1
|
Zhao L, Chen J, Zhang Y, Liu J, Li W, Sun Y, Chen G, Guo Z, Gu L. Loss of DNA Polymerase β Delays Atherosclerosis in ApoE-/- Mice Due to Inhibition of Vascular Smooth Muscle Cell Migration. Int J Mol Sci 2024; 25:11778. [PMID: 39519329 PMCID: PMC11547094 DOI: 10.3390/ijms252111778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Atherosclerosis (AS) is an inflammatory disease characterized by arterial inflammation. One important trigger for AS development is the excessive migration of vascular smooth muscle cells (VSMCs); however, the mechanism underlying this phenomenon remains unclear. Therefore, we investigated the role of DNA polymerase β (Pol β), a crucial enzyme involved in base excision repair, VSMC migration, and subsequent AS development. In this study, we revealed a significant increase in Pol β content within AS plaques in ApoE-/-Pol β+/+ mice. In vitro experiments demonstrated a significant decrease in hCASMC viability and migration ability upon Pol β knockdown, whereas the subsequent recovery of Pol β expression reversed this effect. Moreover, our investigations revealed that Pol β knockdown leads to the inhibition of the POSTN gene transcription by suppressing the YY1/TGF-β1 pathway, resulting in the decreased expression of the protein periostin during VSMC migration. Collectively, our findings provide insights into the role of Pol β in AS development, offering a novel approach for the clinical treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lili Gu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China; (L.Z.); (J.C.); (Y.Z.); (J.L.); (W.L.); (Y.S.); (G.C.); (Z.G.)
| |
Collapse
|
2
|
Moore MP, Wang X, Kennelly JP, Shi H, Ishino Y, Kano K, Aoki J, Cherubini A, Ronzoni L, Guo X, Chalasani NP, Khalid S, Saleheen D, Mitsche MA, Rotter JI, Yates KP, Valenti L, Kono N, Tontonoz P, Tabas I. Low MBOAT7 expression, a genetic risk for MASH, promotes a profibrotic pathway involving hepatocyte TAZ upregulation. Hepatology 2024:01515467-990000000-00886. [PMID: 38776184 DOI: 10.1097/hep.0000000000000933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/03/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND AND AIMS The common genetic variant rs641738 C>T is a risk factor for metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis (MASH), including liver fibrosis, and is associated with decreased expression of the phospholipid-remodeling enzyme MBOAT7 (LPIAT1). However, whether restoring MBOAT7 expression in established metabolic dysfunction-associated steatotic liver disease dampens the progression to liver fibrosis and, importantly, the mechanism through which decreased MBOAT7 expression exacerbates MASH fibrosis remain unclear. APPROACH AND RESULTS We first showed that hepatocyte MBOAT7 restoration in mice with diet-induced steatohepatitis slows the progression to liver fibrosis. Conversely, when hepatocyte-MBOAT7 was silenced in mice with established hepatosteatosis, liver fibrosis but not hepatosteatosis was exacerbated. Mechanistic studies revealed that hepatocyte-MBOAT7 restoration in MASH mice lowered hepatocyte-TAZ (WWTR1), which is known to promote MASH fibrosis. Conversely, hepatocyte-MBOAT7 silencing enhanced TAZ upregulation in MASH. Finally, we discovered that changes in hepatocyte phospholipids due to MBOAT7 loss-of-function promote a cholesterol trafficking pathway that upregulates TAZ and the TAZ-induced profibrotic factor Indian hedgehog (IHH). As evidence for relevance in humans, we found that the livers of individuals with MASH carrying the rs641738-T allele had higher hepatocyte nuclear TAZ, indicating higher TAZ activity and increased IHH mRNA. CONCLUSIONS This study provides evidence for a novel mechanism linking MBOAT7-LoF to MASH fibrosis, adds new insight into an established genetic locus for MASH, and, given the druggability of hepatocyte TAZ for MASH fibrosis, suggests a personalized medicine approach for subjects at increased risk for MASH fibrosis due to inheritance of variants that lower MBOAT7.
Collapse
Affiliation(s)
- Mary P Moore
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - John Paul Kennelly
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Hongxue Shi
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Yuki Ishino
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Alessandro Cherubini
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Luisa Ronzoni
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Naga P Chalasani
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shareef Khalid
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Center for Non-Communicable Disease, Karachi, Karachi City, Sindh, Pakistan
| | - Danish Saleheen
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Center for Non-Communicable Disease, Karachi, Karachi City, Sindh, Pakistan
| | - Matthew A Mitsche
- Center for Human Nutrition and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Katherine P Yates
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Luca Valenti
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
| | - Nozomu Kono
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY
| |
Collapse
|
3
|
Gao Y, Kennelly JP, Xiao X, Whang E, Ferrari A, Bedard AH, Mack JJ, Nguyen AH, Weston T, Uchiyama LF, Lee MS, Young SG, Bensinger SJ, Tontonoz P. T cell cholesterol transport is a metabolic checkpoint that links intestinal immune responses to dietary lipid absorption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584164. [PMID: 38559079 PMCID: PMC10979874 DOI: 10.1101/2024.03.08.584164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The intrinsic pathways that control membrane organization in immune cells and the impact of such pathways on cellular function are not well defined. Here we report that the non-vesicular cholesterol transporter Aster-A links plasma membrane (PM) cholesterol availability in T cells to immune signaling and systemic metabolism. Aster-A is recruited to the PM during T-cell receptor (TCR) activation, where it facilitates the removal of newly generated "accessible" membrane cholesterol. Loss of Aster-A leads to excess PM cholesterol accumulation, resulting in enhanced TCR nano-clustering and signaling, and Th17 cytokine production. Finally, we show that the mucosal Th17 response is restrained by PM cholesterol remodeling. Ablation of Aster-A in T cells leads to enhanced IL-22 production, reduced intestinal fatty acid absorption, and resistance to diet-induced obesity. These findings delineate a multi-tiered regulatory scheme linking immune cell lipid flux to nutrient absorption and systemic physiology.
Collapse
|
4
|
Zhou J, Li C, Lu M, Jiang G, Chen S, Li H, Lu K. Pharmacological induction of autophagy reduces inflammation in macrophages by degrading immunoproteasome subunits. PLoS Biol 2024; 22:e3002537. [PMID: 38447109 PMCID: PMC10917451 DOI: 10.1371/journal.pbio.3002537] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Defective autophagy is linked to proinflammatory diseases. However, the mechanisms by which autophagy limits inflammation remain elusive. Here, we found that the pan-FGFR inhibitor LY2874455 efficiently activated autophagy and suppressed expression of proinflammatory factors in macrophages stimulated by lipopolysaccharide (LPS). Multiplex proteomic profiling identified the immunoproteasome, which is a specific isoform of the 20s constitutive proteasome, as a substrate that is degraded by selective autophagy. SQSTM1/p62 was found to be a selective autophagy-related receptor that mediated this degradation. Autophagy deficiency or p62 knockdown blocked the effects of LY2874455, leading to the accumulation of immunoproteasomes and increases in inflammatory reactions. Expression of proinflammatory factors in autophagy-deficient macrophages could be reversed by immunoproteasome inhibitors, confirming the pivotal role of immunoproteasome turnover in the autophagy-mediated suppression on the expression of proinflammatory factors. In mice, LY2874455 protected against LPS-induced acute lung injury and dextran sulfate sodium (DSS)-induced colitis and caused low levels of proinflammatory cytokines and immunoproteasomes. These findings suggested that selective autophagy of the immunoproteasome was a key regulator of signaling via the innate immune system.
