1
|
Cheng CJ, Wang LC, Chu LJ, Chen KY, Huang CY, Lan KL, Huang KY. Extracellular vesicles from fifth-stage larval Angiostrongylus cantonensis upregulate cholesterol biosynthesis and suppress NLRP2-associated inflammatory responses in mouse astrocytes. mSystems 2025; 10:e0101424. [PMID: 39636121 PMCID: PMC11748502 DOI: 10.1128/msystems.01014-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
Angiostrongylus cantonensis is a zoonotic parasite that causes severe symptoms in humans, including eosinophilic meningitis and eosinophilic meningoencephalitis. Extracellular vesicles (EVs) derived from helminthes have been implicated in regulating host survival and immune response. However, the roles of A. cantonensis EVs in modulating parasite pathogenesis and host immune response remain poorly understood. Herein, we characterized EVs derived from A. cantonensis fifth-stage larvae (L5) and adult worms. Ultrastructural features showed that EVs from adult worms are smaller in size compared with those from L5. Proteomic analysis identified stage-specific proteins packaged in L5 and adult worm EVs. To investigate the crosstalk between L5 EVs and host cells, RNA sequencing analysis was conducted to identify the differentially expressed genes (DEGs) and enriched biological pathways in mouse astrocytes treated with L5 EVs. GO and KEGG enrichment analysis demonstrated that the pathways related to "cholesterol biosynthesis" are significantly upregulated in L5 EV-treated astrocytes. Based on the transcriptomic data, we observed a downregulated trend of NOD-like receptors (NLRs) protein 2 (NLRP2), a key regulator of brain inflammation, in mouse astrocytes treated with L5 EVs. To validate this result, we utilized ATP to induce the expression of NLRP2 inflammasome-related genes and proteins, as well as the secretion of downstream cytokines. Notably, ATP-induced overexpression of NLRP2 inflammasome-related molecules was significantly reduced in mouse astrocytes upon L5 EV treatment. Collectively, our data suggest that A. cantonensis L5 EVs enhance cholesterol synthesis and potentially modulate immune response by reducing NLRP2 inflammasome-related signaling in non-permissive host cells.IMPORTANCEAngiostrongylus cantonensis is a significant causative agent of eosinophilic meningitis and eosinophilic meningoencephalitis in humans. Helminth-derived extracellular vesicles (EVs) are known to play a crucial role in parasite pathogenesis and host immunomodulation. However, the protein compositions of A. cantonensis EVs and their roles in parasite pathogenesis and host immune response remain unclear. Our results demonstrate for the first time the distinct protein compositions in A. cantonensis L5 and adult worm EVs. The highly abundant proteins in L5 EVs that have immunomodulatory or pathogenic potential in the host deserve further investigation. Additionally, the uptake of L5 EVs by mouse astrocytes significantly upregulates cholesterol synthesis and suppresses ATP-induced NLRP2 inflammasome-related signaling. This study highlights the immunomodulatory roles of L5 EVs in non-permissive hosts, suggesting their potential as therapeutic targets and vaccine candidates against A. cantonensis.
Collapse
Affiliation(s)
- Chien-Ju Cheng
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, Taiwan
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Lian-Chen Wang
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Lichieh Julie Chu
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
- Department of Otolaryngology—Head & Neck Surgery, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Kuang-Yao Chen
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Ching-Yun Huang
- Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei City, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City, Taiwan
| | - Kuo-Lun Lan
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Kuo-Yang Huang
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, Taiwan
- Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei City, Taiwan
| |
Collapse
|
2
|
Anselmo S, Bonaccorso E, Gangemi C, Sancataldo G, Conti Nibali V, D’Angelo G. Lipid Rafts in Signalling, Diseases, and Infections: What Can Be Learned from Fluorescence Techniques? MEMBRANES 2025; 15:6. [PMID: 39852247 PMCID: PMC11766618 DOI: 10.3390/membranes15010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025]
Abstract
Lipid rafts are dynamic microdomains in the membrane, rich in cholesterol and sphingolipids, that are critical for biological processes like cell signalling, membrane trafficking, and protein organization. Their essential role is claimed in both physiological and pathological conditions, including cancer, neurodegenerative diseases, and viral infections, making them a key area of research. Fluorescence-based approaches, including super-resolution fluorescence microscopy techniques, enable precise analysis of the organization, dynamics, and interactions of these microdomains, thanks also to the innovative design of appropriate fluorescent probes. Moreover, these non-invasive approaches allow for the study of live cells, facilitating the collection of quantitative data under physiologically relevant conditions. This review synthesizes the latest insights into the role of lipid rafts in biological and pathological processes and underscores how fluorescence techniques have advanced our understanding of these critical microdomains. The findings emphasize the pivotal role of lipid rafts in health and disease, providing a foundation for future research and potential therapeutic interventions.
Collapse
Affiliation(s)
- Sara Anselmo
- Department of Physics and Chemistry-Emilio Segré, University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (S.A.); (G.S.)
| | - Elisa Bonaccorso
- Department of Mathematics, Computer Science, Physics and Earth Science, University of Messina, Viale Stagno D’Alcontres 31, 98166 Messina, Italy (V.C.N.)
| | - Chiara Gangemi
- Department of Mathematics, Computer Science, Physics and Earth Science, University of Messina, Viale Stagno D’Alcontres 31, 98166 Messina, Italy (V.C.N.)
| | - Giuseppe Sancataldo
- Department of Physics and Chemistry-Emilio Segré, University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (S.A.); (G.S.)
| | - Valeria Conti Nibali
- Department of Mathematics, Computer Science, Physics and Earth Science, University of Messina, Viale Stagno D’Alcontres 31, 98166 Messina, Italy (V.C.N.)
| | - Giovanna D’Angelo
- Department of Mathematics, Computer Science, Physics and Earth Science, University of Messina, Viale Stagno D’Alcontres 31, 98166 Messina, Italy (V.C.N.)
| |
Collapse
|
3
|
Yamaji-Hasegawa A, Kobayashi T. Maistero-2, a Novel Probe for Sterols: Application for Visualizing Cellular Cholesterol. Methods Mol Biol 2025; 2888:53-66. [PMID: 39699724 DOI: 10.1007/978-1-0716-4318-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Maistero-2 is a novel, non-toxic cholesterol-binding protein derived from an edible mushroom Grifola frondosa mRNA. Maistero-2 specifically binds to lipid membranes containing 3-hydroxy sterols with a lower cholesterol concentration threshold than cholesterol-binding domain 4 (D4) of perfringolysin O (PFO) and anthrolysin O (ALO). Maistero-2 binding is particularly sensitive to the size and conformation of the A-, B-, and D-ring of sterols but not very sensitive to modifications of the isooctyl side chain commonly found in phytosterols. EGFP-maistero-2 has been demonstrated to be a suitable tool to visualize the increase of cell surface cholesterol during the differentiation of Neuro 2a cells and heterogeneous cholesterol distribution between CD63-positive and LAMP1-positive late endosomes/lysosomes in HeLa cells. This chapter describes the detailed protocols for examining the binding of maistero-2 to lipids and model membranes and the labeling of cell surface and intracellular cholesterol by EGFP-maistero-2.
