1
|
Luo H, Cai Y, Shi H, Ma L, Zhang S, Yung KKL, Zhou P. Repurposing oxiconazole to inhibit STING trafficking via OSBP and alleviate autoimmune pathology in Trex1 -/- mice. Int Immunopharmacol 2025; 157:114742. [PMID: 40319749 DOI: 10.1016/j.intimp.2025.114742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
The cGAS-STING pathway is a critical component of the innate immune response to cytosolic DNA, driving the production of type I interferons (IFNs) and pro-inflammatory cytokines. However, excessive activation of this pathway is associated with various autoimmune and inflammatory diseases. In this study, we evaluated the regulation of FDA-approved azole antifungal drugs on the cGAS-STING pathway. Among these drugs, oxiconazole, miconazole, and itraconazole demonstrate significant inhibitory effects, with oxiconazole showing the strongest activity. Our data demonstrates that oxiconazole significantly suppressed type I IFN production and downstream inflammatory responses in macrophages and fibroblasts stimulated with synthetic DNA or infected with HSV-1. Mechanistically, oxiconazole hindered STING trafficking via oxysterol-binding protein OSBP. Using the Listeria monocytogenes infection model and the Trex1-/- mouse disease model, both representing in vivo models of inflammation driven by excessive cGAS-STING activation, we demonstrate that oxiconazole enhanced bacterial clearance and reduced tissue damage in the Listeria monocytogenes infection model. Moreover, oxiconazole treatment significantly alleviated multi-organ inflammation and normalized aberrant IFN responses in the Trex1-/- autoimmune disease mouse model. These findings highlight the potential of oxiconazole as a promising therapeutic agent for STING-driven autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Hui Luo
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yijing Cai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hanhui Shi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Liang Ma
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shiqing Zhang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Ken Kin Lam Yung
- Provincial Key Laboratory of New Drug Screening and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, Department of Science and Environmental Studies, the Education University of Hong Kong, Hong Kong, SAR 999077, China
| | - Pingzheng Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China.
| |
Collapse
|
2
|
Balla T. Phosphatidylinositol 4-phosphate; A minor lipid with multiple personalities. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159615. [PMID: 40262701 PMCID: PMC12145240 DOI: 10.1016/j.bbalip.2025.159615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
Phosphorylated products of phosphatidylinositol (PI), named Diphosphoinositide (DPI) and triphosphoinositide (TPI) were identified long time ago and found to exhibit high turnover rates based on their rapid 32P-phosphate labeling. The PI kinase activities that were responsible for their production were subsequently identified and found to be associated with different organelle membranes, including the plasma membrane. These activities were then linked with a certain group of cell surface receptors that activated phospholipase C enzymes to hydrolyze PI and used calcium or cGMP as a second messenger. This visionary concept was introduced in the seminal BBA review written by Robert Michell, exactly 50 years ago. The enzymology and functional diversity of PI 4-phosphate (PI4P) (the term that has replaced DPI) has since underwent an expansion that could not have been foreseen. In this review I will attempt to revisit this expansion with some historical reflections celebrating the 50th anniversary of the Michell review.
Collapse
Affiliation(s)
- Tamas Balla
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Ferrari A, Tontonoz P. Nonvesicular cholesterol transport in physiology. J Clin Invest 2025; 135:e188127. [PMID: 40091839 PMCID: PMC11910210 DOI: 10.1172/jci188127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
In mammalian cells cholesterol can be synthesized endogenously or obtained exogenously through lipoprotein uptake. Plasma membrane (PM) is the primary intracellular destination for both sources of cholesterol, and maintaining appropriate membrane cholesterol levels is critical for cellular viability. The endoplasmic reticulum (ER) acts as a cellular cholesterol sensor, regulating synthesis in response to cellular needs and determining the metabolic fates of cholesterol. Upon reaching the ER, cholesterol can be esterified to facilitate its incorporation into lipoproteins and lipid droplets or converted into other molecules such as bile acids and oxysterols. In recent years, it has become clear that the intracellular redistribution of lipids, including cholesterol, is critical for the regulation of various biological processes. This Review highlights physiology and mechanisms of nonvesicular (protein-mediated) intracellular cholesterol trafficking, with a focus on the role of Aster proteins in PM to ER cholesterol transport.
Collapse
|
4
|
Jain BK, Duan HD, Valentine C, Samiha A, Li H, Graham TR. P4-ATPase control over phosphoinositide membrane asymmetry and neomycin resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.641220. [PMID: 40093091 PMCID: PMC11908233 DOI: 10.1101/2025.03.03.641220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Neomycin, an aminoglycoside antibiotic, has robust antibacterial properties, yet its clinical utility is curtailed by its nephrotoxicity and ototoxicity. The mechanism by which the polycationic neomycin enters specific eukaryotic cell types remains poorly understood. In budding yeast, NEO1 is required for neomycin resistance and encodes a phospholipid flippase that establishes membrane asymmetry. Here, we show that mutations altering Neo1 substrate recognition cause neomycin hypersensitivity by exposing phosphatidylinositol-4-phosphate (PI4P) in the plasma membrane extracellular leaflet. Human cells also expose extracellular PI4P upon knockdown of ATP9A, a Neo1 ortholog and ATP9A expression level correlates to neomycin sensitivity. In yeast, the extracellular PI4P is initially produced in the cytosolic leaflet of the plasma membrane and then delivered by Osh6-dependent nonvesicular transport to the endoplasmic reticulum (ER). Here, a portion of PI4P escapes degradation by the Sac1 phosphatase by entering the ER lumenal leaflet. COPII vesicles transport lumenal PI4P to the Golgi where Neo1 flips this substrate back to the cytosolic leaflet. Cryo-EM reveals that PI4P binds Neo1 within the substrate translocation pathway. Loss of Neo1 activity in the Golgi allows secretion of extracellular PI4P, which serves as a neomycin receptor and facilitates its endocytic uptake. These findings unveil novel mechanisms of aminoglycoside sensitivity and phosphoinositide homeostasis, with important implications for signaling by extracellular phosphoinositides.
Collapse
Affiliation(s)
- Bhawik K Jain
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
- These authors contributed equally: Bhawik K. Jain, H. Diessel Duan
| | - H Diessel Duan
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
- These authors contributed equally: Bhawik K. Jain, H. Diessel Duan
| | - Christina Valentine
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Ariana Samiha
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Todd R Graham
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
5
|
Sokolov SS, Zyrina AN, Akimov SA, Severin FF. Interrelationship between the Non-Vesicular Transport of Sterols and Their Distribution between the Rafts and the Non-Raft Phase of the Plasma Membrane. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:321-333. [PMID: 40367076 DOI: 10.1134/s0006297924604313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 05/16/2025]
Abstract
Sterols significantly affect the barrier properties of the membrane, which might explains the fact that their concentration is maximal in the plasma membrane (PM). Together with sphingolipids, sterols form rafts, i.e., bilayer regions whose physicochemical properties differ from those of the surrounding PM. The presence of rafts allows membrane proteins to choose the lipid environment optimal for their functioning (in terms of thickness, rigidity, spontaneous curvature, and lateral pressure profile of the bilayer). The ratio between sterols and sphingolipids in the rafts is close to stoichiometric. Theoretically, excess sterol outside the rafts can critically reduce the degree of order of membrane phospholipids. Sterols are synthesized in the endoplasmic reticulum (ER). The active (against the concentration gradient) transport of sterols from the ER to the PM is driven by proteins of the Osh family, while Lam proteins provide passive reverse transport of sterols from the PM to the ER. Inactivation of Osh proteins does not reduce the total level of sterols in the PM but reduces the rate of their movement inside the PM (the mechanisms underlying this effect remains unclear). Therefore, the vesicular transport of sterols from the ER to the PM is probably more active than the non-vesicular transport carried out by Osh proteins. Since sterols are more rigidly anchored and less sterically accessible in the rafts than outside them, we suggested that Lam proteins transport excess sterols from the non-raft phase of the PM to the ER, and Osh proteins return them back to the PM. In this way, the mutual activity of the Osh and lam proteins provides the rotation of sterols between the non-raft fraction of the PM and rafts, with the enrichment of the latter. It is possible that with a decrease in the sterol concentration in the non-raft fraction of the membrane, the rate of the Lam-dependent transport decreases since the degree of order of phospholipids and, consequently, the strength of retention of sterol molecules in the membrane increases, which might represent a mechanisms maintaining the concentration and distribution of sterols in the PM.
Collapse
Affiliation(s)
- Svyatoslav S Sokolov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - Anna N Zyrina
- Chumakov Federal Scientific Center for Research and Development of Immune and Biological Products (Institute of Poliomyelitis), Russian Academy of Sciences, Moscow, 108819, Russia
| | - Sergey A Akimov
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Fedor F Severin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
6
|
Arce A, Altman R, Badolian A, Low J, Cuaresma AB, Keshet U, Fiehn O, Stahelin RV, Nikolaidis N. Heat Shock-Induced PI(4)P Increase Drives HSPA1A Translocation to the Plasma Membrane in Cancer and Stressed Cells through PI4KIII Alpha Activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.16.638537. [PMID: 40027828 PMCID: PMC11870583 DOI: 10.1101/2025.02.16.638537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
HSPA1A, a major heat shock protein, is known to translocate to the plasma membrane (PM) in response to cellular stress and cancer, where it plays protective roles in membrane integrity and stress resistance. Although phosphatidylinositol 4-phosphate [PI(4)P] is essential in this translocation, the signals that trigger and facilitate HSPA1A's movement remain undefined.Given that membrane lipid composition dynamically shifts during stress, we hypothesized that heat shock-induced PI(4)P changes are crucial for HSPA1A's PM localization. To test this hypothesis, we investigated the mechanisms driving PI(4)P changes and HSPA1A PM localization under heat shock. Lipidomic analysis, enzyme-linked immunosorbent assay (ELISA), and confocal imaging revealed a rapid PI(4)P increase at the PM post-heat shock, with levels peaking at 0 hours and declining by 8 hours. RNA sequencing and protein quantification indicated no transcriptional increase in PI4KIII alpha, the kinase responsible for PI(4)P synthesis, suggesting an alternative regulatory mechanism. Hypothesizing that heat shock enhances PI4KIII alpha activity, we performed ELISA coupled with immunoprecipitation, confirming a significant rise in PI4KIII alpha activity following heat shock. Functional analyses further demonstrated that RNAi-mediated PI4KIII alpha depletion or pharmacological PI(4)P reduction, using GSK-A1, impairs HSPA1A's localization to the PM, confirming that HSPA1A translocation is PI(4)P-dependent. Our findings identify PI4KIII alpha activity as a key regulator of PI(4)P accumulation and subsequent HSPA1A recruitment to the PM in stressed and cancer cells. This lipid-mediated response offers new insights into stress adaptation and potentially modifiable pathways for therapeutic interventions to control HSPA1A function in cancer.
