51
|
Zhang H, Shen L, Li Y, Xu Z, Zhang X, Yu J, Cao Z, Luan P. Genome-wide association study for plasma very low-density lipoprotein concentration in chicken. J Anim Breed Genet 2019; 136:351-361. [PMID: 31037768 DOI: 10.1111/jbg.12397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/20/2019] [Accepted: 04/01/2019] [Indexed: 01/17/2023]
Abstract
The plasma very low-density lipoprotein (VLDL) concentration is an effective blood biochemical indicator that could be used to select lean chicken lines. In the current study, we used Genome-wide association study (GWAS) method to detect SNPs with significant effects on plasma VLDL concentration. As a result, 38 SNPs significantly associated with plasma VLDL concentration were identified using at least one of the three mixed linear model (MLM) packages, including GRAMMAR, EMMAX and GEMMA. Nearly, all these SNPs with significant effects on plasma VLDL concentration (except Gga_rs16160897) have significantly different allele frequencies between lean and fat lines. The 1-Mb regions surrounding these 38 SNPs were extracted, and twelve important regions were obtained after combining the overlaps. A total of 122 genes in these twelve important regions were detected. Among these genes, LRRK2, ABCD2, TLR4, E2F1, SUGP1, NCAN, KLF2 and RAB8A were identified as important genes for plasma VLDL concentration based on their basic functions. The results of this study may supply useful information to select lean chicken lines.
Collapse
Affiliation(s)
- Hui Zhang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Linyong Shen
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yumao Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Zichun Xu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Xinyang Zhang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Jiaqiang Yu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Zhiping Cao
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Peng Luan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
52
|
Ioannou GN, Landis CS, Jin GY, Haigh WG, Farrell GC, Kuver R, Lee SP, Savard C. Cholesterol Crystals in Hepatocyte Lipid Droplets Are Strongly Associated With Human Nonalcoholic Steatohepatitis. Hepatol Commun 2019; 3:776-791. [PMID: 31168512 PMCID: PMC6545865 DOI: 10.1002/hep4.1348] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
It is unclear what drives the development of fibrosing nonalcoholic steatohepatitis (NASH). We aimed to determine whether cholesterol crystallization within hepatocyte lipid droplets (LDs) distinguishes patients with fibrosing NASH from patients with isolated hepatic steatosis and to study pathways leading to cholesterol accumulation in hepatocyte LDs. Patients with fibrosing NASH (n = 16) were compared to patients with isolated steatosis (n = 14). Almost all patients with fibrosing NASH had free cholesterol staining by filipin (16/16) and cholesterol crystals (15/16) in hepatocyte LDs, mostly in association with the LD membrane, compared to only 3/14 with cholesterol crystals and 3/14 with faint filipin staining in patients with isolated steatosis (P < 0.05). We were unable to identify significant differences in the expression of genes in liver tissue related to cholesterol homeostasis or LD proteins between patients with fibrosing NASH and isolated steatosis. Human hepatoma cell line (HepG2) cells were supplemented with low-density lipoprotein (LDL)-cholesterol and oleic acid to develop large LDs, similar to those observed in patients with NASH. Fluorescent markers were used to track the uptake and intracellular trafficking of LDL-cholesterol. LDL-cholesterol was taken up by HepG2 cells and transported through the endosomal-lysosomal compartment directly to LDs, suggesting direct contact sites between late endosomes and LDs. Exposure of HepG2 cells to LDL-cholesterol resulted in a high concentration of cholesterol and cholesterol crystallization in LDs. Conclusion: Excess cholesterol is stored in the liver primarily within hepatocyte LDs where it can crystallize. Our findings are best explained by direct transport of cholesterol from late endosomes/lysosomes to LDs in hepatocytes. We found a strong association between the presence of LD cholesterol crystals and the development of fibrosing NASH in humans, suggesting a causal relationship.
Collapse
Affiliation(s)
- George N Ioannou
- Division of Gastroenterology, Department of Medicine Veterans Affairs Puget Sound Health Care System Seattle WA.,Division of Gastroenterology, Department of Medicine University of Washington Seattle WA.,Research and Development Veterans Affairs Puget Sound Health Care System Seattle WA
| | - Charles S Landis
- Division of Gastroenterology, Department of Medicine University of Washington Seattle WA
| | - Ga-Young Jin
- Research and Development Veterans Affairs Puget Sound Health Care System Seattle WA
| | - W Geoffrey Haigh
- Research and Development Veterans Affairs Puget Sound Health Care System Seattle WA
| | - Geoffrey C Farrell
- Liver Research Group Australian National University Medical School at the Canberra Hospital Garran Australia
| | - Rahul Kuver
- Division of Gastroenterology, Department of Medicine University of Washington Seattle WA
| | - Sum P Lee
- Division of Gastroenterology, Department of Medicine University of Washington Seattle WA
| | - Christopher Savard
- Division of Gastroenterology, Department of Medicine Veterans Affairs Puget Sound Health Care System Seattle WA.,Division of Gastroenterology, Department of Medicine University of Washington Seattle WA.,Research and Development Veterans Affairs Puget Sound Health Care System Seattle WA
| |
Collapse
|
53
|
Vacca F, Vossio S, Mercier V, Moreau D, Johnson S, Scott CC, Montoya JP, Moniatte M, Gruenberg J. Cyclodextrin triggers MCOLN1-dependent endo-lysosome secretion in Niemann-Pick type C cells. J Lipid Res 2019; 60:832-843. [PMID: 30709900 PMCID: PMC6446697 DOI: 10.1194/jlr.m089979] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
In specialized cell types, lysosome-related organelles support regulated secretory pathways, whereas in nonspecialized cells, lysosomes can undergo fusion with the plasma membrane in response to a transient rise in cytosolic calcium. Recent evidence also indicates that lysosome secretion can be controlled transcriptionally and promote clearance in lysosome storage diseases. In addition, evidence is also accumulating that low concentrations of cyclodextrins reduce the cholesterol-storage phenotype in cells and animals with the cholesterol storage disease Niemann-Pick type C, via an unknown mechanism. Here, we report that cyclodextrin triggers the secretion of the endo/lysosomal content in nonspecialized cells and that this mechanism is responsible for the decreased cholesterol overload in Niemann-Pick type C cells. We also find that the secretion of the endo/lysosome content occurs via a mechanism dependent on the endosomal calcium channel mucolipin-1, as well as FYCO1, the AP1 adaptor, and its partner Gadkin. We conclude that endo-lysosomes in nonspecialized cells can acquire secretory functions elicited by cyclodextrin and that this pathway is responsible for the decrease in cholesterol storage in Niemann-Pick C cells.
Collapse
Affiliation(s)
- Fabrizio Vacca
- Department of Biochemistry, University of Geneva, 1211-Geneva-4, Switzerland
| | - Stefania Vossio
- Department of Biochemistry, University of Geneva, 1211-Geneva-4, Switzerland
| | - Vincent Mercier
- Department of Biochemistry, University of Geneva, 1211-Geneva-4, Switzerland
| | - Dimitri Moreau
- Department of Biochemistry, University of Geneva, 1211-Geneva-4, Switzerland
| | - Shem Johnson
- Department of Biochemistry, University of Geneva, 1211-Geneva-4, Switzerland
| | - Cameron C Scott
- Department of Biochemistry, University of Geneva, 1211-Geneva-4, Switzerland
| | - Jonathan Paz Montoya
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Marc Moniatte
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, 1211-Geneva-4, Switzerland.
| |
Collapse
|
54
|
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]
|
55
|
Mechanisms of cellular cholesterol compartmentalization: recent insights. Curr Opin Cell Biol 2018; 53:77-83. [PMID: 29960186 DOI: 10.1016/j.ceb.2018.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/07/2018] [Accepted: 06/05/2018] [Indexed: 11/20/2022]
Abstract
This review discusses advances in understanding how the controlled delivery of cholesterol between subcellular compartments is achieved and what novel experimental strategies are being employed to address this fundamental question. Recent work has focused on cholesterol-binding proteins that can facilitate directional cholesterol transfer between contacts of the ER and Golgi or late endosomal membranes. Increasing structural information on cholesterol-binding proteins, new modules engineered from them as well as improved imaging and gene editing techniques are providing valuable insights. There is also mounting information on how the crosstalk between cholesterol transport and nutrient signaling is orchestrated and how cellular fatty acid metabolism and cholesterol homeostasis are intertwined.
