1
|
Benitez-Amaro A, Garcia E, LaChica Lhoëst MT, Polishchuk A, Zegri-Reiriz I, Vilades D, Guerra JM, Fernández-Del-Rio L, Mirabet S, Samouillan V, Shirihai O, Liesa M, Enrich C, Llorente-Cortés V. LRP1 immunotherapy enhances cardiomyocyte respiration by restricting cholesteryl ester accumulation in mitochondria. J Lipid Res 2025; 66:100783. [PMID: 40122209 PMCID: PMC12051619 DOI: 10.1016/j.jlr.2025.100783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/25/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025] Open
Abstract
Antibodies (Abs) targeting the P3 sequence (Gly1127-Cys1140) of LDL receptor-related protein 1 (anti-P3 Abs) inhibit the interaction between ApoB100 in cholesteryl ester (CE)-enriched lipoproteins and the CR9 domain in LDL receptor-related protein 1, preventing intracellular CE accumulation induced by a high-fat high-cholesterol (HFHC) diet in cardiomyocytes. This study examines (i) whether HFHC induces cholesterol accumulation in mitochondria, and impacts cardiac bioenergetics, and (ii) the effectiveness of anti-P3 Abs in mitigating HFHC-induced mitochondrial alterations. Cardiac tissue was homogenized, and mitochondria were isolated through subcellular fractionation. Thin layer chromatography demonstrated that HFHC induced the accumulation of CE in cardiac mitochondria, and that this process was significantly reduced by anti-P3 Abs. In line, transmission electron microscopy studies revealed that morphological changes induced by HFHC in cardiomyocyte mitochondria were reversed, at least in part, by anti-P3 Abs. Additionally, anti-P3 Abs promoted more extensive interactions between mitochondria and lipid droplets (LDs), accompanied by an increase in LD diameter and electrodensity in cardiomyocytes. Cardiac mitochondrial respiratory capacity assessed by Seahorse analysis showed that HFHC reduced CI/CIV and CII/CIV activity ratios, while anti-P3 Abs restored complex II/IV activity. In conclusion, by blocking CE uptake from lipoproteins, anti-P3 Abs reduce CE accumulation in the cardiomyocyte mitochondria and LDs, enhance bioenergetically favorable mitochondria/LD interactions, and improve cardiomyocyte respiratory function in hypercholesterolemic rabbits. These findings highlight the therapeutic potential of anti-P3 Abs in metabolic diseases by limiting CE loading of mitochondria and LDs in the heart and restoring cardiac bioenergetics.
Collapse
Affiliation(s)
- A Benitez-Amaro
- Lipids and Cardiovascular Pathology Group, Biomedical Research Institut of Barcelona, IIBB-CSIC, Barcelona, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - E Garcia
- Lipids and Cardiovascular Pathology Group, Biomedical Research Institut of Barcelona, IIBB-CSIC, Barcelona, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M T LaChica Lhoëst
- Lipids and Cardiovascular Pathology Group, Biomedical Research Institut of Barcelona, IIBB-CSIC, Barcelona, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departamento de Bioquímica y Biología Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Polishchuk
- Lipids and Cardiovascular Pathology Group, Biomedical Research Institut of Barcelona, IIBB-CSIC, Barcelona, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - I Zegri-Reiriz
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Institut de Recerca Sant Pau (IR SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - D Vilades
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Institut de Recerca Sant Pau (IR SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain; CIBERCV, Institute of Health Carlos III, Madrid, Spain
| | - J M Guerra
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Institut de Recerca Sant Pau (IR SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain; CIBERCV, Institute of Health Carlos III, Madrid, Spain
| | - L Fernández-Del-Rio
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - S Mirabet
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Institut de Recerca Sant Pau (IR SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain; CIBERCV, Institute of Health Carlos III, Madrid, Spain
| | - V Samouillan
- CIRIMAT, Université de Toulouse, Université Paul Sabatier, Equipe PHYPOL, Toulouse, France
| | - O Shirihai
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - M Liesa
- Institut de Biologia Molecular de Barcelona, IBMB, CSIC, Barcelona, Spain; CIBERDEM Institute of Health Carlos III, Madrid, Spain
| | - C Enrich
- Unitat de Biologia Cellular, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - V Llorente-Cortés
- Lipids and Cardiovascular Pathology Group, Biomedical Research Institut of Barcelona, IIBB-CSIC, Barcelona, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; CIBERCV, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
2
|
La Rosa P, Tiberi J, Palermo E, Stefanelli R, Tiano SML, Canterini S, Cortese M, Hiscott J, Fiorenza MT. The inactivation of the Niemann Pick C1 cholesterol transporter restricts SARS-CoV-2 entry into host cells by decreasing ACE2 abundance at the plasma membrane. Cell Biosci 2024; 14:148. [PMID: 39707537 DOI: 10.1186/s13578-024-01331-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The Niemann Pick C1 (NPC1) protein is an intracellular cholesterol transporter located in the late endosome/lysosome (LE/Ly) that is involved in the mobilization of endocytosed cholesterol. Loss-of-function mutations in the NPC1 gene lead to the accumulation of cholesterol and sphingolipids in LE/Ly, resulting in severe fatal NPC1 disease. Cellular alterations associated with NPC1 inactivation affect both the integrity of lipid rafts and the endocytic pathway. Because the angiotensin-converting enzyme 2 (ACE2) and type 2 serine transmembrane protease (TMPRSS2), interactors of the SARS-CoV-2 Spike protein also localize to lipid rafts, we sought to investigate the hypothesis that NPC1 inactivation would generate an intrinsically unfavorable barrier to SARS-CoV-2 entry. RESULTS In this study, we show that inhibition of the cholesterol transporter activity of NPC1 in cells that express both ACE2 and TMPRSS2, considerably reduces SARS-CoV-2 infectivity, evaluated as early as 4 h post-infection. Mechanistically, treatment with NPC1 specific inhibitor U18666A relocalizes ACE2 from the plasma membrane to the autophagosomal/lysosomal compartment, thereby reducing SARS-CoV-2 entry into treated cells. Reduction of viral entry was observed for both fully infectious SARS-CoV-2 virus and with a pseudotyped VSV-Spike-GFP virus. For instance, U18666A-treated Caco-2 cells infected with the pseudotyped VSV-Spike-GFP showed a > threefold and > 40-fold reduction in virus titer when infectivity was measured at 4 h or 24 h post-infection, respectively. A similar effect was observed in CRISP/R-Cas9-edited Caco-2 cells, which were even more resistant to SARS-CoV-2 infection as indicated by a 97% reduction of viral titers. CONCLUSION Overall, this study provides compelling evidence that the inhibition of NPC1 cholesterol transporter activity generates a cellular environment that hinders SARS-CoV-2 entry. ACE2 depletion from the plasma membrane appears to play a major role as limiting factor for viral entry.
Collapse
Affiliation(s)
- Piergiorgio La Rosa
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Jessica Tiberi
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy
- PhD Program in Behavioral Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Enrico Palermo
- Istituto Pasteur Italia-Cenci Bolognetti Foundation, Viale Regina Elena 291, 00161, Rome, Italy
| | - Roberta Stefanelli
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy
| | - Sofia Maria Luigia Tiano
- Telethon Institute of Genetics and Medicine, TIGEM, Via Campi Flegrei, 34, 80078, Pozzuoli, Italy
| | - Sonia Canterini
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Mirko Cortese
- Telethon Institute of Genetics and Medicine, TIGEM, Via Campi Flegrei, 34, 80078, Pozzuoli, Italy
- Universitá della Campania Luigi Vanvitelli, Via Vivaldi, 43, 81100, Caserta, Italy
| | - John Hiscott
- Istituto Pasteur Italia-Cenci Bolognetti Foundation, Viale Regina Elena 291, 00161, Rome, Italy
| | - Maria Teresa Fiorenza
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy.
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy.
| |
Collapse
|
3
|
Rudnik S, Heybrock S, Coyaud E, Xu Z, Neculai D, Raught B, Oorschot V, Heus C, Klumperman J, Saftig P. The lysosomal lipid transporter LIMP-2 is part of lysosome-ER STARD3-VAPB-dependent contact sites. J Cell Sci 2024; 137:jcs261810. [PMID: 39370902 DOI: 10.1242/jcs.261810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 09/26/2024] [Indexed: 10/08/2024] Open
Abstract
LIMP-2 (also known as SCARB2) is an abundant lysosomal membrane protein. Previous studies have shown that LIMP-2 functions as a virus receptor, a chaperone for lysosomal enzyme targeting and a lipid transporter. The large luminal domain of LIMP-2 contains a hydrophobic tunnel that enables transport of phospholipids, sphingosine and cholesterol from the lysosomal lumen to the membrane. The question about the fate of the lipids after LIMP-2-mediated transport is largely unexplored. To elucidate whether LIMP-2 is present at contact sites between lysosomes and the endoplasmic reticulum (ER), we performed a proximity-based interaction screen. This revealed that LIMP-2 interacts with the endosomal protein STARD3 and the ER-resident protein VAPB. Using imaging and co-immunoprecipitation, we demonstrated colocalization and physical interaction between LIMP-2 and these proteins. Moreover, we found that interaction of LIMP-2 with VAPB required the presence of STARD3. Our findings suggest that LIMP-2 is present at ER-lysosome contact sites, possibly facilitating cholesterol transport from the lysosomal to the ER membrane. This suggests a novel mechanism for inter-organelle communication and lipid trafficking mediated by LIMP-2.
Collapse
Affiliation(s)
- Sönke Rudnik
- Institute of Biochemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Saskia Heybrock
- Institute of Biochemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Etienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Zizhen Xu
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322001, China
| | - Dante Neculai
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322001, China
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Viola Oorschot
- Electron Microscopy Core Facility, EMBL Heidelberg, 69117 Heidelberg, Germany
- Center for Molecular Medicine Section Cell Biology, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Cecilia Heus
- Center for Molecular Medicine Section Cell Biology, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Judith Klumperman
- Center for Molecular Medicine Section Cell Biology, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| |
Collapse
|
4
|
Białek W, Hryniewicz-Jankowska A, Czechowicz P, Sławski J, Collawn JF, Czogalla A, Bartoszewski R. The lipid side of unfolded protein response. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159515. [PMID: 38844203 DOI: 10.1016/j.bbalip.2024.159515] [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: 02/28/2024] [Revised: 04/16/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Although our current knowledge of the molecular crosstalk between the ER stress, the unfolded protein response (UPR), and lipid homeostasis remains limited, there is increasing evidence that dysregulation of either protein or lipid homeostasis profoundly affects the other. Most research regarding UPR signaling in human diseases has focused on the causes and consequences of disrupted protein folding. The UPR itself consists of very complex pathways that function to not only maintain protein homeostasis, but just as importantly, modulate lipid biogenesis to allow the ER to adjust and promote cell survival. Lipid dysregulation is known to activate many aspects of the UPR, but the complexity of this crosstalk remains a major research barrier. ER lipid disequilibrium and lipotoxicity are known to be important contributors to numerous human pathologies, including insulin resistance, liver disease, cardiovascular diseases, neurodegenerative diseases, and cancer. Despite their medical significance and continuous research, however, the molecular mechanisms that modulate lipid synthesis during ER stress conditions, and their impact on cell fate decisions, remain poorly understood. Here we summarize the current view on crosstalk and connections between altered lipid metabolism, ER stress, and the UPR.