Collapse
Affiliation(s)
- Alberto Arce
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Rachel Altman
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Allen Badolian
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Jensen Low
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Azalea Blythe Cuaresma
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Uri Keshet
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA
| | - Robert V. Stahelin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute for Cancer Research, Purdue University, 47907, West Lafayette, IN, USA
| | - Nikolas Nikolaidis
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University Fullerton, Fullerton, CA, USA
| |
Collapse
|
7
|
Kawasaki A, Nakatsu F. Measurement of ORP10-Mediated Lipid Countertransport at ER-Endosome Membrane Contact Sites via a Chemically Induced Dimerization Strategy. Methods Mol Biol 2025; 2888:13-22. [PMID: 39699721 DOI: 10.1007/978-1-0716-4318-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Oxysterol-binding protein (OSBP)-related proteins (ORPs) are a large family of lipid transfer proteins (LTPs) in mammals. ORPs mediate the countertransport of two distinct lipids at membrane contact sites (MCSs). ORP10 is localized via binding to ORP9 at the endoplasmic reticulum (ER)-endosome MCSs, where it mediates countertransport of phosphatidylinositol 4-phosphate (PI4P) and phosphatidylserine (PS). To quantitatively monitor the lipid countertransport process mediated by ORP10 in situ, we take advantage of chemically induced dimerization (CID), a strategy of inducing protein-protein interactions by exposure to chemicals. Specifically, we exploit the rapamycin-inducible heterodimerization of FKBP/FRB to acutely recruit the lipid transfer domain of ORP10 to the ER-endosome MCSs and monitor the levels of PI4P and PS on endosomes by their genetic probes in live imaging. This approach enables the measurement of ORP10 activity in lipid countertransport at ER-endosome MCSs and is also beneficial as a versatile method applicable to other LTPs.
Collapse
Affiliation(s)
- Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan.
| |
Collapse
|
8
|
Yu X, Mousley CJ, Bankaitis VA, Iyer P. A budding yeast-centric view of oxysterol binding protein family function. Adv Biol Regul 2025; 95:101061. [PMID: 39613716 DOI: 10.1016/j.jbior.2024.101061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
The Trans Golgi Network (TGN)/endosomal system is a sorting center for cargo brought via the anterograde secretory pathway and the endocytic pathway that internalizes material from the plasma membrane. As many of the cargo that transit this central trafficking hub are components of key homeostatic signaling pathways, TGN/endosomes define a critical signaling hub for cellular growth control. A particularly interesting yet incompletely understood aspect of regulation of TGN/endosome function is control of this system by two families of lipid exchange/lipid transfer proteins. The phosphatidylinositol transfer proteins promote pro-trafficking phosphoinositide (i.e. phosphatidylinositol-4-phosphate) signaling pathways whereas proteins of the oxysterol binding protein family play reciprocal roles in antagonizing those arms of phosphoinositide signaling. The precise mechanisms for how these lipid binding proteins execute their functions remain to be resolved. Moreover, information regarding the coupling of individual members of the oxysterol binding protein family to specific biological activities is particularly sparse. Herein, we review what is being learned regarding functions of the oxysterol binding protein family in the yeast model system. Focus is primarily directed at a discussion of the Kes1/Osh4 protein for which the most information is available.
Collapse
Affiliation(s)
- Xiaohan Yu
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Carl J Mousley
- School of Biomedical Sciences, Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Vytas A Bankaitis
- Department of Cell Biology and Genetics, Texas A&M Health Science Center, College Station, TX, 77843, USA; Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA.
| | - Prasanna Iyer
- Department of Cell Biology and Genetics, Texas A&M Health Science Center, College Station, TX, 77843, USA.
| |
Collapse
|
9
|
Sacher S, Ray A. In Silico Strategies for Characterizing Inner Cavities of Lipid-Binding Proteins. Methods Mol Biol 2025; 2888:305-320. [PMID: 39699739 DOI: 10.1007/978-1-0716-4318-1_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Cavities in proteins perform diverse functions such as substrate binding, enzyme catalysis, passage for transportation of small molecules, and protein oligomerization. Often, the physical properties of these cavities are closely linked to the protein function; such as the hydrophobic lipid-binding cavities in lipid-binding proteins (LBPs) that protect lipid substrates from the larger aqueous milieu. Therefore, the characterization of protein cavities can provide valuable insights into protein structure-function relationships, hinting toward their mechanism of action while aiding in the identification of ligand binding sites that are essential for drug discovery approaches. Several algorithms have historically been designed to identify and characterize the different types of cavities in protein structures. We summarize these algorithms and provide a step-by-step guide for locating and characterizing internal cavities in proteins using CICLOP by using ATP-binding cassette transporter A1 (ABCA1) as an example.
Collapse
Affiliation(s)
- Sukriti Sacher
- Department of Computational Biology, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), New Delhi, India
| | - Arjun Ray
- Department of Computational Biology, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), New Delhi, India.
| |
Collapse
|
10
|
Šťastný D, Balleková A, Tahotná D, Pokorná L, Holič R, Humpolíčková J, Griač P. Characterization of two Plasmodium falciparum lipid transfer proteins of the Sec14/CRAL-TRIO family. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159572. [PMID: 39426587 DOI: 10.1016/j.bbalip.2024.159572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/11/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Invasion of human red blood cells by the malaria parasite Plasmodium falciparum is followed by dramatic modifications of erythrocytes properties, including de novo formation of new membrane systems. Lipid transfer proteins from both the parasite and the host cell are most likely an important part of those membrane remodeling processes. Using bioinformatics and in silico structural analysis, we have identified five P. falciparum potential lipid transfer proteins containing cellular retinaldehyde binding - triple functional domain (CRAL-TRIO). Two of these proteins, C6KTD4, encoded by the PF3D7_0629900 gene and Q8II87, encoded by the PF3D7_1127600 gene, were studied in more detail. In vitro lipid transfer assays using recombinant C6KTD4 and Q8II87 confirmed that these proteins are indeed bona fide lipid transfer proteins. C6KTD4 transfers sterols, phosphatidylinositol 4,5 bisphosphate, and, to some degree, also phosphatidylcholine between two membrane compartments. Q8II87 possesses phosphatidylserine transfer activity in vitro. In the yeast model, the expression of P. falciparumQ8II87 protein partially complements the absence of Sec14p and its closest homologue, Sfh1p. C6KTD4 protein can substitute for the collective essential function of oxysterol-binding related proteins. According to published whole genome studies in P. falciparum, absence of C6KTD4 and Q8II87 proteins has severe consequences for parasite viability. Therefore, CRAL-TRIO lipid transfer proteins of P. falciparum are potential targets of novel antimalarials, in search for which the yeast model expressing these proteins could be a valuable tool.
Collapse
Affiliation(s)
- Dominik Šťastný
- Centre of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Alena Balleková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10 Praha 6, Czech Republic
| | - Dana Tahotná
- Centre of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Lucia Pokorná
- Centre of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Roman Holič
- Centre of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Jana Humpolíčková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10 Praha 6, Czech Republic
| | - Peter Griač
- Centre of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia.
| |
Collapse
|
11
|
Delfosse V, Drin G. Determining the Relative Affinity of ORPs for Lipid Ligands Using Fluorescence and Thermal Shift Assays. Methods Mol Biol 2025; 2888:259-280. [PMID: 39699737 DOI: 10.1007/978-1-0716-4318-1_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Lipid transfer proteins (LTPs) are specialized proteins that convey specific lipids across the cytosol to regulate the lipid composition of organelles and the plasma membrane. Quantifying to which extent these LTPs recognize and transfer various lipid species and subspecies is of prime interest to define their cellular role(s). Here, we describe how to measure in vitro the relative affinity of Osh6p, a yeast phosphatidylserine (PS)/phosphatidylinositol 4-phosphate (PI(4)P) exchanger belonging to the oxysterol-binding protein(OSBP)-related protein (ORP) family, for PS and phosphoinositide subspecies. First, we detail how to produce and purify Osh6p with high purity. Secondly, we describe how to measure its ability to bind PS, PI(4)P, and PI(4,5)P2 by FRET-based and thermal shift assays using liposomes of defined composition. These protocols can allow further analysis of other ORPs or inspire the design of assays to characterize other LTPs. Notably, they can be helpful in defining how LTPs transfer phospholipids subspecies as a function of their acyl chains' length and unsaturation degree and, therefore, whether they can contribute to regulating the acyl chain composition of cell membranes.
Collapse
Affiliation(s)
- Vanessa Delfosse
- Centre de Biologie Structurale (CBS), Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Guillaume Drin
- Université Côte d'Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.
| |
Collapse
|
12
|
Hamaï A, Drin G. Specificity of lipid transfer proteins: An in vitro story. Biochimie 2024; 227:85-110. [PMID: 39304019 DOI: 10.1016/j.biochi.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Lipids, which are highly diverse, are finely distributed between organelle membranes and the plasma membrane (PM) of eukaryotic cells. As a result, each compartment has its own lipid composition and molecular identity, which is essential for the functional fate of many proteins. This distribution of lipids depends on two main processes: lipid synthesis, which takes place in different subcellular regions, and the transfer of these lipids between and across membranes. This review will discuss the proteins that carry lipids throughout the cytosol, called LTPs (Lipid Transfer Proteins). More than the modes of action or biological roles of these proteins, we will focus on the in vitro strategies employed during the last 60 years to address a critical question: What are the lipid ligands of these LTPs? We will describe the extent to which these strategies, combined with structural data and investigations in cells, have made it possible to discover proteins, namely ORPs, Sec14, PITPs, STARDs, Ups/PRELIs, START-like, SMP-domain containing proteins, and bridge-like LTPs, which compose some of the main eukaryotic LTP families, and their lipid ligands. We will see how these approaches have played a central role in cell biology, showing that LTPs can connect distant metabolic branches, modulate the composition of cell membranes, and even create new subcellular compartments.
Collapse
Affiliation(s)
- Amazigh Hamaï
- Université Côte d'Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, 660 route des lucioles, 06560, Valbonne Sophia Antipolis, France
| | - Guillaume Drin
- Université Côte d'Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, 660 route des lucioles, 06560, Valbonne Sophia Antipolis, France.
| |
Collapse
|
13
|
Volpiana MW, Nenadic A, Beh CT. Regulation of yeast polarized exocytosis by phosphoinositide lipids. Cell Mol Life Sci 2024; 81:457. [PMID: 39560727 DOI: 10.1007/s00018-024-05483-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024]
Abstract
Phosphoinositides help steer membrane trafficking routes within eukaryotic cells. In polarized exocytosis, which targets vesicular cargo to sites of polarized growth at the plasma membrane (PM), the two phosphoinositides phosphatidylinositol 4-phosphate (PI4P) and its derivative phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) pave the pathway for vesicle transport from the Golgi to the PM. PI4P is a critical regulator of mechanisms that shape late Golgi membranes for vesicle biogenesis and release. Although enriched in vesicle membranes, PI4P is inexplicably removed from post-Golgi vesicles during their transit to the PM, which drives subsequent steps in exocytosis. At the PM, PI(4,5)P2 recruits effectors that establish polarized membrane sites for targeting the vesicular delivery of secretory cargo. The budding yeast Saccharomyces cerevisiae provides an elegant model to unravel the complexities of phosphoinositide regulation during polarized exocytosis. Here, we review how PI4P and PI(4,5)P2 promote yeast vesicle biogenesis, exocyst complex assembly and vesicle docking at polarized cortical sites, and suggest how these steps might impact related mechanisms of human disease.