Collapse
Affiliation(s)
- Akiko Yamaji-Hasegawa
- Laboratory for Neural Cell Dynamics, RIKEN Center for Brain Science, Saitama, Japan.
| | - Toshihide Kobayashi
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
4
|
Eichler J, Huver S, Knorr CJ, Wendling C, Kobayashi T, Tomasetto C, Alpy F. Methods for Visualizing and Quantifying Cholesterol Distribution in Mammalian Cells Using Filipin and D4 Probes. Methods Mol Biol 2025; 2888:101-118. [PMID: 39699727 DOI: 10.1007/978-1-0716-4318-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Cholesterol is a key component of biological membranes and, like many cellular lipids, is unevenly distributed among organelles. Disruptions in cholesterol trafficking are associated with various pathologies, including lysosomal lipid storage disorders, often characterized by intracellular cholesterol accumulation. A significant challenge in studying cholesterol trafficking is the lack of easy methods to trace this molecule in situ. Fluorescent probes that specifically bind cholesterol have enabled the visualization and imaging of cholesterol distribution within cells. This chapter details optimized methods for visualizing and quantifying free cholesterol at the plasma membrane and intracellulaly, both in individual cells and in large cell populations. These methods use two fluorescent probes: the D4 fragment of perfringolysin O fused to monomeric EGFP (mEGFP-D4 and the more sensitive mutant mEGFP-D4H) and the polyene macrolide filipin. We describe robust methods for quantifying plasma membrane cholesterol by flow cytometry and to visualize intracellular cholesterol pools by light microscopy. Furthermore, we introduce a refined filipin staining protocol that enhances intracellular cholesterol detection. For precise quantification, we developed an automated image analysis pipeline. This chapter provides a comprehensive guide for staining and quantifying cellular cholesterol, offering valuable tools for studying cholesterol dynamics in mammalian cells.
Collapse
Affiliation(s)
- Julie Eichler
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), UMR 7104, CNRS, Inserm, Université de Strasbourg, Illkirch, France
| | - Sophie Huver
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), UMR 7104, CNRS, Inserm, Université de Strasbourg, Illkirch, France
| | - Céline J Knorr
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), UMR 7104, CNRS, Inserm, Université de Strasbourg, Illkirch, France
| | - Corinne Wendling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), UMR 7104, CNRS, Inserm, Université de Strasbourg, Illkirch, France
| | - Toshihide Kobayashi
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Catherine Tomasetto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), UMR 7104, CNRS, Inserm, Université de Strasbourg, Illkirch, France
| | - Fabien Alpy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), UMR 7104, CNRS, Inserm, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
5
|
Baria E, Dallari C, Mattii F, Pavone FS, Credi C, Cicchi R, Morrone A, Capitini C, Calamai M. Evaluating pathological levels of intracellular cholesterol through Raman and surface-enhanced Raman spectroscopies. Sci Rep 2024; 14:28566. [PMID: 39557950 PMCID: PMC11574121 DOI: 10.1038/s41598-024-76621-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Versatile methods for the quantification of intracellular cholesterol are essential for understanding cellular physiology and for diagnosing disorders linked to cholesterol metabolism. Here we used Raman spectroscopy (RS) and surface-enhanced Raman spectroscopy (SERS) to measure changes in cholesterol after incubating human fibroblasts with increasing concentrations of cholesterol-methyl-β-cyclodextrin. RS and SERS were sensitive and accurate enough to detect high levels of cholesterol in fibroblasts from patients affected by type C Niemann-Pick disease (NPC), a lysosomal storage disorder characterized by the primary accumulation of cholesterol. Moreover, SERS was able to distinguish between fibroblasts from different NPC patients, demonstrating higher accuracy than RS and standard fluorescent labeling of cholesterol with filipin III. We show that the type of gold nanoparticles used as signal enhancer surfaces in our SERS measurements are internalized by the cells and are eventually found in lysosomes, the main site of accumulation of cholesterol in NPC fibroblasts. The higher sensitivity of SERS can thus be attributed to the specific trafficking of our gold nanoparticles into these organelles. Our results indicate that RS and SERS can be used as sensitive and accurate methods for the evaluation of intracellular cholesterol content, allowing for the potential development of an optical detection tool for the ex-vivo screening and monitoring of those diseases characterized by abnormal modification in cholesterol levels.
Collapse
Affiliation(s)
- Enrico Baria
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, 50019, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, 50019, Italy
| | - Caterina Dallari
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, 50019, Italy.
- National Institute of Optics - National Research Council, Sesto Fiorentino, 50019, Italy.
| | - Francesco Mattii
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, 50019, Italy
| | - Francesco Saverio Pavone
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, 50019, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, 50019, Italy
- National Institute of Optics - National Research Council, Sesto Fiorentino, 50019, Italy
| | - Caterina Credi
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, 50019, Italy
- National Institute of Optics - National Research Council, Sesto Fiorentino, 50019, Italy
| | - Riccardo Cicchi
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, 50019, Italy
- National Institute of Optics - National Research Council, Sesto Fiorentino, 50019, Italy
| | - Amelia Morrone
- Laboratory of Molecular Genetics of Neurometabolic Diseases, Department of Neuroscience and Medical Genetics, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Claudia Capitini
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, 50019, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, 50019, Italy
| | - Martino Calamai
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, 50019, Italy.
- National Institute of Optics - National Research Council, Sesto Fiorentino, 50019, Italy.
