. Our findings potentially provide a molecular basis for positive outcomes reported in COVID-19 patients taking statins and suggest new strategies for therapeutics targeting the membrane of SARS-CoV-2 and other fusogenic viruses.
Collapse
Affiliation(s)
- David W Sanders
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Chanelle C Jumper
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Paul J Ackerman
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Dan Bracha
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Anita Donlic
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Hahn Kim
- Princeton University Small Molecule Screening Center, Princeton University, Princeton, United States
- Department of Chemistry, Princeton University, Princeton, United States
| | - Devin Kenney
- Department of Microbiology, Boston University School of Medicine, Boston, United States
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
| | - Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - Saori Suzuki
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Tomokazu Tamura
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Alexander H Tavares
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, United States
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - Florian Douam
- Department of Microbiology, Boston University School of Medicine, Boston, United States
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Clifford P Brangwynne
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
- Howard Hughes Medical Institute, Princeton, United States
| |
Collapse
|
137
|
Abe M, Kobayashi T. Imaging cholesterol depletion at the plasma membrane by methyl-β-cyclodextrin. J Lipid Res 2021; 62:100077. [PMID: 33891936 PMCID: PMC8281586 DOI: 10.1016/j.jlr.2021.100077] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Mitsuhiro Abe
- Lipid Biology Laboratory, RIKEN, Wako, Saitama, Japan; Cellular Informatics Laboratory, RIKEN, Wako, Saitama, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, Wako, Saitama, Japan; Cellular Informatics Laboratory, RIKEN, Wako, Saitama, Japan; Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France.
| |
Collapse
|
138
|
Russo-Savage L, Schulman IG. Liver X receptors and liver physiology. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166121. [PMID: 33713792 DOI: 10.1016/j.bbadis.2021.166121] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/29/2022]
Abstract
The liver x receptors LXRα (NR1H3) and LXRβ (NR1H2) are members of the nuclear hormone receptor superfamily of ligand dependent transcription factors that regulate transcription in response to the direct binding of cholesterol derivatives. Studies using genetic knockouts and synthetic ligands have defined the LXRs as important modulators of lipid homeostasis throughout the body. This review focuses on the control of cholesterol and fatty acid metabolism by LXRs in the liver and how modifying LXR activity can influence the pathology of liver diseases.
Collapse
Affiliation(s)
- Lillian Russo-Savage
- Department of Pharmacology, University of Virginia, School of Medicine, United States of America
| | - Ira G Schulman
- Department of Pharmacology, University of Virginia, School of Medicine, United States of America.
| |
Collapse
|
139
|
Gatineau E, Arthur G, Poupeau A, Nichols K, Spear BT, Shelman NR, Graf GA, Temel RE, Yiannikouris FB. The prorenin receptor and its soluble form contribute to lipid homeostasis. Am J Physiol Endocrinol Metab 2021; 320:E609-E618. [PMID: 33459178 PMCID: PMC7988779 DOI: 10.1152/ajpendo.00135.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Obesity is associated with alterations in hepatic lipid metabolism. We previously identified the prorenin receptor (PRR) as a potential contributor to liver steatosis. Therefore, we aimed to determine the relative contribution of PRR and its soluble form, sPRR, to lipid homeostasis. PRR-floxed male mice were treated with an adeno-associated virus with thyroxine-binding globulin promoter-driven Cre to delete PRR in the liver [liver PRR knockout (KO) mice]. Hepatic PRR deletion did not change the body weight but increased liver weights. The deletion of PRR in the liver decreased peroxisome proliferator-activated receptor gamma (PPARγ) and triglyceride levels, but liver PRR KO mice exhibited higher plasma cholesterol levels and lower hepatic low-density lipoprotein receptor (LDLR) and Sortilin 1 (SORT1) proteins than control (CTL) mice. Surprisingly, hepatic PRR deletion elevated hepatic cholesterol, and up-regulated hepatic sterol regulatory element-binding protein 2 (SREBP2) and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG CoA-R) genes. In addition, the plasma levels of sPRR were significantly higher in liver PRR KO mice than in controls. In vitro studies in HepG2 cells demonstrated that sPRR treatment upregulated SREBP2, suggesting that sPRR could contribute to hepatic cholesterol biosynthesis. Interestingly, PRR, total cleaved and noncleaved sPRR contents, furin, and Site-1 protease (S1P) were elevated in the adipose tissue of liver PRR KO mice, suggesting that adipose tissue could contribute to the circulating pool of sPRR. Overall, this work supports previous works and opens a new area of investigation concerning the function of sPRR in lipid metabolism and adipose tissue-liver cross talk.NEW & NOTEWORTHY Hepatic PRR and its soluble form, sPRR, contribute to triglyceride and cholesterol homeostasis and hepatic inflammation. Deletion of hepatic PRR decreased triglyceride levels through a PRR-PPARγ-dependent mechanism but increased hepatic cholesterol synthesis through sPRR-medicated upregulation of SREBP-2. Our study highlighted a new paradigm of cross talk between the liver and the adipose tissue involving cholesterol and sPRR.