Collapse
Affiliation(s)
- Jiao Zhou
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Chunxia Li
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Meng Lu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Gaoyue Jiang
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Shanze Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Huihui Li
- West China Second University Hospital, Sichuan University, Chengdu, China
| | - Kefeng Lu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| |
Collapse
|
5
|
Ru Y, Dong S, Liu J, Liu J, Eyden B. Structural characterization and origin of surface vesicles in monocytes: another membranous pathway from cytoplasm to cell surface. Ultrastruct Pathol 2024; 48:56-65. [PMID: 38037244 DOI: 10.1080/01913123.2023.2286972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
The monocytes in acute monocytic leukemia (AML-M5b) were analyzed by scanning and transmission electron microscopy (SEM and TEM) to understand more fully their structure and origin. By SEM, monocytes exhibited localized expansions of the surface, some of which appeared to bud off as surface vesicles (SVs). Filopodial processes and pseudopodia were also present. TEM demonstrated that the SVs were composed of a double-membrane at the pole away from the cell body, and a single membrane nearer to the cell body. In the peripheral cytoplasm, intracellular vesicles (IVs) had the appearance of vacuoles and were enclosed by single membranes. Most SVs were characterized by a notch as a rER edge and an expanded head. Filopodial processes had the same thickness of 40 nm as the SV walls, which suggested a close developmental relationship between the two. Pseudopodia between SVs were irregular in size. Rod-like rER cisternae were prominent in the peripheral cytoplasm and some showed a close physical juxtaposition as to suggest a transition from rER to IVs to SVs. Ultrastructural cytochemistry demonstrated activity of 5'-nucleotidase over rER, SVs, filopodial processes and pseudopodia, and a patchy reaction over other areas of plasma membrane. Overall, the results indicated that rER transforms into SVs, filopodial processes and pseudopodia, as a way of integrating cytoplasmic membranes into the plasma membrane.
Collapse
Affiliation(s)
- Yongxin Ru
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology&Blood Diseases Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuxu Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology&Blood Diseases Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jing Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology&Blood Diseases Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jinhua Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology&Blood Diseases Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Brian Eyden
- Department of Histopathology, Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
6
|
Wüstner D, Dupont Juhl A, Egebjerg JM, Werner S, McNally J, Schneider G. Kinetic modelling of sterol transport between plasma membrane and endo-lysosomes based on quantitative fluorescence and X-ray imaging data. Front Cell Dev Biol 2023; 11:1144936. [PMID: 38020900 PMCID: PMC10644255 DOI: 10.3389/fcell.2023.1144936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Niemann Pick type C1 and C2 (NPC1 and NPC2) are two sterol-binding proteins which, together, orchestrate cholesterol transport through late endosomes and lysosomes (LE/LYSs). NPC2 can facilitate sterol exchange between model membranes severalfold, but how this is connected to its function in cells is poorly understood. Using fluorescent analogs of cholesterol and quantitative fluorescence microscopy, we have recently measured the transport kinetics of sterol between plasma membrane (PM), recycling endosomes (REs) and LE/LYSs in control and NPC2 deficient fibroblasts. Here, we use kinetic modeling of this data to determine rate constants for sterol transport between intracellular compartments. Our model predicts that sterol is trapped in intraluminal vesicles (ILVs) of LE/LYSs in the absence of NPC2, causing delayed sterol export from LE/LYSs in NPC2 deficient fibroblasts. Using soft X-ray tomography, we confirm, that LE/LYSs of NPC2 deficient cells but not of control cells contain enlarged, carbon-rich intraluminal vesicular structures, supporting our model prediction of lipid accumulation in ILVs. By including sterol export via exocytosis of ILVs as exosomes and by release of vesicles-ectosomes-from the PM, we can reconcile measured sterol efflux kinetics and show that both pathways can be reciprocally regulated by the intraluminal sterol transfer activity of NPC2 inside LE/LYSs. Our results thereby connect the in vitro function of NPC2 as sterol transfer protein between membranes with its in vivo function.
Collapse
Affiliation(s)
- Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Alice Dupont Juhl
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Jacob Marcus Egebjerg
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Stephan Werner
- Department of X-Ray Microscopy, Helmholtz-Zentrum Berlin, Berlin, Germany
| | - James McNally
- Department of X-Ray Microscopy, Helmholtz-Zentrum Berlin, Berlin, Germany
| | - Gerd Schneider
- Department of X-Ray Microscopy, Helmholtz-Zentrum Berlin, Berlin, Germany
| |
Collapse
|
7
|
Jin J, Zhao Q, Wei Z, Chen K, Su Y, Hu X, Peng X. Glycolysis-cholesterol metabolic axis in immuno-oncology microenvironment: emerging role in immune cells and immunosuppressive signaling. Cell Biosci 2023; 13:189. [PMID: 37828561 PMCID: PMC10571292 DOI: 10.1186/s13578-023-01138-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Cell proliferation and function require nutrients, energy, and biosynthesis activity to duplicate repertoires for each daughter. It is therefore not surprising that tumor microenvironment (TME) metabolic reprogramming primarily orchestrates the interaction between tumor and immune cells. Tumor metabolic reprogramming affords bioenergetic, signaling intermediates, and biosynthesis requirements for both malignant and immune cells. Different immune cell subsets are recruited into the TME, and these manifestations have distinct effects on tumor progression and therapeutic outcomes, especially the mutual contribution of glycolysis and cholesterol metabolism. In particularly, glycolysis-cholesterol metabolic axis interconnection plays a critical role in the TME modulation, and their changes in tumor metabolism appear to be a double-edged sword in regulating various immune cell responses and immunotherapy efficacy. Hence, we discussed the signature manifestation of the glycolysis-cholesterol metabolic axis and its pivotal role in tumor immune regulation. We also highlight how hypothetical combinations of immunotherapy and glycolysis/cholesterol-related metabolic interventions unleash the potential of anti-tumor immunotherapies, as well as developing more effective personalized treatment strategies.
Collapse
Affiliation(s)
- Jing Jin
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Qijie Zhao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhigong Wei
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Keliang Chen
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yonglin Su
- Department of Rehabilitation, Cancer Center, West China Hospital, Sichuan University, Sichuan, People's Republic of China.
| | - Xiaolin Hu
- Department of Nursing, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
8
|
Melero A, Jiménez-Rojo N. Cracking the membrane lipid code. Curr Opin Cell Biol 2023; 83:102203. [PMID: 37437490 DOI: 10.1016/j.ceb.2023.102203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/01/2023] [Accepted: 06/16/2023] [Indexed: 07/14/2023]
Abstract
Why has nature acquired such a huge lipid repertoire? Although it would be theoretically possible to make a lipid bilayer fulfilling barrier functions with only one glycerophospholipid, there are diverse and numerous different lipid species. Lipids are heterogeneously distributed across the evolutionary tree with lipidomes evolving in parallel to organismal complexity. Moreover, lipids are different between organs and tissues and even within the same cell, different organelles have characteristic lipid signatures. At the molecular level, membranes are asymmetric and laterally heterogeneous. This lipid asymmetry at different scales indicates that these molecules may play very specific molecular functions in biology. Some of these roles have been recently uncovered: lipids have been shown to be essential in processes such as hypoxia and ferroptosis or in protein sorting and trafficking but many of them remain still unknown. In this review we will discuss the importance of understanding lipid diversity in biology across scales and we will share a toolbox with some of the emerging technologies that are helping us to uncover new lipid molecular functions in cell biology and, step by step, crack the membrane lipid code.