Collapse
Affiliation(s)
- Matthew W Volpiana
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Aleksa Nenadic
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Christopher T Beh
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
14
|
Tanwar S, Kalra S, Bari VK. Insights into the role of sterol metabolism in antifungal drug resistance: a mini-review. Front Microbiol 2024; 15:1409085. [PMID: 39464401 PMCID: PMC11502366 DOI: 10.3389/fmicb.2024.1409085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
Sterols are essential for eukaryotic cells and are crucial in cellular membranes' structure, function, fluidity, permeability, adaptability to environmental stressors, and host-pathogen interactions. Fungal sterol, such as ergosterol metabolism, involves several organelles, including the mitochondria, lipid droplets, endoplasmic reticulum, and peroxisomes that can be regulated mainly by feedback mechanisms and transcriptionally. The majority of sterol transport in yeast occurs via non-vesicular transport pathways mediated by lipid transfer proteins, which determine the quantity of sterol present in the cell membrane. Pathogenic fungi Candida, Aspergillus, and Cryptococcus species can cause a range of superficial to potentially fatal systemic and invasive infections that are more common in immunocompromised patients. There is a significant risk of morbidity and mortality from these infections, which are very difficult to cure. Several antifungal drugs with different modes of action have received clinical approval to treat fungal infections. Antifungal drugs targeting the ergosterol biosynthesis pathway are well-known for their antifungal activity; however, an imbalance in the regulation and transport of ergosterol could lead to resistance to antifungal therapy. This study summarizes how fungal sterol metabolism and regulation can modulate sterol-targeting antifungal drug resistance.
Collapse
|
15
|
Karmakar S, Klauda JB. Proposed dual membrane contact with full-length Osh4. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184365. [PMID: 38960299 DOI: 10.1016/j.bbamem.2024.184365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/03/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Membrane contacts sites (MCSs) play important roles in lipid trafficking across cellular compartments and maintain the widespread structural diversity of organelles. We have utilized microsecond long all-atom (AA) molecular dynamics (MD) simulations and enhanced sampling techniques to unravel the MCS structure targeting by yeast oxysterol binding protein (Osh4) in an environment that mimics the interface of membranes with an increased proportion of anionic lipids using CHARMM36m forcefield with additional CUFIX parameters for lipid-protein electrostatic interactions. In a dual-membrane environment, unbiased MD simulations show that Osh4 briefly interacts with both membranes, before aligning itself with a single membrane, adopting a β-crease-bound conformation similar to observations in a single-membrane scenario. Targeted molecular dynamics simulations followed by microsecond-long AA MD simulations have revealed a distinctive dual-membrane bound state of Osh4 at MCS, wherein the protein interacts with the lower membrane via the β-crease surface, featuring its PHE-239 residue positioned below the phosphate plane of membrane, while concurrently establishing contact with the opposite membrane through the extended α6-α7 region. Osh4 maintains these dual membrane contacts simultaneously over the course of microsecond-long MD simulations. Moreover, binding energy calculations highlighted the essential roles played by the phenylalanine loop and the α6 helix in dynamically stabilizing dual-membrane bound state of Osh4 at MCS. Our computational findings were corroborated through frequency of contact analysis, showcasing excellent agreement with past experimental cross-linking data. Our computational study reveals a dual-membrane bound conformation of Osh4, providing insights into protein-membrane interactions at membrane contact sites and their relevance to lipid transfer processes.
Collapse
Affiliation(s)
- Sharmistha Karmakar
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA
| | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA; Biophysics Graduate Program, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
16
|
Cabukusta B, Borst Pauwels S, Akkermans JJLL, Blomberg N, Mulder AA, Koning RI, Giera M, Neefjes J. The ORP9-ORP11 dimer promotes sphingomyelin synthesis. eLife 2024; 12:RP91345. [PMID: 39106189 PMCID: PMC11302984 DOI: 10.7554/elife.91345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024] Open
Abstract
Numerous lipids are heterogeneously distributed among organelles. Most lipid trafficking between organelles is achieved by a group of lipid transfer proteins (LTPs) that carry lipids using their hydrophobic cavities. The human genome encodes many intracellular LTPs responsible for lipid trafficking and the function of many LTPs in defining cellular lipid levels and distributions is unclear. Here, we created a gene knockout library targeting 90 intracellular LTPs and performed whole-cell lipidomics analysis. This analysis confirmed known lipid disturbances and identified new ones caused by the loss of LTPs. Among these, we found major sphingolipid imbalances in ORP9 and ORP11 knockout cells, two proteins of previously unknown function in sphingolipid metabolism. ORP9 and ORP11 form a heterodimer to localize at the ER-trans-Golgi membrane contact sites, where the dimer exchanges phosphatidylserine (PS) for phosphatidylinositol-4-phosphate (PI(4)P) between the two organelles. Consequently, loss of either protein causes phospholipid imbalances in the Golgi apparatus that result in lowered sphingomyelin synthesis at this organelle. Overall, our LTP knockout library toolbox identifies various proteins in control of cellular lipid levels, including the ORP9-ORP11 heterodimer, which exchanges PS and PI(4)P at the ER-Golgi membrane contact site as a critical step in sphingomyelin synthesis in the Golgi apparatus.
Collapse
Affiliation(s)
- Birol Cabukusta
- Cell and Chemical Biology, Oncode Institute, Leiden University Medical CenterLeidenNetherlands
| | - Shalom Borst Pauwels
- Cell and Chemical Biology, Oncode Institute, Leiden University Medical CenterLeidenNetherlands
| | - Jimmy JLL Akkermans
- Cell and Chemical Biology, Oncode Institute, Leiden University Medical CenterLeidenNetherlands
| | - Niek Blomberg
- Centre for Proteomics and Metabolomics, Leiden University Medical CenterLeidenNetherlands
| | - Aat A Mulder
- Electron Microscopy Facility, Cell and Chemical Biology, Leiden University Medical CenterLeidenNetherlands
| | - Roman I Koning
- Electron Microscopy Facility, Cell and Chemical Biology, Leiden University Medical CenterLeidenNetherlands
| | - Martin Giera
- Centre for Proteomics and Metabolomics, Leiden University Medical CenterLeidenNetherlands
| | - Jacques Neefjes
- Cell and Chemical Biology, Oncode Institute, Leiden University Medical CenterLeidenNetherlands
| |
Collapse
|
17
|
Srinivasan S, Di Luca A, Álvarez D, John Peter AT, Gehin C, Lone MA, Hornemann T, D’Angelo G, Vanni S. The conformational plasticity of structurally unrelated lipid transport proteins correlates with their mode of action. PLoS Biol 2024; 22:e3002737. [PMID: 39159271 PMCID: PMC11361750 DOI: 10.1371/journal.pbio.3002737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 08/29/2024] [Accepted: 07/05/2024] [Indexed: 08/21/2024] Open
Abstract
Lipid transfer proteins (LTPs) are key players in cellular homeostasis and regulation, as they coordinate the exchange of lipids between different cellular organelles. Despite their importance, our mechanistic understanding of how LTPs function at the molecular level is still in its infancy, mostly due to the large number of existing LTPs and to the low degree of conservation at the sequence and structural level. In this work, we use molecular simulations to characterize a representative dataset of lipid transport domains (LTDs) of 12 LTPs that belong to 8 distinct families. We find that despite no sequence homology nor structural conservation, the conformational landscape of LTDs displays common features, characterized by the presence of at least 2 main conformations whose populations are modulated by the presence of the bound lipid. These conformational properties correlate with their mechanistic mode of action, allowing for the interpretation and design of experimental strategies to further dissect their mechanism. Our findings indicate the existence of a conserved, fold-independent mechanism of lipid transfer across LTPs of various families and offer a general framework for understanding their functional mechanism.
Collapse
Affiliation(s)
| | - Andrea Di Luca
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Daniel Álvarez
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Departamento de Química Física y Analítica, Universidad de Oviedo, Oviedo, Spain
| | | | - Charlotte Gehin
- Institute of Bioengineering (IBI) and Global Heath Institute (GHI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Museer A. Lone
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Giovanni D’Angelo
- Institute of Bioengineering (IBI) and Global Heath Institute (GHI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Stefano Vanni
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- National Center of Competence in Research Bio-inspired Materials, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
18
|
Ajiki M, Yoshikawa M, Miyazaki T, Kawasaki A, Aoki K, Nakatsu F, Tsukiji S. ORP9-PH domain-based fluorescent reporters for visualizing phosphatidylinositol 4-phosphate dynamics in living cells. RSC Chem Biol 2024; 5:544-555. [PMID: 38846081 PMCID: PMC11151866 DOI: 10.1039/d3cb00232b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/15/2024] [Indexed: 06/09/2024] Open
Abstract
Fluorescent reporters that visualize phosphatidylinositol 4-phosphate (PI4P) in living cells are indispensable to elucidate the roles of this fundamental lipid in cell physiology. However, currently available PI4P reporters have limitations, such as Golgi-biased localization and low detection sensitivity. Here, we present a series of fluorescent PI4P reporters based on the pleckstrin homology (PH) domain of oxysterol-binding protein-related protein 9 (ORP9). We show that the green fluorescent protein AcGFP1-tagged ORP9-PH domain can be used as a fluorescent PI4P reporter to detect cellular PI4P across its wide distribution at multiple cellular locations, including the plasma membrane (PM), Golgi, endosomes, and lysosomes with high specificity and contrast. We also developed blue, red, and near-infrared fluorescent PI4P reporters suitable for multicolor fluorescence imaging experiments. Finally, we demonstrate the utility of the ORP9-PH domain-based reporter to visualize dynamic changes in the PI4P distribution and level in living cells upon synthetic ER-PM membrane contact manipulation and GPCR stimulation. This work offers a new set of genetically encoded fluorescent PI4P reporters that are practically useful for the study of PI4P biology.