Collapse
Affiliation(s)
- Wojciech Białek
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | - Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Rafał Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.
| |
Collapse
|
5
|
Schiffmann A, Ahlswede L, Gimpl G. Reversible translocation of acyl-CoA:cholesterol acyltransferase (ACAT) between the endoplasmic reticulum and vesicular structures. Front Mol Biosci 2023; 10:1258799. [PMID: 38028547 PMCID: PMC10667705 DOI: 10.3389/fmolb.2023.1258799] [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: 07/14/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
The enzyme acyl-CoA:cholesterol acyltransferase (ACAT) is normally localized in the endoplasmic reticulum (ER) where it can esterify cholesterol for storage in lipid droplets and/or the formation of lipoproteins. Here, we report that ACAT can translocate from the ER into vesicular structures in response to different ACAT inhibitors. The translocation was fast (within minutes), reversible and occurred in different cell types. Interestingly, oleic acid was able to fasten the re-translocation from vesicles back into the reticular ER network. The process of ACAT translocation could also be induced by cyclodextrins, cholesterol, lanosterol (but not 4-cholestene-3 one), 25-hydroxycholesterol, and by certain stress stimuli such as hyperosmolarity (sucrose treatment), temperature change, or high-density cultivation. In vitro esterification showed that ACAT remains fully active after it has been translocated to vesicles in response to hyperosmotic sucrose treatment of the cells. The translocation process was not accompanied by changes in the electrophoretic mobility of ACAT, even after chemical crosslinking. Interestingly, the protein synthesis inhibitor cycloheximide showed a stimulating effect on ACAT activity and prevented the translocation of ACAT from the ER into vesicles.
Collapse
Affiliation(s)
| | | | - Gerald Gimpl
- Department of Chemistry and Biochemistry, Biocenter II, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
6
|
Sidhu SK, Mishra S. A cholesterol-centric outlook on steroidogenesis. VITAMINS AND HORMONES 2023; 124:405-428. [PMID: 38408806 DOI: 10.1016/bs.vh.2023.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Cholesterol, an essential and versatile lipid, is the precursor substrate for the biosynthesis of steroid hormones, and a key structural and functional component of organelle membranes in eukaryotic cells. Consequently, the framework of steroidogenesis across main steroidogenic cell types is built around cholesterol, including its cellular uptake, mobilization from intracellular storage, and finally, its transport to the mitochondria where steroidogenesis begins. This setup, which is controlled by different trophic hormones in their respective target tissues, allows steroidogenic cells to meet their steroidogenic need of cholesterol effectively without impinging on the basic need for organelle membranes and their functions. However, our understanding of the basal steroidogenesis (i.e., independent of trophic hormone stimulation), which is a cell-intrinsic trait, remains poor. Particularly, the role that cholesterol itself plays in the regulation of steroidogenic factors and events in steroid hormone-producing cells remains largely unexplored. This is likely because of challenges in selectively targeting the steroidogenic intracellular cholesterol pool in studies. New evidence suggests that cholesterol plays a role in steroidogenesis. These new findings have created new opportunities to advance our understanding in this field. In this book chapter, we will provide a cholesterol-centric view on steroidogenesis and emphasize the importance of the interplay between cholesterol and the mitochondria in steroidogenic cells. Moreover, we will discuss a novel mitochondrial player, prohibitin-1, in this context. The overall goal is to provide a stimulating perspective on cholesterol as an important regulator of steroidogenesis (i.e., more than just a substrate for steroid hormones) and present the mitochondria as a potential cell-intrinsic factor in regulating steroidogenic cholesterol homeostasis.
Collapse
Affiliation(s)
- Simarjit Kaur Sidhu
- Department of Physiology & Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Suresh Mishra
- Department of Physiology & Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Department of Internal Medicine, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
7
|
Abstract
Most cholesterol in mammalian cells is stored in the plasma membrane (PM). Cholesterol transport from the PM to low-sterol regulatory regions of the endoplasmic reticulum (ER) controls cholesterol synthesis and uptake, and thereby influences the rates of cholesterol flux between tissues of complex organisms. Cholesterol transfer to the ER is also required for steroidogenesis, oxysterol and bile acid synthesis, and cholesterol esterification. The ER-resident Aster proteins (Aster-A, -B, and -C) form contacts with the PM to move cholesterol to the ER in mammals. Mice lacking Aster-B have low adrenal cholesteryl ester stores and impaired steroidogenesis because of a defect in cholesterol transport from high-density lipoprotein (HDL) to the ER. This work reviews the molecular characteristics of Asters, their role in HDL- and low-density lipoprotein (LDL)-cholesterol movement, and how cholesterol transferred to the ER is utilized by cells. The roles of other lipid transporters and of membrane lipid organization in maintaining aspects of cholesterol homeostasis are also highlighted.
Collapse
Affiliation(s)
- John P Kennelly
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
8
|
Palladino END, Bernas T, Green CD, Weigel C, Singh SK, Senkal CE, Martello A, Kennelly JP, Bieberich E, Tontonoz P, Ford DA, Milstien S, Eden ER, Spiegel S. Sphingosine kinases regulate ER contacts with late endocytic organelles and cholesterol trafficking. Proc Natl Acad Sci U S A 2022; 119:e2204396119. [PMID: 36122218 PMCID: PMC9522378 DOI: 10.1073/pnas.2204396119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/16/2022] [Indexed: 11/18/2022] Open
Abstract
Membrane contact sites (MCS), close membrane apposition between organelles, are platforms for interorganellar transfer of lipids including cholesterol, regulation of lipid homeostasis, and co-ordination of endocytic trafficking. Sphingosine kinases (SphKs), two isoenzymes that phosphorylate sphingosine to the bioactive sphingosine-1-phosphate (S1P), have been implicated in endocytic trafficking. However, the physiological functions of SphKs in regulation of membrane dynamics, lipid trafficking and MCS are not known. Here, we report that deletion of SphKs decreased S1P with concomitant increases in its precursors sphingosine and ceramide, and markedly reduced endoplasmic reticulum (ER) contacts with late endocytic organelles. Expression of enzymatically active SphK1, but not catalytically inactive, rescued the deficit of these MCS. Although free cholesterol accumulated in late endocytic organelles in SphK null cells, surprisingly however, cholesterol transport to the ER was not reduced. Importantly, deletion of SphKs promoted recruitment of the ER-resident cholesterol transfer protein Aster-B (also called GRAMD1B) to the plasma membrane (PM), consistent with higher accessible cholesterol and ceramide at the PM, to facilitate cholesterol transfer from the PM to the ER. In addition, ceramide enhanced in vitro binding of the Aster-B GRAM domain to phosphatidylserine and cholesterol liposomes. Our study revealed a previously unknown role for SphKs and sphingolipid metabolites in governing diverse MCS between the ER network and late endocytic organelles versus the PM to control the movement of cholesterol between distinct cell membranes.
Collapse
Affiliation(s)
- Elisa N. D. Palladino
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Tytus Bernas
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Christopher D. Green
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Cynthia Weigel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Sandeep K. Singh
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Can E. Senkal
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Andrea Martello
- UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - John P. Kennelly
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky and Veteran Affairs Medical Center, Lexington, KY 40536
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
| | - David A. Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Sheldon Milstien
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Emily R. Eden
- UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| |
Collapse
|
9
|
Kim S, Coukos R, Gao F, Krainc D. Dysregulation of organelle membrane contact sites in neurological diseases. Neuron 2022; 110:2386-2408. [PMID: 35561676 PMCID: PMC9357093 DOI: 10.1016/j.neuron.2022.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/21/2022] [Accepted: 04/18/2022] [Indexed: 10/18/2022]
Abstract
The defining evolutionary feature of eukaryotic cells is the emergence of membrane-bound organelles. Compartmentalization allows each organelle to maintain a spatially, physically, and chemically distinct environment, which greatly bolsters individual organelle function. However, the activities of each organelle must be balanced and are interdependent for cellular homeostasis. Therefore, properly regulated interactions between organelles, either physically or functionally, remain critical for overall cellular health and behavior. In particular, neuronal homeostasis depends heavily on the proper regulation of organelle function and cross talk, and deficits in these functions are frequently associated with diseases. In this review, we examine the emerging role of organelle contacts in neurological diseases and discuss how the disruption of contacts contributes to disease pathogenesis. Understanding the molecular mechanisms underlying the formation and regulation of organelle contacts will broaden our knowledge of their role in health and disease, laying the groundwork for the development of new therapies targeting interorganelle cross talk and function.
Collapse
Affiliation(s)
- Soojin Kim
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Robert Coukos
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Fanding Gao
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL 60611, USA.
| |
Collapse
|
10
|
Rogers MA, Chang CCY, Maue RA, Melton EM, Peden AA, Garver WS, Lee J, Schroen P, Huang M, Chang TY. Acat1/Soat1 knockout extends the mutant Npc1 mouse lifespan and ameliorates functional deficiencies in multiple organelles of mutant cells. Proc Natl Acad Sci U S A 2022; 119:e2201646119. [PMID: 35507892 PMCID: PMC9170141 DOI: 10.1073/pnas.2201646119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 11/18/2022] Open
Abstract
Multiple membrane organelles require cholesterol for proper function within cells. The Niemann-Pick type C (NPC) proteins export cholesterol from endosomes to other membrane compartments, including the endoplasmic reticulum (ER), plasma membrane (PM), trans-Golgi network (TGN), and mitochondria, to meet their cholesterol requirements. Defects in NPC cause malfunctions in multiple membrane organelles and lead to an incurable neurological disorder. Acyl-coenzyme A:cholesterol acyltransferase 1 (ACAT1), a resident enzyme in the ER, converts cholesterol to cholesteryl esters for storage. In mutant NPC cells, cholesterol storage still occurs in an NPC-independent manner. Here we report the interesting finding that in a mutant Npc1 mouse (Npc1nmf), Acat1 gene (Soat1) knockout delayed the onset of weight loss, motor impairment, and Purkinje neuron death. It also improved hepatosplenic pathology and prolonged lifespan by 34%. In mutant NPC1 fibroblasts, ACAT1 blockade (A1B) increased cholesterol content associated with TGN-rich membranes and mitochondria, while decreased cholesterol content associated with late endosomes. A1B also restored proper localization of syntaxin 6 and golgin 97 (key proteins in membrane trafficking at TGN) and improved the levels of cathepsin D (a key protease in lysosome and requires Golgi/endosome transport for maturation) and ABCA1 (a key protein controlling cholesterol release at PM). This work supports the hypothesis that diverting cholesterol from storage can benefit multiple diseases that involve cholesterol deficiencies in cell membranes.