Collapse
Affiliation(s)
- Alejandro Melero
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Noemi Jiménez-Rojo
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain; Instituto Biofisika (UPV/EHU, CSIC), 48940, Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
9
|
Effects of Cancer Cell-Derived Nanovesicle Vaccines Produced by the Oxidative Stress-Induced Expression of DAMP and Spontaneous Release/Filter Extrusion in the Interplay of Cancer Cells and Macrophages. Biomedicines 2022; 10:biomedicines10081977. [PMID: 36009524 PMCID: PMC9405549 DOI: 10.3390/biomedicines10081977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/18/2022] Open
Abstract
Photodynamic therapy (PDT)-based cancer vaccines are shown to be more effective modalities for treating cancer in animal models compared to other methods used to generate cancer cell-derived vaccines. The higher efficacy seems to stem from the generation of cell membrane nanovesicles or fragments that carry both cancer cell-specific antigens and high surface content of damage-associated molecular pattern (DAMP) molecules induced by oxidative stress. To develop more effective cancer vaccines in this direction, we explored the generation of cancer vaccines by applying different sources of oxidative stress on cancer cell cultures followed by spontaneous release or filter extrusions to produce cancer cell-derived DAMP-expressing nanovesicles. Through an in-vitro test based on the co-culture of cancer cells and macrophages, it was found that the nanovesicle vaccines generated by H2O2 are as effective as those generated by PDT in diminishing cancer cell culture masses, providing a simpler way to manufacture vaccines. In addition, the nanovesicle vaccines produced by filter extrusion are as potent as those produced by spontaneous release, rendering a more stable way for vaccine production.
Collapse
|
10
|
Different Pathways of Cellular Cholesterol Efflux. Cell Biochem Biophys 2022; 80:471-481. [PMID: 35737216 DOI: 10.1007/s12013-022-01081-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 12/22/2022]
Abstract
Cholesterol efflux is the first and rate-limiting step of reverse cholesterol transport (RCT) from peripheric cells to the liver. The involvement of high-density lipoprotein (HDL) in RCT determines the atheroprotective properties of HDL. Cholesterol efflux from different membrane pools includes both passive and energy-dependent processes. The first type of route consists of cholesterol desorption from the cell membrane into the unstirred layer adjacent to the cell surface and diffusion in the water phase. Moreover, the selective uptake and facilitated diffusion of cholesterol and cholesteryl ester molecules through the hydrophobic tunnel in the scavenger receptor BI molecule does not require energy consumption. The second type of route includes active cholesterol export by the ATP-binding cassette transporters A1 (ABCA1) and G1 (ABCG1). Several cholesterol acceptors specifically bind cholesterol and phospholipid molecules, and cholesterol binding to the albumin molecule, which acts as a shuttle, significantly increases cholesterol movement between acceptors and red blood cells, thus functioning as a sink for cholesterol. Cholesterol and phospholipid molecules effluxed from macrophages by ABCA1 are accepted exclusively by the lipid-free apolipoprotein apoA-I, which is the major protein moiety of HDL, whereas those effluxed by ABCG1 are accepted by HDL. ABCA1- and ABCG1-mediated cholesterol transport, together with cholesterol diffusion, largely determine cholesterol turnover at the physiological level of intracellular cholesterol. However, at cholesterol overload, ABCA1-mediated efflux prevails over other routes. The exchange of apoA-I between lipid-free and lipid-associated states and the synergism of nascent and mature HDL contribute to cholesterol efflux efficiency. Moreover, extracellular cholesterol deposits and microvesicles may be involved in RCT.
Collapse
|
11
|
Xiang P, Blanchard V, Francis GA. Smooth Muscle Cell—Macrophage Interactions Leading to Foam Cell Formation in Atherosclerosis: Location, Location, Location. Front Physiol 2022; 13:921597. [PMID: 35795646 PMCID: PMC9251363 DOI: 10.3389/fphys.2022.921597] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Cholesterol-overloaded cells or “foam cells” in the artery wall are the biochemical hallmark of atherosclerosis, and are responsible for much of the growth, inflammation and susceptibility to rupture of atherosclerotic lesions. While it has previously been thought that macrophages are the main contributor to the foam cell population, recent evidence indicates arterial smooth muscle cells (SMCs) are the source of the majority of foam cells in both human and murine atherosclerosis. This review outlines the timeline, site of appearance and proximity of SMCs and macrophages with lipids in human and mouse atherosclerosis, and likely interactions between SMCs and macrophages that promote foam cell formation and removal by both cell types. An understanding of these SMC-macrophage interactions in foam cell formation and regression is expected to provide new therapeutic targets to reduce the burden of atherosclerosis for the prevention of coronary heart disease, stroke and peripheral vascular disease.
Collapse
|
12
|
Extracellular Vesicles as Drivers of Immunoinflammation in Atherothrombosis. Cells 2022; 11:cells11111845. [PMID: 35681540 PMCID: PMC9180657 DOI: 10.3390/cells11111845] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the leading cause of morbidity and mortality all over the world. Extracellular vesicles (EVs), small lipid-bilayer membrane vesicles released by most cellular types, exert pivotal and multifaceted roles in physiology and disease. Emerging evidence emphasizes the importance of EVs in intercellular communication processes with key effects on cell survival, endothelial homeostasis, inflammation, neoangiogenesis, and thrombosis. This review focuses on EVs as effective signaling molecules able to both derail vascular homeostasis and induce vascular dysfunction, inflammation, plaque progression, and thrombus formation as well as drive anti-inflammation, vascular repair, and atheroprotection. We provide a comprehensive and updated summary of the role of EVs in the development or regression of atherosclerotic lesions, highlighting the link between thrombosis and inflammation. Importantly, we also critically describe their potential clinical use as disease biomarkers or therapeutic agents in atherothrombosis.
Collapse
|
13
|
Björkegren JLM, Lusis AJ. Atherosclerosis: Recent developments. Cell 2022; 185:1630-1645. [PMID: 35504280 PMCID: PMC9119695 DOI: 10.1016/j.cell.2022.04.004] [Citation(s) in RCA: 417] [Impact Index Per Article: 139.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is an inflammatory disease of the large arteries that is the major cause of cardiovascular disease (CVD) and stroke. Here, we review the current understanding of the molecular, cellular, genetic, and environmental contributions to atherosclerosis, from both individual pathway and systems perspectives. We place an emphasis on recent developments, some of which have yielded unexpected biology, including previously unknown heterogeneity of inflammatory and smooth muscle cells in atherosclerotic lesions, roles for senescence and clonal hematopoiesis, and links to the gut microbiome.
Collapse
Affiliation(s)
- Johan L M Björkegren
- Department of Genetics and Genomic Sciences, Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics, A2-237 Center for the Health Sciences, University of California, Los Angeles, Los Angeles, CA USA.
| |
Collapse
|
14
|
Juhl AD, Wüstner D. Pathways and Mechanisms of Cellular Cholesterol Efflux-Insight From Imaging. Front Cell Dev Biol 2022; 10:834408. [PMID: 35300409 PMCID: PMC8920967 DOI: 10.3389/fcell.2022.834408] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
Cholesterol is an essential molecule in cellular membranes, but too much cholesterol can be toxic. Therefore, mammalian cells have developed complex mechanisms to remove excess cholesterol. In this review article, we discuss what is known about such efflux pathways including a discussion of reverse cholesterol transport and formation of high-density lipoprotein, the function of ABC transporters and other sterol efflux proteins, and we highlight their role in human diseases. Attention is paid to the biophysical principles governing efflux of sterols from cells. We also discuss recent evidence for cholesterol efflux by the release of exosomes, microvesicles, and migrasomes. The role of the endo-lysosomal network, lipophagy, and selected lysosomal transporters, such as Niemann Pick type C proteins in cholesterol export from cells is elucidated. Since oxysterols are important regulators of cellular cholesterol efflux, their formation, trafficking, and secretion are described briefly. In addition to discussing results obtained with traditional biochemical methods, focus is on studies that use established and novel bioimaging approaches to obtain insight into cholesterol efflux pathways, including fluorescence and electron microscopy, atomic force microscopy, X-ray tomography as well as mass spectrometry imaging.