Collapse
Affiliation(s)
- Moeka Ajiki
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
| | - Masaru Yoshikawa
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
| | - Tomoki Miyazaki
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University 1-757 Asahimachi, Chuo-ku Niigata 951-8510 Japan
| | - Kazuhiro Aoki
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences 5-1 Higashiyama, Myodaiji-cho Okazaki Aichi 444-8787 Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences 5-1 Higashiyama, Myodaiji-cho Okazaki Aichi 444-8787 Japan
- Department of Basic Biology, Faculty of Life Science, SOKENDAI (The Graduate University for Advanced Studies) 5-1 Higashiyama, Myodaiji-cho Okazaki Aichi 444-8787 Japan
| | - Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University 1-757 Asahimachi, Chuo-ku Niigata 951-8510 Japan
| | - Shinya Tsukiji
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
| |
Collapse
|
19
|
Li YE, Norris DM, Xiao FN, Pandzic E, Whan RM, Fok S, Zhou M, Du G, Liu Y, Du X, Yang H. Phosphatidylserine regulates plasma membrane repair through tetraspanin-enriched macrodomains. J Cell Biol 2024; 223:e202307041. [PMID: 38530252 PMCID: PMC10964951 DOI: 10.1083/jcb.202307041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/30/2023] [Accepted: 03/11/2024] [Indexed: 03/27/2024] Open
Abstract
The integrity of the plasma membrane is critical to cell function and survival. Cells have developed multiple mechanisms to repair damaged plasma membranes. A key process during plasma membrane repair is to limit the size of the damage, which is facilitated by the presence of tetraspanin-enriched rings surrounding damage sites. Here, we identify phosphatidylserine-enriched rings surrounding damaged sites of the plasma membrane, resembling tetraspanin-enriched rings. Importantly, the formation of both the phosphatidylserine- and tetraspanin-enriched rings requires phosphatidylserine and its transfer proteins ORP5 and ORP9. Interestingly, ORP9, but not ORP5, is recruited to the damage sites, suggesting cells acquire phosphatidylserine from multiple sources upon plasma membrane damage. We further demonstrate that ORP9 contributes to efficient plasma membrane repair. Our results thus unveil a role for phosphatidylserine and its transfer proteins in facilitating the formation of tetraspanin-enriched macrodomains and plasma membrane repair.
Collapse
Affiliation(s)
- Yang E. Li
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Dougall M. Norris
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Fanqian N. Xiao
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Elvis Pandzic
- Katerina Gaus Light Microscopy Facility, Mark Wainwright Analytical Center, University of New South Wales, Sydney, Australia
| | - Renee M. Whan
- Katerina Gaus Light Microscopy Facility, Mark Wainwright Analytical Center, University of New South Wales, Sydney, Australia
| | - Sandra Fok
- Katerina Gaus Light Microscopy Facility, Mark Wainwright Analytical Center, University of New South Wales, Sydney, Australia
| | - Ming Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yang Liu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ximing Du
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
20
|
Zhang Y, Lin C. Lipid osmosis, membrane tension, and other mechanochemical driving forces of lipid flow. Curr Opin Cell Biol 2024; 88:102377. [PMID: 38823338 PMCID: PMC11193448 DOI: 10.1016/j.ceb.2024.102377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/03/2024]
Abstract
Nonvesicular lipid transport among different membranes or membrane domains plays crucial roles in lipid homeostasis and organelle biogenesis. However, the forces that drive such lipid transport are not well understood. We propose that lipids tend to flow towards the membrane area with a higher membrane protein density in a process termed lipid osmosis. This process lowers the membrane tension in the area, resulting in a membrane tension difference called osmotic membrane tension. We examine the thermodynamic basis and experimental evidence of lipid osmosis and osmotic membrane tension. We predict that lipid osmosis can drive bulk lipid flows between different membrane regions through lipid transfer proteins, scramblases, or similar barriers that selectively pass lipids but not membrane proteins. We also speculate on the biological functions of lipid osmosis. Finally, we explore other driving forces for lipid transfer and describe potential methods and systems to further test our theory.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA.
| | - Chenxiang Lin
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Nanobiology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
21
|
Zhang Y, Lin C. Lipid osmosis, membrane tension, and other mechanochemical driving forces of lipid flow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574656. [PMID: 38260424 PMCID: PMC10802412 DOI: 10.1101/2024.01.08.574656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Nonvesicular lipid transport among different membranes or membrane domains plays crucial roles in lipid homeostasis and organelle biogenesis. However, the forces that drive such lipid transport are not well understood. We propose that lipids tend to flow towards the membrane area with a higher membrane protein density in a process termed lipid osmosis. This process lowers the membrane tension in the area, resulting in a membrane tension difference called osmotic membrane tension. We examine the thermodynamic basis and experimental evidence of lipid osmosis and osmotic membrane tension. We predict that lipid osmosis can drive bulk lipid flows between different membrane regions through lipid transfer proteins, scramblases, or other similar barriers that selectively pass lipids but not membrane proteins. We also speculate on the biological functions of lipid osmosis. Finally, we explore other driving forces for lipid transfer and describe potential methods and systems to further test our theory.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Chenxiang Lin
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
22
|
Panagiotou S, Tan KW, Nguyen PM, Müller A, Oqua AI, Tomas A, Wendt A, Eliasson L, Tengholm A, Solimena M, Idevall-Hagren O. OSBP-mediated PI(4)P-cholesterol exchange at endoplasmic reticulum-secretory granule contact sites controls insulin secretion. Cell Rep 2024; 43:113992. [PMID: 38536815 DOI: 10.1016/j.celrep.2024.113992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/07/2024] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
Insulin is packaged into secretory granules that depart the Golgi and undergo a maturation process that involves changes in the protein and lipid composition of the granules. Here, we show that insulin secretory granules form physical contacts with the endoplasmic reticulum and that the lipid exchange protein oxysterol-binding protein (OSBP) is recruited to these sites in a Ca2+-dependent manner. OSBP binding to insulin granules is positively regulated by phosphatidylinositol-4 (PI4)-kinases and negatively regulated by the PI4 phosphate (PI(4)P) phosphatase Sac2. Loss of Sac2 results in excess accumulation of cholesterol on insulin granules that is normalized when OSBP expression is reduced, and both acute inhibition and small interfering RNA (siRNA)-mediated knockdown of OSBP suppress glucose-stimulated insulin secretion without affecting insulin production or intracellular Ca2+ signaling. In conclusion, we show that lipid exchange at endoplasmic reticulum (ER)-granule contact sites is involved in the exocytic process and propose that these contacts act as reaction centers with multimodal functions during insulin granule maturation.
Collapse
Affiliation(s)
| | - Kia Wee Tan
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Phuoc My Nguyen
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Andreas Müller
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Affiong Ika Oqua
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Anna Wendt
- Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Diabetes Center (LUDC), Lund, Sweden
| | - Lena Eliasson
- Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Diabetes Center (LUDC), Lund, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Michele Solimena
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | |
Collapse
|
23
|
Hollingsworth LR, Veeraraghavan P, Paulo JA, Harper JW. Spatiotemporal proteomic profiling of cellular responses to NLRP3 agonists. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590338. [PMID: 38659763 PMCID: PMC11042255 DOI: 10.1101/2024.04.19.590338] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Nucleotide-binding domain and leucine-rich repeat pyrin-domain containing protein 3 (NLRP3) is an innate immune sensor that forms an inflammasome in response to various cellular stressors. Gain-of-function mutations in NLRP3 cause autoinflammatory diseases and NLRP3 signalling itself exacerbates the pathogenesis of many other human diseases. Despite considerable therapeutic interest, the primary drivers of NLRP3 activation remain controversial due to the diverse array of signals that are integrated through NLRP3. Here, we mapped subcellular proteome changes to lysosomes, mitochondrion, EEA1-positive endosomes, and Golgi caused by the NLRP3 inflammasome agonists nigericin and CL097. We identified several common disruptions to retrograde trafficking pathways, including COPI and Shiga toxin-related transport, in line with recent studies. We further characterized mouse NLRP3 trafficking throughout its activation using temporal proximity proteomics, which supports a recent model of NLRP3 recruitment to endosomes during inflammasome activation. Collectively, these findings provide additional granularity to our understanding of the molecular events driving NLRP3 activation and serve as a valuable resource for cell biological research. We have made our proteomics data accessible through an open-access Shiny browser to facilitate future research within the community, available at: https://harperlab.connect.hms.harvard.edu/inflame/. We will display anonymous peer review for this manuscript on pubpub.org (https://harperlab.pubpub.org/pub/nlrp3/) rather than a traditional journal. Moreover, we invite community feedback on the pubpub version of this manuscript, and we will address criticisms accordingly.
Collapse
Affiliation(s)
- L. Robert Hollingsworth
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| | | | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| | - J. Wade Harper
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| |
Collapse
|
24
|
Fuggetta N, Rigolli N, Magdeleine M, Hamaï A, Seminara A, Drin G. Reconstitution of ORP-mediated lipid exchange coupled to PI4P metabolism. Proc Natl Acad Sci U S A 2024; 121:e2315493121. [PMID: 38408242 PMCID: PMC10927502 DOI: 10.1073/pnas.2315493121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/24/2024] [Indexed: 02/28/2024] Open
Abstract
Oxysterol-binding protein-related proteins (ORPs) play key roles in the distribution of lipids in eukaryotic cells by exchanging sterol or phosphatidylserine for PI4P between the endoplasmic reticulum (ER) and other cell regions. However, it is unclear how their exchange capacity is coupled to PI4P metabolism. To address this question quantitatively, we analyze the activity of a representative ORP, Osh4p, in an ER/Golgi interface reconstituted with ER- and Golgi-mimetic membranes functionalized with PI4P phosphatase Sac1p and phosphatidylinositol (PI) 4-kinase, respectively. Using real-time assays, we demonstrate that upon adenosine triphosphate (ATP) addition, Osh4p creates a sterol gradient between these membranes, relying on the spatially distant synthesis and hydrolysis of PI4P, and quantify how much PI4P is needed for this process. Then, we develop a quantitatively accurate kinetic model, validated by our data, and extrapolate this to estimate to what extent PI4P metabolism can drive ORP-mediated sterol transfer in cells. Finally, we show that Sec14p can support PI4P metabolism and Osh4p activity by transferring PI between membranes. This study establishes that PI4P synthesis drives ORP-mediated lipid exchange and that ATP energy is needed to generate intermembrane lipid gradients. Furthermore, it defines to what extent ORPs can distribute lipids in the cell and reassesses the role of PI-transfer proteins in PI4P metabolism.
Collapse
Affiliation(s)
- Nicolas Fuggetta
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne06560, France
| | - Nicola Rigolli
- Department of Physics, École Normale Supérieure (LPENS), Paris75005, France
| | - Maud Magdeleine
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne06560, France
| | - Amazigh Hamaï
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne06560, France
| | - Agnese Seminara
- Malga, Department of Civil, Chemical and Environmental Engineering, University of Genoa, Genoa16145, Italy
| | - Guillaume Drin
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne06560, France
| |
Collapse
|
25
|
Doyle CP, Timple L, Hammond GRV. OSBP is a major determinant of Golgi phosphatidylinositol 4-phosphate homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.21.572879. [PMID: 38187665 PMCID: PMC10769437 DOI: 10.1101/2023.12.21.572879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The lipid phosphatidylinositol 4-phosphate (PI4P) plays a master regulatory role at Golgi membranes, orchestrating membrane budding, non-vesicular lipid transport and membrane organization. It follows that harmonious Golgi function requires strictly maintained PI4P homeostasis. One of the most abundant PI4P effector proteins is the oxysterol binding protein (OSBP), a lipid transfer protein that exchanges trans Golgi PI4P for ER cholesterol. Although this protein consumes PI4P as part of its lipid anti-porter function, whether it actively contributes to Golgi PI4P homeostasis has been questioned. Here, we employed a series of acute and chronic genetic manipulations, together with orthogonal targeting of OSBP, to interrogate its control over Golgi PI4P abundance. Modulating OSBP levels at ER:Golgi membrane contact sites produces reciprocal changes in PI4P levels. Additionally, we observe that OSBP has a high capacity for PI4P turnover, even at orthogonal organelle membranes. However, despite also visiting the plasma membrane, endogenous OSBP makes no impact on PI4P levels in this compartment. We conclude that OSBP is a major determinant of Golgi PI4P homeostasis.