Collapse
Affiliation(s)
- Maximillian A. Rogers
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Catherine C. Y. Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Robert A. Maue
- Department of Biology, Dartmouth College, Hanover, NH 03755
| | - Elaina M. Melton
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Andrew A. Peden
- Department of Biomedical Science, Centre for Membrane Interactions and Dynamics, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - William S. Garver
- Department of Chemistry & Chemical Biology, University of New Mexico, Albuquerque, NM 87131
| | - Junghoon Lee
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Peter Schroen
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Mitchell Huang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Ta-Yuan Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
11
|
Papatheodorou P, Kindig S, Badilla-Lobo A, Fischer S, Durgun E, Thuraisingam T, Witte A, Song S, Aktories K, Chaves-Olarte E, Rodríguez C, Barth H. The Compound U18666A Inhibits the Intoxication of Cells by Clostridioides difficile Toxins TcdA and TcdB. Front Microbiol 2021; 12:784856. [PMID: 34912322 PMCID: PMC8667575 DOI: 10.3389/fmicb.2021.784856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
The intestinal pathogen Clostridioides (C.) difficile is a major cause of diarrhea both in hospitals and outpatient in industrialized countries. This bacterium produces two large exotoxins, toxin A (TcdA) and toxin B (TcdB), which are directly responsible for the onset of clinical symptoms of C. difficile-associated diseases (CDADs), such as antibiotics-associated diarrhea and the severe, life-threatening pseudomembranous colitis. Both toxins are multidomain proteins and taken up into host eukaryotic cells via receptor-mediated endocytosis. Within the cell, TcdA and TcdB inactivate Rho and/or Ras protein family members by glucosylation, which eventually results in cell death. The cytotoxic mode of action of the toxins is the main reason for the disease. Thus, compounds capable of inhibiting the cellular uptake and/or mode-of-action of both toxins are of high therapeutic interest. Recently, we found that the sterol regulatory element-binding protein 2 (SREBP-2) pathway, which regulates cholesterol content in membranes, is crucial for the intoxication of cells by TcdA and TcdB. Furthermore, it has been shown that membrane cholesterol is required for TcdA- as well as TcdB-mediated pore formation in endosomal membranes, which is a key step during the translocation of the glucosyltransferase domain of both toxins from endocytic vesicles into the cytosol of host cells. In the current study, we demonstrate that intoxication by TcdA and TcdB is diminished in cultured cells preincubated with the compound U18666A, an established inhibitor of cholesterol biosynthesis and/or intracellular transport. U18666A-pretreated cells were also less sensitive against TcdA and TcdB variants from the epidemic NAP1/027 C. difficile strain. Our study corroborates the crucial role of membrane cholesterol for cell entry of TcdA and TcdB, thus providing a valuable basis for the development of novel antitoxin strategies in the context of CDADs.
Collapse
Affiliation(s)
| | - Selina Kindig
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Adriana Badilla-Lobo
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Stephan Fischer
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Ebru Durgun
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Tharani Thuraisingam
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Alexander Witte
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Shuo Song
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - César Rodríguez
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Holger Barth
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
12
|
Ikhlef S, Lipp NF, Delfosse V, Fuggetta N, Bourguet W, Magdeleine M, Drin G. Functional analyses of phosphatidylserine/PI(4)P exchangers with diverse lipid species and membrane contexts reveal unanticipated rules on lipid transfer. BMC Biol 2021; 19:248. [PMID: 34801011 PMCID: PMC8606082 DOI: 10.1186/s12915-021-01183-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/04/2021] [Indexed: 11/14/2022] Open
Abstract
Background Lipid species are accurately distributed in the eukaryotic cell so that organelle and plasma membranes have an adequate lipid composition to support numerous cellular functions. In the plasma membrane, a precise regulation of the level of lipids such as phosphatidylserine, PI(4)P, and PI(4,5)P2, is critical for maintaining the signaling competence of the cell. Several lipid transfer proteins of the ORP/Osh family contribute to this fine-tuning by delivering PS, synthesized in the endoplasmic reticulum, to the plasma membrane in exchange for PI(4)P. To get insights into the role of these PS/PI(4)P exchangers in regulating plasma membrane features, we question how they selectively recognize and transfer lipid ligands with different acyl chains, whether these proteins exchange PS exclusively for PI(4)P or additionally for PI(4,5)P2, and how sterol abundance in the plasma membrane impacts their activity. Results We measured in vitro how the yeast Osh6p and human ORP8 transported PS and PI(4)P subspecies of diverse length and unsaturation degree between membranes by fluorescence-based assays. We established that the exchange activity of Osh6p and ORP8 strongly depends on whether these ligands are saturated or not, and is high with representative cellular PS and PI(4)P subspecies. Unexpectedly, we found that the speed at which these proteins individually transfer lipid ligands between membranes is inversely related to their affinity for them and that high-affinity ligands must be exchanged to be transferred more rapidly. Next we determined that Osh6p and ORP8 cannot use PI(4,5)P2 for exchange processes, because it is a low-affinity ligand, and do not transfer more PS into sterol-rich membranes. Conclusions Our study provides new insights into PS/PI(4)P exchangers by indicating the degree to which they can regulate the acyl chain composition of the PM, and how they control PM phosphoinositide levels. Moreover, we establish general rules on how the activity of lipid transfer proteins relates to their affinity for ligands. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01183-1.
Collapse
Affiliation(s)
- Souade Ikhlef
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France
| | - Nicolas-Frédéric Lipp
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France.,Current position: Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Vanessa Delfosse
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Nicolas Fuggetta
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France
| | - William Bourguet
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Maud Magdeleine
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France
| | - Guillaume Drin
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 06560, Valbonne, France.
| |
Collapse
|
13
|
Schoop V, Martello A, Eden ER, Höglinger D. Cellular cholesterol and how to find it. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158989. [PMID: 34118431 DOI: 10.1016/j.bbalip.2021.158989] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 01/06/2023]
Abstract
Cholesterol is an essential component of eukaryotic cellular membranes. Information about its subcellular localization and transport pathways inside cells are key for the understanding and treatment of cholesterol-related diseases. In this review we give an overview over the most commonly used methods that contributed to our current understanding of subcellular cholesterol localization and transport routes. First, we discuss methods that provide insights into cholesterol metabolism based on readouts of downstream effects such as esterification. Subsequently, we focus on the use of cholesterol-binding molecules as probes that facilitate visualization and quantification of sterols inside of cells. Finally, we explore different analogues of cholesterol which, when taken up by living cells, are integrated and transported in a similar fashion as endogenous sterols. Taken together, we highlight the challenges and advantages of each method such that researchers studying aspects of cholesterol transport may choose the most pertinent approach for their problem.
Collapse
Affiliation(s)
- Valentin Schoop
- Heidelberg University Biochemistry Center (BZH), 69120 Heidelberg, Germany
| | - Andrea Martello
- University College London (UCL), Institute of Ophthalmology, EC1V 9EL London, United Kingdom
| | - Emily R Eden
- University College London (UCL), Institute of Ophthalmology, EC1V 9EL London, United Kingdom
| | - Doris Höglinger
- Heidelberg University Biochemistry Center (BZH), 69120 Heidelberg, Germany.
| |
Collapse
|
14
|
Selective Aster inhibitors distinguish vesicular and nonvesicular sterol transport mechanisms. Proc Natl Acad Sci U S A 2021; 118:2024149118. [PMID: 33376205 DOI: 10.1073/pnas.2024149118] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Aster proteins (encoded by the Gramd1a-c genes) contain a ligand-binding fold structurally similar to a START domain and mediate nonvesicular plasma membrane (PM) to endoplasmic reticulum (ER) cholesterol transport. In an effort to develop small molecule modulators of Asters, we identified 20α-hydroxycholesterol (HC) and U18666A as lead compounds. Unfortunately, both 20α-HC and U18666A target other sterol homeostatic proteins, limiting their utility. 20α-HC inhibits sterol regulatory element-binding protein 2 (SREBP2) processing, and U18666A is an inhibitor of the vesicular trafficking protein Niemann-Pick C1 (NPC1). To develop potent and selective Aster inhibitors, we synthesized a series of compounds by modifying 20α-HC and U18666A. Among these, AI (Aster inhibitor)-1l, which has a longer side chain than 20α-HC, selectively bound to Aster-C. The crystal structure of Aster-C in complex with AI-1l suggests that sequence and flexibility differences in the loop that gates the binding cavity may account for the ligand specificity for Aster C. We further identified the U18666A analog AI-3d as a potent inhibitor of all three Aster proteins. AI-3d blocks the ability of Asters to bind and transfer cholesterol in vitro and in cells. Importantly, AI-3d also inhibits the movement of low-density lipoprotein (LDL) cholesterol to the ER, although AI-3d does not block NPC1. This finding positions the nonvesicular Aster pathway downstream of NPC1-dependent vesicular transport in the movement of LDL cholesterol to the ER. Selective Aster inhibitors represent useful chemical tools to distinguish vesicular and nonvesicular sterol transport mechanisms in mammalian cells.
Collapse
|
15
|
Teixeira da Costa LF. On the possible existence of a liver LDL-ostat, and its malfunctioning in familial hypercholesterolemia. Med Hypotheses 2021; 147:110500. [PMID: 33515861 DOI: 10.1016/j.mehy.2021.110500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/29/2020] [Accepted: 01/08/2021] [Indexed: 11/16/2022]
Abstract
The investigation of familial hypercholesterolemia (FH) and its relationship to atherosclerosis has led to enormous scientific and medical progress, including the identification of genetic defects underlying FH, the elucidation of molecular mechanisms crucial for cellular cholesterol homeostasis and the development of current pharmaceutical tools for FH treatment (which are directed at increasing LDL uptake). These successes also led to the establishment of a model centered on cellular rather than whole organism processes, and a view of FH as resulting from a deficiency in LDL uptake. On the other hand, whole organism fluxes of cholesterol (like those of other nutrients) are centered on the liver, LDL (ultimately derived from the liver) is the main cholesterol transporter in plasma, and there is evidence of evolutionary pressure favoring mechanisms to maintain LDL plasma concentrations. Furthermore, the alterations in cellular metabolism observed in FH are consistent with a coordinated response by the liver to increase the levels of plasma LDL, suggesting that a signaling defect (rather than an uptake deficiency) is the fundamental problem underlying hypercholesterolemia - an hypothesis that explains the occurrence of hypercholesterolemia in CESD, despite normal LDL binding and uptake. I therefore propose that the liver contains a mechanism to assess and regulate plasma levels of LDL (an "LDL-ostat"), and that hypercholesterolemia is caused by defects in it. This model has implications for future research directions, and suggests alternative therapeutic approaches, particularly centered on efforts to restore LDL measurement/signaling (rather than its uptake), some of which are in stark contrast to those currently in use.
Collapse
Affiliation(s)
- Luís Filipe Teixeira da Costa
- Unit for Cardiac and Cardiovascular Genetics, Department of Medical Genetics, Oslo University Hospital, P.O. Box 4956 Nydalen, 0424 Oslo, Norway.
| |
Collapse
|
16
|
Vos DY, van de Sluis B. Function of the endolysosomal network in cholesterol homeostasis and metabolic-associated fatty liver disease (MAFLD). Mol Metab 2021; 50:101146. [PMID: 33348067 PMCID: PMC8324686 DOI: 10.1016/j.molmet.2020.101146] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/26/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
Background Metabolic-associated fatty liver disease (MAFLD), also known as non-alcoholic fatty liver disease, has become the leading cause of chronic liver disease worldwide. In addition to hepatic accumulation of triglycerides, dysregulated cholesterol metabolism is an important contributor to the pathogenesis of MAFLD. Maintenance of cholesterol homeostasis is highly dependent on cellular cholesterol uptake and, subsequently, cholesterol transport to other membrane compartments, such as the endoplasmic reticulum (ER). Scope of review The endolysosomal network is key for regulating cellular homeostasis and adaptation, and emerging evidence has shown that the endolysosomal network is crucial to maintain metabolic homeostasis. In this review, we will summarize our current understanding of the role of the endolysosomal network in cholesterol homeostasis and its implications in MAFLD pathogenesis. Major conclusions Although multiple endolysosomal proteins have been identified in the regulation of cholesterol uptake, intracellular transport, and degradation, their physiological role is incompletely understood. Further research should elucidate their role in controlling metabolic homeostasis and development of fatty liver disease. The intracellular cholesterol transport is tightly regulated by the endocytic and lysosomal network. Dysfunction of the endolysosomal network affects hepatic lipid homeostasis. The endosomal sorting of lipoprotein receptors is precisely regulated and is not a bulk process.