Collapse
|
56
|
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
|
57
|
Jin X, Dimitriadis EK, Liu Y, Combs CA, Chang J, Varsano N, Stempinski E, Flores R, Jackson SN, Muller L, Woods AS, Addadi L, Kruth HS. Macrophages Shed Excess Cholesterol in Unique Extracellular Structures Containing Cholesterol Microdomains. Arterioscler Thromb Vasc Biol 2018; 38:1504-1518. [PMID: 29853567 PMCID: PMC6023747 DOI: 10.1161/atvbaha.118.311269] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/16/2018] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Cells use various mechanisms to maintain cellular cholesterol homeostasis including efflux of cholesterol from the cellular plasma membrane to cholesterol acceptors such as HDLs (high-density lipoproteins). Little is known about the transfer of cholesterol from cells into the extracellular matrix. Using a unique monoclonal antibody that detects ordered cholesterol arrays (ie, cholesterol micro[or nano]-domains), we previously identified that particles containing these cholesterol domains accumulate in the extracellular matrix during cholesterol enrichment of human monocyte-derived macrophages and are found in atherosclerotic lesions. In this study, we further investigate these deposited particles containing cholesterol microdomains and discover their unexpected morphology. Approach and Results— Although appearing spherical at the resolution of the conventional fluorescence microscope, super-resolution immunofluorescence and atomic force microscopy of in situ cholesterol microdomains, and immunoelectron microscopy of isolated cholesterol microdomains revealed that the microdomains are not vesicles or 3-dimensional crystals but rather appear as branching irregularly shaped deposits of varying size. These cholesterol microdomain-containing deposits are shed from the plasma membrane into the extracellular matrix. Conclusions— To date, research on cellular excretion of excess cholesterol has demonstrated cellular cholesterol efflux in the form of membranous vesicles and discoidal HDL particles released into the fluid-phase medium. Shedding of plasma membrane cholesterol microdomains provides an additional mechanism for cells such as macrophages to maintain plasma membrane cholesterol homeostasis. Furthermore, recognition that macrophages shed cholesterol microdomains into the extracellular matrix is important to our understanding of extracellular buildup of cholesterol in atherosclerosis.
Collapse
Affiliation(s)
- Xueting Jin
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| | - Emilios K Dimitriadis
- Scanning Probe Microscopy Unit, National Institute of Biomedical Imaging and Bioengineering (E.K.D.)
| | - Ying Liu
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| | - Christian A Combs
- Light Microscopy Core, National Heart, Lung, and Blood Institute (C.A.C.)
| | - Janet Chang
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| | - Neta Varsano
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (N.V., L.A.)
| | - Erin Stempinski
- Electron Microscopy Core, National Heart, Lung, and Blood Institute (E.S.)
| | - Rhonda Flores
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| | - Shelley N Jackson
- Structural Biology Core, National Institute of Drug Abuse (S.N.J., L.M., A.S.W.), National Institutes of Health, Baltimore, MD
| | - Ludovic Muller
- Structural Biology Core, National Institute of Drug Abuse (S.N.J., L.M., A.S.W.), National Institutes of Health, Baltimore, MD
| | - Amina S Woods
- Structural Biology Core, National Institute of Drug Abuse (S.N.J., L.M., A.S.W.), National Institutes of Health, Baltimore, MD
| | - Lia Addadi
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (N.V., L.A.)
| | - Howard S Kruth
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| |
Collapse
|
58
|
Sobajima T, Yoshimura SI, Maeda T, Miyata H, Miyoshi E, Harada A. The Rab11-binding protein RELCH/KIAA1468 controls intracellular cholesterol distribution. J Cell Biol 2018; 217:1777-1796. [PMID: 29514919 PMCID: PMC5940305 DOI: 10.1083/jcb.201709123] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/20/2018] [Accepted: 02/16/2018] [Indexed: 12/29/2022] Open
Abstract
Cholesterol, which is endocytosed to the late endosome (LE)/lysosome, is delivered to other organelles through vesicular and nonvesicular transport mechanisms. In this study, we discuss a novel mechanism of cholesterol transport from recycling endosomes (REs) to the trans-Golgi network (TGN) through RELCH/KIAA1468, which is newly identified in this study as a Rab11-GTP- and OSBP-binding protein. After treating cells with 25-hydroxycholesterol to induce OSBP relocation from the cytoplasm to the TGN, REs accumulated around the TGN area, but this accumulation was diminished in RELCH- or OSBP-depleted cells. Cholesterol content in the TGN was decreased in Rab11-, RELCH-, and OSBP-depleted cells and increased in the LE/lysosome. According to in vitro reconstitution experiments, RELCH tethers Rab11-bound RE-like and OSBP-bound TGN-like liposomes and promotes OSBP-dependent cholesterol transfer from RE-like to TGN-like liposomes. These data suggest that RELCH promotes nonvesicular cholesterol transport from REs to the TGN through membrane tethering.
Collapse
Affiliation(s)
- Tomoaki Sobajima
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shin-Ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomomi Maeda
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Haruhiko Miyata
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
59
|
Hussain SS, Harris MT, Kreutzberger AJB, Inouye CM, Doyle CA, Castle AM, Arvan P, Castle JD. Control of insulin granule formation and function by the ABC transporters ABCG1 and ABCA1 and by oxysterol binding protein OSBP. Mol Biol Cell 2018. [PMID: 29540530 PMCID: PMC5935073 DOI: 10.1091/mbc.e17-08-0519] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In pancreatic β-cells, insulin granule membranes are enriched in cholesterol and are both recycled and newly generated. Cholesterol’s role in supporting granule membrane formation and function is poorly understood. ATP binding cassette transporters ABCG1 and ABCA1 regulate intracellular cholesterol and are important for insulin secretion. RNAi interference–induced depletion in cultured pancreatic β-cells shows that ABCG1 is needed to stabilize newly made insulin granules against lysosomal degradation; ABCA1 is also involved but to a lesser extent. Both transporters are also required for optimum glucose-stimulated insulin secretion, likely via complementary roles. Exogenous cholesterol addition rescues knockdown-induced granule loss (ABCG1) and reduced secretion (both transporters). Another cholesterol transport protein, oxysterol binding protein (OSBP), appears to act proximally as a source of endogenous cholesterol for granule formation. Its knockdown caused similar defective stability of young granules and glucose-stimulated insulin secretion, neither of which were rescued with exogenous cholesterol. Dual knockdowns of OSBP and ABC transporters support their serial function in supplying and concentrating cholesterol for granule formation. OSBP knockdown also decreased proinsulin synthesis consistent with a proximal endoplasmic reticulum defect. Thus, membrane cholesterol distribution contributes to insulin homeostasis at production, packaging, and export levels through the actions of OSBP and ABCs G1 and A1.
Collapse
Affiliation(s)
- Syed Saad Hussain
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Megan T Harris
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Alex J B Kreutzberger
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22908.,Center for Membrane and Cell Physiology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Candice M Inouye
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Catherine A Doyle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Anna M Castle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Peter Arvan
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - J David Castle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908.,Center for Membrane and Cell Physiology, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
60
|
Modzel M, Solanko KA, Szomek M, Hansen SK, Dupont A, Nåbo LJ, Kongsted J, Wüstner D. Live-cell imaging of new polyene sterols for improved analysis of intracellular cholesterol transport. J Microsc 2018. [PMID: 29516493 DOI: 10.1111/jmi.12691] [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: 11/27/2022]
Abstract
Analysis of intracellular cholesterol transport by fluorescence microscopy requires suitable fluorescent analogues of cholesterol. Most existing cholesterol analogues contain lipophilic dyes which can compromise the sterol properties in membranes. An alternative strategy is to introduce additional double bonds into the sterol ring system resulting in intrinsic fluorescence, while at the same time keeping the cholesterol-like properties of the analogues. Existing polyene sterols, such as dehydroergosterol (DHE) or cholestatrienol (CTL), however, contain only three double bonds and suffer from low brightness, significant photobleaching and excitation/emission in the ultraviolet region. Thus, special equipment is required to image such sterols. Here, we describe synthesis, characterization and intracellular imaging of new polyene sterols containing four conjugated double bonds in the sterol ring system. We show that such analogues have red-shifted excitation and emission by ∼20 nm compared to DHE or CTL. The red shift was even more pronounced when preventing keto-enol tautomer equilibration by protecting the 3'-hydroxy group with acetate. We show that the latter analogue can be imaged on a conventional wide field microscope with a DAPI/filipin filter cube. The new polyene sterols show reduced photobleaching compared to DHE or CTL allowing for improved deconvolution microscopy of sterol containing cellular membranes.