| |
Collapse
|
6
|
Wang SH, Chiang PM, Su YY, Yu YT, Chen YP, Chen TY, Medeiros LJ, Chu CY, Chen PC, Chang KC. Cytoplasmic Lipid Droplets Predict Worse Prognosis in Diffuse Large B-Cell Lymphoma: Next-Generation Sequencing Deciphering Lipogenic Genes. Am J Surg Pathol 2024; 48:1425-1438. [PMID: 38979928 DOI: 10.1097/pas.0000000000002280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Burkitt lymphoma is characterized by high cell turnover and numerous cytoplasmic vacuoles that are demonstrated to be lipid droplets (LDs) decorated by adipophilin. By contrast, cytoplasmic vacuoles are variably observed in diffuse large B-cell lymphoma (DLBCL) and less well characterized. In this study, we first validated in DLBCL that cytoplasmic vacuoles are indeed LDs by Oil-red-O stain, Bodipy fluorescent stain, and electron microscopy. Second, in a cohort of DLBCL patients (n=52) we showed that LDs in effusional lymphoma cells were associated with a poorer prognosis ( P =0.029, log-rank test) and higher International Prognostic Index (IPI) score (94% vs. 66%, P =0.026) than those without. Moreover, using adipophilin as a surrogate marker for LDs, we found in another cohort of biopsy specimen (n=85) that expression of adipophilin by lymphoma cells predicted a poorer prognosis ( P =0.007, log-rank test) and higher IPI score (63% vs. 30%, P =0.005). In addition, whole exome sequencing of effusional DLBCL cells showed LD-positive DLBCL shared genetic features with the MCD ( MYD88 and CD79B mutations) subtype and highlighted OSBPL10 and CUBN as the most frequently mutated genes involved in lipogenesis. Whole transcriptome analysis by comparing effusional DLBCL cells with versus without LDs showed upregulation of EHHADH , SLC1A1 , CD96 , INPP4B , and RNF183 relevant for lymphoma lipogenesis and upregulation of epithelial-mesenchymal transition and KRAS signaling pathways. Higher expression of EHHADH and CD96 were validated in LD-positive clinical samples and LD-rich cell lines than LD-poor cells along with the known lipogenic gene, FASN . Our findings highlight the roles of LDs and adipophilin expression in DLBCL, suggest that these markers may predict prognosis and show that lipogenic genes may be potential therapeutic targets.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lipid Droplets/metabolism
- Male
- Lipogenesis/genetics
- Female
- Perilipin-2/genetics
- Perilipin-2/analysis
- Perilipin-2/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/analysis
- Middle Aged
- Prognosis
- High-Throughput Nucleotide Sequencing
- Aged
- Adult
- Mutation
- Predictive Value of Tests
- Fatty Acid Synthase, Type I/genetics
- Fatty Acid Synthase, Type I/metabolism
- Fatty Acid Synthase, Type I/analysis
- Aged, 80 and over
Collapse
Affiliation(s)
| | - Po-Min Chiang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University
| | - Yung-Yeh Su
- Oncology
- National Institute of Cancer Research, National Health Research Institutes
| | - Yu-Ting Yu
- Department of Pathology, School of Medicine, Chung Shan Medical University
- Department of Pathology, Chung Shan Medical University Hospital, Taichung
| | - Ya-Ping Chen
- Department of Internal Medicine, Division of Hematology, National Cheng Kung University Hospital
| | - Tsai-Yun Chen
- Department of Internal Medicine, Division of Hematology, National Cheng Kung University Hospital
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Chang-Yao Chu
- Department of Pathology, Chi-Mei Medical Center, Tainan
- School of Medicine, College of Medicine, National Sun Yat-sen University
| | - Peng-Chieh Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University
| | - Kung-Chao Chang
- Departments of Pathology
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
7
|
Dai S, Luo M, Jiang T, Lu M, Zhou X, Zhu S, Han X, Yang F, Wang H, Xu D. Dexamethasone as an emerging environmental pollutant: Disruption of cholesterol-dependent synaptogenesis in the hippocampus and subsequent neurobehavioral impacts in offspring. ENVIRONMENT INTERNATIONAL 2024; 192:109064. [PMID: 39413532 DOI: 10.1016/j.envint.2024.109064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
When fetuses are exposed to abnormally high levels of glucocorticoids in utero, irreversible damage to neuronal synaptogenesis occurs, leading to long-term cognitive and emotional behavioral abnormalities after birth. In this study, we investigated how maternal exposure to a novel environmental pollutant-synthetic glucocorticoid dexamethasone-affects offspring cognitive and emotional behaviors enduringly. We noted that offspring subjected to maternal dexamethasone exposure (MDE) displayed cognitive and emotional neurobehavioral deficits beginning in infancy, and these impairments persisted into adulthood. The principal mechanism involves MDE-induced damage to hippocampal neuronal synapse formation in the offspring, primarily due to a cholesterol deficiency which destabilizes neuronal membranes, thereby affecting normal synapse formation and ultimately leading to cognitive and emotional deficiencies. Specifically, we demonstrated abnormal activation of glucocorticoid receptors in hippocampal astroglial cells of MDE offspring, which triggers changes in the miR-450a-3p/HAT1/ABCG1 signaling axis, causing impaired cholesterol efflux in astroglial cells and insufficient cholesterol supply to neurons, further impairing synaptogenesis. This research not only underscores the significant impact of prenatal environmental pollutants on long-term health outcomes in offspring but also broadens our understanding of how prenatal exposure to glucocorticoids affects brain development in the progeny, providing new insights for interventions in neurodevelopmental and psychiatric disorders of fetal origin.
Collapse
Affiliation(s)
- Shiyun Dai
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; National Health Commission Key Laboratory of Clinical Research for Cardiovascular Medications, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingcui Luo
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Tao Jiang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Mengxi Lu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinli Zhou
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Sen Zhu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiaoyi Han
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Fang Yang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Dan Xu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
8
|
Ahn JH, Johnny CL, Chenoweth DM. Duo-Chol: A Photoconvertible Live Cell Imaging Tool for Tracking Cholesterol. Bioconjug Chem 2024; 35:890-896. [PMID: 38913976 DOI: 10.1021/acs.bioconjchem.4c00207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Investigating cholesterol trafficking pathways continues to be of significant scientific interest owing to its homeostasis being associated with several debilitating cardiovascular and neurodegenerative diseases including atherosclerosis, Niemann-Pick's disease, Alzheimer's disease, and Parkinson's disease. To further our understanding of cholesterol trafficking, it is imperative to develop new fluorescent probes that possess improved photostability, low efflux, and high spatial and temporal resolution for live-cell imaging. In this study, we developed a photoconvertible fluorescent cholesterol analog, Duo-Chol, enabling the improved spatiotemporal fluorescence imaging of the dynamic localization of cholesterol in live cells. This tool provides a unique and powerful approach to interrogating cholesterol dynamics, addressing the limitations of existing methods, and expanding our ability to probe the biological role of sterols in living cells.
Collapse
Affiliation(s)
- June H Ahn
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Christopher L Johnny
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - David M Chenoweth
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
9
|
Palmulli R, Couty M, Piontek MC, Ponnaiah M, Dingli F, Verweij FJ, Charrin S, Tantucci M, Sasidharan S, Rubinstein E, Kontush A, Loew D, Lhomme M, Roos WH, Raposo G, van Niel G. CD63 sorts cholesterol into endosomes for storage and distribution via exosomes. Nat Cell Biol 2024; 26:1093-1109. [PMID: 38886558 DOI: 10.1038/s41556-024-01432-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 05/01/2024] [Indexed: 06/20/2024]
Abstract
Extracellular vesicles such as exosomes are now recognized as key players in intercellular communication. Their role is influenced by the specific repertoires of proteins and lipids, which are enriched when they are generated as intraluminal vesicles (ILVs) in multivesicular endosomes. Here we report that a key component of small extracellular vesicles, the tetraspanin CD63, sorts cholesterol to ILVs, generating a pool that can be mobilized by the NPC1/2 complex, and exported via exosomes to recipient cells. In the absence of CD63, cholesterol is retrieved from the endosomes by actin-dependent vesicular transport, placing CD63 and cholesterol at the centre of a balance between inward and outward budding of endomembranes. These results establish CD63 as a lipid-sorting mechanism within endosomes, and show that ILVs and exosomes are alternative providers of cholesterol.