Collapse
Affiliation(s)
- Dyonne Y Vos
- Department of Pediatrics, section Molecular Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, section Molecular Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
17
|
Dubey V, Bozorg B, Wüstner D, Khandelia H. Cholesterol binding to the sterol-sensing region of Niemann Pick C1 protein confines dynamics of its N-terminal domain. PLoS Comput Biol 2020; 16:e1007554. [PMID: 33021976 PMCID: PMC7537887 DOI: 10.1371/journal.pcbi.1007554] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Lysosomal accumulation of cholesterol is a hallmark of Niemann Pick type C (NPC) disease caused by mutations primarily in the lysosomal membrane protein NPC1. NPC1 contains a transmembrane sterol-sensing domain (SSD), which is supposed to regulate protein activity upon cholesterol binding, but the mechanisms underlying this process are poorly understood. Using atomistic simulations, we show that in the absence of cholesterol in the SSD, the luminal domains of NPC1 are highly dynamic, resulting in the disengagement of the NTD from the rest of the protein. The disengaged NPC1 adopts a flexed conformation that approaches the lipid bilayer, and could represent a conformational state primed to receive a sterol molecule from the soluble lysosomal cholesterol carrier NPC2. The binding of cholesterol to the SSD of NPC1 allosterically suppresses the conformational dynamics of the luminal domains resulting in an upright NTD conformation. The presence of an additional 20% cholesterol in the membrane has negligible impact on this process. The additional presence of an NTD-bound cholesterol suppresses the flexing of the NTD. We propose that cholesterol acts as an allosteric effector, and the modulation of NTD dynamics by the SSD-bound cholesterol constitutes an allosteric feedback mechanism in NPC1 that controls cholesterol abundance in the lysosomal membrane. Cholesterol is absorbed from LDL particles in esterified form, and is broken down to free cholesterol in the lysosomes of cells, from where cholesterol must be transported to other cellular compartments such as the plasma membrane. The Niemann Pick type C (NPC) diseases arise from deficient cholesterol transport and result from mutations in the cholesterol transport protein NPC1. Using computer simulations, we show that cholesterol, when bound to one part of NPC1, can control the structural transitions of an 8-nm distant, different part of NPC1 protein called the N-terminal domain (NTD). Such long-range control of protein conformations (allostery), controls a wide range of cellular functions mediated by proteins. Fundamental molecular insights into the function of the NPC1 protein can potentially lead to better pharmaceutical interventions for the NPC diseases.
Collapse
Affiliation(s)
- Vikas Dubey
- PhyLife Physical Life Sciences, Department of Physics Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Physics Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Behruz Bozorg
- PhyLife Physical Life Sciences, Department of Physics Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Physics Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Daniel Wüstner
- PhyLife Physical Life Sciences, Department of Physics Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Himanshu Khandelia
- PhyLife Physical Life Sciences, Department of Physics Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Physics Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- MEMPHYS: Center for Biomembrane Physics, Odense M, Denmark
- * E-mail:
| |
Collapse
|
18
|
Lange Y, Steck TL. Active cholesterol 20 years on. Traffic 2020; 21:662-674. [PMID: 32930466 DOI: 10.1111/tra.12762] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
This review considers the following hypotheses, some well-supported and some speculative. Almost all of the sterol molecules in plasma membranes are associated with bilayer phospholipids in complexes of varied strength and stoichiometry. These complexes underlie many of the material properties of the bilayer. The small fraction of cholesterol molecules exceeding the binding capacity of the phospholipids is thermodynamically active and serves diverse functions. It circulates briskly among the cell membranes, particularly through contact sites linking the organelles. Active cholesterol provides the upstream feedback signal to multiple mechanisms governing plasma membrane homeostasis, pegging the sterol level to a threshold set by its phospholipids. Active cholesterol could also be the cargo for various inter-organelle transporters and the form excreted from cells by reverse transport. Furthermore, it is integral to the function of caveolae; a mediator of Hedgehog regulation; and a ligand for the binding of cytolytic toxins to membranes. Active cholesterol modulates a variety of plasma membrane proteins-receptors, channels and transporters-at least in vitro.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
19
|
Sturley SL, Rajakumar T, Hammond N, Higaki K, Márka Z, Márka S, Munkacsi AB. Potential COVID-19 therapeutics from a rare disease: weaponizing lipid dysregulation to combat viral infectivity. J Lipid Res 2020; 61:972-982. [PMID: 32457038 PMCID: PMC7328045 DOI: 10.1194/jlr.r120000851] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/21/2020] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2 has resulted in the death of more than 328,000 persons worldwide in the first 5 months of 2020. Herculean efforts to rapidly design and produce vaccines and other antiviral interventions are ongoing. However, newly evolving viral mutations, the prospect of only temporary immunity, and a long path to regulatory approval pose significant challenges and call for a common, readily available, and inexpensive treatment. Strategic drug repurposing combined with rapid testing of established molecular targets could provide a pause in disease progression. SARS-CoV-2 shares extensive structural and functional conservation with SARS-CoV-1, including engagement of the same host cell receptor (angiotensin-converting enzyme 2) localized in cholesterol-rich microdomains. These lipid-enveloped viruses encounter the endosomal/lysosomal host compartment in a critical step of infection and maturation. Niemann-Pick type C (NP-C) disease is a rare monogenic neurodegenerative disease caused by deficient efflux of lipids from the late endosome/lysosome (LE/L). The NP-C disease-causing gene (NPC1) has been strongly associated with viral infection, both as a filovirus receptor (e.g., Ebola) and through LE/L lipid trafficking. This suggests that NPC1 inhibitors or NP-C disease mimetics could serve as anti-SARS-CoV-2 agents. Fortunately, there are such clinically approved molecules that elicit antiviral activity in preclinical studies, without causing NP-C disease. Inhibition of NPC1 may impair viral SARS-CoV-2 infectivity via several lipid-dependent mechanisms, which disturb the microenvironment optimum for viral infectivity. We suggest that known mechanistic information on NPC1 could be utilized to identify existing and future drugs to treat COVID-19.
Collapse
MESH Headings
- Androstenes/therapeutic use
- Angiotensin-Converting Enzyme 2
- Anticholesteremic Agents/therapeutic use
- Antiviral Agents/therapeutic use
- Betacoronavirus/drug effects
- Betacoronavirus/metabolism
- Betacoronavirus/pathogenicity
- COVID-19
- Cholesterol/metabolism
- Coronavirus Infections/diagnosis
- Coronavirus Infections/drug therapy
- Coronavirus Infections/epidemiology
- Drug Repositioning/methods
- Humans
- Hydroxychloroquine/therapeutic use
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Lysosomes/drug effects
- Lysosomes/metabolism
- Lysosomes/virology
- Niemann-Pick C1 Protein
- Niemann-Pick Disease, Type C/drug therapy
- Niemann-Pick Disease, Type C/genetics
- Niemann-Pick Disease, Type C/metabolism
- Niemann-Pick Disease, Type C/pathology
- Pandemics
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- Pneumonia, Viral/diagnosis
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/epidemiology
- Protein Binding
- Receptors, Virus/antagonists & inhibitors
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/metabolism
Collapse
Affiliation(s)
| | - Tamayanthi Rajakumar
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| | - Natalie Hammond
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| | - Katsumi Higaki
- Division of Functional Genomics,
Tottori University, Yonago 683-8503,
Japan
| | - Zsuzsa Márka
- Department of Physics,
Columbia University, New York,
NY 10027
| | - Szabolcs Márka
- Department of Physics,
Columbia University, New York,
NY 10027
| | - Andrew B. Munkacsi
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| |
Collapse
|
20
|
Martello A, Platt FM, Eden ER. Staying in touch with the endocytic network: The importance of contacts for cholesterol transport. Traffic 2020; 21:354-363. [PMID: 32129938 PMCID: PMC8650999 DOI: 10.1111/tra.12726] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/21/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022]
Abstract
Cholesterol homeostasis is critical for cell function and human health. Cholesterol is heterogeneously distributed among cellular membranes, with the redistribution of endocytosed dietary cholesterol playing a pivotal role in the regulation of cholesterol homeostasis. While gaps remain in our understanding of intracellular dietary cholesterol transport, a highly complex network of pathways is starting to emerge, often involving inter‐dependent vesicular and non‐vesicular transport mechanisms. The last decade has seen a surge in interest in non‐vesicular transport and inter‐organellar communication at membrane contact sites. By providing platforms for protein interactions, signalling events, lipid exchange and calcium flux, membrane contact sites (MCS) are now appreciated as controlling the fate of large amounts of lipid and play central roles in the regulation and co‐ordination of endocytic trafficking. Here, we review the role of MCS in multiple pathways for cholesterol export from the endocytic pathway and highlight the intriguing interplay between vesicular and non‐vesicular transport mechanisms and relationship with neurodegenerative disease.
Collapse
Affiliation(s)
| | - Fran M Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | |
Collapse
|
21
|
Meng Y, Heybrock S, Neculai D, Saftig P. Cholesterol Handling in Lysosomes and Beyond. Trends Cell Biol 2020; 30:452-466. [PMID: 32413315 DOI: 10.1016/j.tcb.2020.02.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/14/2020] [Accepted: 02/21/2020] [Indexed: 01/06/2023]
Abstract
Lysosomes are of major importance for the regulation of cellular cholesterol homeostasis. Food-derived cholesterol and cholesterol esters contained within lipoproteins are delivered to lysosomes by endocytosis. From the lysosomal lumen, cholesterol is transported to the inner surface of the lysosomal membrane through the glycocalyx; this shuttling requires Niemann-Pick C (NPC) 1 and NPC2 proteins. The lysosomal membrane proteins lysosomal-associated membrane protein (LAMP)-2 and lysosomal integral membrane protein (LIMP)-2/SCARB2 also bind cholesterol. LAMP-2 may serve as a cholesterol reservoir, whereas LIMP-2, like NPC1, is able to transport cholesterol through a transglycocalyx tunnel. Contact sites and fusion events between lysosomes and other organelles mediate the distribution of cholesterol. Lysosomal cholesterol content is sensed thereby regulating mammalian target of rapamycin complex (mTORC)-dependent signaling. This review summarizes our understanding of the major steps in cholesterol handling from the moment it enters the lysosome until it leaves this compartment.
Collapse
Affiliation(s)
- Ying Meng
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Saskia Heybrock
- Biochemisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Dante Neculai
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Paul Saftig
- Biochemisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany.
| |
Collapse
|
22
|
Johnson KA, Endapally S, Vazquez DC, Infante RE, Radhakrishnan A. Ostreolysin A and anthrolysin O use different mechanisms to control movement of cholesterol from the plasma membrane to the endoplasmic reticulum. J Biol Chem 2019; 294:17289-17300. [PMID: 31597703 DOI: 10.1074/jbc.ra119.010393] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/08/2019] [Indexed: 01/30/2023] Open
Abstract
Recent studies using two cholesterol-binding bacterial toxin proteins, perfringolysin O (PFO) and domain 4 of anthrolysin O (ALOD4), have shown that cholesterol in the plasma membranes (PMs) of animal cells resides in three distinct pools. The first pool comprises mobile cholesterol, accessible to both PFO and ALOD4, that is rapidly transported to the endoplasmic reticulum (ER) to signal cholesterol excess and maintain cholesterol homeostasis. The second is a sphingomyelin (SM)-sequestered pool inaccessible to PFO and ALOD4 but that becomes accessible by treatment with SM-degrading sphingomyelinase (SMase). The third is an essential pool also inaccessible to PFO and ALOD4 that cannot be liberated by SMase treatment. The accessible cholesterol pool can be trapped on PMs of live cells by nonlytic ALOD4, blocking its transport to the ER. However, studies of the two other pools have been hampered by a lack of available tools. Here, we used ostreolysin A (OlyA), which specifically binds SM/cholesterol complexes in membranes, to study the SM-sequestered cholesterol pool. Binding of nonlytic OlyA to SM/cholesterol complexes in PMs of live cells depleted the accessible PM cholesterol pool detectable by ALOD4. Consequently, transport of accessible cholesterol from PM to ER ceased, thereby activating SREBP transcription factors and increasing cholesterol synthesis. Thus, OlyA and ALOD4 both control movement of PM cholesterol, but through different lipid-binding mechanisms. We also found that PM-bound OlyA was rapidly internalized into cells, whereas PM-bound ALOD4 remained on the cell surface. Our findings establish OlyA and ALOD4 as complementary tools to investigate cellular cholesterol transport.