Collapse
Affiliation(s)
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, PhyLife, Physical Life Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
15
|
Kinnebrew M, Johnson KA, Radhakrishnan A, Rohatgi R. Measuring and Manipulating Membrane Cholesterol for the Study of Hedgehog Signaling. Methods Mol Biol 2022; 2374:73-87. [PMID: 34562244 PMCID: PMC8819901 DOI: 10.1007/978-1-0716-1701-4_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cholesterol is an abundant lipid in mammalian plasma membranes that regulates the reception of the Hedgehog (Hh) signal in target cells. In vertebrates, cell-surface organelles called primary cilia function as compartments for the propagation of Hh signals. Recent structural, biochemical, and cell-biological studies have led to the model that Patched-1 (PTCH1), the receptor for Hh ligands, uses its transporter-like activity to lower cholesterol accessibility in the membrane surrounding primary cilia. Cholesterol restriction at cilia may represent the long-sought-after mechanism by which PTCH1 inhibits Smoothened (SMO), a cholesterol-responsive transmembrane protein of the G protein-coupled receptor superfamily that transmits the Hh signal across the membrane.Protein probes based on microbial cholesterol-binding proteins revealed that PTCH1 controls only a subset of the total cholesterol molecules, a biochemically defined fraction called accessible cholesterol. The accessible cholesterol pool coexists (and exchanges) with a pool of sequestered cholesterol, which is bound to phospholipids like sphingomyelin. In this chapter, we describe how to measure the accessible and sequestered cholesterol pools in live cells with protein-based probes. We discuss how to purify and fluorescently label these probes for use in flow cytometry and microscopy-based measurements of the cholesterol pools. Additionally, we describe how to modulate accessible cholesterol levels to determine if this pool regulates Hh signaling (or any other cellular process of interest).
Collapse
Affiliation(s)
- Maia Kinnebrew
- Department of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristen A Johnson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rajat Rohatgi
- Department of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
16
|
Li X, Ge J, Li Y, Cai Y, Zheng Q, Huang N, Gu Y, Han Q, Li Y, Sun R, Liu R. Integrative lipidomic and transcriptomic study unravels the therapeutic effects of saikosaponins A and D on non-alcoholic fatty liver disease. Acta Pharm Sin B 2021; 11:3527-3541. [PMID: 34900534 PMCID: PMC8642447 DOI: 10.1016/j.apsb.2021.03.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become one of the most prominent causes of chronic liver diseases and malignancies. However, few therapy has been approved. Radix Bupleuri (RB) is the most frequently used herbal medicine for the treatment of liver diseases. In the current study, we aim to systemically evaluate the therapeutic effects of saikosaponin A (SSa) and saikosaponin D (SSd), the major bioactive monomers in RB, against NAFLD and to investigate the underlying mechanisms. Our results demonstrated that both SSa and SSd improved diet-induced NAFLD. Integrative lipidomic and transcriptomic analysis revealed that SSa and SSd modulated glycerolipid metabolism by regulating related genes, like Lipe and Lipg. SSd profoundly suppressed the fatty acid biosynthesis by downregulating Fasn and Acaca expression and promoted fatty acid degradation by inducing Acox1 and Cpt1a expression. Bioinformatic analysis further predicted the implication of master transcription factors, including peroxisome proliferator-activated receptor alpha (PPARα), in the protective effects of SSa and SSd. These results were further confirmed in vitro in mouse primary hepatocytes. In summary, our study uncoded the complicated mechanisms underlying the promising anti-steatosis activities of saikosaponins (SSs), and provided critical evidence inspiring the discovery of innovative therapies based on SSa and SSd for the treatment of NAFLD and related complications.
Collapse
|
17
|
El-Kenawi A, Dominguez-Viqueira W, Liu M, Awasthi S, Abraham-Miranda J, Keske A, Steiner KK, Noel L, Serna AN, Dhillon J, Gillies RJ, Yu X, Koomen JM, Yamoah K, Gatenby RA, Ruffell B. Macrophage-derived cholesterol contributes to therapeutic resistance in prostate cancer. Cancer Res 2021; 81:5477-5490. [PMID: 34301759 DOI: 10.1158/0008-5472.can-20-4028] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/16/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022]
Abstract
Castration-resistant prostate cancer (CRPC) is a lethal stage of disease in which androgen receptor (AR) signaling is persistent despite androgen deprivation therapy (ADT). Most studies have focused on investigating cell-autonomous alterations in CRPC, while the contributions of the tumor microenvironment are less well understood. Here we sought to determine the role of tumor-associated macrophages in CRPC, based upon their role in cancer progression and therapeutic resistance. In a syngeneic model that reflected the mutational landscape of CRPC, macrophage depletion resulted in a reduced transcriptional signature for steroid and bile acid synthesis, indicating potential perturbation of cholesterol metabolism. As cholesterol is the precursor of the five major types of steroid hormones, we hypothesized that macrophages were regulating androgen biosynthesis within the prostate tumor microenvironment. Macrophage depletion reduced androgen levels within prostate tumors and restricted androgen receptor (AR) nuclear localization in vitro and in vivo. Macrophages were also cholesterol-rich and were able to transfer cholesterol to tumor cells in vitro. AR nuclear translocation was inhibited by activation of Liver X Receptor (LXR)-β, the master regulator of cholesterol homeostasis. Consistent with these data, macrophage depletion extended survival during ADT and the presence of macrophages correlated with therapeutic resistance in patient-derived explants. Taken together, these findings support the therapeutic targeting of macrophages in CRPC.
Collapse
Affiliation(s)
- Asmaa El-Kenawi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Min Liu
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Shivanshu Awasthi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Julieta Abraham-Miranda
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aysenur Keske
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - KayLee K Steiner
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Leenil Noel
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amparo N Serna
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jasreman Dhillon
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kosj Yamoah
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Robert A Gatenby
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
18
|
Ikonen E, Zhou X. Cholesterol transport between cellular membranes: A balancing act between interconnected lipid fluxes. Dev Cell 2021; 56:1430-1436. [PMID: 34004151 DOI: 10.1016/j.devcel.2021.04.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022]
Abstract
Cholesterol represents the most abundant single lipid in mammalian cells. How its asymmetric distribution between subcellular membranes is achieved and maintained attracts considerable interest. One of the challenges is that cholesterol rarely is transported alone, but rather is coupled with heterotypic transport and metabolism of other lipids, in particular phosphoinositides, phosphatidylserine, and sphingolipids. This perspective summarizes the major exo- and endocytic cholesterol transport routes and how lipid transfer proteins at membrane contacts and membrane transport intersect along these routes. It discusses the co-transport of cholesterol with other lipids in mammalian cells and reviews emerging evidence related to the physiological relevance of this process.
Collapse
Affiliation(s)
- Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Xin Zhou
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| |
Collapse
|
19
|
Sanders DW, Jumper CC, Ackerman PJ, Bracha D, Donlic A, Kim H, Kenney D, Castello-Serrano I, Suzuki S, Tamura T, Tavares AH, Saeed M, Holehouse AS, Ploss A, Levental I, Douam F, Padera RF, Levy BD, Brangwynne CP. SARS-CoV-2 requires cholesterol for viral entry and pathological syncytia formation. eLife 2021; 10:e65962. [PMID: 33890572 PMCID: PMC8104966 DOI: 10.7554/elife.65962] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/01/2021] [Indexed: 12/27/2022] Open
Abstract
Many enveloped viruses induce multinucleated cells (syncytia), reflective of membrane fusion events caused by the same machinery that underlies viral entry. These syncytia are thought to facilitate replication and evasion of the host immune response. Here, we report that co-culture of human cells expressing the receptor ACE2 with cells expressing SARS-CoV-2 spike, results in synapse-like intercellular contacts that initiate cell-cell fusion, producing syncytia resembling those we identify in lungs of COVID-19 patients. To assess the mechanism of spike/ACE2-driven membrane fusion, we developed a microscopy-based, cell-cell fusion assay to screen ~6000 drugs and >30 spike variants. Together with quantitative cell biology approaches, the screen reveals an essential role for biophysical aspects of the membrane, particularly cholesterol-rich regions, in spike-mediated fusion, which extends to replication-competent SARS-CoV-2 isolates. Our findings potentially provide a molecular basis for positive outcomes reported in COVID-19 patients taking statins and suggest new strategies for therapeutics targeting the membrane of SARS-CoV-2 and other fusogenic viruses.