Collapse
Affiliation(s)
- Roberta Palmulli
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Mickaël Couty
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
- CRCI2NA, Nantes Université, Inserm UMR1307, CNRS UMR6075, Université d'Angers, Nantes, France
| | - Melissa C Piontek
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013, Paris, France
| | - Florent Dingli
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Frederik J Verweij
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Stéphanie Charrin
- Centre d'Immunologie et des Maladies Infectieuses (CIMI), Sorbonne Université, Inserm, Paris, France
| | - Matteo Tantucci
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Sajitha Sasidharan
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Eric Rubinstein
- Centre d'Immunologie et des Maladies Infectieuses (CIMI), Sorbonne Université, Inserm, Paris, France
| | - Anatol Kontush
- ICAN, National Institute for Health and Medical Research, Paris, France
| | - Damarys Loew
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013, Paris, France
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248, Paris Cedex 05, France
| | - Guillaume van Niel
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France.
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France.
- CRCI2NA, Nantes Université, Inserm UMR1307, CNRS UMR6075, Université d'Angers, Nantes, France.
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France.
| |
Collapse
|
10
|
Hu Y, Luo H, Zhao L, Guo X, Wang S, Hu R, Yang G. A chalcone-based ESIPT and AIE fluorophore for β-gal imaging in living cells. Org Biomol Chem 2024; 22:1850-1858. [PMID: 38345427 DOI: 10.1039/d3ob01953e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
β-Galactosidase (β-gal), which is responsible for the hydrolysis of the glycosidic bond of lactose to galactose, has been recognized as an important biomarker of cell or organism status, especially cell senescence and primary ovarian cancer. Extensive efforts have been devoted to develop probes for detecting and visualizing β-gal in cells. Herein, a fluorescent probe gal-HCA which possesses both excited-state intramolecular proton transfer (ESIPT) and aggregation-induced emission (AIE) properties was prepared to monitor β-gal in living cells. The probe consists of 2-hydroxy-4'-dimethylamino-chalcone (HCA) capped with a D-galactose group. The cleavage of the glycosidic bond in gal-HCA triggered by β-gal releases HCA, which results in a significant bathochromic shift in fluorescence from 532 to 615 nm. The probe exhibited high selectivity and sensitivity toward β-gal with a detection limit as low as 0.0122 U mL-1. The confocal imaging investigation demonstrated the potential of gal-HCA in monitoring the endocellular overexpressed β-gal in senescent cells and ovarian cancer cells. This study provides a straightforward approach for the development of fluorescent probes to monitor β-gal and detection of β-gal-associated diseases.
Collapse
Affiliation(s)
- Yiran Hu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China.
| | - Haiyan Luo
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China.
| | - Luyao Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China.
| | - Xudong Guo
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China.
| | - Shuangqing Wang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China.
| | - Rui Hu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China.
| | - Guoqiang Yang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
11
|
Bruno F, Camuso S, Capuozzo E, Canterini S. The Antifungal Antibiotic Filipin as a Diagnostic Tool of Cholesterol Alterations in Lysosomal Storage Diseases and Neurodegenerative Disorders. Antibiotics (Basel) 2023; 12:antibiotics12010122. [PMID: 36671323 PMCID: PMC9855188 DOI: 10.3390/antibiotics12010122] [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: 12/02/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 01/10/2023] Open
Abstract
Cholesterol is the most considerable member of a family of polycyclic compounds understood as sterols, and represents an amphipathic molecule, such as phospholipids, with the polar hydroxyl group located in position 3 and the rest of the molecule is completely hydrophobic. In cells, it is usually present as free, unesterified cholesterol, or as esterified cholesterol, in which the hydroxyl group binds to a carboxylic acid and thus generates an apolar molecule. Filipin is a naturally fluorescent antibiotic that exerts a primary antifungal effect with low antibacterial activity, interfering with the sterol stabilization of the phospholipid layers and favoring membrane leakage. This polyene macrolide antibiotic does not bind to esterified sterols, but only to non-esterified cholesterol, and it is commonly used as a marker to label and quantify free cholesterol in cells and tissues. Several lines of evidence have indicated that filipin staining could be a good diagnostic tool for the cholesterol alterations present in neurodegenerative (e.g., Alzheimer's Disease and Huntington Disease) and lysosomal storage diseases (e.g., Niemann Pick type C Disease and GM1 gangliosidosis). Here, we have discussed the uses and applications of this fluorescent molecule in lipid storage diseases and neurodegenerative disorders, exploring not only the diagnostic strength of filipin staining, but also its limitations, which over the years have led to the development of new diagnostic tools to combine with filipin approach.
Collapse
Affiliation(s)
- Francesco Bruno
- Regional Neurogenetic Centre (CRN), Department of Primary Care, ASP Catanzaro, 88046 Lamezia Terme, Italy
- Association for Neurogenetic Research (ARN), 88046 Lamezia Terme, Italy
| | - Serena Camuso
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy
| | - Elisabetta Capuozzo
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (E.C.); (S.C.)
| | - Sonia Canterini
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (E.C.); (S.C.)
| |
Collapse
|
12
|
Hocquemiller M, Giersch L, Mei X, Gross AL, Randle AN, Gray-Edwards HL, Hudson JA, Todeasa S, Stoica L, Martin DR, Sena-Esteves M, Aiach K, Laufer R. AAVrh10 vector corrects pathology in animal models of GM1 gangliosidosis and achieves widespread distribution in the CNS of nonhuman primates. Mol Ther Methods Clin Dev 2022; 27:281-292. [PMID: 36320411 PMCID: PMC9594110 DOI: 10.1016/j.omtm.2022.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/05/2022] [Indexed: 12/13/2022]
Abstract
GM1 gangliosidosis is a rare, inherited neurodegenerative disorder caused by mutations in the GLB1 gene, which encodes the lysosomal hydrolase acid β-galactosidase (β-gal). β-gal deficiency leads to toxic accumulation of GM1 ganglioside, predominantly in the central nervous system (CNS), resulting in progressive neurodegeneration. LYS-GM101 is an AAVrh.10-based gene therapy vector carrying the human GLB1 cDNA. The efficacy of intra-cerebrospinal fluid injection of LYS-GM101 analogs was demonstrated in GM1 mouse and cat models with widespread diffusion of β-gal and correction of GM1 ganglioside accumulation in the CNS without observable adverse effects. Clinical dose selection was performed, based on a good-laboratory-practice study, in nonhuman primates (NHPs) using the clinical LYS-GM101 vector. A broadly distributed increase of β-gal activity was observed in NHP brain 3 months after intra-cisterna magna injection of LYS-GM101 at 1.0 × 1012 vg/mL CSF and 4.0 × 1012 vg/mL CSF, with 20% and 60% increases compared with vehicle-treated animals, respectively. Histopathologic examination revealed asymptomatic adverse changes in the sensory pathways of the spinal cord and dorsal root ganglia in both sexes and at both doses. Taken as a whole, these pre-clinical data support the initiation of a clinical study with LYS-GM101 for the treatment of GM1 gangliosidosis.