Collapse
Affiliation(s)
- Colleen P Doyle
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Liz Timple
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
26
|
Doyle CP, Rectenwald A, Timple L, Hammond GRV. Orthogonal targeting of SAC1 to mitochondria implicates ORP2 as a major player in PM PI4P turnover. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.28.555163. [PMID: 37693626 PMCID: PMC10491111 DOI: 10.1101/2023.08.28.555163] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Oxysterol binding protein (OSBP)-related proteins (ORPs) 5 and 8 have been shown to deplete the lipid phosphatidylinositol 4-phosphate (PI4P) at sites of membrane contact between the endoplasmic reticulum (ER) and plasma membrane (PM). This is believed to be caused by transport of PI4P from the PM to the ER, where PI4P is degraded by an ER-localized SAC1 phosphatase. This is proposed to power the anti-port of phosphatidylserine (PS) lipids from ER to PM, up their concentration gradient. Alternatively, ORPs have been proposed to sequester PI4P, dependent on the concentration of their alternative lipid ligand. Here, we aimed to distinguish these possibilities in living cells by orthogonal targeting of PI4P transfer and degradation to PM-mitochondria contact sites. Surprisingly, we found that orthogonal targeting of SAC1 to mitochondria enhanced PM PI4P turnover independent of targeting to contact sites with the PM. This turnover could be slowed by knock-down of soluble ORP2, which also has a major impact on PM PI4P levels even without SAC1 over-expression. The data reveal a role for contact site-independent modulation of PM PI4P levels and lipid antiport.
Collapse
Affiliation(s)
- Colleen P Doyle
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Andrew Rectenwald
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Liz Timple
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
27
|
Doyle CP, Rectenwald A, Timple L, Hammond GRV. Orthogonal Targeting of SAC1 to Mitochondria Implicates ORP2 as a Major Player in PM PI4P Turnover. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241229272. [PMID: 38327560 PMCID: PMC10848804 DOI: 10.1177/25152564241229272] [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: 08/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
Oxysterol-binding protein (OSBP)-related proteins (ORPs) 5 and 8 have been shown to deplete the lipid phosphatidylinositol 4-phosphate (PI4P) at sites of membrane contact between the endoplasmic reticulum (ER) and plasma membrane (PM). This is believed to be caused by transport of PI4P from the PM to the ER, where PI4P is degraded by an ER-localized SAC1 phosphatase. This is proposed to power the anti-port of phosphatidylserine (PS) lipids from ER to PM, up their concentration gradient. Alternatively, ORPs have been proposed to sequester PI4P, dependent on the concentration of their alternative lipid ligand. Here, we aimed to distinguish these possibilities in living cells by orthogonal targeting of PI4P transfer and degradation to PM-mitochondria contact sites. Surprisingly, we found that orthogonal targeting of SAC1 to mitochondria enhanced PM PI4P turnover independent of targeting to contact sites with the PM. This turnover could be slowed by knock-down of soluble ORP2, which also has a major impact on PM PI4P levels even without SAC1 over-expression. The data reveal a role for contact site-independent modulation of PM PI4P levels and lipid antiport.
Collapse
Affiliation(s)
- Colleen P. Doyle
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Andrew Rectenwald
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Liz Timple
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Gerald R. V. Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
28
|
Mizuike A, Hanada K. DGARM/C10orf76/ARMH3 for Ceramide Transfer Zone at the Endoplasmic Reticulum-Distal Golgi Contacts. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241239443. [PMID: 38515862 PMCID: PMC10956147 DOI: 10.1177/25152564241239443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Phosphatidylinositol 4-monophosphate (PtdIns(4)P) is one of the key membrane components which mark the membrane contact sites. In the mammalian Golgi complex, PtdIns(4)P is produced at various subregions via specific mechanisms for each site. Particularly, PtdIns(4)P pools generated at the distal Golgi regions are pivotal for the determination of membrane contacts between the endoplasmic reticulum (ER) and Golgi, at which inter-organelle lipid transport takes place. In this short review, we will focus on C10orf76 (or ARMH3), which we propose to rename as DGARM after a distal Golgi armadillo repeat protein, for its function in generating a PtdIns(4)P pool crucial for ER-to-distal Golgi ceramide transport. We further discuss from the viewpoint of the evolutionary conservation of DGARM.
Collapse
Affiliation(s)
- Aya Mizuike
- Department of Quality Assurance, Radiation Safety and Information System, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kentaro Hanada
- Department of Quality Assurance, Radiation Safety and Information System, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
29
|
Doyle CP, Timple L, Hammond GRV. OSBP is a Major Determinant of Golgi Phosphatidylinositol 4-Phosphate Homeostasis. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241232196. [PMID: 38405037 PMCID: PMC10893830 DOI: 10.1177/25152564241232196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/27/2024]
Abstract
The lipid phosphatidylinositol 4-phosphate (PI4P) plays a master regulatory role at Golgi membranes, orchestrating membrane budding, non-vesicular lipid transport and membrane organization. It follows that harmonious Golgi function requires strictly maintained PI4P homeostasis. One of the most abundant PI4P effector proteins is the oxysterol binding protein (OSBP), a lipid transfer protein that exchanges trans-Golgi PI4P for ER cholesterol. Although this protein consumes PI4P as part of its lipid anti-porter function, whether it actively contributes to Golgi PI4P homeostasis has been questioned. Here, we employed a series of acute and chronic genetic manipulations, together with orthogonal targeting of OSBP, to interrogate its control over Golgi PI4P abundance. Modulating OSBP levels at ER:Golgi membrane contact sites produces reciprocal changes in PI4P levels. Additionally, we observe that OSBP has a high capacity for PI4P turnover, even at orthogonal organelle membranes. However, despite also visiting the plasma membrane, endogenous OSBP makes no impact on PI4P levels in this compartment. We conclude that OSBP is a major determinant of Golgi PI4P homeostasis.
Collapse
Affiliation(s)
- Colleen P. Doyle
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Liz Timple
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gerald R. V. Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Olkkonen VM, Ikonen E. Getting to Grips with the Oxysterol-Binding Protein Family - a Forty Year Perspective. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241273598. [PMID: 39210909 PMCID: PMC11359446 DOI: 10.1177/25152564241273598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
This review discusses how research around the oxysterol-binding protein family has evolved. We briefly summarize how this protein family, designated OSBP-related (ORP) or OSBP-like (OSBPL) proteins, was discovered, how protein domains highly conserved among family members between taxa paved the way for understanding their mechanisms of action, and how insights into protein structural and functional features help to understand their versatility as lipid transporters. We also discuss questions and future avenues of research opened by these findings. The investigations on oxysterol-binding protein family serve as a real-life example of the notion that science often advances as a collective effort of multiple lines of enquiry, including serendipitous routes. While original articles invariably explain the motivation of the research undertaken in rational terms, the actual paths to findings may be less intentional. Fortunately, this does not reduce the impact of the discoveries made. Besides hopefully providing a useful account of ORP family proteins, we aim to convey this message.
Collapse
Affiliation(s)
- Vesa M. Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Elina Ikonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Faculty of Medicine, Dept of Anatomy and Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Heckle LA, Kozminski KG. Osh-dependent and -independent Regulation of PI4P Levels During Polarized Growth of Saccharomyces cerevisiae. Mol Biol Cell 2023; 34:ar104. [PMID: 37556206 PMCID: PMC10559303 DOI: 10.1091/mbc.e23-03-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/03/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
Polarized secretion facilitates polarized cell growth. For a secretory vesicle to dock at the plasma membrane, it must mature with a progressive association or dissociation of molecules that are, respectively, necessary for or inhibitory to vesicle docking, including an exchange of Rab GTPases. In current models, oxysterol-binding protein homologue 4 (Osh4p) establishes a phosphatidylinositol 4-phosphate (PI4P) gradient along the secretory trafficking pathway such that vesicles have higher PI4P levels after budding from the trans-Golgi relative to when vesicles arrive at the plasma membrane. In this study, using the lipid-binding domain P4M and live-cell imaging, we show that secretory vesicle-associated PI4P levels remain constant when vesicles traffic from the trans-Golgi to the plasma membrane. We also show that deletion of OSH4 does not alter vesicle-associated PI4P levels, though loss of any individual member of the OSH family or complete loss of OSH family function alters the intracellular distribution of PI4P. We propose a model in which the Rab GTPases Ypt32p and Sec4p remain associated with a secretory vesicle during trafficking, independent of PI4P levels and Osh4p. Together these data indicate the necessity of experiments revealing the location and timing of events required for vesicle maturation.
Collapse
Affiliation(s)
- Lindsay A. Heckle
- Department of Biology, University of Virginia, Charlottesville, VA 22904
| | - Keith G. Kozminski
- Department of Biology, University of Virginia, Charlottesville, VA 22904
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
32
|
Wang X, Liu J, Azoitei A, Eiseler T, Meessen S, Jiang W, Zheng X, Makori AW, Eckstein M, Hartmann A, Stilgenbauer S, Elati M, Hohwieler M, Kleger A, John A, Zengerling F, Wezel F, Bolenz C, Günes C. Loss of ORP3 induces aneuploidy and promotes bladder cancer cell invasion through deregulated microtubule and actin dynamics. Cell Mol Life Sci 2023; 80:299. [PMID: 37740130 PMCID: PMC10516806 DOI: 10.1007/s00018-023-04959-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/24/2023]
Abstract
We have recently shown that loss of ORP3 leads to aneuploidy induction and promotes tumor formation. However, the specific mechanisms by which ORP3 contributes to ploidy-control and cancer initiation and progression is still unknown. Here, we report that ORP3 is highly expressed in ureter and bladder epithelium while its expression is downregulated in invasive bladder cancer cell lines and during tumor progression, both in human and in mouse bladder cancer. Moreover, we observed an increase in the incidence of N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN)-induced invasive bladder carcinoma in the tissue-specific Orp3 knockout mice. Experimental data demonstrate that ORP3 protein interacts with γ-tubulin at the centrosomes and with components of actin cytoskeleton. Altering the expression of ORP3 induces aneuploidy and genomic instability in telomerase-immortalized urothelial cells with a stable karyotype and influences the migration and invasive capacity of bladder cancer cell lines. These findings demonstrate a crucial role of ORP3 in ploidy-control and indicate that ORP3 is a bona fide tumor suppressor protein. Of note, the presented data indicate that ORP3 affects both cell invasion and migration as well as genome stability through interactions with cytoskeletal components, providing a molecular link between aneuploidy and cell invasion and migration, two crucial characteristics of metastatic cells.