Collapse
Affiliation(s)
- M Modzel
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | - K A Solanko
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | - M Szomek
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | - S K Hansen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | - A Dupont
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | - L J Nåbo
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense, Denmark
| | - J Kongsted
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense, Denmark
| | - D Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| |
Collapse
|
61
|
Viaud M, Ivanov S, Vujic N, Duta-Mare M, Aira LE, Barouillet T, Garcia E, Orange F, Dugail I, Hainault I, Stehlik C, Marchetti S, Boyer L, Guinamard R, Foufelle F, Bochem A, Hovingh KG, Thorp EB, Gautier EL, Kratky D, Dasilva-Jardine P, Yvan-Charvet L. Lysosomal Cholesterol Hydrolysis Couples Efferocytosis to Anti-Inflammatory Oxysterol Production. Circ Res 2018. [PMID: 29523554 DOI: 10.1161/circresaha.117.312333] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Macrophages face a substantial amount of cholesterol after the ingestion of apoptotic cells, and the LIPA (lysosomal acid lipase) has a major role in hydrolyzing cholesteryl esters in the endocytic compartment. OBJECTIVE Here, we directly investigated the role of LIPA-mediated clearance of apoptotic cells both in vitro and in vivo. METHODS AND RESULTS We show that LIPA inhibition causes a defective efferocytic response because of impaired generation of 25-hydroxycholesterol and 27-hydroxycholesterol. Reduced synthesis of 25-hydroxycholesterol after LIPA inhibition contributed to defective mitochondria-associated membrane leading to mitochondrial oxidative stress-induced NLRP3 (NOD-like receptor family, pyrin domain containing) inflammasome activation and caspase-1-dependent Rac1 (Ras-related C3 botulinum toxin substrate 1) degradation. A secondary event consisting of failure to appropriately activate liver X receptor-mediated pathways led to mitigation of cholesterol efflux and apoptotic cell clearance. In mice, LIPA inhibition caused defective clearance of apoptotic lymphocytes and stressed erythrocytes by hepatic and splenic macrophages, culminating in splenomegaly and splenic iron accumulation under hypercholesterolemia. CONCLUSIONS Our findings position lysosomal cholesterol hydrolysis as a critical process that prevents metabolic inflammation by enabling efficient macrophage apoptotic cell clearance.
Collapse
Affiliation(s)
- Manon Viaud
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Stoyan Ivanov
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Nemanja Vujic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Madalina Duta-Mare
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Lazaro-Emilio Aira
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | | | - Elsa Garcia
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Francois Orange
- UFR Sciences, Faculté des Sciences de l'Université de Nice-Sophia Antipolis, France (F.O.)
| | - Isabelle Dugail
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Pierre & Marie Curie University, ICAN Institute of Cardiometabolism & Nutrition, Hôpital de la Pitié, Boulevard de l'Hôpital, Paris, France (I.D., E.L.G.)
| | - Isabelle Hainault
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMRS 1138, Centre de Recherche des Cordeliers, Paris, France (I.H., F.F.)
| | - Christian Stehlik
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL (C.S., E.B.T.)
| | - Sandrine Marchetti
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Laurent Boyer
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Rodolphe Guinamard
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Fabienne Foufelle
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMRS 1138, Centre de Recherche des Cordeliers, Paris, France (I.H., F.F.)
| | | | | | - Edward B Thorp
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL (C.S., E.B.T.)
| | - Emmanuel L Gautier
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Pierre & Marie Curie University, ICAN Institute of Cardiometabolism & Nutrition, Hôpital de la Pitié, Boulevard de l'Hôpital, Paris, France (I.D., E.L.G.)
| | - Dagmar Kratky
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Paul Dasilva-Jardine
- Academic Medical Center, Amsterdam, The Netherlands; and Staten Biotechnology, Nijmegen, The Netherlands (P.D.-J.)
| | - Laurent Yvan-Charvet
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| |
Collapse
|
62
|
Yamauchi Y, Rogers MA. Sterol Metabolism and Transport in Atherosclerosis and Cancer. Front Endocrinol (Lausanne) 2018; 9:509. [PMID: 30283400 PMCID: PMC6157400 DOI: 10.3389/fendo.2018.00509] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/14/2018] [Indexed: 01/22/2023] Open
Abstract
Cholesterol is a vital lipid molecule for mammalian cells, regulating fluidity of biological membranes, and serving as an essential constituent of lipid rafts. Mammalian cells acquire cholesterol from extracellular lipoproteins and from de novo synthesis. Cholesterol biosynthesis generates various precursor sterols. Cholesterol undergoes metabolic conversion into oxygenated sterols (oxysterols), bile acids, and steroid hormones. Cholesterol intermediates and metabolites have diverse and important cellular functions. A network of molecular machineries including transcription factors, protein modifiers, sterol transporters/carriers, and sterol sensors regulate sterol homeostasis in mammalian cells and tissues. Dysfunction in metabolism and transport of cholesterol, sterol intermediates, and oxysterols occurs in various pathophysiological settings such as atherosclerosis, cancers, and neurodegenerative diseases. Here we review the cholesterol, intermediate sterol, and oxysterol regulatory mechanisms and intracellular transport machineries, and discuss the roles of sterols and sterol metabolism in human diseases.
Collapse
Affiliation(s)
- Yoshio Yamauchi
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- *Correspondence: Yoshio Yamauchi
| | - Maximillian A. Rogers
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
63
|
Abstract
Signaling pathways direct organogenesis, often through concentration-dependent effects on cells. The hedgehog pathway enables cells to sense and respond to hedgehog ligands, of which the best studied is sonic hedgehog. Hedgehog signaling is essential for development, proliferation, and stem cell maintenance, and it is a driver of certain cancers. Lipid metabolism has a profound influence on both hedgehog signal transduction and the properties of the ligands themselves, leading to changes in the strength of hedgehog signaling and cellular functions. Here we review the evolving understanding of the relationship between lipids and hedgehog signaling.
Collapse
Affiliation(s)
- Robert Blassberg
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - John Jacob
- Nuffield Department of Clinical Neurosciences (NDCN), Level 6, West Wing, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK. .,Department of Neurology, West Wing, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK. .,Milton Keynes University Hospital, Standing Way, Eaglestone, Milton Keynes, MK6 5LD, UK.
| |
Collapse
|
64
|
Luo J, Jiang L, Yang H, Song BL. Routes and mechanisms of post-endosomal cholesterol trafficking: A story that never ends. Traffic 2017; 18:209-217. [DOI: 10.1111/tra.12471] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Jie Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Wuhan University; Wuhan China
| | - Luyi Jiang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Wuhan University; Wuhan China
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences; The University of New South Wales; Sydney Australia
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Wuhan University; Wuhan China
| |
Collapse
|
65
|
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
|
66
|
Nagasato AI, Yamashita H, Matsuo M, Ueda K, Kioka N. The distribution of vinculin to lipid rafts plays an important role in sensing stiffness of extracellular matrix. Biosci Biotechnol Biochem 2017; 81:1136-1147. [PMID: 28485208 DOI: 10.1080/09168451.2017.1289074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Extracellular matrix (ECM) stiffness regulates cell differentiation, survival, and migration. Our previous study has shown that the interaction of the focal adhesion protein vinculin with vinexin α plays a critical role in sensing ECM stiffness and regulating stiffness-dependent cell migration. However, the mechanism how vinculin-vinexin α interaction affects stiffness-dependent cell migration is unclear. Lipid rafts are membrane microdomains that are known to affect ECM-induced signals and cell behaviors. Here, we show that vinculin and vinexin α can localize to lipid rafts. Cell-ECM adhesion, intracellular tension, and a rigid ECM promote vinculin distribution to lipid rafts. The disruption of lipid rafts with Methyl-β-cyclodextrin impaired the ECM stiffness-mediated regulation of vinculin behavior and rapid cell migration on rigid ECM. These results indicate that lipid rafts play an important role in ECM-stiffness regulation of cell migration via vinculin.