Collapse
Affiliation(s)
- Kristen A Johnson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Shreya Endapally
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Danya C Vazquez
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Rodney E Infante
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390 .,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
23
|
NPC1 regulates ER contacts with endocytic organelles to mediate cholesterol egress. Nat Commun 2019; 10:4276. [PMID: 31537798 PMCID: PMC6753064 DOI: 10.1038/s41467-019-12152-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 08/19/2019] [Indexed: 12/19/2022] Open
Abstract
Transport of dietary cholesterol from endocytic organelles to the endoplasmic reticulum (ER) is essential for cholesterol homoeostasis, but the mechanism and regulation of this transport remains poorly defined. Membrane contact sites (MCS), microdomains of close membrane apposition, are gaining attention as important platforms for non-vesicular, inter-organellar communication. Here we investigate the impact of ER-endocytic organelle MCS on cholesterol transport. We report a role for Niemann-Pick type C protein 1 (NPC1) in tethering ER-endocytic organelle MCS where it interacts with the ER-localised sterol transport protein Gramd1b to regulate cholesterol egress. We show that artificially tethering MCS rescues the cholesterol accumulation that characterises NPC1-deficient cells, consistent with direct lysosome to ER cholesterol transport across MCS. Finally, we identify an expanded population of lysosome-mitochondria MCS in cells depleted of NPC1 or Gramd1b that is dependent on the late endosomal sterol-binding protein STARD3, likely underlying the mitochondrial cholesterol accumulation in NPC1-deficient cells. Though endocytosed dietary cholesterol is transferred to the endoplasmic reticulum (ER), how this is regulated is unclear. Here, the authors report a role for Niemann-Pick Type C Protein 1 (NPC1) in tethering endocytic organelles to the ER, which may contribute to interorganelle cholesterol transport.
Collapse
|
24
|
Rodriguez-Agudo D, Malacrida L, Kakiyama G, Sparrer T, Fortes C, Maceyka M, Subler MA, Windle JJ, Gratton E, Pandak WM, Gil G. StarD5: an ER stress protein regulates plasma membrane and intracellular cholesterol homeostasis. J Lipid Res 2019; 60:1087-1098. [PMID: 31015253 PMCID: PMC6547630 DOI: 10.1194/jlr.m091967] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/08/2019] [Indexed: 01/01/2023] Open
Abstract
How plasma membrane (PM) cholesterol is controlled is poorly understood. Ablation of the gene encoding the ER stress steroidogenic acute regulatory-related lipid transfer domain (StarD)5 leads to a decrease in PM cholesterol content, a decrease in cholesterol efflux, and an increase in intracellular neutral lipid accumulation in macrophages, the major cell type that expresses StarD5. ER stress increases StarD5 expression in mouse hepatocytes, which results in an increase in accessible PM cholesterol in WT but not in StarD5-/- hepatocytes. StarD5-/- mice store higher levels of cholesterol and triglycerides, which leads to altered expression of cholesterol-regulated genes. In vitro, a recombinant GST-StarD5 protein transfers cholesterol between synthetic liposomes. StarD5 overexpression leads to a marked increase in PM cholesterol. Phasor analysis of 6-dodecanoyl-2-dimethylaminonaphthalene fluorescence lifetime imaging microscopy data revealed an increase in PM fluidity in StarD5-/- macrophages. Taken together, these studies show that StarD5 is a stress-responsive protein that regulates PM cholesterol and intracellular cholesterol homeostasis.
Collapse
Affiliation(s)
- Daniel Rodriguez-Agudo
- Departments of Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
- McGuire Veterans Affairs Medical Center, Richmond, VA 23248
| | - Leonel Malacrida
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California at Irvine, Irvine, CA 92697
- Area de Investigación Respiratoria, Departamento de Fisiopatologia, Hospital de Clinicas, Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Genta Kakiyama
- Departments of Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
- McGuire Veterans Affairs Medical Center, Richmond, VA 23248
| | - Tavis Sparrer
- Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Carolina Fortes
- Departments of Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
- Departmento de Biologia Molecular y Bioquimica, Universidad de Malaga, Malaga, Spain
| | - Michael Maceyka
- Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Mark A Subler
- Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Jolene J Windle
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
- Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California at Irvine, Irvine, CA 92697
| | - William M Pandak
- Departments of Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
- McGuire Veterans Affairs Medical Center, Richmond, VA 23248
| | - Gregorio Gil
- Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| |
Collapse
|
25
|
Cholesterol transport through the peroxisome-ER membrane contacts tethered by PI(4,5)P 2 and extended synaptotagmins. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1117-1135. [PMID: 31144242 DOI: 10.1007/s11427-019-9569-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 10/26/2022]
Abstract
Most mammalian cells take up cholesterol from low-density lipoproteins (LDLs) via receptor-mediated endocytosis. After reaching lysosomes, LDL-derived cholesterol continues to transport to downstream organelles including the ER for specific structural and functional needs. Peroxisomes are recently found to receive cholesterol from lysosomes through lysosome-peroxisome membrane contacts. However, whether and how cholesterol is conveyed from peroxisomes to the ER remain unknown. Here, by combining high-resolution microscopic analyses and in vitro reconstitution of highly purified organelles or artificial liposomes, we demonstrate that peroxisomes form membrane contacts with the ER through the interaction between peroxisomal PI(4,5)P2 and ER-resident extended synaptotagmin-1, 2 and 3 (E-Syts). Depletion of peroxisomal PI(4,5)P2 or E-Syts markedly decreases peroxisome-ER membrane contacts and induces cholesterol accumulation in lysosomes. Furthermore, we show that cholesterol is delivered from 3H-labeled peroxisomes or PI(4,5)P2-containing liposomes to the ER in vitro, and that the presence of peroxisomes augments cholesterol transfer from lysosomes to the ER. Together, our study reveals a new cholesterol transport pathway along the lysosome-peroxisome-ER membrane contacts in the cell.
Collapse
|
26
|
Kanda T, Sugihara T, Takata T, Mae Y, Kinoshita H, Sakaguchi T, Hasegawa T, Kurumi H, Ikebuchi Y, Murakami T, Isomoto H. Low-density lipoprotein receptor expression is involved in the beneficial effect of photodynamic therapy using talaporfin sodium on gastric cancer cells. Oncol Lett 2019; 17:3261-3266. [PMID: 30867758 PMCID: PMC6396135 DOI: 10.3892/ol.2019.10004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/17/2019] [Indexed: 01/05/2023] Open
Abstract
Photodynamic therapy (PDT) is a therapeutic method used to destroy tumor tissue via reactive oxygen. Notably, reactive oxygen is induced by a combination of photosensitizers, including talaporfin sodium (TS) and laser light. Gastric cancer cell lines, MKN45 and MKN74, were used to evaluate the effect of TS-PDT in vitro. The antitumor effect of TS-PDT, which was evaluated via cellular viability assay, on MKN74 was weaker than that on MKN45 cells, suggesting that MKN74 cell could be resistant to TS-PDT. However, using a higher TS concentration or setting a longer treatment time (24 h) resulted in effective TS-PDT treatment on MKN74 cells. In addition, when irradiation power of LED was raised up to 5.06 J/cm2, TS-PDT was able to induce an antitumor effect on MKN74 cells. This suggested that the difference in TS-PDT efficacy between MKN45 and MKN74 cells is based on the difference in cellular uptake of TS. As expected, uptake of TS by MKN74 cells was lower than that by MKN45 cells. The expression levels of low-density lipoprotein (LDL) receptor in MKN74 cells were lower than those in MKN45 cells. With GW3965 treatment, an agonist/activator of Liver X Receptor, LDL receptor expression was reduced, weakening the TS-PDT effect. Furthermore, as a hydroxymethylglutaryl-Coenzyme A reductase inhibitor, treatment using simvastatin increased LDL receptor expression, leading to enhancement of the TS-PDT effect on MKN74 cells. In conclusion, the difference in LDL receptor expression between the two gastric cell lines could influence TS-PDT efficacy; simvastatin may enhance the antitumor effect of TS-PDT through upregulating the LDL receptor even on PDT-resistant gastric cancer cells.
Collapse
Affiliation(s)
- Tsutomu Kanda
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Takaaki Sugihara
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Tomoaki Takata
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Yukari Mae
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Hidehito Kinoshita
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Takuki Sakaguchi
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Takashi Hasegawa
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Hiroki Kurumi
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Yuichiro Ikebuchi
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| | - Takashi Murakami
- Department of Microbiology, Faculty of Medicine Saitama Medical University, Moroyama, Saitama 350-0495, Japan
| | - Hajime Isomoto
- Division of Medicine and Clinical Science, Faculty of Medicine Tottori University, Yonago, Tottori 683-8504, Japan
| |
Collapse
|
27
|
Luo J, Jiang LY, Yang H, Song BL. Intracellular Cholesterol Transport by Sterol Transfer Proteins at Membrane Contact Sites. Trends Biochem Sci 2019; 44:273-292. [DOI: 10.1016/j.tibs.2018.10.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/06/2018] [Accepted: 10/10/2018] [Indexed: 12/20/2022]
|
28
|
Hanada K. Lipid transfer proteins rectify inter-organelle flux and accurately deliver lipids at membrane contact sites. J Lipid Res 2018; 59:1341-1366. [PMID: 29884707 PMCID: PMC6071762 DOI: 10.1194/jlr.r085324] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/24/2018] [Indexed: 12/22/2022] Open
Abstract
The endoplasmic reticulum (ER) is the main center for the synthesis of various lipid types in cells, and newly synthesized lipids are delivered from the ER to other organelles. In the past decade, various lipid transfer proteins (LTPs) have been recognized as mediators of lipid transport from the ER to other organelles; inter-organelle transport occurs at membrane contact sites (MCSs) and in a nonvesicular manner. Although the intermembrane transfer reaction catalyzed by LTPs is an equilibrium reaction, various types of newly synthesized lipids are transported unidirectionally in cells. This review provides a brief history of the inter-organelle trafficking of lipids and summarizes the structural and biochemical characteristics of the ceramide transport protein (CERT) as a typical LTP acting at MCSs. In addition, this review compares several LTP-mediated inter-organelle lipid trafficking systems and proposes that LTPs generate unidirectional fluxes of specific lipids between different organelles by indirect coupling with the metabolic reactions that occur in specific organelles. Moreover, the available data also suggest that the major advantage of LTP-mediated lipid transport at MCSs may be the accuracy of delivery. Finally, how cholesterol is enriched in the plasma membrane is discussed from a thermodynamic perspective.