Collapse
Affiliation(s)
- Xue Wang
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Junnan Liu
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Anca Azoitei
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine I, University Hospital, Ulm, Germany
| | - Sabine Meessen
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
- Division of Hepatology, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Wencheng Jiang
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
| | - Xi Zheng
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
- Department of Urology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
- Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Arika W Makori
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
| | - Markus Eckstein
- Institute of Pathology, Friedrich-Alexander University, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander University, Erlangen, Germany
| | | | - Mohamed Elati
- CANTHER, ONCOLille Institute, University of Lille, CNRS UMR 1277, Inserm U9020, 59045, Lille Cedex, France
| | - Meike Hohwieler
- Institute of Mol. Oncology and Stem Cell Biology, University Hospital, Ulm, Germany
| | - Alexander Kleger
- Institute of Mol. Oncology and Stem Cell Biology, University Hospital, Ulm, Germany
| | - Axel John
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
| | - Friedemann Zengerling
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
| | - Felix Wezel
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
| | - Christian Bolenz
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany
| | - Cagatay Günes
- Department of Urology, Ulm University Hospital, Helmholtzstr. 10, 89081, Ulm, Germany.
| |
Collapse
|
33
|
Hempelmann P, Lolicato F, Graziadei A, Brown RDR, Spiegel S, Rappsilber J, Nickel W, Höglinger D, Jamecna D. The sterol transporter STARD3 transports sphingosine at ER-lysosome contact sites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.557036. [PMID: 37790546 PMCID: PMC10542139 DOI: 10.1101/2023.09.18.557036] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Sphingolipids are important structural components of membranes. Additionally, simple sphingolipids such as sphingosine are highly bioactive and participate in complex subcellular signaling. Sphingolipid deregulation is associated with many severe diseases including diabetes, Parkinson's and cancer. Here, we focus on how sphingosine, generated from sphingolipid catabolism in late endosomes/lysosomes, is reintegrated into the biosynthetic machinery at the endoplasmic reticulum (ER). We characterized the sterol transporter STARD3 as a sphingosine transporter acting at lysosome-ER contact sites. Experiments featuring crosslinkable sphingosine probes, supported by unbiased molecular dynamics simulations, exposed how sphingosine binds to the lipid-binding domain of STARD3. Following the metabolic fate of pre-localized lysosomal sphingosine showed the importance of STARD3 and its actions at contact sites for the integration of sphingosine into ceramide in a cellular context. Our findings provide the first example of interorganellar sphingosine transfer and pave the way for a better understanding of sphingolipid - sterol co-regulation.
Collapse
Affiliation(s)
- Pia Hempelmann
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
| | - Fabio Lolicato
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
- Department of Physics, University of Helsinki, FI-00014 Helsinki, Finland
| | - Andrea Graziadei
- Institute for Biotechnology, Technical University Berlin, Gustav Mayer Allee 25, 13355 Berlin
| | - Ryan D R Brown
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Juri Rappsilber
- Institute for Biotechnology, Technical University Berlin, Gustav Mayer Allee 25, 13355 Berlin
| | - Walter Nickel
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
| | - Doris Höglinger
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
| | - Denisa Jamecna
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
| |
Collapse
|
34
|
Nenadic A, Zaman MF, Johansen J, Volpiana MW, Beh CT. Increased Phospholipid Flux Bypasses Overlapping Essential Requirements for the Yeast Sac1p Phosphoinositide Phosphatase and ER-PM Membrane Contact Sites. J Biol Chem 2023; 299:105092. [PMID: 37507017 PMCID: PMC10470028 DOI: 10.1016/j.jbc.2023.105092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
In budding yeast cells, much of the inner surface of the plasma membrane (PM) is covered with the endoplasmic reticulum (ER). This association is mediated by seven ER membrane proteins that confer cortical ER-PM association at membrane contact sites (MCSs). Several of these membrane "tether" proteins are known to physically interact with the phosphoinositide phosphatase Sac1p. However, it is unclear how or if these interactions are necessary for their interdependent functions. We find that SAC1 inactivation in cells lacking the homologous synaptojanin-like genes INP52 and INP53 results in a significant increase in cortical ER-PM MCSs. We show in sac1Δ, sac1tsinp52Δ inp53Δ, or Δ-super-tether (Δ-s-tether) cells lacking all seven ER-PM tethering genes that phospholipid biosynthesis is disrupted and phosphoinositide distribution is altered. Furthermore, SAC1 deletion in Δ-s-tether cells results in lethality, indicating a functional overlap between SAC1 and ER-PM tethering genes. Transcriptomic profiling indicates that SAC1 inactivation in either Δ-s-tether or inp52Δ inp53Δ cells induces an ER membrane stress response and elicits phosphoinositide-dependent changes in expression of autophagy genes. In addition, by isolating high-copy suppressors that rescue sac1Δ Δ-s-tether lethality, we find that key phospholipid biosynthesis genes bypass the overlapping function of SAC1 and ER-PM tethers and that overexpression of the phosphatidylserine/phosphatidylinositol-4-phosphate transfer protein Osh6 also provides limited suppression. Combined with lipidomic analysis and determinations of intracellular phospholipid distributions, these results suggest that Sac1p and ER phospholipid flux controls lipid distribution to drive Osh6p-dependent phosphatidylserine/phosphatidylinositol-4-phosphate counter-exchange at ER-PM MCSs.
Collapse
Affiliation(s)
- Aleksa Nenadic
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Mohammad F Zaman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Jesper Johansen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Matthew W Volpiana
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Christopher T Beh
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada; Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada.
| |
Collapse
|
35
|
Fuggetta N, Rigolli N, Magdeleine M, Seminara A, Drin G. Reconstitution of ORP-mediated lipid exchange process coupled to PI(4)P metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.551917. [PMID: 37577629 PMCID: PMC10418177 DOI: 10.1101/2023.08.04.551917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Lipid distribution in the eukaryotic cells depends on tight couplings between lipid transfer and lipid metabolism. Yet these couplings remain poorly described. Notably, it is unclear to what extent lipid exchangers of the OSBP-related proteins (ORPs) family, coupled to PI(4)P metabolism, contribute to the formation of sterol and phosphatidylserine gradient between the endoplasmic reticulum (ER) and other cell regions. To address this question, we have examined in vitro the activity of Osh4p, a representative ORP, between Golgi mimetic membranes in which PI(4)P is produced by a PI 4-kinase and ER mimetic membranes in which PI(4)P is hydrolyzed by the phosphatase Sac1p. Using quantitative, real-time assays, we demonstrate that Osh4p creates a sterol gradient between the two membranes by sterol/PI(4)P exchange as soon as a PI(4)P gradient is generated at this interface following ATP addition, and define how much PI(4)P must be synthesized for this process. Then, using a kinetic model supported by our in vitro data, we estimate to what extent PI(4)P metabolism can drive lipid transfer in cells. Finally, we show that Sec14p, by transferring phosphatidylinositol between membranes, can support the synthesis of PI(4)P and the creation of a sterol gradient by Osh4p. These results indicate to what extent ORPs, under the control of PI(4)P metabolism, can distribute lipids in the cell.
Collapse
Affiliation(s)
- 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
| | - Nicola Rigolli
- Laboratoire de Physique, École Normale Supérieure (LPENS), 75005 Paris, 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
| | - Agnese Seminara
- Malga, Department of Civil, Chemical and Environmental Engineering, University of Genoa, Villa Cambiaso 1, 16145 Genoa, Italy
| | - 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
|
36
|
Abstract
Cholesterol is an essential lipid species of mammalian cells. Cells acquire it through synthesis in the endoplasmic reticulum (ER) and uptake from lipoprotein particles. Newly synthesized cholesterol is efficiently distributed from the ER to other organelles via lipid-binding/transfer proteins concentrated at membrane contact sites (MCSs) to reach the trans-Golgi network, endosomes, and plasma membrane. Lipoprotein-derived cholesterol is exported from the plasma membrane and endosomal compartments via a combination of vesicle/tubule-mediated membrane transport and transfer through MCSs. In this review, we provide an overview of intracellular cholesterol trafficking pathways, including cholesterol flux from the ER to other membranes, cholesterol uptake from lipoprotein donors and transport from the plasma membrane to the ER, cellular cholesterol efflux to lipoprotein acceptors, as well as lipoprotein cholesterol secretion from enterocytes, hepatocytes, and astrocytes. We also briefly discuss human diseases caused by defects in these processes and therapeutic strategies available in such conditions.
Collapse
Affiliation(s)
- Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00100 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| |
Collapse
|
37
|
Khaddaj R, Kukulski W. Piecing together the structural organisation of lipid exchange at membrane contact sites. Curr Opin Cell Biol 2023; 83:102212. [PMID: 37515839 DOI: 10.1016/j.ceb.2023.102212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/31/2023]
Abstract
Membrane contact sites (MCSs) are areas of close proximity between organelles, implicated in transport of small molecules and in organelle biogenesis. Lipid transfer proteins at MCSs facilitate the distribution of lipid species between organelle membranes. Such exchange processes rely on the apposition of two different membranes delimiting distinct compartments and a cytosolic intermembrane space. Maintaining organelle identity while transferring molecules therefore implies control over MCS architecture both on the ultrastructural and molecular levels. Factors including intermembrane distance, density of resident proteins, and contact surface area fine-tune MCS function. Furthermore, the structural arrangement of lipid transfer proteins and associated proteins underpins the molecular mechanisms of lipid fluxes at MCSs. Thus, the architecture of MCSs emerges as an essential aspect of their function.
Collapse
Affiliation(s)
- Rasha Khaddaj
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Wanda Kukulski
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| |
Collapse
|
38
|
Limar S, Körner C, Martínez-Montañés F, Stancheva VG, Wolf VN, Walter S, Miller EA, Ejsing CS, Galassi VV, Fröhlich F. Yeast Svf1 binds ceramides and contributes to sphingolipid metabolism at the ER cis-Golgi interface. J Cell Biol 2023; 222:e202109162. [PMID: 36897280 PMCID: PMC10038888 DOI: 10.1083/jcb.202109162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2022] [Accepted: 02/03/2023] [Indexed: 03/11/2023] Open
Abstract
Ceramides are essential precursors of complex sphingolipids and act as potent signaling molecules. Ceramides are synthesized in the endoplasmic reticulum (ER) and receive their head-groups in the Golgi apparatus, yielding complex sphingolipids (SPs). Transport of ceramides between the ER and the Golgi is executed by the essential ceramide transport protein (CERT) in mammalian cells. However, yeast cells lack a CERT homolog, and the mechanism of ER to Golgi ceramide transport remains largely elusive. Here, we identified a role for yeast Svf1 in ceramide transport between the ER and the Golgi. Svf1 is dynamically targeted to membranes via an N-terminal amphipathic helix (AH). Svf1 binds ceramide via a hydrophobic binding pocket that is located in between two lipocalin domains. We showed that Svf1 membrane-targeting is important to maintain flux of ceramides into complex SPs. Together, our results show that Svf1 is a ceramide binding protein that contributes to sphingolipid metabolism at Golgi compartments.