Collapse
Affiliation(s)
- David W Sanders
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Chanelle C Jumper
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Paul J Ackerman
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Dan Bracha
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Anita Donlic
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Hahn Kim
- Princeton University Small Molecule Screening Center, Princeton University, Princeton, United States
- Department of Chemistry, Princeton University, Princeton, United States
| | - Devin Kenney
- Department of Microbiology, Boston University School of Medicine, Boston, United States
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
| | - Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - Saori Suzuki
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Tomokazu Tamura
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Alexander H Tavares
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, United States
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - Florian Douam
- Department of Microbiology, Boston University School of Medicine, Boston, United States
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Clifford P Brangwynne
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
- Howard Hughes Medical Institute, Princeton, United States
| |
Collapse
|
20
|
He C, Migawa MT, Chen K, Weston TA, Tanowitz M, Song W, Guagliardo P, Iyer KS, Bennett CF, Fong LG, Seth PP, Young SG, Jiang H. High-resolution visualization and quantification of nucleic acid-based therapeutics in cells and tissues using Nanoscale secondary ion mass spectrometry (NanoSIMS). Nucleic Acids Res 2021; 49:1-14. [PMID: 33275144 PMCID: PMC7797060 DOI: 10.1093/nar/gkaa1112] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/27/2020] [Accepted: 11/04/2020] [Indexed: 01/19/2023] Open
Abstract
Nucleic acid therapeutics (NATs) have proven useful in promoting the degradation of specific transcripts, modifying gene expression, and regulating mRNA splicing. In each situation, efficient delivery of nucleic acids to cells, tissues and intracellular compartments is crucial—both for optimizing efficacy and reducing side effects. Despite successes in NATs, our understanding of their cellular uptake and distribution in tissues is limited. Current methods have yielded insights into distribution of NATs within cells and tissues, but the sensitivity and resolution of these approaches are limited. Here, we show that nanoscale secondary ion mass spectrometry (NanoSIMS) imaging can be used to define the distribution of 5-bromo-2′-deoxythymidine (5-BrdT) modified antisense oligonucleotides (ASO) in cells and tissues with high sensitivity and spatial resolution. This approach makes it possible to define ASO uptake and distribution in different subcellular compartments and to quantify the impact of targeting ligands designed to promote ASO uptake by cells. Our studies showed that phosphorothioate ASOs are associated with filopodia and the inner nuclear membrane in cultured cells, and also revealed substantial cellular and subcellular heterogeneity of ASO uptake in mouse tissues. NanoSIMS imaging represents a significant advance in visualizing uptake and distribution of NATs; this approach will be useful in optimizing efficacy and delivery of NATs for treating human disease.
Collapse
Affiliation(s)
- Cuiwen He
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | - Kai Chen
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia
| | - Thomas A Weston
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | - Wenxin Song
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Paul Guagliardo
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia
| | - K Swaminathan Iyer
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia
| | | | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Punit P Seth
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, CA 90095, USA.,Department of Human Genetics, University of California, Los Angeles, CA 90095, USA
| | - Haibo Jiang
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia.,Department of Chemistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
21
|
Juhl AD, Lund FW, Jensen MLV, Szomek M, Heegaard CW, Guttmann P, Werner S, McNally J, Schneider G, Kapishnikov S, Wüstner D. Niemann Pick C2 protein enables cholesterol transfer from endo-lysosomes to the plasma membrane for efflux by shedding of extracellular vesicles. Chem Phys Lipids 2021; 235:105047. [PMID: 33422548 DOI: 10.1016/j.chemphyslip.2020.105047] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
The Niemann-Pick C2 protein (NPC2) is a sterol transfer protein in the lumen of late endosomes and lysosomes (LE/LYSs). Absence of functional NPC2 leads to endo-lysosomal buildup of cholesterol and other lipids. How NPC2's known capacity to transport cholesterol between model membranes is linked to its function in living cells is not known. Using quantitative live-cell imaging combined with modeling of the efflux kinetics, we show that NPC2-deficient human fibroblasts can export the cholesterol analog dehydroergosterol (DHE) from LE/LYSs. Internalized NPC2 accelerated sterol efflux extensively, accompanied by reallocation of LE/LYSs containing fluorescent NPC2 and DHE to the cell periphery. Using quantitative fluorescence loss in photobleaching of TopFluor-cholesterol (TF-Chol), we estimate a residence time for a rapidly exchanging sterol pool in LE/LYSs localized in close proximity to the plasma membrane (PM), of less than one min and observed non-vesicular sterol exchange between LE/LYSs and the PM. Excess sterol was released from the PM by shedding of cholesterol-rich vesicles. The ultrastructure of such vesicles was analyzed by combined fluorescence and cryo soft X-ray tomography (SXT), revealing that they can contain lysosomal cargo and intraluminal vesicles. Treating cells with apoprotein A1 and with nuclear receptor liver X-receptor (LXR) agonists to upregulate expression of ABC transporters enhanced cholesterol efflux from the PM, at least partly by accelerating vesicle release. We conclude that NPC2 inside LE/LYSs facilitates non-vesicular sterol exchange with the PM for subsequent sterol efflux to acceptor proteins and for shedding of sterol-rich vesicles from the cell surface.
Collapse
Affiliation(s)
- Alice Dupont Juhl
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark
| | - Frederik W Lund
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark
| | - Maria Louise V Jensen
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark
| | - Maria Szomek
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark
| | - Christian W Heegaard
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000, Aarhus C, Denmark
| | - Peter Guttmann
- Department X-Ray Microscopy, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, 12489, Berlin, Germany
| | - Stephan Werner
- Department X-Ray Microscopy, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, 12489, Berlin, Germany
| | - James McNally
- Department X-Ray Microscopy, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, 12489, Berlin, Germany
| | - Gerd Schneider
- Department X-Ray Microscopy, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, 12489, Berlin, Germany
| | - Sergey Kapishnikov
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark.
| |
Collapse
|
22
|
Abrams ME, Johnson KA, Radhakrishnan A, Alto NM. Accessible cholesterol is localized in bacterial plasma membrane protrusions. J Lipid Res 2020; 61:1538. [PMID: 32661016 PMCID: PMC7707182 DOI: 10.1194/jlr.ilr120000891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Michael E Abrams
- Departments of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kristen A Johnson
- Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arun Radhakrishnan
- Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Neal M Alto
- Departments of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
23
|
Radhakrishnan A, Rohatgi R, Siebold C. Cholesterol access in cellular membranes controls Hedgehog signaling. Nat Chem Biol 2020; 16:1303-1313. [PMID: 33199907 PMCID: PMC7872078 DOI: 10.1038/s41589-020-00678-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022]
Abstract
The Hedgehog (Hh) signaling pathway coordinates cell-cell communication in development and regeneration. Defects in this pathway underlie diseases ranging from birth defects to cancer. Hh signals are transmitted across the plasma membrane by two proteins, Patched 1 (PTCH1) and Smoothened (SMO). PTCH1, a transporter-like tumor-suppressor protein, binds to Hh ligands, but SMO, a G-protein-coupled-receptor family oncoprotein, transmits the Hh signal across the membrane. Recent structural, biochemical and cell-biological studies have converged at the surprising model that a specific pool of plasma membrane cholesterol, termed accessible cholesterol, functions as a second messenger that conveys the signal between PTCH1 and SMO. Beyond solving a central puzzle in Hh signaling, these studies are revealing new principles in membrane biology: how proteins respond to and remodel cholesterol accessibility in membranes and how the cholesterol composition of organelle membranes is used to regulate protein function.