Collapse
Affiliation(s)
- Eva Gatineau
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Gertrude Arthur
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Audrey Poupeau
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Kellea Nichols
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Brett T Spear
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky
| | - Nathan R Shelman
- Department of Pathology & Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Gregory A Graf
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky
| | - Ryan E Temel
- Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, Kentucky
| | | |
Collapse
|
140
|
Gan ES, Tan HC, Le DHT, Huynh TT, Wills B, Seidah NG, Ooi EE, Yacoub S. Dengue virus induces PCSK9 expression to alter antiviral responses and disease outcomes. J Clin Invest 2021; 130:5223-5234. [PMID: 32644974 PMCID: PMC7524462 DOI: 10.1172/jci137536] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022] Open
Abstract
Dengue virus (DENV) infection requires cholesterol as a proviral factor, although statin treatment did not show antiviral efficacy in patients with dengue. Here, we show that DENV infection manipulated cholesterol metabolism in cells residing in low-oxygen microenvironments (hypoxia) such as in the liver, spleen, and lymph nodes. DENV infection induced expression of proprotein convertase subtilisin/kexin type 9 (PCSK9), which reduces low-density lipoprotein receptor (LDLR) recycling and hence cholesterol uptake. We found that, whereas LDLR uptake would have distributed cholesterol throughout the various cell compartments, de novo cholesterol synthesis enriched this lipid in the endoplasmic reticulum (ER). With cholesterol enrichment in the ER, ER-resident STING and type I IFN (IFN) activation was repressed during DENV infection. Our in vitro findings were further supported by the detection of elevated plasma PCSK9 levels in patients with dengue with high viremia and increased severity of plasma leakage. Our findings therefore suggest that PCSK9 plays a hitherto unrecognized role in dengue pathogenesis and that PCSK9 inhibitors could be a suitable host-directed treatment for patients with dengue.