Collapse
Affiliation(s)
- Michaël Hocquemiller
- Lysogene, 18–20 rue Jacques Dulud, 92200 Neuilly-sur-Seine, France,Corresponding author Michaël Hocquemiller, Lysogene, 18–20 rue Jacques Dulud, 92200 Neuilly-sur-Seine, France.
| | - Laura Giersch
- Lysogene, 18–20 rue Jacques Dulud, 92200 Neuilly-sur-Seine, France
| | - Xin Mei
- Lysogene, 18–20 rue Jacques Dulud, 92200 Neuilly-sur-Seine, France
| | - Amanda L. Gross
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL 36849, USA
| | - Ashley N. Randle
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL 36849, USA
| | - Heather L. Gray-Edwards
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL 36849, USA
| | - Judith A. Hudson
- Department of Clinical Sciences, Auburn University College of Veterinary Medicine, Auburn, AL 36849, USA
| | - Sophia Todeasa
- Department of Neurology, Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lorelei Stoica
- Department of Neurology, Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Douglas R. Martin
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL 36849, USA,Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL 36849, USA
| | - Miguel Sena-Esteves
- Department of Neurology, Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Karen Aiach
- Lysogene, 18–20 rue Jacques Dulud, 92200 Neuilly-sur-Seine, France
| | - Ralph Laufer
- Lysogene, 18–20 rue Jacques Dulud, 92200 Neuilly-sur-Seine, France,Corresponding author Ralph Laufer, Lysogene, 18–20 rue Jacques Dulud, 92200 Neuilly-sur-Seine, France.
| |
Collapse
|
13
|
Yamaji-Hasegawa A, Murate M, Inaba T, Dohmae N, Sato M, Fujimori F, Sako Y, Greimel P, Kobayashi T. A novel sterol-binding protein reveals heterogeneous cholesterol distribution in neurite outgrowth and in late endosomes/lysosomes. Cell Mol Life Sci 2022; 79:324. [PMID: 35644822 PMCID: PMC11072113 DOI: 10.1007/s00018-022-04339-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
Abstract
We identified a mushroom-derived protein, maistero-2 that specifically binds 3-hydroxy sterol including cholesterol (Chol). Maistero-2 bound lipid mixture in Chol-dependent manner with a binding threshold of around 30%. Changing lipid composition did not significantly affect the threshold concentration. EGFP-maistero-2 labeled cell surface and intracellular organelle Chol with higher sensitivity than that of well-established Chol probe, D4 fragment of perfringolysin O. EGFP-maistero-2 revealed increase of cell surface Chol during neurite outgrowth and heterogeneous Chol distribution between CD63-positive and LAMP1-positive late endosomes/lysosomes. The absence of strictly conserved Thr-Leu pair present in Chol-dependent cytolysins suggests a distinct Chol-binding mechanism for maistero-2.
Collapse
Affiliation(s)
| | - Motohide Murate
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- UMR 7021, CNRS, Université de Strasbourg, 67401, Illkirch, France
| | - Takehiko Inaba
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN CSRS, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Masayuki Sato
- Yukiguni Maitake Co, Ltd. Yokawa 89, Minamiuonuma, Niigata, 949-6695, Japan
| | - Fumihiro Fujimori
- Laboratory of Biological Science and Technology, Tokyo Kasei University, 1-18-1 Kaga, Itabashi, Tokyo, 173-8062, Japan
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Peter Greimel
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- UMR 7021, CNRS, Université de Strasbourg, 67401, Illkirch, France.
| |
Collapse
|
14
|
Goicoechea L, Arenas F, Castro F, Nuñez S, Torres S, Garcia-Ruiz C, Fernandez-Checa JC. GST-Perfringolysin O production for the localization and quantification of membrane cholesterol in human and mouse brain and liver. STAR Protoc 2022; 3:101068. [PMID: 35024626 PMCID: PMC8728529 DOI: 10.1016/j.xpro.2021.101068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Abnormal cholesterol metabolism is linked to many neurodegenerative disorders. Here, we present a protocol for the production of a recombinant protein consisting of a Glutathione-S-Transferase tag fused with the Perfringolysin O (PFO). The GST-PFO tag enables analysis of the localization of cholesterol in subcellular membranes of human and mice brain and liver tissues. We have used this approach for samples from Niemann-Pick type C disease and non-alcoholic steatohepatitis models. The construct may also have applications for the diagnosis of cholesterol-accumulating disorders. For complete details on the use and execution of this protocol, please refer to Kwiatkowska et al. (2014).
Collapse
Affiliation(s)
- Leire Goicoechea
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Fabian Arenas
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fernanda Castro
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Susana Nuñez
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
- Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - José C. Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
- Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
15
|
Nieto-Garai JA, Lorizate M, Contreras FX. Shedding light on membrane rafts structure and dynamics in living cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2022; 1864:183813. [PMID: 34748743 DOI: 10.1016/j.bbamem.2021.183813] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022]
Abstract
Cellular membranes are fundamental building blocks regulating an extensive repertoire of biological functions. These structures contain lipids and membrane proteins that are known to laterally self-aggregate in the plane of the membrane, forming defined membrane nanoscale domains essential for protein activity. Membrane rafts are described as heterogeneous, dynamic, and short-lived cholesterol- and sphingolipid-enriched membrane nanodomains (10-200 nm) induced by lipid-protein and lipid-lipid interactions. Those membrane nanodomains have been extensively characterized using model membranes and in silico methods. However, despite the development of advanced fluorescence microscopy techniques, undoubted nanoscale visualization by imaging techniques of membrane rafts in the membrane of unperturbed living cells is still uncompleted, increasing the skepticism about their existence. Here, we broadly review recent biochemical and microscopy techniques used to investigate membrane rafts in living cells and we enumerate persistent open questions to answer before unlocking the mystery of membrane rafts in living cells.
Collapse
Affiliation(s)
- Jon Ander Nieto-Garai
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Bilbao, Spain.
| | - Maier Lorizate
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Bilbao, Spain; Instituto Biofisika (UPV/EHU, CSIC), Barrio Sarriena s/n, 48940 Bilbao, Spain
| | - F-Xabier Contreras
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Bilbao, Spain; Instituto Biofisika (UPV/EHU, CSIC), Barrio Sarriena s/n, 48940 Bilbao, Spain; IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain.
| |
Collapse
|
16
|
Barrantes FJ. Fluorescence sensors for imaging membrane lipid domains and cholesterol. CURRENT TOPICS IN MEMBRANES 2021; 88:257-314. [PMID: 34862029 DOI: 10.1016/bs.ctm.2021.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lipid membrane domains are supramolecular lateral heterogeneities of biological membranes. Of nanoscopic dimensions, they constitute specialized hubs used by the cell as transient signaling platforms for a great variety of biologically important mechanisms. Their property to form and dissolve in the bulk lipid bilayer endow them with the ability to engage in highly dynamic processes, and temporarily recruit subpopulations of membrane proteins in reduced nanometric compartments that can coalesce to form larger mesoscale assemblies. Cholesterol is an essential component of these lipid domains; its unique molecular structure is suitable for interacting intricately with crevices and cavities of transmembrane protein surfaces through its rough β face while "talking" to fatty acid acyl chains of glycerophospholipids and sphingolipids via its smooth α face. Progress in the field of membrane domains has been closely associated with innovative improvements in fluorescence microscopy and new fluorescence sensors. These advances enabled the exploration of the biophysical properties of lipids and their supramolecular platforms. Here I review the rationale behind the use of biosensors over the last few decades and their contributions towards elucidation of the in-plane and transbilayer topography of cholesterol-enriched lipid domains and their molecular constituents. The challenges introduced by super-resolution optical microscopy are discussed, as well as possible scenarios for future developments in the field, including virtual ("no staining") staining.