Collapse
Affiliation(s)
- Ayaka Ichikawa Nagasato
- a Division of Applied Life Sciences, Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Hiroshi Yamashita
- a Division of Applied Life Sciences, Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Michinori Matsuo
- a Division of Applied Life Sciences, Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Kazumitsu Ueda
- a Division of Applied Life Sciences, Graduate School of Agriculture , Kyoto University , Kyoto , Japan.,b Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University , Kyoto , Japan
| | - Noriyuki Kioka
- a Division of Applied Life Sciences, Graduate School of Agriculture , Kyoto University , Kyoto , Japan.,b Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University , Kyoto , Japan
| |
Collapse
|
67
|
Li H, Ou L, Fan J, Xiao M, Kuang C, Liu X, Sun Y, Xu Y. Rab8A regulates insulin-stimulated GLUT4 translocation in C2C12 myoblasts. FEBS Lett 2017; 591:491-499. [DOI: 10.1002/1873-3468.12555] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/10/2016] [Accepted: 01/03/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Hanbing Li
- Department of Biomedical Engineering; Key Laboratory for Biomedical Engineering of Ministry of Education; Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal; Zhejiang University; Hangzhou China
- College of Pharmaceutical Science; Zhejiang University of Technology; Hangzhou China
| | - Liting Ou
- College of Pharmaceutical Science; Zhejiang University of Technology; Hangzhou China
| | - Jiannan Fan
- Department of Biomedical Engineering; Key Laboratory for Biomedical Engineering of Ministry of Education; Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal; Zhejiang University; Hangzhou China
| | - Mei Xiao
- College of Pharmaceutical Science; Zhejiang University of Technology; Hangzhou China
| | - Cuifang Kuang
- State Key Laboratory of Modern Optical Instrumentation; Department of Optical Engineering; Zhejiang University; Hangzhou China
| | - Xu Liu
- State Key Laboratory of Modern Optical Instrumentation; Department of Optical Engineering; Zhejiang University; Hangzhou China
| | - Yonghong Sun
- Department of Biomedical Engineering; Key Laboratory for Biomedical Engineering of Ministry of Education; Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal; Zhejiang University; Hangzhou China
| | - Yingke Xu
- Department of Biomedical Engineering; Key Laboratory for Biomedical Engineering of Ministry of Education; Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal; Zhejiang University; Hangzhou China
| |
Collapse
|
68
|
Grewal T, Hoque M, Conway JRW, Reverter M, Wahba M, Beevi SS, Timpson P, Enrich C, Rentero C. Annexin A6-A multifunctional scaffold in cell motility. Cell Adh Migr 2017; 11:288-304. [PMID: 28060548 DOI: 10.1080/19336918.2016.1268318] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Annexin A6 (AnxA6) belongs to a highly conserved protein family characterized by their calcium (Ca2+)-dependent binding to phospholipids. Over the years, immunohistochemistry, subcellular fractionations, and live cell microscopy established that AnxA6 is predominantly found at the plasma membrane and endosomal compartments. In these locations, AnxA6 acts as a multifunctional scaffold protein, recruiting signaling proteins, modulating cholesterol and membrane transport and influencing actin dynamics. These activities enable AnxA6 to contribute to the formation of multifactorial protein complexes and membrane domains relevant in signal transduction, cholesterol homeostasis and endo-/exocytic membrane transport. Hence, AnxA6 has been implicated in many biological processes, including cell proliferation, survival, differentiation, inflammation, but also membrane repair and viral infection. More recently, we and others identified roles for AnxA6 in cancer cell migration and invasion. This review will discuss how the multiple scaffold functions may enable AnxA6 to modulate migratory cell behavior in health and disease.
Collapse
Affiliation(s)
- Thomas Grewal
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Monira Hoque
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - James R W Conway
- b The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine , University of New South Wales , Sydney , NSW , Australia
| | - Meritxell Reverter
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| | - Mohamed Wahba
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Syed S Beevi
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Paul Timpson
- b The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine , University of New South Wales , Sydney , NSW , Australia
| | - Carlos Enrich
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| | - Carles Rentero
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| |
Collapse
|
69
|
Ferri N, Marchianò S, Tibolla G, Baetta R, Dhyani A, Ruscica M, Uboldi P, Catapano AL, Corsini A. PCSK9 knock-out mice are protected from neointimal formation in response to perivascular carotid collar placement. Atherosclerosis 2016; 253:214-224. [DOI: 10.1016/j.atherosclerosis.2016.07.910] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/25/2022]
|
70
|
Intracellular cholesterol transport proteins: roles in health and disease. Clin Sci (Lond) 2016; 130:1843-59. [DOI: 10.1042/cs20160339] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/12/2016] [Indexed: 12/13/2022]
Abstract
Effective cholesterol homoeostasis is essential in maintaining cellular function, and this is achieved by a network of lipid-responsive nuclear transcription factors, and enzymes, receptors and transporters subject to post-transcriptional and post-translational regulation, whereas loss of these elegant, tightly regulated homoeostatic responses is integral to disease pathologies. Recent data suggest that sterol-binding sensors, exchangers and transporters contribute to regulation of cellular cholesterol homoeostasis and that genetic overexpression or deletion, or mutations, in a number of these proteins are linked with diseases, including atherosclerosis, dyslipidaemia, diabetes, congenital lipoid adrenal hyperplasia, cancer, autosomal dominant hearing loss and male infertility. This review focuses on current evidence exploring the function of members of the ‘START’ (steroidogenic acute regulatory protein-related lipid transfer) and ‘ORP’ (oxysterol-binding protein-related proteins) families of sterol-binding proteins in sterol homoeostasis in eukaryotic cells, and the evidence that they represent valid therapeutic targets to alleviate human disease.
Collapse
|
71
|
Hölttä‐Vuori M, Sezgin E, Eggeling C, Ikonen E. Use of BODIPY-Cholesterol (TF-Chol) for Visualizing Lysosomal Cholesterol Accumulation. Traffic 2016; 17:1054-7. [PMID: 27187581 PMCID: PMC4988379 DOI: 10.1111/tra.12414] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 05/13/2016] [Accepted: 05/13/2016] [Indexed: 02/03/2023]
Abstract
Dipyrromethene difluoride-cholesterol (TopFluor-Cholesterol, TF-Chol) is a widely used cholesterol analogue due to its excellent fluorescence properties and considerable similarity with natural cholesterol in terms of membrane partitioning. However, the suitability of TF-Chol for detecting lysosomal cholesterol deposition has recently been questioned. Here, we highlight the fact that the method of lipid delivery and the analysis of time-point both affect the membrane distribution and labeling pattern of TF-Chol, similarly as with radiolabeled cholesterol. Lysosomal sterol accumulation characteristic to a lysosomal storage disease is most readily detected when the probe is introduced via the physiological route, i.e. as a sterol fatty acid ester in low-density lipoprotein particles. When administered to cells from solvent, lysosomal sterol sequestration becomes evident after an overnight equilibration between membranes.
Collapse
Affiliation(s)
- Maarit Hölttä‐Vuori
- Faculty of Medicine, Department of AnatomyUniversity of Helsinki00014 HelsinkiFinland
- Minerva Foundation Institute for Medical Research00290 HelsinkiFinland
| | - Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular MedicineUniversity of OxfordOX39DS OxfordUK
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular MedicineUniversity of OxfordOX39DS OxfordUK
| | - Elina Ikonen
- Faculty of Medicine, Department of AnatomyUniversity of Helsinki00014 HelsinkiFinland
- Minerva Foundation Institute for Medical Research00290 HelsinkiFinland
| |
Collapse
|
72
|
Chung S, Ghelfi M, Atkinson J, Parker R, Qian J, Carlin C, Manor D. Vitamin E and Phosphoinositides Regulate the Intracellular Localization of the Hepatic α-Tocopherol Transfer Protein. J Biol Chem 2016; 291:17028-39. [PMID: 27307040 DOI: 10.1074/jbc.m116.734210] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Indexed: 12/12/2022] Open
Abstract
α-Tocopherol (vitamin E) is an essential nutrient for all vertebrates. From the eight naturally occurring members of the vitamin E family, α-tocopherol is the most biologically active species and is selectively retained in tissues. The hepatic α-tocopherol transfer protein (TTP) preferentially selects dietary α-tocopherol and facilitates its transport through the hepatocyte and its secretion to the circulation. In doing so, TTP regulates body-wide levels of α-tocopherol. The mechanisms by which TTP facilitates α-tocopherol trafficking in hepatocytes are poorly understood. We found that the intracellular localization of TTP in hepatocytes is dynamic and responds to the presence of α-tocopherol. In the absence of the vitamin, TTP is localized to perinuclear vesicles that harbor CD71, transferrin, and Rab8, markers of the recycling endosomes. Upon treatment with α-tocopherol, TTP- and α-tocopherol-containing vesicles translocate to the plasma membrane, prior to secretion of the vitamin to the exterior of the cells. The change in TTP localization is specific to α-tocopherol and is time- and dose-dependent. The aberrant intracellular localization patterns of lipid binding-defective TTP mutants highlight the importance of protein-lipid interaction in the transport of α-tocopherol. These findings provide the basis for a proposed mechanistic model that describes TTP-facilitated trafficking of α-tocopherol through hepatocytes.