Collapse
Affiliation(s)
| | - Hwee Cheng Tan
- Duke-National University of Singapore Medical School, Singapore
| | - Duyen Huynh Thi Le
- Oxford University Clinical Research Unit (OUCRU), Ho Chi Minh City, Vietnam
| | - Trieu Trung Huynh
- Oxford University Clinical Research Unit (OUCRU), Ho Chi Minh City, Vietnam
| | - Bridget Wills
- Oxford University Clinical Research Unit (OUCRU), Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute, Université de Montréal, Montréal, Québec, Canada
| | - Eng Eong Ooi
- Duke-National University of Singapore Medical School, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore.,SingHealth Duke-National University of Singapore Global Health Institute, Singapore.,Antimicrobial Resistance Interdisciplinary Research Group, Singapore MIT Alliance in Research and Technology, Singapore
| | - Sophie Yacoub
- Oxford University Clinical Research Unit (OUCRU), Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom.,Antimicrobial Resistance Interdisciplinary Research Group, Singapore MIT Alliance in Research and Technology, Singapore
| |
Collapse
|
141
|
Ercan B, Naito T, Koh DHZ, Dharmawan D, Saheki Y. Molecular basis of accessible plasma membrane cholesterol recognition by the GRAM domain of GRAMD1b. EMBO J 2021; 40:e106524. [PMID: 33604931 PMCID: PMC7957428 DOI: 10.15252/embj.2020106524] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/16/2020] [Accepted: 01/08/2021] [Indexed: 12/25/2022] Open
Abstract
Cholesterol is essential for cell physiology. Transport of the "accessible" pool of cholesterol from the plasma membrane (PM) to the endoplasmic reticulum (ER) by ER-localized GRAMD1 proteins (GRAMD1a/1b/1c) contributes to cholesterol homeostasis. However, how cells detect accessible cholesterol within the PM remains unclear. We show that the GRAM domain of GRAMD1b, a coincidence detector for anionic lipids, including phosphatidylserine (PS), and cholesterol, possesses distinct but synergistic sites for sensing accessible cholesterol and anionic lipids. We find that a mutation within the GRAM domain of GRAMD1b that is associated with intellectual disability in humans specifically impairs cholesterol sensing. In addition, we identified another point mutation within this domain that enhances cholesterol sensitivity without altering its PS sensitivity. Cell-free reconstitution and cell-based assays revealed that the ability of the GRAM domain to sense accessible cholesterol regulates membrane tethering and determines the rate of cholesterol transport by GRAMD1b. Thus, cells detect the codistribution of accessible cholesterol and anionic lipids in the PM and fine-tune the non-vesicular transport of PM cholesterol to the ER via GRAMD1s.
Collapse
Affiliation(s)
- Bilge Ercan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Tomoki Naito
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | | - Dennis Dharmawan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
142
|
Martin LJ, Banister SD, Bowen MT. Understanding the complex pharmacology of cannabidiol: Mounting evidence suggests a common binding site with cholesterol. Pharmacol Res 2021; 166:105508. [PMID: 33610721 DOI: 10.1016/j.phrs.2021.105508] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/28/2021] [Accepted: 02/16/2021] [Indexed: 12/19/2022]
Abstract
Cannabidiol is claimed to bind to a large number of protein targets based on in vitro assays. This suggests opportunities for a wide range of therapeutic applications. On the other hand, the existence of phytochemical 'nuisance compounds' suggests some measure of caution - these compounds are capable of altering membrane biophysical properties and changing protein function without directly contacting a binding site. Like cannabidiol, cholesterol alters membrane properties, but it also binds directly to membrane proteins through abundant cholesterol recognition sites. We present the evidence that cannabidiol and cholesterol may bind to the same site on some proteins. As a starting point for further research, we also used blind docking to show that cannabidiol binds to a cholesterol binding site on the CB1 receptor. Elucidation of the mechanism(s) of action of cannabidiol will assist the prioritisation of in vitro hits across targets, improve the success rate of medicinal chemistry campaigns, and ultimately benefit patient populations by focusing resources on programs with the most translational potential.
Collapse
Affiliation(s)
- Lewis J Martin
- The University of Sydney, Brain and Mind Centre, The Lambert Initiative for Cannabinoid Therapeutics, Sydney, NSW, Australia; The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia; The University of Sydney, Faculty of Science, School of Psychology, NSW, Australia
| | - Samuel D Banister
- The University of Sydney, Brain and Mind Centre, The Lambert Initiative for Cannabinoid Therapeutics, Sydney, NSW, Australia; The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia
| | - Michael T Bowen
- The University of Sydney, Brain and Mind Centre, The Lambert Initiative for Cannabinoid Therapeutics, Sydney, NSW, Australia; The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia; The University of Sydney, Faculty of Science, School of Psychology, NSW, Australia.