Collapse
Affiliation(s)
- Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
24
|
Lange Y, Steck TL. Active cholesterol 20 years on. Traffic 2020; 21:662-674. [PMID: 32930466 DOI: 10.1111/tra.12762] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
This review considers the following hypotheses, some well-supported and some speculative. Almost all of the sterol molecules in plasma membranes are associated with bilayer phospholipids in complexes of varied strength and stoichiometry. These complexes underlie many of the material properties of the bilayer. The small fraction of cholesterol molecules exceeding the binding capacity of the phospholipids is thermodynamically active and serves diverse functions. It circulates briskly among the cell membranes, particularly through contact sites linking the organelles. Active cholesterol provides the upstream feedback signal to multiple mechanisms governing plasma membrane homeostasis, pegging the sterol level to a threshold set by its phospholipids. Active cholesterol could also be the cargo for various inter-organelle transporters and the form excreted from cells by reverse transport. Furthermore, it is integral to the function of caveolae; a mediator of Hedgehog regulation; and a ligand for the binding of cytolytic toxins to membranes. Active cholesterol modulates a variety of plasma membrane proteins-receptors, channels and transporters-at least in vitro.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
25
|
Aster Proteins Regulate the Accessible Cholesterol Pool in the Plasma Membrane. Mol Cell Biol 2020; 40:MCB.00255-20. [PMID: 32719109 DOI: 10.1128/mcb.00255-20] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/23/2020] [Indexed: 01/03/2023] Open
Abstract
Recent studies have demonstrated the existence of a discrete pool of cholesterol in the plasma membranes (PM) of mammalian cells-referred to as the accessible cholesterol pool-that can be detected by the binding of modified versions of bacterial cytolysins (e.g., anthrolysin O). When the amount of accessible cholesterol in the PM exceeds a threshold level, the excess cholesterol moves to the endoplasmic reticulum (ER), where it regulates the SREBP2 pathway and undergoes esterification. We reported previously that the Aster/Gramd1 family of sterol transporters mediates nonvesicular movement of cholesterol from the PM to the ER in multiple mammalian cell types. Here, we investigated the PM pool of accessible cholesterol in cholesterol-loaded fibroblasts with a knockdown of Aster-A and in mouse macrophages from Aster-B and Aster-A/B-deficient mice. Nanoscale secondary ion mass spectrometry (NanoSIMS) analyses revealed expansion of the accessible cholesterol pool in cells lacking Aster expression. The increased accessible cholesterol pool in the PM was accompanied by reduced cholesterol movement to the ER, evidenced by increased expression of SREBP2-regulated genes. Cosedimentation experiments with liposomes revealed that the Aster-B GRAM domain binds to membranes in a cholesterol concentration-dependent manner and that the binding is facilitated by the presence of phosphatidylserine. These studies revealed that the Aster-mediated nonvesicular cholesterol transport pathway controls levels of accessible cholesterol in the PM, as well as the activity of the SREBP pathway.
Collapse
|
26
|
Lee KS, Lee J, Lee P, Kim CU, Kim DJ, Jeong YJ, Park YJ, Tesh VL, Lee MS. Exosomes released from Shiga toxin 2a-treated human macrophages modulate inflammatory responses and induce cell death in toxin receptor expressing human cells. Cell Microbiol 2020; 22:e13249. [PMID: 32772454 DOI: 10.1111/cmi.13249] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/02/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022]
Abstract
Shiga toxins (Stxs) produced by Stx-producing Escherichia coli are the primarily virulence factors of hemolytic uremic syndrome and central nervous system (CNS) impairment. Although the precise mechanisms of toxin dissemination remain unclear, Stxs bind to extracellular vesicles (EVs). Exosomes, a subset of EVs, may play a key role in Stx-mediated renal injury. To test this hypothesis, we isolated exosomes from monocyte-derived macrophages in the presence of Stx2a or Stx2 toxoids. Macrophage-like differentiated THP-1 cells treated with Stxs secreted Stx-associated exosomes (Stx-Exo) of 90-130 nm in diameter, which induced cytotoxicity in recipient cells in a toxin receptor globotriaosylceramide (Gb3 )-dependent manner. Stx2-Exo engulfed by Gb3 -positive cells were translocated to the endoplasmic reticulum in the human proximal tubule epithelial cell line HK-2. Stx2-Exo contained pro-inflammatory cytokine mRNAs and proteins and induced more severe inflammation than purified Stx2a accompanied by greater death of target cells such as human renal or retinal pigment epithelial cells. Blockade of exosome biogenesis using the pharmacological inhibitor GW4869 reduced Stx2-Exo-mediated human renal cell death. Stx2-Exo isolated from human primary monocyte-derived macrophages activated caspase 3/7 and resulted in significant cell death in primary human renal cortical epithelial cells. Based on these results, we speculate that Stx-containing exosomes derived from macrophages may exacerbate cytotoxicity and inflammation and trigger cell death in toxin-sensitive cells. Therapeutic interventions targeting Stx-containing exosomes may prevent or ameliorate Stx-mediated acute vascular dysfunction.
Collapse
Affiliation(s)
- Kyung-Soo Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Jieun Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Pureum Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Yu-Jin Jeong
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Young-Jun Park
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College of Medicine, Bryan, Texas, USA
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| |
Collapse
|
27
|
Shao S, Fang H, Li Q, Wang G. Extracellular vesicles in Inflammatory Skin Disorders: from Pathophysiology to Treatment. Am J Cancer Res 2020; 10:9937-9955. [PMID: 32929326 PMCID: PMC7481415 DOI: 10.7150/thno.45488] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/31/2020] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs), naturally secreted by almost all known cell types into extracellular space, can transfer their bioactive cargos of nucleic acids and proteins to recipient cells, mediating cell-cell communication. Thus, they participate in many pathogenic processes including immune regulation, cell proliferation and differentiation, cell death, angiogenesis, among others. Cumulative evidence has shown the important regulatory effects of EVs on the initiation and progression of inflammation, autoimmunity, and cancer. In dermatology, recent studies indicate that EVs play key immunomodulatory roles in inflammatory skin disorders, including psoriasis, atopic dermatitis, lichen planus, bullous pemphigoid, systemic lupus erythematosus, and wound healing. Importantly, EVs can be used as biomarkers of pathophysiological states and/or therapeutic agents, both as carriers of drugs or even as a drug by themselves. In this review, we will summarize current research advances of EVs from different cells and their implications in inflammatory skin disorders, and further discuss their future applications, updated techniques, and challenges in clinical translational medicine.