Collapse
Affiliation(s)
- Francisco J Barrantes
- Biomedical Research Institute (BIOMED), Catholic University of Argentina (UCA)-National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
17
|
Jurášek M, Valečka J, Novotný I, Kejík Z, Fähnrich J, Marešová A, Tauchen J, Bartůněk P, Dolenský B, Jakubek M, Drašar PB, Králová J. Synthesis and biological evaluation of cationic TopFluor cholesterol analogues. Bioorg Chem 2021; 117:105410. [PMID: 34700109 DOI: 10.1016/j.bioorg.2021.105410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/25/2021] [Accepted: 10/03/2021] [Indexed: 12/17/2022]
Abstract
Cholesterol is not only a major component of the cell membrane, but also plays an important role in a wide range of biological processes and pathologies. It is therefore crucial to develop appropriate tools for visualizing intracellular cholesterol transport. Here, we describe new cationic analogues of BODIPY-Cholesterol (TopFluor-Cholesterol, TF-Chol), which combine a positive charge on the sterol side chain and a BODIPY group connected via a C-4 linker. In contrast to TF-Chol, the new analogues TF-1 and TF-3 possessing acetyl groups on the A ring (C-3 position on steroid) internalized much faster and displayed slightly different levels of intracellular localization. Their applicability for cholesterol monitoring was indicated by the fact that they strongly label compartments with accumulated cholesterol in cells carrying a mutation of the Niemann-Pick disease-associated cholesterol transporter, NPC1.
Collapse
Affiliation(s)
- Michal Jurášek
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jan Valečka
- Light microscopy core facility, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Ivan Novotný
- Light microscopy core facility, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Zdeněk Kejík
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jan Fähnrich
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Anna Marešová
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jan Tauchen
- Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague 6, Czech Republic
| | - Petr Bartůněk
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Bohumil Dolenský
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Milan Jakubek
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Pavel B Drašar
- University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jarmila Králová
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vídeňská 1083, 142 20 Prague 4, Czech Republic.
| |
Collapse
|
18
|
Schoop V, Martello A, Eden ER, Höglinger D. Cellular cholesterol and how to find it. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158989. [PMID: 34118431 DOI: 10.1016/j.bbalip.2021.158989] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 01/06/2023]
Abstract
Cholesterol is an essential component of eukaryotic cellular membranes. Information about its subcellular localization and transport pathways inside cells are key for the understanding and treatment of cholesterol-related diseases. In this review we give an overview over the most commonly used methods that contributed to our current understanding of subcellular cholesterol localization and transport routes. First, we discuss methods that provide insights into cholesterol metabolism based on readouts of downstream effects such as esterification. Subsequently, we focus on the use of cholesterol-binding molecules as probes that facilitate visualization and quantification of sterols inside of cells. Finally, we explore different analogues of cholesterol which, when taken up by living cells, are integrated and transported in a similar fashion as endogenous sterols. Taken together, we highlight the challenges and advantages of each method such that researchers studying aspects of cholesterol transport may choose the most pertinent approach for their problem.
Collapse
Affiliation(s)
- Valentin Schoop
- Heidelberg University Biochemistry Center (BZH), 69120 Heidelberg, Germany
| | - Andrea Martello
- University College London (UCL), Institute of Ophthalmology, EC1V 9EL London, United Kingdom
| | - Emily R Eden
- University College London (UCL), Institute of Ophthalmology, EC1V 9EL London, United Kingdom
| | - Doris Höglinger
- Heidelberg University Biochemistry Center (BZH), 69120 Heidelberg, Germany.
| |
Collapse
|
19
|
Visualisation of cholesterol and ganglioside GM1 in zebrafish models of Niemann-Pick type C disease and Smith-Lemli-Opitz syndrome using light sheet microscopy. Histochem Cell Biol 2020; 154:565-578. [PMID: 33079236 PMCID: PMC7609433 DOI: 10.1007/s00418-020-01925-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 12/20/2022]
Abstract
Lysosomal storage diseases are the most common cause of neurodegeneration in children. They are characterised at the cellular level by the accumulation of storage material within lysosomes. There are very limited therapeutic options, and the search for novel therapies has been hampered as few good small animal models are available. Here, we describe the use of light sheet microscopy to assess lipid storage in drug and morpholino induced zebrafish models of two diseases of cholesterol homeostasis with lysosomal dysfunction: First, Niemann–Pick type C disease (NPC), caused by mutations in the lysosomal transmembrane protein NPC1, characterised by intralysosomal accumulation of cholesterol and several other lipids. Second, Smith–Lemli–Opitz syndrome (SLOS), caused by mutations in 7-dehydrocholesterol reductase, which catalyses the last step of cholesterol biosynthesis and is characterised by intralysosomal accumulation of dietary cholesterol. This is the first description of a zebrafish SLOS model. We find that zebrafish accurately model lysosomal storage and disease-specific phenotypes in both diseases. Increased cholesterol and ganglioside GM1 were observed in sections taken from NPC model fish, and decreased cholesterol in SLOS model fish, but these are of limited value as resolution is poor, and accurate anatomical comparisons difficult. Using light sheet microscopy, we were able to observe lipid changes in much greater detail and identified an unexpected accumulation of ganglioside GM1 in SLOS model fish. Our data demonstrate, for the first time in zebrafish, the immense potential that light sheet microscopy has in aiding the resolution of studies involving lysosomal and lipid disorders.
Collapse
|
20
|
Salinas ML, Fuentes NR, Choate R, Wright RC, McMurray DN, Chapkin RS. AdipoRon Attenuates Wnt Signaling by Reducing Cholesterol-Dependent Plasma Membrane Rigidity. Biophys J 2020; 118:885-897. [PMID: 31630812 PMCID: PMC7036725 DOI: 10.1016/j.bpj.2019.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/28/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023] Open
Abstract
The increasing prevalence of adult and adolescent obesity and its associated risk of colorectal cancer reinforces the urgent need to elucidate the underlying mechanisms contributing to the promotion of colon cancer in obese individuals. Adiponectin is an adipose tissue-derived adipokine, whose levels are reduced during obesity. Both epidemiological and preclinical data indicate that adiponectin suppresses colon tumorigenesis. We have previously demonstrated that both adiponectin and AdipoRon, a small-molecule adiponectin receptor agonist, suppress colon cancer risk in part by reducing the number of Lgr5+ stem cells in mouse colonic organoids. However, the mechanism by which the adiponectin signaling pathway attenuates colon cancer risk remains to be addressed. Here, we have hypothesized that adiponectin signaling supports colonic stem cell maintenance through modulation of the biophysical properties of the plasma membrane (PM). Specifically, we investigated the effects of adiponectin receptor activation by AdipoRon on the biophysical perturbations linked to the attenuation of Wnt-driven signaling and cell proliferation as determined by LEF luciferase reporter assay and colonic organoid proliferation, respectively. Using physicochemical sensitive dyes, Di-4-ANEPPDHQ and C-laurdan, we demonstrated that AdipoRon decreased the rigidity of the colonic cell PM. The decrease in membrane rigidity was associated with a reduction in PM free cholesterol levels and the intracellular accumulation of free cholesterol in lysosomes. These results suggest that adiponectin signaling plays a role in modulating cellular cholesterol homeostasis, PM biophysical properties, and Wnt-driven signaling. These findings are noteworthy because they may in part explain how obesity drives colon cancer progression.