| |
Collapse
|
143
|
Johnson KA, Radhakrishnan A. The use of anthrolysin O and ostreolysin A to study cholesterol in cell membranes. Methods Enzymol 2021; 649:543-566. [PMID: 33712199 DOI: 10.1016/bs.mie.2021.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cholesterol is a major component of the plasma membranes (PMs) of animal cells, comprising 35-40mol% of total PM lipids. Recent studies using cholesterol-binding bacterial toxins such as domain 4 of Anthrolysin O (ALOD4) and fungal toxins such as Ostreolysin A (OlyA) have revealed new insights into the organization of PM cholesterol. These studies have defined three distinct pools of PM cholesterol-a fixed pool that is essential for membrane integrity, a sphingomyelin (SM)-sequestered pool that can be detected by OlyA, and a third pool that is accessible and can be detected by ALOD4. Accessible cholesterol is available to interact with proteins and transport to the endoplasmic reticulum (ER), and controls many cellular signaling processes including cholesterol homeostasis, Hedgehog signaling, and bacterial and viral infection. Here, we provide detailed descriptions for the use of ALOD4 and OlyA, both of which are soluble and non-lytic proteins, to study cholesterol organization in the PMs of animal cells. Furthermore, we describe two new versions of ALOD4 that we have developed to increase the versatility of this probe in cellular studies. One is a dual His6 and FLAG epitope-tagged version and the other is a fluorescent version where ALOD4 is fused to Neon, a monomeric fluorescent protein. These new forms of ALOD4 together with previously described OlyA provide an expanded collection of tools to sense, visualize, and modulate levels of accessible and SM-sequestered cholesterol on PMs and study the role of these cholesterol pools in diverse membrane signaling events.
Collapse
Affiliation(s)
- Kristen A Johnson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
144
|
Abstract
Pore forming proteins are released as water-soluble monomers that form-mostly oligomeric-pores in target membranes. Our understanding of such pore formation relies in part on the direct visualization of their assemblies on and in the membrane. Here, we discuss the application of atomic force microscopy (AFM) to visualize and understand membrane pore formation, illustrated specifically by studies of proteins of the MACPF/CDC superfamily on supported lipid bilayers. Besides detailed protocols, we also point out common imaging artefacts and strategies to avoid them, and briefly outline how AFM can be effectively used in conjunction with other methods.
Collapse
Affiliation(s)
- Adrian W Hodel
- Killer Cell Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Katharine Hammond
- National Physical Laboratory, Teddington, United Kingdom; London Centre for Nanotechnology, University College London, London, United Kingdom; Department of Physics & Astronomy, University College London, London, United Kingdom
| | - Bart W Hoogenboom
- London Centre for Nanotechnology, University College London, London, United Kingdom; Department of Physics & Astronomy, University College London, London, United Kingdom.
| |
Collapse
|
145
|
Zhou Y, Prakash PS, Liang H, Gorfe AA, Hancock JF. The KRAS and other prenylated polybasic domain membrane anchors recognize phosphatidylserine acyl chain structure. Proc Natl Acad Sci U S A 2021; 118:e2014605118. [PMID: 33526670 PMCID: PMC8017956 DOI: 10.1073/pnas.2014605118] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
KRAS interacts with the inner leaflet of the plasma membrane (PM) using a hybrid anchor that comprises a lysine-rich polybasic domain (PBD) and a C-terminal farnesyl chain. Electrostatic interactions have been envisaged as the primary determinant of interactions between KRAS and membranes. Here, we integrated molecular dynamics (MD) simulations and superresolution spatial analysis in mammalian cells and systematically compared four equally charged KRAS anchors: the wild-type farnesyl hexa-lysine and engineered mutants comprising farnesyl hexa-arginine, geranylgeranyl hexa-lysine, and geranylgeranyl hexa-arginine. MD simulations show that these equally charged KRAS mutant anchors exhibit distinct interactions and