Collapse
Affiliation(s)
- Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| |
Collapse
|
29
|
Khosravi M, Hosseini-Fard R, Najafi M. Circulating low density lipoprotein (LDL). Horm Mol Biol Clin Investig 2018; 35:/j/hmbci.ahead-of-print/hmbci-2018-0024/hmbci-2018-0024.xml. [PMID: 30059347 DOI: 10.1515/hmbci-2018-0024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/22/2018] [Indexed: 12/13/2022]
Abstract
Low-density lipoprotein (LDL) particles are known as atherogenic agents in coronary artery diseases. They modify to other electronegative forms and may be the subject for improvement of inflammatory events in vessel subendothelial spaces. The circulating LDL value is associated with the plasma PCSK-9 level. They internalize into macrophages using the lysosomal receptor-mediated pathways. LDL uptake is related to the membrane scavenger receptors, modifications of lipid and protein components of LDL particles, vesicular maturation and lipid stores of cells. Furthermore, LDL vesicular trafficking is involved with the function of some proteins such as Rab and Lamp families. These proteins also help in the transportation of free cholesterol from lysosome into the cytosol. The aggregation of lipids in the cytosol is a starting point for the formation of foam cells so that they may participate in the primary core of atherosclerosis plaques. The effects of macrophage subclasses are different in the formation and remodeling of plaques. This review is focused on the cellular and molecular events involved in cholesterol homeostasis.
Collapse
Affiliation(s)
- Mohsen Khosravi
- Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Hosseini-Fard
- Biochemistry Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Cellular and Molecular Research Center, Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran, Phone: 09155192401
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Cholesterol is delivered to the limiting membrane of late endosomes by Niemann-Pick Type C1 and C2 proteins. This review summarizes recent evidence that cholesterol transfer from endosomes to the endoplasmic reticulum and other organelles is mediated by lipid-binding proteins that localize to membrane contact sites (MCS). RECENT FINDINGS LDL-cholesterol in the late endosomal/lysosomes is exported to the plasma membrane, where most cholesterol resides, and the endoplasmic reticulum, which harbors the regulatory complexes and enzymes that control the synthesis and esterification of cholesterol. A major advance in dissecting these cholesterol transport pathways was identification of frequent and dynamic MCS between endosomes and the endoplasmic reticulum, peroxisomes and plasma membrane. Positioned at these MCS are members of the oxysterol-binding protein (OSBP) and steroidogenic acute regulatory protein-related lipid-transfer family of lipid transfer proteins that bridge the opposing membranes and directly or indirectly mediate cholesterol transfer. OSBP-related protein 1L (ORP1L), ORP5 and ORP6 mediate cholesterol transfer to the endoplasmic reticulum that regulates cholesterol homeostasis. ORP1L and STARD3 also move cholesterol from the endoplasmic reticulum-to-late endosomal/lysosomes under low-cholesterol conditions to facilitate intraluminal vesicle formation. Cholesterol transport also occurs at MCS with peroxisomes and possibly the plasma membrane. SUMMARY Frequent contacts between organelles and the endo-lysosomal vesicles are sites for bidirectional transfer of cholesterol.
Collapse
Affiliation(s)
- Neale D Ridgway
- Department of Biochemistry & Molecular Biology
- Department of Pediatrics, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kexin Zhao
- Department of Biochemistry & Molecular Biology
- Department of Pediatrics, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
31
|
Quantitative Measurement of Cholesterol in Cell Populations Using Flow Cytometry and Fluorescent Perfringolysin O. Methods Mol Biol 2018; 1583:85-95. [PMID: 28205169 DOI: 10.1007/978-1-4939-6875-6_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Methods to quantify intracellular cholesterol are valuable for the study of its trafficking and storage in normal cells and in lysosomal storage disorders. Traditionally, cholesterol has been tracked using the small molecule, filipin. Filipin can be difficult to visualize and visualization can be cytotoxic as it requires UV illumination. Here we describe a method to measure cholesterol using a fluorescently labeled, mutant form of Perfringolysin O, a soluble protein toxin that binds cholesterol specifically. This approach has been used to measure the impact of NPC1 deficiency on lysosomal cholesterol levels and monitor the rescue of cholesterol export under conditions that reduce the thickness of the lysosomal glycocalyx.
Collapse
|
32
|
Zhang S, Glukhova SA, Caldwell KA, Caldwell GA. NCEH-1 modulates cholesterol metabolism and protects against α-synuclein toxicity in a C. elegans model of Parkinson's disease. Hum Mol Genet 2018; 26:3823-3836. [PMID: 28934392 DOI: 10.1093/hmg/ddx269] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/06/2017] [Indexed: 02/03/2023] Open
Abstract
Parkinson's disease (PD) is an aging-associated neurodegenerative disease affecting millions worldwide. Misfolding, oligomerization and accumulation of the human α-synuclein protein is a key pathological hallmark of PD and is associated with the progressive loss of dopaminergic neurons over the course of aging. Lifespan extension via the suppression of IGF-1/insulin-like signaling (IIS) offers a possibility to retard disease onset through induction of metabolic changes that provide neuroprotection. The nceh-1 gene of Caenorhabditis elegans encodes an ortholog of neutral cholesterol ester hydrolase 1 (NCEH-1), an IIS downstream protein that was identified in a screen as a modulator of α-synuclein accumulation in vivo. The mechanism whereby cholesterol metabolism functionally impacts neurodegeneration induced by α-synuclein is undefined. Here we report that NCEH-1 protects dopaminergic neurons from α-synuclein-dependent neurotoxicity in C. elegans via a mechanism that is independent of lifespan extension. We discovered that the presence of cholesterol, LDLR-mediated cholesterol endocytosis, and cholesterol efflux are all essential to NCEH-1-mediated neuroprotection. In protecting from α-synuclein neurotoxicity, NCEH-1 also stimulates cholesterol-derived neurosteroid formation and lowers cellular reactive oxygen species in mitochondria. Collectively, this study augments our understanding of how cholesterol metabolism can modulate a neuroprotective mechanism that attenuates α-synuclein neurotoxicity, thereby pointing toward regulation of neuronal cholesterol turnover as a potential therapeutic avenue for PD.
Collapse
Affiliation(s)
- Siyuan Zhang
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Samantha A Glukhova
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA.,Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama School of Medicine at Birmingham, Birmingham, AL 35294, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA.,Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama School of Medicine at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
33
|
The Role of Mitochondria in the Activation/Maintenance of SOCE: Membrane Contact Sites as Signaling Hubs Sustaining Store-Operated Ca2+ Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:277-296. [DOI: 10.1007/978-3-319-57732-6_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
|
34
|
Infante RE, Radhakrishnan A. Continuous transport of a small fraction of plasma membrane cholesterol to endoplasmic reticulum regulates total cellular cholesterol. eLife 2017; 6. [PMID: 28414269 PMCID: PMC5433840 DOI: 10.7554/elife.25466] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/16/2017] [Indexed: 01/15/2023] Open
Abstract
Cells employ regulated transport mechanisms to ensure that their plasma membranes (PMs) are optimally supplied with cholesterol derived from uptake of low-density lipoproteins (LDL) and synthesis. To date, all inhibitors of cholesterol transport block steps in lysosomes, limiting our understanding of post-lysosomal transport steps. Here, we establish the cholesterol-binding domain 4 of anthrolysin O (ALOD4) as a reversible inhibitor of cholesterol transport from PM to endoplasmic reticulum (ER). Using ALOD4, we: (1) deplete ER cholesterol without altering PM or overall cellular cholesterol levels; (2) demonstrate that LDL-derived cholesterol travels from lysosomes first to PM to meet cholesterol needs, and subsequently from PM to regulatory domains of ER to suppress activation of SREBPs, halting cholesterol uptake and synthesis; and (3) determine that continuous PM-to-ER cholesterol transport allows ER to constantly monitor PM cholesterol levels, and respond rapidly to small declines in cellular cholesterol by activating SREBPs, increasing cholesterol uptake and synthesis.
Collapse
Affiliation(s)
- Rodney Elwood Infante
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States.,Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Arun Radhakrishnan
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
35
|
Chang TY, Yamauchi Y, Hasan MT, Chang C. Cellular cholesterol homeostasis and Alzheimer's disease. J Lipid Res 2017; 58:2239-2254. [PMID: 28298292 DOI: 10.1194/jlr.r075630] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/14/2017] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia in older adults. Currently, there is no cure for AD. The hallmark of AD is the accumulation of extracellular amyloid plaques composed of amyloid-β (Aβ) peptides (especially Aβ1-42) and neurofibrillary tangles, composed of hyperphosphorylated tau and accompanied by chronic neuroinflammation. Aβ peptides are derived from the amyloid precursor protein (APP). The oligomeric form of Aβ peptides is probably the most neurotoxic species; its accumulation eventually forms the insoluble and aggregated amyloid plaques. ApoE is the major apolipoprotein of the lipoprotein(s) present in the CNS. ApoE has three alleles, of which the Apoe4 allele constitutes the major risk factor for late-onset AD. Here we describe the complex relationship between ApoE4, oligomeric Aβ peptides, and cholesterol homeostasis. The review consists of four parts: 1) key elements involved in cellular cholesterol metabolism and regulation; 2) key elements involved in intracellular cholesterol trafficking; 3) links between ApoE4, Aβ peptides, and disturbance of cholesterol homeostasis in the CNS; 4) potential lipid-based therapeutic targets to treat AD. At the end, we recommend several research topics that we believe would help in better understanding the connection between cholesterol and AD for further investigations.
Collapse
Affiliation(s)
- Ta-Yuan Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Yoshio Yamauchi
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Mazahir T Hasan
- Laboratory of Memory Circuits, Achucarro Basque Center for Neuroscience, Zamudio, Spain
| | - Catherine Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
| |
Collapse
|
36
|
Kuzu OF, Toprak M, Noory MA, Robertson GP. Effect of lysosomotropic molecules on cellular homeostasis. Pharmacol Res 2017; 117:177-184. [DOI: 10.1016/j.phrs.2016.12.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/13/2016] [Indexed: 01/01/2023]
|
37
|
Li J, Wang X, Yuan Y, Long X, Bao J, Li X. An in vitro test system for evaluation of SCAP–SREBP pathway inhibitory activities of Traditional Chinese Medicines. RSC Adv 2017. [DOI: 10.1039/c7ra09521j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In vitro assay system demonstrates that Rhizoma Alismatis and Semen Cassiae show beneficial effects on inhibition of SCAP–SREBP pathway activities.