Collapse
Affiliation(s)
- Sergej Limar
- Department of Biology/Chemistry Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
| | - Carolin Körner
- Department of Biology/Chemistry Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
| | - Fernando Martínez-Montañés
- Department of Biochemistry and Molecular Biology Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | | | - Verena N. Wolf
- Department of Biology/Chemistry Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
| | - Stefan Walter
- Osnabrück University Center of Cellular Nanoanalytic Osnabrück (CellNanOs), Osnabrück, Germany
| | | | - Christer S. Ejsing
- Department of Biochemistry and Molecular Biology Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vanesa Viviana Galassi
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
- Instituto Interdisciplinario de Ciencias Básicas (ICB), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Florian Fröhlich
- Department of Biology/Chemistry Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
- Osnabrück University Center of Cellular Nanoanalytic Osnabrück (CellNanOs), Osnabrück, Germany
| |
Collapse
|
39
|
Vormittag S, Ende RJ, Derré I, Hilbi H. Pathogen vacuole membrane contact sites - close encounters of the fifth kind. MICROLIFE 2023; 4:uqad018. [PMID: 37223745 PMCID: PMC10117887 DOI: 10.1093/femsml/uqad018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 05/25/2023]
Abstract
Vesicular trafficking and membrane fusion are well-characterized, versatile, and sophisticated means of 'long range' intracellular protein and lipid delivery. Membrane contact sites (MCS) have been studied in far less detail, but are crucial for 'short range' (10-30 nm) communication between organelles, as well as between pathogen vacuoles and organelles. MCS are specialized in the non-vesicular trafficking of small molecules such as calcium and lipids. Pivotal MCS components important for lipid transfer are the VAP receptor/tether protein, oxysterol binding proteins (OSBPs), the ceramide transport protein CERT, the phosphoinositide phosphatase Sac1, and the lipid phosphatidylinositol 4-phosphate (PtdIns(4)P). In this review, we discuss how these MCS components are subverted by bacterial pathogens and their secreted effector proteins to promote intracellular survival and replication.
Collapse
Affiliation(s)
| | | | - Isabelle Derré
- Corresponding author. Department of Microbiology, Immunology and Cancer Biology, University of Virginia, 1340 Jefferson Park Ave, Charlottesville, VA 22908, United States. Tel: +1-434-924-2330; E-mail:
| | - Hubert Hilbi
- Corresponding author. Institute of Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006 Zürich, Switzerland. Tel: +41-44-634-2650; E-mail:
| |
Collapse
|
40
|
Guillén-Samander A, De Camilli P. Endoplasmic Reticulum Membrane Contact Sites, Lipid Transport, and Neurodegeneration. Cold Spring Harb Perspect Biol 2023; 15:a041257. [PMID: 36123033 PMCID: PMC10071438 DOI: 10.1101/cshperspect.a041257] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The Endoplasmic Reticulum (ER) is an endomembrane system that plays a multiplicity of roles in cell physiology and populates even the most distal cell compartments, including dendritic tips and axon terminals of neurons. Some of its functions are achieved by a cross talk with other intracellular membranous organelles and with the plasma membrane at membrane contacts sites (MCSs). As the ER synthesizes most membrane lipids, lipid exchanges mediated by lipid transfer proteins at MCSs are a particularly important aspect of this cross talk, which synergizes with the cross talk mediated by vesicular transport. Several mutations of genes that encode proteins localized at ER MCSs result in familial neurodegenerative diseases, emphasizing the importance of the normal lipid traffic within cells for a healthy brain. Here, we provide an overview of such diseases, with a specific focus on proteins that directly or indirectly impact lipid transport.
Collapse
Affiliation(s)
- Andrés Guillén-Samander
- Departments of Neuroscience and of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Pietro De Camilli
- Departments of Neuroscience and of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
41
|
Liu X, Li C, Chen Y, Xue Z, Miao J, Liu X. Untargeted lipidomics reveals lipid metabolism disorders induced by oxathiapiprolin in Phytophthora sojae. PEST MANAGEMENT SCIENCE 2023; 79:1593-1603. [PMID: 36562252 DOI: 10.1002/ps.7334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/12/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Oxathiapiprolin, an oxysterol-binding protein inhibitor (OSBPI), shows unexceptionable inhibitory activity against plant pathogenic oomycetes. FRAC (Fungicide Resistance Action Committee) classifies it into the mode of action group F9 (lipid homeostasis and transfer/storage), but very little is known about the lipid metabolism of oomycete pathogens when subjected to oxathiapiprolin. RESULTS In this study, seven lipid categories and 1435 lipid molecules were identified in Phytophthora sojae, among which glycerolipids, glycerophospholipids, and sphingolipids account for 30.10%, 50.59%, and 7.28%, respectively. These lipids were categorized into 31 subclasses, which varied to different extents when treated with oxathiapiprolin. A total of 11 lipid subclasses showed significant changes. Among them, 10 lipid subclasses, lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), phosphatidylcholine (PC), phosphatidylserine (PS), ceramide (Cer), triglyceride (TG), (o-acyl)-1-hydroxy fatty acid, diglycosylceramide, sphingoshine (So), and sitosterol ester, were significantly up-regulated, while digalactosyldiacylglycerol was the only lipid that was significantly down-regulated by a factor of almost three. These lipid molecules were further analyzed at the lipid species level. A total of 542 species were significantly altered when treated with oxathiapiprolin, including 212 glycerolipids [186 TG and 26 diglycerides (DG)], 167 glycerophospholipids (38 PC, 15 LPC, 19 LPE, seven PS, etc.), 156 sphingolipids (146 Cer, four So, etc.), and some other lipid molecules. Finally, from the orthogonal partial least-squares discrimination analysis model, variable importance for the projection score analysis showed that Cer, TG, and some glycerophospholipids contribute to the metabolic disorder when subjected to oxathiapiprolin. CONCLUSION Glycerolipids, glycerophospholipids, and sphingolipids in P. sojae undergo significant changes with oxathiapiprolin treatment. These results provided valuable information for further understanding the function of the target protein and the mode of action of OSBPIs in oomycetes. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaofei Liu
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Chengcheng Li
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Yue Chen
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Ziwei Xue
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Jianqiang Miao
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Xili Liu
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
42
|
Fang R, Jiang Q, Jia X, Jiang Z. ARMH3-mediated recruitment of PI4KB directs Golgi-to-endosome trafficking and activation of the antiviral effector STING. Immunity 2023; 56:500-515.e6. [PMID: 36921576 DOI: 10.1016/j.immuni.2023.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/30/2022] [Accepted: 01/26/2023] [Indexed: 03/15/2023]
Abstract
The cGAS-STING pathway mediates cytoplasmic DNA-triggered innate immunity. STING activation is initiated by cyclic-GMP-AMP (cGAMP)-induced translocation from the endoplasmic reticulum and sulfated glycosaminoglycans-induced polymerization at the Golgi. Here, we examine the mechanisms underlying STING transport and activation beyond the Golgi. A genome-wide CRISPR-Cas9 screen identified Armadillo-like helical domain-containing protein 3 (ARMH3) as critical for STING activation. Upon cGAMP-triggered translocation, ARMH3 interacted with STING at the Golgi and recruited phosphatidylinositol 4-kinase beta (PI4KB) to synthesize PI4P, which directed STING Golgi-to-endosome trafficking via PI4P-binding proteins AP-1 and GGA2. Disrupting PI4P-dependent lipid transport through RNAi of other PI4P-binding proteins impaired STING activation. Consistently, disturbed lipid composition inhibited STING activation, whereas aberrantly elevated cellular PI4P led to cGAS-independent STING activation. Armh3fl/fllLyzCre/Cre mice were susceptible to DNA virus challenge in vivo. Thus, ARMH3 bridges STING and PIK4B to generate PI4P for STING transportation and activation, an interaction conserved in all eukaryotes.
Collapse
Affiliation(s)
- Run Fang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Qifei Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xinying Jia
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
43
|
Singh RP, Poh YP, Sinha SD, Wideman JG. Evolutionary History of Oxysterol-Binding Proteins Reveals Complex History of Duplication and Loss in Animals and Fungi. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2023; 6:25152564221150428. [PMID: 37366416 PMCID: PMC10243569 DOI: 10.1177/25152564221150428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 06/28/2023]
Abstract
Cells maintain the specific lipid composition of distinct organelles by vesicular transport as well as non-vesicular lipid trafficking via lipid transport proteins. Oxysterol-binding proteins (OSBPs) are a family of lipid transport proteins that transfer lipids at various membrane contact sites (MCSs). OSBPs have been extensively investigated in human and yeast cells where 12 have been identified in Homo sapiens and 7 in Saccharomyces cerevisiae. The evolutionary relationship between these well-characterized OSBPs is still unclear. By reconstructing phylogenies of eukaryote OSBPs, we show that the ancestral Saccharomycotina had four OSBPs, the ancestral fungus had five OSBPs, and the ancestral animal had six OSBPs, whereas the shared ancestor of animals and fungi as well as the ancestral eukaryote had only three OSBPs. Our analyses identified three undescribed ancient OSBP orthologues, one fungal OSBP (Osh8) lost in the lineage leading to yeast, one animal OSBP (ORP12) lost in the lineage leading to vertebrates, and one eukaryotic OSBP (OshEu) lost in both the animal and fungal lineages.
Collapse
Affiliation(s)
- Rohan P. Singh
- Center for Mechanisms of Evolution, Biodesign Institute,
School of Life Sciences, Arizona State University, Tempe, USA
| | - Yu-Ping Poh
- Center for Mechanisms of Evolution, Biodesign Institute,
School of Life Sciences, Arizona State University, Tempe, USA
| | - Savar D. Sinha
- Center for Mechanisms of Evolution, Biodesign Institute,
School of Life Sciences, Arizona State University, Tempe, USA
| | - Jeremy G. Wideman
- Center for Mechanisms of Evolution, Biodesign Institute,
School of Life Sciences, Arizona State University, Tempe, USA
| |
Collapse
|
44
|
Balla T, Gulyas G, Mandal A, Alvarez-Prats A, Niu Y, Kim YJ, Pemberton J. Roles of Phosphatidylinositol 4-Phosphorylation in Non-vesicular Cholesterol Trafficking. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:327-352. [PMID: 36988887 PMCID: PMC11135459 DOI: 10.1007/978-3-031-21547-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Cholesterol (Chol) is an essential component of all eukaryotic cell membranes that affects the function of numerous peripheral as well as integral membrane proteins. Chol is synthesized in the ER, but it is selectively enriched within the plasma membrane (PM) and other endomembranes, which requires Chol to cross the aqueous phase of the cytoplasm. In addition to the classical vesicular trafficking pathways that are known to facilitate the bulk transport of membrane intermediates, Chol is also transported via non-vesicular lipid transfer proteins that work primarily within specialized membrane contact sites. Some of these transport pathways work against established concentration gradients and hence require energy. Recent studies highlight the unique role of phosphoinositides (PPIns), and phosphatidylinositol 4-phosphate (PI4P) in particular, for the control of non-vesicular Chol transport. In this chapter, we will review the emerging connection between Chol, PPIns, and lipid transfer proteins that include the important family of oxysterol-binding protein related proteins, or ORPs.