Collapse
Affiliation(s)
- Michael L Salinas
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas; Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Natividad R Fuentes
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas; Department of Nutrition and Food Science, Texas A&M University, College Station, Texas; Interdisciplinary Faculty of Toxicology Program, Texas A&M University, College Station, Texas
| | - Rachel Choate
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Rachel C Wright
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas; Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - David N McMurray
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas; Department of Nutrition and Food Science, Texas A&M University, College Station, Texas; Interdisciplinary Faculty of Toxicology Program, Texas A&M University, College Station, Texas; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas; Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas; Center for Environmental Health Research, Texas A&M University, College Station, Texas.
| |
Collapse
|
21
|
Králová J, Král V. Fluorescent Probes for Monitoring Cholesterol Trafficking in Cells. Folia Biol (Praha) 2019; 65:1-10. [PMID: 31171077 DOI: 10.14712/fb2019065010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Cellular cholesterol plays fundamental and diverse roles in many biological processes and affects the pathology of various diseases. Comprehensive and detailed understanding of the cellular functions and characteristics of cholesterol requires visualization of its subcellular distribution, which can be achieved by fluorescence microscopy. Many attempts have been made to develop fluorescent cholesterol reporters, but so far, none of them seems to be ideal for studying all aspects of cholesterol management. To meet the requirements for the right probe remains a great challenge, and progress in this field continues. The main objective of this review is to not only present the current state of the art, but also critically evaluate the applicability of individual probes and for what purpose they can be used to obtain relevant data. Hence, the data obtained with different probes might provide complementary information to build an integrated picture about the cellular cholesterol.
Collapse
Affiliation(s)
- J Králová
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - V Král
- University of Chemistry and Technology, Prague, Czech Republic
| |
Collapse
|
22
|
Heterocyclic sterol probes for live monitoring of sterol trafficking and lysosomal storage disorders. Sci Rep 2018; 8:14428. [PMID: 30258093 PMCID: PMC6158244 DOI: 10.1038/s41598-018-32776-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 09/14/2018] [Indexed: 12/13/2022] Open
Abstract
The monitoring of intracellular cholesterol homeostasis and trafficking is of great importance because their imbalance leads to many pathologies. Reliable tools for cholesterol detection are in demand. This study presents the design and synthesis of fluorescent probes for cholesterol recognition and demonstrates their selectivity by a variety of methods. The construction of dedicated library of 14 probes was based on heterocyclic (pyridine)-sterol derivatives with various attached fluorophores. The most promising probe, a P1-BODIPY conjugate FP-5, was analysed in detail and showed an intensive labelling of cellular membranes followed by intracellular redistribution into various cholesterol rich organelles and vesicles. FP-5 displayed a stronger signal, with faster kinetics, than the commercial TF-Chol probe. In addition, cells with pharmacologically disrupted cholesterol transport, or with a genetic mutation of cholesterol transporting protein NPC1, exhibited strong and fast FP-5 signal in the endo/lysosomal compartment, co-localizing with filipin staining of cholesterol. Hence, FP-5 has high potential as a new probe for monitoring cholesterol trafficking and its disorders.
Collapse
|
23
|
Jung SH, Lee M, Park HA, Lee HC, Kang D, Hwang HJ, Park C, Yu DM, Jung YR, Hong MN, Kim YN, Park HJ, Ko YG, Lee JS. Integrin α6β4-Src-AKT signaling induces cellular senescence by counteracting apoptosis in irradiated tumor cells and tissues. Cell Death Differ 2018; 26:245-259. [PMID: 29786073 DOI: 10.1038/s41418-018-0114-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 03/07/2018] [Accepted: 03/22/2018] [Indexed: 02/06/2023] Open
Abstract
Cellular senescence refers to an irreversible growth arrest that is triggered by various intrinsic and extrinsic stresses. Many recent studies have demonstrated that cellular senescence plays a crucial role in the regression of tumors exposed to ionizing radiation (IR), but the underlying mechanism remains unknown. Here we show that the activation of integrin β4 is essential for IR-induced cellular senescence. IR treatment results in the phosphorylation of integrin β4 at tyrosine residue 1510, leading to activation of the integrin α6β4-Src-AKT signaling pathway. We further reveal that the IR-induced phosphorylation of integrin β4 is regulated by the cholesterol content and membrane fluidity. We also find that IR-induced p53-caspase signaling is independent of integrin α6β4-Src-AKT signaling. Finally, we show that siRNA- or inhibitor-mediated blockade of integrin α6β4-Src-AKT signaling switches the post-irradiation fate from senescence to apoptosis, under p53 activated condition, in both cancer cells and tumor tissues of xenograft mice. On the basis of our finding that, integrin α6β4 is specifically activated and acts primarily to induce premature senescence in irradiated cancer cells, we propose that this integrin may be a valuable target and biomarker for radiotherapy.
Collapse
Affiliation(s)
- Seung Hee Jung
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Minyoung Lee
- Radiation Non-clinical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hyun A Park
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Hyung Chul Lee
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Donghee Kang
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Hyun Jung Hwang
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Chanho Park
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Dong-Min Yu
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Yu Ri Jung
- Radiation Non-clinical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Mi-Na Hong
- Radiation Non-clinical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Yong-Nyun Kim
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, Korea
| | - Heon Joo Park
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea.,Department of Microbiology, Inha University College of Medicine, Incheon, Korea
| | - Young-Gyu Ko
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Jae-Seon Lee
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea. .,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea.
| |
Collapse
|
24
|
Bader CA, Carter EA, Safitri A, Simpson PV, Wright P, Stagni S, Massi M, Lay PA, Brooks DA, Plush SE. Unprecedented staining of polar lipids by a luminescent rhenium complex revealed by FTIR microspectroscopy in adipocytes. MOLECULAR BIOSYSTEMS 2017; 12:2064-8. [PMID: 27170554 DOI: 10.1039/c6mb00242k] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Fourier transform infrared (FTIR) microspectroscopy and confocal imaging have been used to demonstrate that the neutral rhenium(i) tricarbonyl 1,10-phenanthroline complex bound to 4-cyanophenyltetrazolate as the ancillary ligand is able to localise in regions with high concentrations of polar lipids such as phosphatidylethanolamine (PE), sphingomyelin, sphingosphine and lysophosphatidic acid (LPA) in mammalian adipocytes.