Collapse
Affiliation(s)
- Jianzong Li
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education
- College of Life Sciences
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Xin Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education
- College of Life Sciences
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Yuan Yuan
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education
- College of Life Sciences
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Xin Long
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education
- College of Life Sciences
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Jinku Bao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education
- College of Life Sciences
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Xin Li
- State Key Laboratory of Oral Diseases
- National Clinical Research Center for Oral Diseases
- West China Hospital of Stomatology
- Sichuan University
- Chengdu
| |
Collapse
|
38
|
Eden ER. The formation and function of ER-endosome membrane contact sites. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1861:874-879. [PMID: 26898183 PMCID: PMC4917889 DOI: 10.1016/j.bbalip.2016.01.020] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/27/2016] [Accepted: 01/29/2016] [Indexed: 02/02/2023]
Abstract
Recent advances in membrane contact site (MCS) biology have revealed key roles for MCSs in inter-organellar exchange, the importance of which is becoming increasingly apparent. Roles for MCSs in many essential physiological processes including lipid transfer, calcium exchange, receptor tyrosine kinase signalling, lipid droplet formation, autophagosome formation, organelle dynamics and neurite outgrowth have been reported. The ER forms an extensive and dynamic network of MCSs with a diverse range of functionally distinct organelles. MCSs between the ER and endocytic pathway are particularly abundant, suggesting important physiological roles. Here, our current knowledge of the formation and function of ER contact sites with endocytic organelles from studies in mammalian systems is reviewed. Their relatively poorly defined molecular composition and recently identified functions are discussed. In addition, likely, but yet to be established, roles for these contacts in lipid transfer and calcium signalling are considered. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
|
39
|
Ortiz-Sandoval CG, Hughes SC, Dacks JB, Simmen T. Interaction with the effector dynamin-related protein 1 (Drp1) is an ancient function of Rab32 subfamily proteins. CELLULAR LOGISTICS 2014; 4:e986399. [PMID: 25767741 PMCID: PMC4355727 DOI: 10.4161/21592799.2014.986399] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/06/2014] [Indexed: 01/21/2023]
Abstract
The mitochondria-associated membrane (MAM) is an endoplasmic reticulum (ER) domain that forms contacts with mitochondria and accommodates Ca2+ transfer between the two organelles. The GTPase Rab32 regulates this function of the MAM via determining the localization of the Ca2+ regulatory transmembrane protein calnexin to the MAM. Another function of the MAM is the regulation of mitochondrial dynamics mediated by GTPases such as dynamin-related protein 1 (Drp1). Consistent with the importance of the MAM for mitochondrial dynamics and the role of Rab32 in MAM enrichment, the inactivation of Rab32 leads to mitochondrial collapse around the nucleus. However, Rab32 and related Rabs also perform intracellular functions at locations other than the MAM including melanosomal trafficking, autophagosome formation and maturation, and retrograde trafficking to the trans-Golgi network (TGN). This plethora of functions raises questions concerning the original cellular role of Rab32 in the last common ancestor of animals and its possible role in the last eukaryotic common ancestor (LECA). Our results now shed light on this conundrum and identify a role in Drp1-mediated mitochondrial dynamics as one common denominator of this group of Rabs, which includes the paralogues Rab32A and Rab32B, as well as the more recently derived Rab29 and Rab38 proteins. Moreover, we provide evidence that this mitochondrial function is dictated by the extent of ER-association of Rab32 family proteins.
Collapse
Affiliation(s)
- Carolina G Ortiz-Sandoval
- Faculty of Medicine and Dentistry; Department of Cell Biology; University of Alberta ; Edmonton, Alberta, Canada
| | - Sarah C Hughes
- Faculty of Medicine and Dentistry; Department of Cell Biology; University of Alberta ; Edmonton, Alberta, Canada ; Faculty of Medicine and Dentistry; Department of Medical Genetics; University of Alberta ; Edmonton, Alberta, Canada
| | - Joel B Dacks
- Faculty of Medicine and Dentistry; Department of Cell Biology; University of Alberta ; Edmonton, Alberta, Canada
| | - Thomas Simmen
- Faculty of Medicine and Dentistry; Department of Cell Biology; University of Alberta ; Edmonton, Alberta, Canada
| |
Collapse
|
40
|
Akpovi CD, Murphy BD, Erickson RP, Pelletier RM. Dysregulation of testicular cholesterol metabolism following spontaneous mutation of the niemann-pick c1 gene in mice. Biol Reprod 2014; 91:42. [PMID: 25009206 DOI: 10.1095/biolreprod.114.119412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The Niemann-Pick-type C1 (Npc1) protein mobilizes LDL-derived cholesterol from lysosomes. Npc1 deficiency disease is a panethnic autosomal recessive disorder of intracellular cholesterol trafficking, leading to accumulation of cholesterol in endosomes/lysosomes. This report assesses the effects of a spontaneous inactivating mutation of the Npc1 gene on spermatogenesis and cholesterol homeostasis in mice. We quantified 1) free and esterified cholesterol levels by enzymatic analysis, 2) cholesterol enzymes and transporter protein expression by Western blotting, and 3) the number of Apostain-labeled apoptotic germ cells and apoptosis levels by ELISA in seminiferous tubule-enriched fractions. In wild-type (WT) mice, esterified cholesterol was elevated when Npc1 expression was low during puberty, while in adulthood, the levels were low (P < 0.05) when Npc1 expression was high (P < 0.01). In Npc1-/- mice, free and esterified cholesterol were significantly elevated. The abundance of cholesterol regulatory proteins, HMGR ACAT1, ACAT2, SR-BI, and ABCA1 was significantly higher in Npc1-/- than in WT mice. The level of apoptosis determined by ELISA and the number of Apostain-labeled cells/tubule were higher in Npc1-/- than in WT mice. Circulating testosterone levels in the Npc1-/- males were threefold lower than those observed in the WT. Deleting the Npc1 gene is accompanied by an increase in germ cell apoptosis and compensatory imbalances in the expression of cholesterol enzymatic and transporter factors and is associated with esterified cholesterol accumulation in seminiferous tubules.
Collapse
Affiliation(s)
- Casimir D Akpovi
- Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Bruce D Murphy
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, St.-Hyacinthe, Québec, Canada
| | - Robert P Erickson
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, Arizona
| | - R-Marc Pelletier
- Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
41
|
Das A, Brown MS, Anderson DD, Goldstein JL, Radhakrishnan A. Three pools of plasma membrane cholesterol and their relation to cholesterol homeostasis. eLife 2014; 3:e02882. [PMID: 24920391 PMCID: PMC4086274 DOI: 10.7554/elife.02882] [Citation(s) in RCA: 272] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/10/2014] [Indexed: 01/08/2023] Open
Abstract
When human fibroblasts take up plasma low density lipoprotein (LDL), its cholesterol is liberated in lysosomes and eventually reaches the endoplasmic reticulum (ER) where it inhibits cholesterol synthesis by blocking activation of SREBPs. This feedback protects against cholesterol overaccumulation in the plasma membrane (PM). But how does ER know whether PM is saturated with cholesterol? In this study, we define three pools of PM cholesterol: (1) a pool accessible to bind 125I-PFO*, a mutant form of bacterial Perfringolysin O, which binds cholesterol in membranes; (2) a sphingomyelin(SM)-sequestered pool that binds 125I-PFO* only after SM is destroyed by sphingomyelinase; and (3) a residual pool that does not bind 125I-PFO* even after sphingomyelinase treatment. When LDL-derived cholesterol leaves lysosomes, it expands PM's PFO-accessible pool and, after a short lag, it also increases the ER's PFO-accessible regulatory pool. This regulatory mechanism allows cells to ensure optimal cholesterol levels in PM while avoiding cholesterol overaccumulation.
Collapse
Affiliation(s)
- Akash Das
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Michael S Brown
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Donald D Anderson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Joseph L Goldstein
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
42
|
Brandis KA, Gale S, Jinn S, Langmade SJ, Dudley-Rucker N, Jiang H, Sidhu R, Ren A, Goldberg A, Schaffer JE, Ory DS. Box C/D small nucleolar RNA (snoRNA) U60 regulates intracellular cholesterol trafficking. J Biol Chem 2013; 288:35703-13. [PMID: 24174535 DOI: 10.1074/jbc.m113.488577] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mobilization of plasma membrane (PM) cholesterol to the endoplasmic reticulum is essential for cellular cholesterol homeostasis. The mechanisms regulating this retrograde, intermembrane cholesterol transfer are not well understood. Because mutant cells with defects in PM to endoplasmic reticulum cholesterol trafficking can be isolated on the basis of resistance to amphotericin B, we conducted an amphotericin B loss-of-function screen in Chinese hamster ovary (CHO) cells using insertional mutagenesis to identify genes that regulate this trafficking mechanism. Mutant line A1 displayed reduced cholesteryl ester formation from PM-derived cholesterol and increased de novo cholesterol synthesis, indicating a deficiency in retrograde cholesterol transport. Genotypic analysis revealed that the A1 cell line contained one disrupted allele of the U60 small nucleolar RNA (snoRNA) host gene, resulting in haploinsufficiency of the box C/D snoRNA U60. Complementation and mutational studies revealed the U60 snoRNA to be the essential feature from this locus that affects cholesterol trafficking. Lack of alteration in predicted U60-mediated site-directed methylation of 28 S rRNA in the A1 mutant suggests that the U60 snoRNA modulates cholesterol trafficking by a mechanism that is independent of this canonical function. Our study adds to a growing body of evidence for participation of small noncoding RNAs in cholesterol homeostasis and is the first to implicate a snoRNA in this cellular function.
Collapse
Affiliation(s)
- Katrina A Brandis
- From the Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, Missouri 63110
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Use of mutant 125I-perfringolysin O to probe transport and organization of cholesterol in membranes of animal cells. Proc Natl Acad Sci U S A 2013; 110:10580-5. [PMID: 23754385 DOI: 10.1073/pnas.1309273110] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Animal cells strictly control the distribution of cholesterol in their organelle membranes. This regulation requires an efficient machinery to transport insoluble cholesterol between organelles. In the present study, we generate an (125)I-labeled mutant version of Perfringolysin O (PFO), a cholesterol-binding protein, and use it to measure cholesterol in the plasma membrane of intact cells. We also purify plasma membranes from the same cells, which allows us to directly relate cholesterol concentration to (125)I-PFO binding. We show that cholesterol is organized in the plasma membrane in a manner that makes it inaccessible to PFO until its concentration exceeds a threshold of 35 mol% of total lipids. This 35% threshold is in striking contrast to the 5% threshold previously found for PFO binding to endoplasmic reticulum membranes. The (125)I-PFO probe also proved useful in monitoring the movement of LDL-derived cholesterol from lysosomes to plasma membranes. Using three different mutant cell lines, we show that this transport requires receptor-mediated uptake of LDL, hydrolysis of LDL-cholesteryl esters in lysosomes, and transfer of the liberated cholesterol to the plasma membrane.
Collapse
|
44
|
Amritraj A, Wang Y, Revett TJ, Vergote D, Westaway D, Kar S. Role of cathepsin D in U18666A-induced neuronal cell death: potential implication in Niemann-Pick type C disease pathogenesis. J Biol Chem 2012; 288:3136-52. [PMID: 23250759 DOI: 10.1074/jbc.m112.412460] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cathepsin D is an aspartyl protease that plays a crucial role in normal cellular functions and in a variety of neurodegenerative disorders, including Niemann-Pick type C (NPC) disease, which is characterized by intracellular accumulation of cholesterol and glycosphingolipids in many tissues, including the brain. There is evidence that the level and activity of cathepsin D increased markedly in vulnerable neurons in NPC pathology, but its involvement in neurodegeneration remains unclear. In the present study, using mouse hippocampal cultured neurons, we evaluated the significance of cathepsin D in toxicity induced by U18666A, a class II amphiphile, which triggers cell death by impairing the trafficking of cholesterol, as observed in NPC pathology. Our results showed that U18666A-mediated toxicity is accompanied by an increase in cathepsin D mRNA and enzyme activity but a decrease in the total peptide content. The cytosolic level of cathepsin D, on the other hand, was increased along with cytochrome c and activated caspase-3 in U18666A-treated neurons. The cathepsin D inhibitor, pepstatin A, partially protected neurons against toxicity by attenuating these signaling mechanisms. Additionally, down-regulation of cathepsin D level prevented, whereas overexpression of the protease increased, vulnerability of cultured N2a cells to U18666A-induced toxicity. We also showed that extracellular cathepsin D from U18666A-treated neurons or application of exogenous enzyme can induce neurotoxicity by activating the autophagic pathway. These results suggest that increased release/activation of cathepsin D can trigger neurodegeneration and possibly development of NPC pathology. Thus, targeting cathepsin D level/activity may provide a new therapeutic opportunity for the treatment of NPC pathology.