Collapse
Affiliation(s)
| | - Mikel Ghelfi
- the Department of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, Canada, and
| | - Jeffrey Atkinson
- the Department of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, Canada, and
| | - Robert Parker
- the Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853
| | - Jinghui Qian
- the Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853
| | - Cathleen Carlin
- Microbiology and Molecular Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Danny Manor
- From the Departments of Nutrition, Pharmacology, and
| |
Collapse
|
73
|
Nakajo A, Yoshimura SI, Togawa H, Kunii M, Iwano T, Izumi A, Noguchi Y, Watanabe A, Goto A, Sato T, Harada A. EHBP1L1 coordinates Rab8 and Bin1 to regulate apical-directed transport in polarized epithelial cells. J Cell Biol 2016; 212:297-306. [PMID: 26833786 PMCID: PMC4739609 DOI: 10.1083/jcb.201508086] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The highly conserved Rab guanosine triphosphatase (GTPase) Rab8 plays a role in exocytosis toward the polarized plasma membrane in eukaryotic cells. In murine Rab8-deficient small intestine cells, apical proteins are missorted into lysosomes. In this study, we identified a novel Rab8-interacting protein complex containing an EH domain-binding protein 1-like 1 (EHBP1L1), Bin1/amphiphysin II, and dynamin. Biochemical analyses showed that EHBP1L1 directly bound to GTP-loaded Rab8 and Bin1. The spatial dependency of these complexes at the endocytic recycling compartment (ERC) was demonstrated through overexpression and knockdown experiments. EHBP1L1- or Bin1-depleted or dynamin-inhibited small intestine organoids significantly accumulated apical membrane proteins but not basolateral membrane proteins in lysosomes. Furthermore, in EHBP1L1-deficient mice, small intestine cells displayed truncated and sparse microvilli, suggesting that EHBP1L1 maintains the apical plasma membrane by regulating apical transport. In summary, our data demonstrate that EHBP1L1 links Rab8 and the Bin1-dynamin complex, which generates membrane curvature and excises the vesicle at the ERC for apical transport.
Collapse
Affiliation(s)
- Atsuhiro Nakajo
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Shin-ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Hiroko Togawa
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Masataka Kunii
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Tomohiko Iwano
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Ayaka Izumi
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yuria Noguchi
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Ayako Watanabe
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Ayako Goto
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
74
|
Solanko KA, Modzel M, Solanko LM, Wüstner D. Fluorescent Sterols and Cholesteryl Esters as Probes for Intracellular Cholesterol Transport. Lipid Insights 2016; 8:95-114. [PMID: 27330304 PMCID: PMC4902042 DOI: 10.4137/lpi.s31617] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 12/20/2022] Open
Abstract
Cholesterol transport between cellular organelles comprised vesicular trafficking and nonvesicular exchange; these processes are often studied by quantitative fluorescence microscopy. A major challenge for using this approach is producing analogs of cholesterol with suitable brightness and structural and chemical properties comparable with those of cholesterol. This review surveys currently used fluorescent sterols with respect to their behavior in model membranes, their photophysical properties, as well as their transport and metabolism in cells. In the first part, several intrinsically fluorescent sterols, such as dehydroergosterol or cholestatrienol, are discussed. These polyene sterols (P-sterols) contain three conjugated double bonds in the steroid ring system, giving them slight fluorescence in ultraviolet light. We discuss the properties of P-sterols relative to cholesterol, outline their chemical synthesis, and explain how to image them in living cells and organisms. In particular, we show that P-sterol esters inserted into low-density lipoprotein can be tracked in the fibroblasts of Niemann–Pick disease using high-resolution deconvolution microscopy. We also describe fluorophore-tagged cholesterol probes, such as BODIPY-, NBD-, Dansyl-, or Pyrene-tagged cholesterol, and eventual esters of these analogs. Finally, we survey the latest developments in the synthesis and use of alkyne cholesterol analogs to be labeled with fluorophores by click chemistry and discuss the potential of all approaches for future applications.
Collapse
Affiliation(s)
- Katarzyna A Solanko
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Maciej Modzel
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Lukasz M Solanko
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| |
Collapse
|
75
|
Osati S, Ali H, van Lier JE. BODIPY–steroid conjugates: Syntheses and biological applications. J PORPHYR PHTHALOCYA 2016. [DOI: 10.1142/s1088424616300019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BODIPY and aza-BODIPY fluorophores linked to steroids are being developed as multimodal-imaging agents to monitor the mechanism of action of biologically active components in living systems.
Collapse
Affiliation(s)
- Samira Osati
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke QC J1H 5N4, Canada
| | - Hasrat Ali
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke QC J1H 5N4, Canada
| | - Johan E. van Lier
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke QC J1H 5N4, Canada
| |
Collapse
|
76
|
Abstract
PURPOSE OF REVIEW In this article, we summarize the present information related to the export of LDL-derived cholesterol from late endosomes, with a focus on Nieman-Pick disease, type C1 (NPC1) cholesterol delivery toward the endoplasmic reticulum (ER). We review data suggesting that several pathways may operate in parallel, including membrane transport routes and membrane contact sites (MCSs). RECENT FINDINGS There is increasing appreciation that MCSs provide an important mechanism for intermembrane lipid transfer. In late endosome-ER contacts, three protein bridges involving oxysterol binding protein related protein (ORP)1L-vesicle associated membrane protein-associated protein (VAP), steroidogenic acute regulatory protein (StAR)D3-VAP and ORP5-NPC1 proteins have been reported. How much they contribute to the flux of LDL-cholesterol to the ER is currently open. Studies for lipid transfer via MCSs have been most advanced in Saccharomyces cerevisiae. Recently, a new sterol-binding protein family conserved between yeast and man was identified. Its members localize at MCSs and were named lipid transfer protein anchored at membrane contact sites (Lam) proteins. In yeast, sterol transfer between the ER and the yeast lysosome may be facilitated by a Lam protein. SUMMARY Increasing insights into the role of MCSs in directional sterol delivery between membranes propose that they might provide routes for LDL-cholesterol transfer to the ER. Future work should reveal which specific contacts may operate for this, and how they are controlled by cholesterol homeostatic machineries.
Collapse
Affiliation(s)
- Simon G Pfisterer
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Finland and Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | | | | |
Collapse
|
77
|
Kishimoto T, Ishitsuka R, Kobayashi T. Detectors for evaluating the cellular landscape of sphingomyelin- and cholesterol-rich membrane domains. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:812-829. [PMID: 26993577 DOI: 10.1016/j.bbalip.2016.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/09/2016] [Accepted: 03/09/2016] [Indexed: 12/11/2022]
Abstract
Although sphingomyelin and cholesterol are major lipids of mammalian cells, the detailed distribution of these lipids in cellular membranes remains still obscure. However, the recent development of protein probes that specifically bind sphingomyelin and/or cholesterol provides new information about the landscape of the lipid domains that are enriched with sphingomyelin or cholesterol or both. Here, we critically summarize the tools to study distribution and dynamics of sphingomyelin and cholesterol. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
Affiliation(s)
| | - Reiko Ishitsuka
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan; INSERM U1060, Université Lyon 1, Villeurbanne 69621, France.
| |
Collapse
|
78
|
The counterflow transport of sterols and PI4P. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:940-951. [PMID: 26928592 DOI: 10.1016/j.bbalip.2016.02.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 02/23/2016] [Accepted: 02/24/2016] [Indexed: 02/03/2023]
Abstract
Cholesterol levels in intracellular membranes are constantly adjusted to match with specific organelle functions. Cholesterol is kept high in the plasma membrane (PM) because it is essential for its barrier function, while low levels are found in the endoplasmic reticulum (ER) where cholesterol mediates feedback control of its own synthesis by sterol-sensor proteins. The ER→Golgi→PM concentration gradient of cholesterol in mammalian cells, and ergosterol in yeast, appears to be sustained by specific intracellular transport processes, which are mostly mediated by lipid transfer proteins (LTPs). Here we review a recently described function of two LTPs, OSBP and its yeast homolog Osh4p, which consists in creating a sterol gradient between membranes by vectorial transport. OSBP also contributes to the formation of ER/Golgi membrane contact sites, which are important hubs for the transfer of several lipid species. OSBP and Osh4p organize a counterflow transport of lipids whereby sterols are exchanged for the phosphoinositide PI4P, which is used as a fuel to drive sterol transport. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
|
79
|
Lange Y, Steck TL. Active membrane cholesterol as a physiological effector. Chem Phys Lipids 2016; 199:74-93. [PMID: 26874289 DOI: 10.1016/j.chemphyslip.2016.02.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 02/05/2023]
Abstract
Sterols associate preferentially with plasma membrane sphingolipids and saturated phospholipids to form stoichiometric complexes. Cholesterol in molar excess of the capacity of these polar bilayer lipids has a high accessibility and fugacity; we call this fraction active cholesterol. This review first considers how active cholesterol serves as an upstream regulator of cellular sterol homeostasis. The mechanism appears to utilize the redistribution of active cholesterol down its diffusional gradient to the endoplasmic reticulum and mitochondria, where it binds multiple effectors and directs their feedback activity. We have also reviewed a broad literature in search of a role for active cholesterol (as opposed to bulk cholesterol or lipid domains such as rafts) in the activity of diverse membrane proteins. Several systems provide such evidence, implicating, in particular, caveolin-1, various kinds of ABC-type cholesterol transporters, solute transporters, receptors and ion channels. We suggest that this larger role for active cholesterol warrants close attention and can be tested easily.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, 1653 W. Congress Parkway, Chicago, IL 60612, USA.