Collapse
|
28
|
Zhou QD, Chi X, Lee MS, Hsieh WY, Mkrtchyan JJ, Feng AC, He C, York AG, Bui VL, Kronenberger EB, Ferrari A, Xiao X, Daly AE, Tarling EJ, Damoiseaux R, Scumpia PO, Smale ST, Williams KJ, Tontonoz P, Bensinger SJ. Interferon-mediated reprogramming of membrane cholesterol to evade bacterial toxins. Nat Immunol 2020; 21:746-755. [PMID: 32514064 PMCID: PMC7778040 DOI: 10.1038/s41590-020-0695-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022]
Abstract
Plasma membranes of animal cells are enriched for cholesterol. Cholesterol-dependent cytolysins (CDCs) are pore-forming toxins secreted by bacteria that target membrane cholesterol for their effector function. Phagocytes are essential for clearance of CDC-producing bacteria; however, the mechanisms by which these cells evade the deleterious effects of CDCs are largely unknown. Here, we report that interferon (IFN) signals convey resistance to CDC-induced pores on macrophages and neutrophils. We traced IFN-mediated resistance to CDCs to the rapid modulation of a specific pool of cholesterol in the plasma membrane of macrophages without changes to total cholesterol levels. Resistance to CDC-induced pore formation requires the production of the oxysterol 25-hydroxycholesterol (25HC), inhibition of cholesterol synthesis and redistribution of cholesterol to an esterified cholesterol pool. Accordingly, blocking the ability of IFN to reprogram cholesterol metabolism abrogates cellular protection and renders mice more susceptible to CDC-induced tissue damage. These studies illuminate targeted regulation of membrane cholesterol content as a host defense strategy.
Collapse
Affiliation(s)
- Quan D Zhou
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Xun Chi
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Min Sub Lee
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wei Yuan Hsieh
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonathan J Mkrtchyan
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - An-Chieh Feng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Cuiwen He
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Autumn G York
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University, New Haven, CT, USA
| | - Viet L Bui
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eliza B Kronenberger
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alessandra Ferrari
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xu Xiao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Allison E Daly
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elizabeth J Tarling
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Philip O Scumpia
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stephen T Smale
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kevin J Williams
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Steven J Bensinger
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA. .,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
29
|
|
30
|
Cultured macrophages transfer surplus cholesterol into adjacent cells in the absence of serum or high-density lipoproteins. Proc Natl Acad Sci U S A 2020; 117:10476-10483. [PMID: 32354992 DOI: 10.1073/pnas.1922879117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cholesterol-laden macrophage foam cells are a hallmark of atherosclerosis. For that reason, cholesterol metabolism in macrophages has attracted considerable scrutiny, particularly the mechanisms by which macrophages unload surplus cholesterol (a process referred to as "cholesterol efflux"). Many studies of cholesterol efflux in macrophages have focused on the role of ABC transporters in moving cholesterol onto high-density lipoproteins (HDLs), but other mechanisms for cholesterol efflux likely exist. We hypothesized that macrophages have the capacity to unload cholesterol directly onto adjacent cells. To test this hypothesis, we used methyl-β-cyclodextrin (MβCD) to load mouse peritoneal macrophages with [13C]cholesterol. We then plated the macrophages (in the absence of serum or HDL) onto smooth muscle cells (SMCs) that had been metabolically labeled with [15N]choline. After incubating the cells overnight in the absence of HDL or serum, we visualized 13C and 15N distribution by nanoscale secondary ion mass spectrometry (NanoSIMS). We observed substantial 13C enrichment in SMCs that were adjacent to [13C]cholesterol-loaded macrophages-including in cytosolic lipid droplets of SMCs. In follow-up studies, we depleted "accessible cholesterol" from the plasma membrane of [13C]cholesterol-loaded macrophages with MβCD before plating the macrophages onto the SMCs. After an overnight incubation, we again observed substantial 13C enrichment in the SMCs adjacent to macrophages. Thus, macrophages transfer cholesterol to adjacent cells in the absence of serum or HDL. We suspect that macrophages within tissues transfer cholesterol to adjacent cells, thereby contributing to the ability to unload surplus cholesterol.
Collapse
|
31
|
Leussink S, Aranda-Pardos I, A-Gonzalez N. Lipid metabolism as a mechanism of immunomodulation in macrophages: the role of liver X receptors. Curr Opin Pharmacol 2020; 53:18-26. [PMID: 32361182 DOI: 10.1016/j.coph.2020.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/21/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Macrophages are immune myeloid cells with an extreme ability to modulate their phenotype in response to insults and/or pathogens. The immunomodulatory capacity of macrophages is also patent during development as they adapt their phenotype to the host tissue environment establishing the heterogeneous populations of tissue-resident macrophages. An important mechanism of immunomodulation in macrophages occurs through the regulation of transcriptional activity. Numerous transcription factors are associated with macrophage plasticity, among them, several nuclear receptors. The nuclear receptors Liver X Receptors (LXRα and LXRβ) have also revealed as active players during macrophage adaptations in diverse scenarios. This review will address the different mechanisms by which LXRs contribute to immunomodulation in macrophages by connecting lipid metabolism and immunity through transcriptional regulation.
Collapse
Affiliation(s)
- Sophia Leussink
- Institute of Immunology, Westfälische Wilhelms Universität Münster, Germany
| | | | - Noelia A-Gonzalez
- Institute of Immunology, Westfälische Wilhelms Universität Münster, Germany; Cells-in-Motion Interfaculty Center, University of Münster, Germany.
| |
Collapse
|
32
|
Wang K, Wei Y, Zhang P, Wang J, Hu J, Wang L, Li B. [Progress in extracellular vesicle imaging methods]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:279-286. [PMID: 32376541 DOI: 10.12122/j.issn.1673-4254.2020.02.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are lipid bilayer-enclosed nanovesicles secreted by cells. These EVs are important mediators of intercellular communication by serving as vehicles for transfer of proteins, mRNA, miRNA and lipids between cells. Various visualization methods have been established to explore the characteristics of EVs and their role in physiological and pathological processes. The nanoscale size and high heterogeneity of EVs hamper the identification of their biological characteristics and functions. This review presents a comprehensive overview of EV imaging methods in light of the origin, separation and dynamic tracking of EVs, and the advantages and disadvantages of different imaging strategies are discussed. We believe that studies at the levels of single vesicles and single cells will become the frontier of future researches of EVs.
Collapse
Affiliation(s)
- Kaizhe Wang
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuhui Wei
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Ping Zhang
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianhua Wang
- School of Physics Science and Technology, Ningbo University, Ningbo 315211, China
| | - Jun Hu
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Lihua Wang
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Bin Li
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| |
Collapse
|
33
|
Naito T, Ercan B, Krshnan L, Triebl A, Koh DHZ, Wei FY, Tomizawa K, Torta FT, Wenk MR, Saheki Y. Movement of accessible plasma membrane cholesterol by the GRAMD1 lipid transfer protein complex. eLife 2019; 8:51401. [PMID: 31724953 PMCID: PMC6905856 DOI: 10.7554/elife.51401] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/13/2019] [Indexed: 12/18/2022] Open
Abstract
Cholesterol is a major structural component of the plasma membrane (PM). The majority of PM cholesterol forms complexes with other PM lipids, making it inaccessible for intracellular transport. Transition of PM cholesterol between accessible and inaccessible pools maintains cellular homeostasis, but how cells monitor the accessibility of PM cholesterol remains unclear. We show that endoplasmic reticulum (ER)-anchored lipid transfer proteins, the GRAMD1s, sense and transport accessible PM cholesterol to the ER. GRAMD1s bind to one another and populate ER-PM contacts by sensing a transient expansion of the accessible pool of PM cholesterol via their GRAM domains. They then facilitate the transport of this cholesterol via their StART-like domains. Cells that lack all three GRAMD1s exhibit striking expansion of the accessible pool of PM cholesterol as a result of less efficient PM to ER transport of accessible cholesterol. Thus, GRAMD1s facilitate the movement of accessible PM cholesterol to the ER in order to counteract an acute increase of PM cholesterol, thereby activating non-vesicular cholesterol transport. The human body contains trillions of cells. At the outer edge of each cell is the plasma membrane, which protects the cell from the external environment. This membrane is mostly made of fatty molecules known as lipids and about half of these lipids are specifically cholesterol. Human cells can either take up cholesterol that were obtained via the diet or produce it within a compartment of the cell called the endoplasmic reticulum. Cells need to monitor the cholesterol levels in both the endoplasmic reticulum and the plasma membrane in order to regulate the uptake or production of this lipid. For example, if there is too much of cholesterol in the plasma membrane, then the cell transports some to the endoplasmic reticulum to tell it to shut down cholesterol production. However, how these different areas of the cell communicate with each other, and transport cholesterol, has remained unclear. Naito et al. set out to look for key regulators of cholesterol transport and identified a group of endoplasmic reticulum proteins called GRAMD1 proteins. Cholesterol in the plasma membrane is either accessible or inaccessible, meaning it either can or cannot be moved back into the cell. The GRAMD1 proteins sense accessible cholesterol, and experiments with human cells grown in the laboratory showed that, specifically, the GRAMD1 proteins work together in a complex to sense accessible cholesterol at or near the plasma membrane. One particular part of the protein senses when the amount of accessible cholesterol reaches a certain level at the plasma membrane; when this threshold is reached, the complex flips a switch to start the transport of cholesterol to the endoplasmic reticulum and tell it to shut down cholesterol production. This coupling of sensing and transporting lipids by one protein complex also helps maintain the right ratio of accessible and inaccessible cholesterol in the plasma membrane to prevent cells from activating unwanted cell-signaling events. Getting rid of the GRAMD1 proteins in cells, or removing sensing part of these proteins, leads to inefficient transport of cholesterol. A better understanding of how GRAMD1 proteins sense the accessibility of cholesterol could potentially help identify new approaches to control cholesterol transport inside cells. This may in turn eventually lead to new treatments that counteract the defects in cholesterol metabolism seen in some forms of neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease.