packing patterns with different phosphatidylserine (PtdSer) species, indicating that prenylated PBD-bilayer interactions extend beyond electrostatics. Similar observations were apparent in intact cells, where each anchor exhibited binding specificities for PtdSer species with distinct acyl chain compositions. Acyl chain composition determined responsiveness of the spatial organization of different PtdSer species to diverse PM perturbations, including transmembrane potential, cholesterol depletion, and PM curvature. In consequence, the spatial organization and PM binding of each KRAS anchor precisely reflected the behavior of its preferred PtdSer ligand to these same PM perturbations. Taken together these results show that small GTPase PBD-prenyl anchors, such as that of KRAS, have the capacity to encode binding specificity for specific acyl chains as well as lipid headgroups, which allow differential responses to biophysical perturbations that may have biological and signaling consequences for the anchored GTPase.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030
- Graduate School of Biological Sciences, MD Anderson Cancer Center and University of Texas Health Science Center, Houston, TX 77030
| | - Priyanka S Prakash
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030
| | - Hong Liang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030
| | - Alemayehu A Gorfe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030
- Graduate School of Biological Sciences, MD Anderson Cancer Center and University of Texas Health Science Center, Houston, TX 77030
| | - John F Hancock
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030;
- Graduate School of Biological Sciences, MD Anderson Cancer Center and University of Texas Health Science Center, Houston, TX 77030
| |
Collapse
|
146
|
Zinöcker MK, Svendsen K, Dankel SN. The homeoviscous adaptation to dietary lipids (HADL) model explains controversies over saturated fat, cholesterol, and cardiovascular disease risk. Am J Clin Nutr 2021; 113:277-289. [PMID: 33471045 DOI: 10.1093/ajcn/nqaa322] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
SFAs play the leading role in 1 of the greatest controversies in nutrition science. Relative to PUFAs, SFAs generally increase circulating concentrations of LDL cholesterol, a risk factor for atherosclerotic cardiovascular disease (ASCVD). However, the purpose of regulatory mechanisms that control the diet-induced lipoprotein cholesterol dynamics is rarely discussed in the context of human adaptive biology. We argue that better mechanistic explanations can help resolve lingering controversies, with the potential to redefine aspects of research, clinical practice, dietary advice, public health management, and food policy. In this paper we propose a novel model, the homeoviscous adaptation to dietary lipids (HADL) model, which explains changes in lipoprotein cholesterol as adaptive homeostatic adjustments that serve to maintain cell membrane fluidity and hence optimal cell function. Due to the highly variable intake of fatty acids in humans and other omnivore species, we propose that circulating lipoproteins serve as a buffer to enable the rapid redistribution of cholesterol molecules between specific cells and tissues that is necessary with changes in dietary fatty acid supply. Hence, circulating levels of LDL cholesterol may change for nonpathological reasons. Accordingly, an SFA-induced raise in LDL cholesterol in healthy individuals could represent a normal rather than a pathologic response. These regulatory mechanisms may become disrupted secondarily to pathogenic processes in association with insulin resistance and the presence of other ASCVD risk factors, as supported by evidence showing diverging lipoprotein responses in healthy individuals as opposed to those with metabolic disorders such as insulin resistance and obesity. Corresponding with the model, we suggest alternative contributing factors to the association between elevated LDL cholesterol concentrations and ASCVD, involving dietary factors beyond SFAs, such as an increased endotoxin load from diet-gut microbiome interactions and subsequent chronic low-grade inflammation that interferes with fine-tuned signaling pathways.