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
80
|
Wüstner D, Lund FW, Röhrl C, Stangl H. Potential of BODIPY-cholesterol for analysis of cholesterol transport and diffusion in living cells. Chem Phys Lipids 2016; 194:12-28. [DOI: 10.1016/j.chemphyslip.2015.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/07/2015] [Accepted: 08/12/2015] [Indexed: 01/04/2023]
|
81
|
Bravo-Cordero JJ, Cordani M, Soriano SF, Díez B, Muñoz-Agudo C, Casanova-Acebes M, Boullosa C, Guadamillas MC, Ezkurdia I, González-Pisano D, del Pozo MA, Montoya MC. A novel high content analysis tool reveals Rab8-driven actin and FA reorganization through Rho GTPases and calpain/MT1. J Cell Sci 2016; 129:1734-49. [DOI: 10.1242/jcs.174920] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 02/29/2016] [Indexed: 01/31/2023] Open
Abstract
Rab8 is a small Ras-related GTPase that regulates polarized membrane transport to the plasma membrane. A high content analysis (HCA) tool developed to dissect Rab8-mediated actin and focal adhesion (FA) reorganization revealed that Rab8 activation significantly induced Rac1/Tiam1 to mediate cortical actin (CA) formation and RhoA-dependent stress fibre (SF) disassembly. Rab8 activation increased Rac1 activity, while its depletion activated RhoA, which led to reorganization of the actin cytoskeleton. Rab8 was also associated with FA, promoting their disassembly in a microtubule dependent manner. This Rab8 effect involved Calpain, MT1-MMP and Rho GTPases. Moreover, we demonstrate the role of Rab8 in the cell migration process. Indeed, Rab8 is required for EGF-induced cell polarization and chemotaxis as well as for the directional persistency of intrinsic cell motility. These data reveal that Rab8 drives cell motility by mechanisms both dependent and independent of Rho GTPases, thereby regulating the establishment of cell polarity, turnover of FA, and actin cytoskeleton rearrangements, thus determining the directionality of cell migration.
Collapse
Affiliation(s)
- José J. Bravo-Cordero
- Current Address: Division of Hematology and Oncology, Department of Medicine, Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY, Box 1079, USA
| | - Marco Cordani
- Integrin Signaling Laboratory, Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid, E28029, Spain
| | - Silvia F. Soriano
- Integrin Signaling Laboratory, Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid, E28029, Spain
| | - Begoña Díez
- Cellomics Unit. Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares CNIC. C/ Melchor Fernandez Almagro, 3. Madrid, E28029, Spain
| | - Carmen Muñoz-Agudo
- Cellomics Unit. Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares CNIC. C/ Melchor Fernandez Almagro, 3. Madrid, E28029, Spain
| | - María Casanova-Acebes
- Cellomics Unit. Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares CNIC. C/ Melchor Fernandez Almagro, 3. Madrid, E28029, Spain
| | - César Boullosa
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro 3, Madrid E28029, Spain
| | - Marta C. Guadamillas
- Integrin Signaling Laboratory, Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid, E28029, Spain
| | - Iakes Ezkurdia
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro 3, Madrid E28029, Spain
| | - David González-Pisano
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro 3, Madrid E28029, Spain
| | - Miguel A. del Pozo
- Integrin Signaling Laboratory, Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid, E28029, Spain
| | - María C. Montoya
- Cellomics Unit. Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares CNIC. C/ Melchor Fernandez Almagro, 3. Madrid, E28029, Spain
| |
Collapse
|
82
|
A reflection of the lasting contributions from Dr. Robert Bittman to sterol trafficking, sphingolipid and phospholipid research. Prog Lipid Res 2015; 61:19-29. [PMID: 26584871 DOI: 10.1016/j.plipres.2015.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/21/2015] [Accepted: 10/28/2015] [Indexed: 10/22/2022]
Abstract
With the passing of Dr. Robert Bittman from pancreatic cancer on the 1st October 2014, the lipid research field lost one of the most influential and significant personalities. Robert Bittman's genius was in chemical design and his contribution to the lipid research field was truly immense. The reagents and chemicals he designed and synthesised allowed interrogation of the role of lipids in constituting complex biophysical membranes, sterol transfer and in cellular communication networks. Here we provide a review of these works which serve as a lasting memory to his life.
Collapse
|
83
|
García-Melero A, Reverter M, Hoque M, Meneses-Salas E, Koese M, Conway JRW, Johnsen CH, Alvarez-Guaita A, Morales-Paytuvi F, Elmaghrabi YA, Pol A, Tebar F, Murray RZ, Timpson P, Enrich C, Grewal T, Rentero C. Annexin A6 and Late Endosomal Cholesterol Modulate Integrin Recycling and Cell Migration. J Biol Chem 2015; 291:1320-35. [PMID: 26578516 DOI: 10.1074/jbc.m115.683557] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Indexed: 01/01/2023] Open
Abstract
Annexins are a family of proteins that bind to phospholipids in a calcium-dependent manner. Earlier studies implicated annexin A6 (AnxA6) to inhibit secretion and participate in the organization of the extracellular matrix. We recently showed that elevated AnxA6 levels significantly reduced secretion of the extracellular matrix protein fibronectin (FN). Because FN is directly linked to the ability of cells to migrate, this prompted us to investigate the role of AnxA6 in cell migration. Up-regulation of AnxA6 in several cell models was associated with reduced cell migration in wound healing, individual cell tracking and three-dimensional migration/invasion assays. The reduced ability of AnxA6-expressing cells to migrate was associated with decreased cell surface expression of αVβ3 and α5β1 integrins, both FN receptors. Mechanistically, we found that elevated AnxA6 levels interfered with syntaxin-6 (Stx6)-dependent recycling of integrins to the cell surface. AnxA6 overexpression caused mislocalization and accumulation of Stx6 and integrins in recycling endosomes, whereas siRNA-mediated AnxA6 knockdown did not modify the trafficking of integrins. Given our recent findings that inhibition of cholesterol export from late endosomes (LEs) inhibits Stx6-dependent integrin recycling and that elevated AnxA6 levels cause LE cholesterol accumulation, we propose that AnxA6 and blockage of LE cholesterol transport are critical for endosomal function required for Stx6-mediated recycling of integrins in cell migration.
Collapse
Affiliation(s)
- Ana García-Melero
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Meritxell Reverter
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Monira Hoque
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Elsa Meneses-Salas
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Meryem Koese
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales 2006, Australia
| | - James R W Conway
- Garvan Institute of Medical Research and Kinghorn Cancer Centre, Cancer Research Program, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Camilla H Johnsen
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Anna Alvarez-Guaita
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Frederic Morales-Paytuvi
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Yasmin A Elmaghrabi
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Albert Pol
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain, and
| | - Francesc Tebar
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain, and
| | - Rachael Z Murray
- Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4095, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research and Kinghorn Cancer Centre, Cancer Research Program, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Carlos Enrich
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain, and
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales 2006, Australia,
| | - Carles Rentero
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain, and
| |
Collapse
|
84
|
Hoque M, Rentero C, Conway JR, Murray RZ, Timpson P, Enrich C, Grewal T. The cross-talk of LDL-cholesterol with cell motility: insights from the Niemann Pick Type C1 mutation and altered integrin trafficking. Cell Adh Migr 2015; 9:384-91. [PMID: 26366834 DOI: 10.1080/19336918.2015.1019996] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cholesterol is considered indispensible for the recruitment and functioning of integrins in focal adhesions for cell migration. However, the physiological cholesterol pools that control integrin trafficking and focal adhesion assembly remain unclear. Using Niemann Pick Type C1 (NPC) mutant cells, which accumulate Low Density lipoprotein (LDL)-derived cholesterol in late endosomes (LE), several recent studies indicate that LDL-cholesterol has multiple roles in regulating focal adhesion dynamics. Firstly, targeting of endocytosed LDL-cholesterol from LE to focal adhesions controls their formation at the leading edge of migrating cells. Other newly emerging literature suggests that this may be coupled to vesicular transport of integrins, Src kinase and metalloproteases from the LE compartment to focal adhesions. Secondly, our recent work identified LDL-cholesterol as a key factor that determines the distribution and ability of several Soluble NSF Attachment Protein (SNAP) Receptor (SNARE) proteins, key players in vesicle transport, to control integrin trafficking to the cell surface and extracellular matrix (ECM) secretion. Collectively, dietary, genetic and pathological changes in cholesterol metabolism may link with efficiency and speed of integrin and ECM cell surface delivery in metastatic cancer cells. This commentary will summarize how direct and indirect pathways enable LDL-cholesterol to modulate cell motility.