Collapse
Affiliation(s)
- Tomoki Naito
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Bilge Ercan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Logesvaran Krshnan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Alexander Triebl
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dylan Hong Zheng Koh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Fan-Yan Wei
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Federico Tesio Torta
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
34
|
Hu X, Weston TA, He C, Jung RS, Heizer PJ, Young BD, Tu Y, Tontonoz P, Wohlschlegel JA, Jiang H, Young SG, Fong LG. Release of cholesterol-rich particles from the macrophage plasma membrane during movement of filopodia and lamellipodia. eLife 2019; 8:50231. [PMID: 31486771 PMCID: PMC6750930 DOI: 10.7554/elife.50231] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022] Open
Abstract
Cultured mouse peritoneal macrophages release large numbers of ~30-nm cholesterol-rich particles. Here, we show that those particles represent fragments of the plasma membrane that are pulled away and left behind during the projection and retraction of filopodia and lamellipodia. Consistent with this finding, the particles are enriched in proteins found in focal adhesions, which attach macrophages to the substrate. The release of particles is abolished by blocking cell movement (either by depolymerizing actin with latrunculin A or by inhibiting myosin II with blebbistatin). Confocal microscopy and NanoSIMS imaging studies revealed that the plasma membrane-derived particles are enriched in 'accessible cholesterol' (a mobile pool of cholesterol detectable with the modified cytolysin ALO-D4) but not in sphingolipid-sequestered cholesterol [a pool detectable with ostreolysin A (OlyA)]. The discovery that macrophages release cholesterol-rich particles during cellular locomotion is likely relevant to cholesterol efflux and could contribute to extracellular cholesterol deposition in atherosclerotic plaques.
Collapse
Affiliation(s)
- Xuchen Hu
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Thomas A Weston
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Cuiwen He
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Rachel S Jung
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Patrick J Heizer
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Brian D Young
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, United States
| | - Yiping Tu
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, United States
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, United States
| | - Haibo Jiang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,School of Molecular Sciences, University of Western Australia, Perth, Australia
| | - Stephen G Young
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Loren G Fong
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
35
|
Hu X, Matsumoto K, Jung RS, Weston TA, Heizer PJ, He C, Sandoval NP, Allan CM, Tu Y, Vinters HV, Liau LM, Ellison RM, Morales JE, Baufeld LJ, Bayley NA, He L, Betsholtz C, Beigneux AP, Nathanson DA, Gerhardt H, Young SG, Fong LG, Jiang H. GPIHBP1 expression in gliomas promotes utilization of lipoprotein-derived nutrients. eLife 2019; 8:e47178. [PMID: 31169500 PMCID: PMC6594755 DOI: 10.7554/elife.47178] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/05/2019] [Indexed: 12/25/2022] Open
Abstract
GPIHBP1, a GPI-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) within the subendothelial spaces and shuttles it to the capillary lumen. GPIHBP1-bound LPL is essential for the margination of triglyceride-rich lipoproteins (TRLs) along capillaries, allowing the lipolytic processing of TRLs to proceed. In peripheral tissues, the intravascular processing of TRLs by the GPIHBP1-LPL complex is crucial for the generation of lipid nutrients for adjacent parenchymal cells. GPIHBP1 is absent from the capillaries of the brain, which uses glucose for fuel; however, GPIHBP1 is expressed in the capillaries of mouse and human gliomas. Importantly, the GPIHBP1 in glioma capillaries captures locally produced LPL. We use NanoSIMS imaging to show that TRLs marginate along glioma capillaries and that there is uptake of TRL-derived lipid nutrients by surrounding glioma cells. Thus, GPIHBP1 expression in gliomas facilitates TRL processing and provides a source of lipid nutrients for glioma cells.
Collapse
Affiliation(s)
- Xuchen Hu
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Ken Matsumoto
- VIB-KU Leuven Center for Cancer Biology (CCB)LeuvenBelgium
| | - Rachel S Jung
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Thomas A Weston
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Patrick J Heizer
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Cuiwen He
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Norma P Sandoval
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Christopher M Allan
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Yiping Tu
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Rochelle M Ellison
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Jazmin E Morales
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Lynn J Baufeld
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Nicholas A Bayley
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsalaSweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsalaSweden
- Integrated Cardio Metabolic Centre (ICMC)Karolinska InstitutetHuddingeSweden
| | - Anne P Beigneux
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Holger Gerhardt
- VIB-KU Leuven Center for Cancer Biology (CCB)LeuvenBelgium
- Max Delbrück Center for Molecular MedicineBerlinGermany
| | - Stephen G Young
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Loren G Fong
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Haibo Jiang
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- School of Molecular SciencesUniversity of Western AustraliaPerthAustralia
| |
Collapse
|
36
|
Loewith R, Riezman H, Winssinger N. Sphingolipids and membrane targets for therapeutics. Curr Opin Chem Biol 2019; 50:19-28. [PMID: 30897494 DOI: 10.1016/j.cbpa.2019.02.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 12/20/2022]
Abstract
Lipids and membranes are often strongly altered in various diseases and pathologies, but are not often targeted for therapeutic advantage. In particular, the sphingolipids are particularly sensitive to altered physiology and have been implicated as important players in not only several rare hereditary diseases, but also other major pathologies, including cancer. This review discusses some potential targets in the sphingolipid pathway and describes how the initial drug compounds have been evolved to create potentially improved therapeutics. This reveals how lipids and their interactions with proteins can be used for therapeutic advantage. We also discuss the possibility that modification of the physical properties of membranes could also affect intracellular signaling and be of therapeutic interest.
Collapse
Affiliation(s)
- Robbie Loewith
- Department of Molecular Biology, NCCR Chemical Biology, University of Geneva, 30 quai Ernest Ansermet, CH-1205 Geneva, Switzerland.
| | - Howard Riezman
- Department of Biochemistry, NCCR Chemical Biology, University of Geneva, 30 quai Ernest Ansermet, CH-1205 Geneva, Switzerland.
| | - Nicolas Winssinger
- Department of Organic Chemistry, NCCR Chemical Biology, University of Geneva, 30 quai Ernest Ansermet, CH-1205 Geneva, Switzerland.
| |
Collapse
|