Collapse
Affiliation(s)
- Asha Amritraj
- Department of Psychiatry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2M8, Canada
| | | | | | | | | | | |
Collapse
|
45
|
Cholesterol accumulation inhibits ER to Golgi transport and protein secretion: studies of apolipoprotein E and VSVGt. Biochem J 2012; 447:51-60. [PMID: 22747346 DOI: 10.1042/bj20111891] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cholesterol excess is typical of various diseases including atherosclerosis. We have investigated whether cholesterol accumulation in the ER (endoplasmic reticulum) can inhibit exit of vesicular cargo and secretion of proteins by studying apoE (apolipoprotein E), a significant glycoprotein in human health and disease. CHO (Chinese hamster ovary) cells expressing human apoE under a cholesterol-independent promoter incubated with cholesterol-cyclodextrin complexes showed increased levels of cellular free and esterified cholesterol, inhibition of SREBP-2 (sterol-regulatory-element-binding protein 2) processing, and a mild induction of ER stress, indicating significant accumulation of cholesterol in the ER. Secretion of apoE was markedly inhibited by cholesterol accumulation, and similar effects were observed in cells enriched with lipoprotein-derived cholesterol and in primary human macrophages. Removal of excess cholesterol by a cyclodextrin vehicle restored apoE secretion, indicating that the transport defect was reversible. That cholesterol impaired protein trafficking was supported by the cellular accumulation of less sialylated apoE glycoforms, and by direct visualization of altered ER to Golgi transport of thermo-reversible VSVG (vesicular stomatitis virus glycoprotein) linked to GFP (green fluorescent protein). We conclude that intracellular accumulation of cholesterol in the ER reversibly inhibits protein transport and secretion. Strategies to correct ER cholesterol may restore homoeostatic processes and intracellular protein transport in conditions characterized by cholesterol excess.
Collapse
|
46
|
Sorci-Thomas MG, Owen JS, Fulp B, Bhat S, Zhu X, Parks JS, Shah D, Jerome WG, Gerelus M, Zabalawi M, Thomas MJ. Nascent high density lipoproteins formed by ABCA1 resemble lipid rafts and are structurally organized by three apoA-I monomers. J Lipid Res 2012; 53:1890-909. [PMID: 22750655 PMCID: PMC3413229 DOI: 10.1194/jlr.m026674] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 06/29/2012] [Indexed: 12/29/2022] Open
Abstract
This report details the lipid composition of nascent HDL (nHDL) particles formed by the action of the ATP binding cassette transporter A1 (ABCA1) on apolipoprotein A-I (apoA-I). nHDL particles of different size (average diameters of ∼ 12, 10, 7.5, and <6 nm) and composition were purified by size-exclusion chromatography. Electron microscopy suggested that the nHDL were mostly spheroidal. The proportions of the principal nHDL lipids, free cholesterol, glycerophosphocholine, and sphingomyelin were similar to that of lipid rafts, suggesting that the lipid originated from a raft-like region of the cell. Smaller amounts of glucosylceramides, cholesteryl esters, and other glycerophospholipid classes were also present. The largest particles, ∼ 12 nm and 10 nm diameter, contained ∼ 43% free cholesterol, 2-3% cholesteryl ester, and three apoA-I molecules. Using chemical cross-linking chemistry combined with mass spectrometry, we found that three molecules of apoA-I in the ∼ 9-14 nm nHDL adopted a belt-like conformation. The smaller (7.5 nm diameter) spheroidal nHDL particles carried 30% free cholesterol and two molecules of apoA-I in a twisted, antiparallel, double-belt conformation. Overall, these new data offer fresh insights into the biogenesis and structural constraints involved in forming nascent HDL from ABCA1.
Collapse
Affiliation(s)
- Mary G Sorci-Thomas
- Department of Pathology, Section on Lipid Sciences, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Shen WJ, Zaidi SK, Patel S, Cortez Y, Ueno M, Azhar R, Azhar S, Kraemer FB. Ablation of vimentin results in defective steroidogenesis. Endocrinology 2012; 153:3249-57. [PMID: 22535769 PMCID: PMC3380307 DOI: 10.1210/en.2012-1048] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In steroidogenic tissues, cholesterol must be transported to the inner mitochondrial membrane to be converted to pregnenolone as the first step of steroidogenesis. Whereas steroidogenic acute regulatory protein has been shown to be responsible for the transport of cholesterol from the outer to the inner mitochondrial membrane, the process of how cholesterol moves to mitochondria from the cytoplasm is not clearly defined. The involvement of the cytoskeleton has been suggested; however, no specific mechanism has been confirmed. In this paper, using genetic ablation of an intermediate filament protein in mice, we present data demonstrating a marked defect in adrenal and ovarian steroidogenesis in the absence of vimentin. Cosyntropin-stimulated corticosterone production is decreased 35 and 50% in male and female Vimentin null (Vim(-/-)) mice, respectively, whereas progesterone production is decreased 70% in female Vim(-/-) mice after pregnant mare's serum gonadotropin and human chorionic gonadotropin stimulation, but no abnormalities in human chorionic gonadotropin-stimulated testosterone production is observed in male Vim(-/-) mice. These defects in steroid production are also seen in isolated adrenal and granulosa cells in vitro. Further studies show a defect in the movement of cholesterol from the cytosol to mitochondria in Vim(-/-) cells. Because the mobilization of cholesterol from lipid droplets and its transport to mitochondria is a preferred pathway for the initiation of steroid production in the adrenal and ovary but not the testis and vimentin is a droplet-associated protein, our results suggest that vimentin is involved in the movement of cholesterol from its storage in lipid droplets to mitochondria for steroidogenesis.
Collapse
Affiliation(s)
- Wen-Jun Shen
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University, Palo Alto, California 94304, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Appelqvist H, Nilsson C, Garner B, Brown AJ, Kågedal K, Ollinger K. Attenuation of the lysosomal death pathway by lysosomal cholesterol accumulation. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:629-39. [PMID: 21281795 DOI: 10.1016/j.ajpath.2010.10.030] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/24/2010] [Accepted: 10/21/2010] [Indexed: 10/18/2022]
Abstract
In the past decade, lysosomal membrane permeabilization (LMP) has emerged as a significant component of cell death signaling. The mechanisms by which lysosomal stability is regulated are not yet fully understood, but changes in the lysosomal membrane lipid composition have been suggested to be involved. Our aim was to investigate the importance of cholesterol in the regulation of lysosomal membrane permeability and its potential impact on apoptosis. Treatment of normal human fibroblasts with U18666A, an amphiphilic drug that inhibits cholesterol transport and causes accumulation of cholesterol in lysosomes, rescued cells from lysosome-dependent cell death induced by the lysosomotropic detergent O-methyl-serine dodecylamide hydrochloride (MSDH), staurosporine (STS), or cisplatin. LMP was decreased by pretreating cells with U18666A, and there was a linear relationship between the cholesterol content of lysosomes and their resistance to permeabilization induced by MSDH. U18666A did not induce changes in expression or localization of 70-kDa heat shock proteins (Hsp70) or antiapoptotic Bcl-2 proteins known to protect the lysosomal membrane. Induction of autophagy also was excluded as a contributor to the protective mechanism. By using Chinese hamster ovary (CHO) cells with lysosomal cholesterol overload due to a mutation in the cholesterol transporting protein Niemann-Pick type C1 (NPC1), the relationship between lysosomal cholesterol accumulation and protection from lysosome-dependent cell death was confirmed. Cholesterol accumulation in lysosomes attenuates apoptosis by increasing lysosomal membrane stability.
Collapse
Affiliation(s)
- Hanna Appelqvist
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | | | | | | | | | | |
Collapse
|
49
|
Sorokina EM, Feinstein SI, Zhou S, Fisher AB. Intracellular targeting of peroxiredoxin 6 to lysosomal organelles requires MAPK activity and binding to 14-3-3ε. Am J Physiol Cell Physiol 2011; 300:C1430-41. [PMID: 21346153 DOI: 10.1152/ajpcell.00285.2010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peroxiredoxin 6 (Prdx6), a bifunctional protein with GSH peroxidase and lysosomal-type phospholipase A(2) activities, has been localized to both cytosolic and acidic compartments (lamellar bodies and lysosomes) in lung alveolar epithelium. We postulate that Prdx6 subcellular localization affects the balance between the two activities. Immunostaining localized Prdx6 to lysosome-related organelles in the MLE12 and A549 alveolar epithelial cell lines. Inhibition of trafficking by brefeldin A indicated processing of the protein through the vesicular pathway. Trafficking of Prdx6 was decreased by inhibitors of PKC, ERK, and p38 MAPK. Immunocytochemistry, immunoprecipitation, and an in situ proximity ligation assay (Duolink) showed that binding of the lysosomal targeting sequence of Prdx6 (amino acids 31-40) to 14-3-3ε was dependent on activity of PKC, ERK, and p38 MAPK. Knockdown of 14-3-3ε with siRNA inhibited the lysosomal targeting of Prdx6. In vitro study with recombinant proteins by pull-down assay and surface plasmon resonance confirmed the interaction of Prdx6 and 14-3-3ε. These findings suggest that ERK and p38 MAPK regulate subcellular localization of Prdx6 by activation of 14-3-3ε as a chaperone protein, resulting in its translocation to acidic organelles.
Collapse
Affiliation(s)
- Elena M Sorokina
- Institute for Environmental Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
50
|
Tong F, Billheimer J, Shechtman CF, Liu Y, Crooke R, Graham M, Cohen DE, Sturley SL, Rader DJ. Decreased expression of ARV1 results in cholesterol retention in the endoplasmic reticulum and abnormal bile acid metabolism. J Biol Chem 2010; 285:33632-41. [PMID: 20663892 PMCID: PMC2962461 DOI: 10.1074/jbc.m110.165761] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Indexed: 11/06/2022] Open
Abstract
Endoplasmic reticulum (ER) membrane cholesterol is maintained at an optimal concentration of ∼5 mol % by the net impact of sterol synthesis, modification, and export. Arv1p was first identified in the yeast Saccharomyces cerevisiae as a key component of this homeostasis due to its probable role in intracellular sterol transport. Mammalian ARV1, which can fully complement the yeast lesion, encodes a ubiquitously expressed, resident ER protein. Repeated dosing of specific antisense oligonucleotides to ARV1 produced a marked reduction of ARV1 transcripts in liver, adipose, and to a lesser extent, intestine. This resulted in marked hypercholesterolemia, elevated serum bile acids, and activation of the hepatic farnesoid X receptor (FXR) regulatory pathway. Knockdown of ARV1 in murine liver and HepG2 cells was associated with accumulation of cholesterol in the ER at the expense of the plasma membrane and suppression of sterol regulatory element-binding proteins and their targets. These studies indicate a critical role of mammalian Arv1p in sterol movement from the ER and in the ensuing regulation of hepatic cholesterol and bile acid metabolism.
Collapse
Affiliation(s)
- Fumin Tong
- From the Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| | - Jeffrey Billheimer
- From the Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| | - Caryn F. Shechtman
- the Institute of Human Nutrition and Department of Pediatrics, Columbia University Medical Center, New York, New York 10032
| | - Ying Liu
- the Institute of Human Nutrition and Department of Pediatrics, Columbia University Medical Center, New York, New York 10032
| | - Roseann Crooke
- ISIS Pharmaceuticals, Inc., Carlsbad, California 92008, and
| | - Mark Graham
- ISIS Pharmaceuticals, Inc., Carlsbad, California 92008, and
| | - David E. Cohen
- the Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Stephen L. Sturley
- the Institute of Human Nutrition and Department of Pediatrics, Columbia University Medical Center, New York, New York 10032
| | - Daniel J. Rader
- From the Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| |
Collapse
|