Collapse
Affiliation(s)
- Monira Hoque
- a Faculty of Pharmacy; University of Sydney ; Sydney , Australia
| | - Carles Rentero
- b Departament de Biologia Cellular ; Immunologia i Neurociències; Centre de Recerca Biomèdica CELLEX; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Facultat de Medicina; Universitat de Barcelona ; Barcelona , Spain
| | - James R Conway
- c Cancer Research Program; The Kinghorn Cancer Center; Garvan Institute of Medical Research ; Darlinghurst , Australia
| | - Rachael Z Murray
- d Tissue Repair and Regeneration Program; Institute of Health and Biomedical Innovation; Queensland University of Technology ; Brisbane , Australia
| | - Paul Timpson
- c Cancer Research Program; The Kinghorn Cancer Center; Garvan Institute of Medical Research ; Darlinghurst , Australia
| | - Carlos Enrich
- b Departament de Biologia Cellular ; Immunologia i Neurociències; Centre de Recerca Biomèdica CELLEX; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Facultat de Medicina; Universitat de Barcelona ; Barcelona , Spain
| | - Thomas Grewal
- a Faculty of Pharmacy; University of Sydney ; Sydney , Australia
| |
Collapse
|
85
|
Ikonen E, Blom T. Lipoprotein-mediated delivery of BODIPY-labeled sterol and sphingolipid analogs reveals lipid transport mechanisms in mammalian cells. Chem Phys Lipids 2015; 194:29-36. [PMID: 26343174 DOI: 10.1016/j.chemphyslip.2015.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 01/29/2023]
Abstract
Lipids are often introduced into cell membranes directly from solvent or from lipophilic artificial carriers, such as cyclodextrins. A physiological lipid entry route into mammalian cells is via lipoprotein mediated uptake. In this review, we discuss the introduction of BODIPY-labeled sterol and sphingolipid analogs into mammalian cells via high- or low-density lipoproteins, and the novel findings made by using this strategy. Lipoprotein mediated delivery favors endocytic uptake and initial incorporation of the lipid into membranes of the endosomal compartments. This routing can therefore highlight physiological mechanisms of lipid entry into and exit from the endo-lysosomal membrane system. The underlying principles are of key importance for instance in controlling plasma cholesterol levels and in the development and regression of lysosomal lipid storage diseases. A common denominator for the BODIPY-labeled lipid analogs discussed in this review is that they were synthesized by late Robert Bittman, whose scientific impact radiates far beyond his lifework in organic chemistry.
Collapse
Affiliation(s)
- Elina Ikonen
- Dept of Anatomy, Faculty of Medicine, University of Helsinki and Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Tomas Blom
- Dept of Anatomy, Faculty of Medicine, University of Helsinki and Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| |
Collapse
|
86
|
Wüstner D, Solanko K. How cholesterol interacts with proteins and lipids during its intracellular transport. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1908-26. [DOI: 10.1016/j.bbamem.2015.05.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 04/14/2015] [Accepted: 05/13/2015] [Indexed: 12/13/2022]
|
87
|
Du X, Brown AJ, Yang H. Novel mechanisms of intracellular cholesterol transport: oxysterol-binding proteins and membrane contact sites. Curr Opin Cell Biol 2015; 35:37-42. [DOI: 10.1016/j.ceb.2015.04.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/12/2015] [Accepted: 04/13/2015] [Indexed: 01/08/2023]
|
88
|
Vassilev B, Sihto H, Li S, Hölttä-Vuori M, Ilola J, Lundin J, Isola J, Kellokumpu-Lehtinen PL, Joensuu H, Ikonen E. Elevated levels of StAR-related lipid transfer protein 3 alter cholesterol balance and adhesiveness of breast cancer cells: potential mechanisms contributing to progression of HER2-positive breast cancers. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:987-1000. [PMID: 25681734 DOI: 10.1016/j.ajpath.2014.12.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 11/07/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
Abstract
The STARD3 gene belongs to the minimal amplicon in HER2-positive breast cancers and encodes a cholesterol-binding membrane protein. To study how elevated StAR-related lipid transfer protein 3 (StARD3) expression affects breast cancer cells, we generated MCF-7 cells stably overexpressing StARD3-green fluorescent protein. We found that StARD3-overexpressing cells exhibited nonadherent morphological features, had increased Src levels, and had altered cholesterol balance, as evidenced by elevated mRNA levels of the cholesterol biosynthesis rate-limiting enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase, and increased plasma membrane cholesterol content. On removal of serum and insulin from the culture medium, the morphological characteristics of the StARD3-overexpressing cells changed, the cells became adherent, and they developed enlarged focal adhesions. Under these conditions, the StARD3-overexpressing cells maintained elevated Src and plasma membrane cholesterol content and showed increased phosphorylation of focal adhesion kinase. In two Finnish nationwide patient cohorts, approximately 10% (212/2220) breast cancers exhibited high StARD3 protein levels, which was strongly associated with HER2 amplification; several factors related to poor disease outcome and poor breast cancer-specific survival. In addition, high StARD3 levels in breast cancers were associated with elevated 3-hydroxy-3-methylglutaryl-coenzyme A reductase mRNA levels and anti-Src-Tyr416 immunoreactivity. These results provide evidence that StARD3 overexpression results in increased cholesterol biosynthesis and Src kinase activity in breast cancer cells and suggest that elevated StARD3 expression may contribute to breast cancer aggressiveness by increasing membrane cholesterol and enhancing oncogenic signaling.
Collapse
Affiliation(s)
- Boris Vassilev
- Faculty of Medicine, Department of Anatomy, University of Helsinki, Helsinki, Finland
| | - Harri Sihto
- Laboratory of Molecular Oncology, Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Shiqian Li
- Faculty of Medicine, Department of Anatomy, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Maarit Hölttä-Vuori
- Faculty of Medicine, Department of Anatomy, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Jaakko Ilola
- Faculty of Medicine, Department of Anatomy, University of Helsinki, Helsinki, Finland
| | - Johan Lundin
- Institute for Molecular Medicine Finland, University of Helsinki, Biomedicum Helsinki 2U, Helsinki, Finland
| | - Jorma Isola
- Institute of Medical Technology, University of Tampere and Tampere University Central Hospital, Tampere, Finland
| | | | - Heikki Joensuu
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
| | - Elina Ikonen
- Faculty of Medicine, Department of Anatomy, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland.
| |
Collapse
|
89
|
Abstract
Integrins are a family of transmembrane cell surface molecules that constitute the principal adhesion receptors for the extracellular matrix (ECM) and are indispensable for the existence of multicellular organisms. In vertebrates, 24 different integrin heterodimers exist with differing substrate specificity and tissue expression. Integrin–extracellular-ligand interaction provides a physical anchor for the cell and triggers a vast array of intracellular signalling events that determine cell fate. Dynamic remodelling of adhesions, through rapid endocytic and exocytic trafficking of integrin receptors, is an important mechanism employed by cells to regulate integrin–ECM interactions, and thus cellular signalling, during processes such as cell migration, invasion and cytokinesis. The initial concept of integrin traffic as a means to translocate adhesion receptors within the cell has now been expanded with the growing appreciation that traffic is intimately linked to the cell signalling apparatus. Furthermore, endosomal pathways are emerging as crucial regulators of integrin stability and expression in cells. Thus, integrin traffic is relevant in a number of pathological conditions, especially in cancer. Nearly a decade ago we wrote a Commentary in Journal of Cell Science entitled ‘Integrin traffic’. With the advances in the field, we felt it would be appropriate to provide the growing number of researchers interested in integrin traffic with an update.