Collapse
Affiliation(s)
| | - Karianne Svendsen
- Department of Nutrition, University of Oslo, Oslo, Norway.,The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Simon Nitter Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
147
|
Hitching a ride to the top: peroxisomes fuel cilium with cholesterol. SCIENCE CHINA-LIFE SCIENCES 2021; 64:478-481. [PMID: 33420924 DOI: 10.1007/s11427-020-1866-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/10/2020] [Indexed: 10/22/2022]
|
148
|
Vos DY, van de Sluis B. Function of the endolysosomal network in cholesterol homeostasis and metabolic-associated fatty liver disease (MAFLD). Mol Metab 2021; 50:101146. [PMID: 33348067 PMCID: PMC8324686 DOI: 10.1016/j.molmet.2020.101146] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/26/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
Background Metabolic-associated fatty liver disease (MAFLD), also known as non-alcoholic fatty liver disease, has become the leading cause of chronic liver disease worldwide. In addition to hepatic accumulation of triglycerides, dysregulated cholesterol metabolism is an important contributor to the pathogenesis of MAFLD. Maintenance of cholesterol homeostasis is highly dependent on cellular cholesterol uptake and, subsequently, cholesterol transport to other membrane compartments, such as the endoplasmic reticulum (ER). Scope of review The endolysosomal network is key for regulating cellular homeostasis and adaptation, and emerging evidence has shown that the endolysosomal network is crucial to maintain metabolic homeostasis. In this review, we will summarize our current understanding of the role of the endolysosomal network in cholesterol homeostasis and its implications in MAFLD pathogenesis. Major conclusions Although multiple endolysosomal proteins have been identified in the regulation of cholesterol uptake, intracellular transport, and degradation, their physiological role is incompletely understood. Further research should elucidate their role in controlling metabolic homeostasis and development of fatty liver disease. The intracellular cholesterol transport is tightly regulated by the endocytic and lysosomal network. Dysfunction of the endolysosomal network affects hepatic lipid homeostasis. The endosomal sorting of lipoprotein receptors is precisely regulated and is not a bulk process.
Collapse
Affiliation(s)
- Dyonne Y Vos
- Department of Pediatrics, section Molecular Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, section Molecular Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
149
|
Zheng Koh DH, Saheki Y. Regulation of Plasma Membrane Sterol Homeostasis by Nonvesicular Lipid Transport. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2021; 4:25152564211042451. [PMID: 37366378 PMCID: PMC10259818 DOI: 10.1177/25152564211042451] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Sterol contributes to the structural integrity of cellular membranes and plays an important role in the regulation of cell signaling in eukaryotes. It is either produced in the endoplasmic reticulum or taken up from the extracellular environment. In most eukaryotic cells, however, the majority of sterol is enriched in the plasma membrane. Thus, the transport of sterol between the plasma membrane and other organelles, including the endoplasmic reticulum, is crucial for maintaining sterol homeostasis. While vesicular transport that relies on membrane budding and fusion reactions plays an important role in bulk sterol transport, this mode of transport is slow and non-selective. Growing evidence suggests a critical role of nonvesicular transport mediated by evolutionarily conserved families of lipid transfer proteins in more rapid and selective delivery of sterol. Some lipid transfer proteins act primarily at the sites of contacts formed between the endoplasmic reticulum and other organelles or the plasma membrane without membrane fusion. In this review, we describe the similarities and differences of sterol biosynthesis and uptake in mammals and yeast and discuss the role of their lipid transfer proteins in maintaining plasma membrane sterol homeostasis.
Collapse
Affiliation(s)
- Dylan Hong Zheng Koh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Institute of Resource Development and
Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
150
|
Radhakrishnan A, Rohatgi R, Siebold C. Cholesterol access in cellular membranes controls Hedgehog signaling. Nat Chem Biol 2020; 16:1303-1313. [PMID: 33199907 PMCID: PMC7872078 DOI: 10.1038/s41589-020-00678-2] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022]
Abstract
The Hedgehog (Hh) signaling pathway coordinates cell-cell communication in development and regeneration. Defects in this pathway underlie diseases ranging from birth defects to cancer. Hh signals are transmitted across the plasma membrane by two proteins, Patched 1 (PTCH1) and Smoothened (SMO). PTCH1, a transporter-like tumor-suppressor protein, binds to Hh ligands, but SMO, a G-protein-coupled-receptor family oncoprotein, transmits the Hh signal across the membrane. Recent structural, biochemical and cell-biological studies have converged at the surprising model that a specific pool of plasma membrane cholesterol, termed accessible cholesterol, functions as a second messenger that conveys the signal between PTCH1 and SMO. Beyond solving a central puzzle in Hh signaling, these studies are revealing new principles in membrane biology: how proteins respond to and remodel cholesterol accessibility in membranes and how the cholesterol composition of organelle membranes is used to regulate protein function.
Collapse
Affiliation(s)
- Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|