Collapse
Affiliation(s)
- Tamas Balla
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD, USA.
| | | | - Amrita Mandal
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro Alvarez-Prats
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD, USA
| | | | - Yeun Ju Kim
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD, USA
| | - Joshua Pemberton
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
45
|
El Atab O, Schneiter R. Measuring the Interaction of Sterols and Steroids with Proteins by Microscale Thermophoresis. Methods Mol Biol 2023; 2704:173-181. [PMID: 37642844 DOI: 10.1007/978-1-0716-3385-4_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Determining the affinity and specificity of protein-lipid interactions is crucial for understanding the physiological function and mode of action of signaling lipids, including steroids. Here we describe a method that relies on microscale thermophoresis (MST) to monitor the binding of sterols and steroids to proteins. The protein of interest is expressed as a polyhistidine-tagged fusion in E. coli and purified by affinity chromatography on a nickel-based resin. The purified protein is then labeled with a fluorescent dye and incubated with a serial dilution of the lipid ligand. The protein-ligand interaction is monitored by MST, which detects the fraction of the protein bound to the ligand based on its altered mobility in a thermal gradient. A binding curve fitted to the measured data points is then used to calculate the corresponding dissociation constant.
Collapse
Affiliation(s)
- Ola El Atab
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
46
|
Enrich C, Lu A, Tebar F, Rentero C, Grewal T. Ca 2+ and Annexins - Emerging Players for Sensing and Transferring Cholesterol and Phosphoinositides via Membrane Contact Sites. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:393-438. [PMID: 36988890 DOI: 10.1007/978-3-031-21547-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Maintaining lipid composition diversity in membranes from different organelles is critical for numerous cellular processes. However, many lipids are synthesized in the endoplasmic reticulum (ER) and require delivery to other organelles. In this scenario, formation of membrane contact sites (MCS) between neighbouring organelles has emerged as a novel non-vesicular lipid transport mechanism. Dissecting the molecular composition of MCS identified phosphoinositides (PIs), cholesterol, scaffolding/tethering proteins as well as Ca2+ and Ca2+-binding proteins contributing to MCS functioning. Compelling evidence now exists for the shuttling of PIs and cholesterol across MCS, affecting their concentrations in distinct membrane domains and diverse roles in membrane trafficking. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) at the plasma membrane (PM) not only controls endo-/exocytic membrane dynamics but is also critical in autophagy. Cholesterol is highly concentrated at the PM and enriched in recycling endosomes and Golgi membranes. MCS-mediated cholesterol transfer is intensely researched, identifying MCS dysfunction or altered MCS partnerships to correlate with de-regulated cellular cholesterol homeostasis and pathologies. Annexins, a conserved family of Ca2+-dependent phospholipid binding proteins, contribute to tethering and untethering events at MCS. In this chapter, we will discuss how Ca2+ homeostasis and annexins in the endocytic compartment affect the sensing and transfer of cholesterol and PIs across MCS.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.
| | - Albert Lu
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
47
|
Basante-Bedoya MA, Bogliolo S, Garcia-Rodas R, Zaragoza O, Arkowitz RA, Bassilana M. Two distinct lipid transporters together regulate invasive filamentous growth in the human fungal pathogen Candida albicans. PLoS Genet 2022; 18:e1010549. [PMID: 36516161 PMCID: PMC9797089 DOI: 10.1371/journal.pgen.1010549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/28/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
Flippases transport lipids across the membrane bilayer to generate and maintain asymmetry. The human fungal pathogen Candida albicans has 5 flippases, including Drs2, which is critical for filamentous growth and phosphatidylserine (PS) distribution. Furthermore, a drs2 deletion mutant is hypersensitive to the antifungal drug fluconazole and copper ions. We show here that such a flippase mutant also has an altered distribution of phosphatidylinositol 4-phosphate [PI(4)P] and ergosterol. Analyses of additional lipid transporters, i.e. the flippases Dnf1-3, and all the oxysterol binding protein (Osh) family lipid transfer proteins, i.e. Osh2-4 and Osh7, indicate that they are not critical for filamentous growth. However, deletion of Osh4 alone, which exchanges PI(4)P for sterol, in a drs2 mutant can bypass the requirement for this flippase in invasive filamentous growth. In addition, deletion of the lipid phosphatase Sac1, which dephosphorylates PI(4)P, in a drs2 mutant results in a synthetic growth defect, suggesting that Drs2 and Sac1 function in parallel pathways. Together, our results indicate that a balance between the activities of two putative lipid transporters regulates invasive filamentous growth, via PI(4)P. In contrast, deletion of OSH4 in drs2 does not restore growth on fluconazole, nor on papuamide A, a toxin that binds PS in the outer leaflet of the plasma membrane, suggesting that Drs2 has additional role(s) in plasma membrane organization, independent of Osh4. As we show that C. albicans Drs2 localizes to different structures, including the Spitzenkörper, we investigated if a specific localization of Drs2 is critical for different functions, using a synthetic physical interaction approach to restrict/stabilize Drs2 at the Spitzenkörper. Our results suggest that the localization of Drs2 at the plasma membrane is critical for C. albicans growth on fluconazole and papuamide A, but not for invasive filamentous growth.
Collapse
Affiliation(s)
| | | | - Rocio Garcia-Rodas
- Université Côte d’Azur, CNRS, INSERM, iBV, Parc Valrose, Nice, FRANCE
- Mycology Reference Laboratory, National Centre for Microbiology, Health Institute Carlos III, Majadahonda, Madrid, Spain
| | - Oscar Zaragoza
- Mycology Reference Laboratory, National Centre for Microbiology, Health Institute Carlos III, Majadahonda, Madrid, Spain
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC-CB21/13/00105), Health Institute Carlos III, Madrid, Spain
| | | | - Martine Bassilana
- Université Côte d’Azur, CNRS, INSERM, iBV, Parc Valrose, Nice, FRANCE
- * E-mail:
| |
Collapse
|
48
|
Creating and sensing asymmetric lipid distributions throughout the cell. Emerg Top Life Sci 2022; 7:7-19. [PMID: 36373850 DOI: 10.1042/etls20220028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
A key feature of eukaryotic cells is the asymmetric distribution of lipids along their secretory pathway. Because of the biological significance of these asymmetries, it is crucial to define the mechanisms which create them. Extensive studies have led to the identification of lipid transfer proteins (LTPs) that work with lipid-synthesizing enzymes to carry lipids between two distinct membranes in a directional manner, and are thus able to create asymmetries in lipid distribution throughout the cell. These networks are often in contact sites where two organelle membranes are in close proximity for reasons we have only recently started to understand. A question is whether these networks transfer lipids en masse within the cells or adjust the lipid composition of organelle membranes. Finally, recent data have confirmed that some networks organized around LTPs do not generate lipid asymmetries between membranes but sense them and rectify the lipid content of the cell.
Collapse
|
49
|
Zhang Y, Ge J, Bian X, Kumar A. Quantitative Models of Lipid Transfer and Membrane Contact Formation. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2022; 5:1-21. [PMID: 36120532 DOI: 10.1177/25152564221096024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid transfer proteins (LTPs) transfer lipids between different organelles, and thus play key roles in lipid homeostasis and organelle dynamics. The lipid transfer often occurs at the membrane contact sites (MCS) where two membranes are held within 10-30 nm. While most LTPs act as a shuttle to transfer lipids, recent experiments reveal a new category of eukaryotic LTPs that may serve as a bridge to transport lipids in bulk at MCSs. However, the molecular mechanisms underlying lipid transfer and MCS formation are not well understood. Here, we first review two recent studies of extended synaptotagmin (E-Syt)-mediated membrane binding and lipid transfer using novel approaches. Then we describe mathematical models to quantify the kinetics of lipid transfer by shuttle LTPs based on a lipid exchange mechanism. We find that simple lipid mixing among membranes of similar composition and/or lipid partitioning among membranes of distinct composition can explain lipid transfer against a concentration gradient widely observed for LTPs. We predict that selective transport of lipids, but not membrane proteins, by bridge LTPs leads to osmotic membrane tension by analogy to the osmotic pressure across a semipermeable membrane. A gradient of such tension and the conventional membrane tension may drive bulk lipid flow through bridge LTPs at a speed consistent with the fast membrane expansion observed in vivo. Finally, we discuss the implications of membrane tension and lipid transfer in organelle biogenesis. Overall, the quantitative models may help clarify the mechanisms of LTP-mediated MCS formation and lipid transfer.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.,Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Jinghua Ge
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Xin Bian
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.,Present address: State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Avinash Kumar
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
50
|
Kobayashi J, Arita M, Sakai S, Kojima H, Senda M, Senda T, Hanada K, Kato R. Ligand Recognition by the Lipid Transfer Domain of Human OSBP Is Important for Enterovirus Replication. ACS Infect Dis 2022; 8:1161-1170. [PMID: 35613096 DOI: 10.1021/acsinfecdis.2c00108] [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] [Indexed: 12/14/2022]
Abstract
Oxysterol-binding protein (OSBP), which transports cholesterol and phosphatidylinositol 4-monophosphate (PtdIns[4]P) between different organelles, serves as a conserved host factor for the replication of various viruses, and OSBP inhibitors exhibit antiviral effects. Here, we determined the crystal structure of the lipid transfer domain of human OSBP in complex with endogenous cholesterol. The hydrocarbon tail and tetracyclic ring of cholesterol interact with the hydrophobic tunnel of OSBP, and the hydroxyl group of cholesterol forms a hydrogen bond network at the bottom of the tunnel. Systematic mutagenesis of the ligand-binding region revealed that M446W and L590W substitutions confer functional tolerance to an OSBP inhibitor, T-00127-HEV2. Employing the M446W variant as a functional replacement for the endogenous OSBP in the presence of T-00127-HEV2, we have identified previously unappreciated amino acid residues required for viral replication. The combined use of the inhibitor and the OSBP variant will be useful in elucidating the enigmatic in vivo functions of OSBP.
Collapse
Affiliation(s)
- Jun Kobayashi
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| | - Minetaro Arita
- Department of Virology II, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Shota Sakai
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Hirotatsu Kojima
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Miki Senda
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| | - Toshiya Senda
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Ryuichi Kato
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| |
Collapse
|