Collapse
Affiliation(s)
| | - Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku, Turku 20521, Finland
| | - Jonna Alanko
- Turku Centre for Biotechnology, University of Turku, Turku 20521, Finland
| | - Pranshu Sahgal
- Turku Centre for Biotechnology, University of Turku, Turku 20521, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku, Turku 20521, Finland
| |
Collapse
|
90
|
Enrich C, Rentero C, Hierro A, Grewal T. Role of cholesterol in SNARE-mediated trafficking on intracellular membranes. J Cell Sci 2015; 128:1071-81. [PMID: 25653390 DOI: 10.1242/jcs.164459] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The cell surface delivery of extracellular matrix (ECM) and integrins is fundamental for cell migration in wound healing and during cancer cell metastasis. This process is not only driven by several soluble NSF attachment protein (SNAP) receptor (SNARE) proteins, which are key players in vesicle transport at the cell surface and intracellular compartments, but is also tightly modulated by cholesterol. Cholesterol-sensitive SNAREs at the cell surface are relatively well characterized, but it is less well understood how altered cholesterol levels in intracellular compartments impact on SNARE localization and function. Recent insights from structural biology, protein chemistry and cell microscopy have suggested that a subset of the SNAREs engaged in exocytic and retrograde pathways dynamically 'sense' cholesterol levels in the Golgi and endosomal membranes. Hence, the transport routes that modulate cellular cholesterol distribution appear to trigger not only a change in the location and functioning of SNAREs at the cell surface but also in endomembranes. In this Commentary, we will discuss how disrupted cholesterol transport through the Golgi and endosomal compartments ultimately controls SNARE-mediated delivery of ECM and integrins to the cell surface and, consequently, cell migration.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biologia Cellular, Immunologia i Neurociències, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Facultat de Medicina, Universitat de Barcelona, 08036-Barcelona, Spain
| | - Carles Rentero
- Departament de Biologia Cellular, Immunologia i Neurociències, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Facultat de Medicina, Universitat de Barcelona, 08036-Barcelona, Spain
| | - Aitor Hierro
- Structural Biology Unit, CIC bioGUNE, Bizkaia Technology Park, 48160 Derio; IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
91
|
Wu L, Xu D, Zhou L, Xie B, Yu L, Yang H, Huang L, Ye J, Deng H, Yuan YA, Chen S, Li P. Rab8a-AS160-MSS4 regulatory circuit controls lipid droplet fusion and growth. Dev Cell 2014; 30:378-93. [PMID: 25158853 DOI: 10.1016/j.devcel.2014.07.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/06/2014] [Accepted: 07/10/2014] [Indexed: 11/15/2022]
Abstract
Rab GTPases, by targeting to specific membrane compartments, play essential roles in membrane trafficking. Lipid droplets (LDs) are dynamic subcellular organelles whose growth is closely linked to obesity and hepatic steatosis. Fsp27 is shown to be required for LD fusion and growth by enriching at LD-LD contact sites. Here, we identify Rab8a as a direct interactor and regulator of Fsp27 in mediating LD fusion in adipocytes. Knockdown of Rab8a in the livers of ob/ob mice results in the accumulation of smaller LDs and lower hepatic lipid levels. Surprisingly, it is the GDP-bound form of Rab8a that exhibits fusion-promoting activity. We further discover AS160 as the GTPase activating protein (GAP) for Rab8a, which forms a ternary complex with Fsp27 and Rab8a to positively regulate LD fusion. MSS4 antagonizes Fsp27-mediated LD fusion activity through Rab8a. Our results have thus revealed a mechanistic signaling circuit controlling LD fusion and fatty liver formation.
Collapse
Affiliation(s)
- Lizhen Wu
- MOE Key Laboratory of Bioinformatics and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Dijin Xu
- MOE Key Laboratory of Bioinformatics and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Linkang Zhou
- MOE Key Laboratory of Bioinformatics and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bingxian Xie
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Li Yu
- MOE Key Laboratory of Bioinformatics and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney NSW 2052, Australia
| | - Lei Huang
- Cell Biology Core Facility and Proteomics Facility, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jing Ye
- Department of Pathology, The Fourth Military Medical University, Xi'an 710032, China
| | - Haiteng Deng
- Cell Biology Core Facility and Proteomics Facility, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Y Adam Yuan
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Peng Li
- MOE Key Laboratory of Bioinformatics and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
92
|
Neuvonen M, Manna M, Mokkila S, Javanainen M, Rog T, Liu Z, Bittman R, Vattulainen I, Ikonen E. Enzymatic oxidation of cholesterol: properties and functional effects of cholestenone in cell membranes. PLoS One 2014; 9:e103743. [PMID: 25157633 PMCID: PMC4144813 DOI: 10.1371/journal.pone.0103743] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/01/2014] [Indexed: 11/19/2022] Open
Abstract
Bacterial cholesterol oxidase is commonly used as an experimental tool to reduce cellular cholesterol content. That the treatment also generates the poorly degradable metabolite 4-cholesten-3-one (cholestenone) has received less attention. Here, we investigated the membrane partitioning of cholestenone using simulations and cell biological experiments and assessed the functional effects of cholestenone in human cells. Atomistic simulations predicted that cholestenone reduces membrane order, undergoes faster flip-flop and desorbs more readily from membranes than cholesterol. In primary human fibroblasts, cholestenone was released from membranes to physiological extracellular acceptors more avidly than cholesterol, but without acceptors it remained in cells over a day. To address the functional effects of cholestenone, we studied fibroblast migration during wound healing. When cells were either cholesterol oxidase treated or part of cellular cholesterol was exchanged for cholestenone with cyclodextrin, cell migration during 22 h was markedly inhibited. Instead, when a similar fraction of cholesterol was removed using cyclodextrin, cells replenished their cholesterol content in 3 h and migrated similarly to control cells. Thus, cholesterol oxidation produces long-term functional effects in cells and these are in part due to the generated membrane active cholestenone.
Collapse
Affiliation(s)
- Maarit Neuvonen
- Institute of Biomedicine, Anatomy, University of Helsinki, Helsinki, Finland
| | - Moutusi Manna
- Department of Physics, Tampere University of Technology, Tampere, Finland
| | - Sini Mokkila
- Department of Physics, Tampere University of Technology, Tampere, Finland
| | - Matti Javanainen
- Department of Physics, Tampere University of Technology, Tampere, Finland
| | - Tomasz Rog
- Department of Physics, Tampere University of Technology, Tampere, Finland
| | - Zheng Liu
- Department of Chemistry and Biochemistry, Queens College, The City University of New York, Flushing, NY, United States of America
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College, The City University of New York, Flushing, NY, United States of America
| | - Ilpo Vattulainen
- Department of Physics, Tampere University of Technology, Tampere, Finland
- MEMPHYS – Center of Biomembrane Physics, University of Southern Denmark, Odense, Denmark
| | - Elina Ikonen
- Institute of Biomedicine, Anatomy, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- * E-mail:
| |
Collapse
|
93
|
Reverter M, Rentero C, Garcia-Melero A, Hoque M, Vilà de Muga S, Alvarez-Guaita A, Conway JRW, Wood P, Cairns R, Lykopoulou L, Grinberg D, Vilageliu L, Bosch M, Heeren J, Blasi J, Timpson P, Pol A, Tebar F, Murray RZ, Grewal T, Enrich C. Cholesterol regulates Syntaxin 6 trafficking at trans-Golgi network endosomal boundaries. Cell Rep 2014; 7:883-97. [PMID: 24746815 DOI: 10.1016/j.celrep.2014.03.043] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 12/30/2013] [Accepted: 03/17/2014] [Indexed: 12/27/2022] Open
Abstract
Inhibition of cholesterol export from late endosomes causes cellular cholesterol imbalance, including cholesterol depletion in the trans-Golgi network (TGN). Here, using Chinese hamster ovary (CHO) Niemann-Pick type C1 (NPC1) mutant cell lines and human NPC1 mutant fibroblasts, we show that altered cholesterol levels at the TGN/endosome boundaries trigger Syntaxin 6 (Stx6) accumulation into VAMP3, transferrin, and Rab11-positive recycling endosomes (REs). This increases Stx6/VAMP3 interaction and interferes with the recycling of αVβ3 and α5β1 integrins and cell migration, possibly in a Stx6-dependent manner. In NPC1 mutant cells, restoration of cholesterol levels in the TGN, but not inhibition of VAMP3, restores the steady-state localization of Stx6 in the TGN. Furthermore, elevation of RE cholesterol is associated with increased amounts of Stx6 in RE. Hence, the fine-tuning of cholesterol levels at the TGN-RE boundaries together with a subset of cholesterol-sensitive SNARE proteins may play a regulatory role in cell migration and invasion.
Collapse
Affiliation(s)
- Meritxell Reverter
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Carles Rentero
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Ana Garcia-Melero
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Monira Hoque
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Sandra Vilà de Muga
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Anna Alvarez-Guaita
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - James R W Conway
- Garvan Institute of Medical Research and Kinghorn Cancer Centre, Cancer Research Program, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Peta Wood
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Rose Cairns
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Lilia Lykopoulou
- First Department of Pediatrics, University of Athens, Aghia Sofia Children's Hospital, 11527 Athens, Greece
| | - Daniel Grinberg
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, CIBERER, IBUB, 08028 Barcelona, Spain
| | - Lluïsa Vilageliu
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, CIBERER, IBUB, 08028 Barcelona, Spain
| | - Marta Bosch
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Joerg Heeren
- Department of Biochemistry and Molecular Biology II. Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Juan Blasi
- Department of Pathology and Experimental Therapeutics, IDIBELL-University of Barcelona, L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Paul Timpson
- Garvan Institute of Medical Research and Kinghorn Cancer Centre, Cancer Research Program, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Albert Pol
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain; Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avaçats (ICREA), 08010 Barcelona, Spain
| | - Francesc Tebar
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain; Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Rachael Z Murray
- Tissue Repair and Regeneration Program, Institute of Health and Biomedical, Innovation, Queensland University of Technology, Brisbane, QLD 4095, Australia
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia.
| | - Carlos Enrich
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain; Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
| |
Collapse
|