Collapse
Affiliation(s)
- C A Bader
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences/Sansom Institute for Health Research, University of South Australia, Adelaide, Australia.
| | - E A Carter
- Vibrational Spectroscopy Core Facility and School of Chemistry, The University of Sydney, Sydney, Australia
| | - A Safitri
- Vibrational Spectroscopy Core Facility and School of Chemistry, The University of Sydney, Sydney, Australia
| | - P V Simpson
- School of Chemistry, Curtin University, Perth, Australia
| | - P Wright
- School of Chemistry, Curtin University, Perth, Australia
| | - S Stagni
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Bologna, Italy
| | - M Massi
- School of Chemistry, Curtin University, Perth, Australia
| | - P A Lay
- Vibrational Spectroscopy Core Facility and School of Chemistry, The University of Sydney, Sydney, Australia
| | - D A Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences/Sansom Institute for Health Research, University of South Australia, Adelaide, Australia.
| | - S E Plush
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences/Sansom Institute for Health Research, University of South Australia, Adelaide, Australia.
| |
Collapse
|
25
|
Fujimoto T, Parmryd I. Interleaflet Coupling, Pinning, and Leaflet Asymmetry-Major Players in Plasma Membrane Nanodomain Formation. Front Cell Dev Biol 2017; 4:155. [PMID: 28119914 PMCID: PMC5222840 DOI: 10.3389/fcell.2016.00155] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 12/27/2016] [Indexed: 01/26/2023] Open
Abstract
The plasma membrane has a highly asymmetric distribution of lipids and contains dynamic nanodomains many of which are liquid entities surrounded by a second, slightly different, liquid environment. Contributing to the dynamics is a continuous repartitioning of components between the two types of liquids and transient links between lipids and proteins, both to extracellular matrix and cytoplasmic components, that temporarily pin membrane constituents. This make plasma membrane nanodomains exceptionally challenging to study and much of what is known about membrane domains has been deduced from studies on model membranes at equilibrium. However, living cells are by definition not at equilibrium and lipids are distributed asymmetrically with inositol phospholipids, phosphatidylethanolamines and phosphatidylserines confined mostly to the inner leaflet and glyco- and sphingolipids to the outer leaflet. Moreover, each phospholipid group encompasses a wealth of species with different acyl chain combinations whose lateral distribution is heterogeneous. It is becoming increasingly clear that asymmetry and pinning play important roles in plasma membrane nanodomain formation and coupling between the two lipid monolayers. How asymmetry, pinning, and interdigitation contribute to the plasma membrane organization is only beginning to be unraveled and here we discuss their roles and interdependence.
Collapse
Affiliation(s)
- Toyoshi Fujimoto
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Ingela Parmryd
- Science for Life Laboratory, Medical Cell Biology, Uppsala University Uppsala, Sweden
| |
Collapse
|
26
|
Duval R, Duplais C. Fluorescent natural products as probes and tracers in biology. Nat Prod Rep 2017; 34:161-193. [DOI: 10.1039/c6np00111d] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fluorescence is a remarkable property of many natural products in addition to their medicinal and biological value. Herein, we provide a review of these peculiar secondary metabolites to stimulate prospecting of them as original fluorescent tracers, endowed with unique photophysical properties and with applications in most fields of biology.
Collapse
Affiliation(s)
- Romain Duval
- IRD
- UMR 216 IRD MERIT (Mère et Enfant face aux Infections Tropicales)
- Université Paris-Descartes
- 75006 Paris
- France
| | - Christophe Duplais
- CNRS
- UMR 8172 EcoFoG (Ecologie des Forêts de Guyane)
- AgroParisTech
- Cirad
- INRA
| |
Collapse
|
27
|
Van Blerkom J, Zimmermann S. Ganglioside-enriched microdomains define an oolemma that is functionally polarized with respect to fertilizability in the mouse. Reprod Biomed Online 2016; 33:458-475. [DOI: 10.1016/j.rbmo.2016.06.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 10/21/2022]
|
28
|
|
29
|
Bader CA, Shandala T, Carter EA, Ivask A, Guinan T, Hickey SM, Werrett MV, Wright PJ, Simpson PV, Stagni S, Voelcker NH, Lay PA, Massi M, Plush SE, Brooks DA. A Molecular Probe for the Detection of Polar Lipids in Live Cells. PLoS One 2016; 11:e0161557. [PMID: 27551717 PMCID: PMC4994960 DOI: 10.1371/journal.pone.0161557] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 08/07/2016] [Indexed: 01/08/2023] Open
Abstract
Lipids have an important role in many aspects of cell biology, including membrane architecture/compartment formation, intracellular traffic, signalling, hormone regulation, inflammation, energy storage and metabolism. Lipid biology is therefore integrally involved in major human diseases, including metabolic disorders, neurodegenerative diseases, obesity, heart disease, immune disorders and cancers, which commonly display altered lipid transport and metabolism. However, the investigation of these important cellular processes has been limited by the availability of specific tools to visualise lipids in live cells. Here we describe the potential for ReZolve-L1™ to localise to intracellular compartments containing polar lipids, such as for example sphingomyelin and phosphatidylethanolamine. In live Drosophila fat body tissue from third instar larvae, ReZolve-L1™ interacted mainly with lipid droplets, including the core region of these organelles. The presence of polar lipids in the core of these lipid droplets was confirmed by Raman mapping and while this was consistent with the distribution of ReZolve-L1™ it did not exclude that the molecular probe might be detecting other lipid species. In response to complete starvation conditions, ReZolve-L1™ was detected mainly in Atg8-GFP autophagic compartments, and showed reduced staining in the lipid droplets of fat body cells. The induction of autophagy by Tor inhibition also increased ReZolve-L1™ detection in autophagic compartments, whereas Atg9 knock down impaired autophagosome formation and altered the distribution of ReZolve-L1™. Finally, during Drosophila metamorphosis fat body tissues showed increased ReZolve-L1™ staining in autophagic compartments at two hours post puparium formation, when compared to earlier developmental time points. We concluded that ReZolve-L1™ is a new live cell imaging tool, which can be used as an imaging reagent for the detection of polar lipids in different intracellular compartments.
Collapse
Affiliation(s)
- Christie A. Bader
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Tetyana Shandala
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Elizabeth A. Carter
- Vibrational Spectroscopy Core Facility, The University of Sydney, Sydney, New South Wales, Australia
| | - Angela Ivask
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Taryn Guinan
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Shane M. Hickey
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Melissa V. Werrett
- Department of Chemistry and Nanochemistry Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Phillip J. Wright
- Department of Chemistry and Nanochemistry Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Peter V. Simpson
- Department of Chemistry and Nanochemistry Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Stefano Stagni
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, Bologna, Italy
| | - Nicolas H. Voelcker
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Peter A. Lay
- Vibrational Spectroscopy Core Facility, The University of Sydney, Sydney, New South Wales, Australia
| | - Massimiliano Massi
- Department of Chemistry and Nanochemistry Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Sally E. Plush
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Douglas A. Brooks
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
- * E-mail:
| |
Collapse
|
30
|
Fareez IM, Lim SM, Lim FT, Mishra RK, Ramasamy K. Microencapsulation of Lactobacillus
SP. Using Chitosan-Alginate-Xanthan Gum-β-Cyclodextrin and Characterization of its Cholesterol Reducing Potential and Resistance Against pH, Temperature and Storage. J FOOD PROCESS ENG 2016. [DOI: 10.1111/jfpe.12458] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Ismail M. Fareez
- Faculty of Pharmacy; University Teknologi MARA (UiTM); Bandar Puncak Alam Selangor Darul Ehsan 42300 Malaysia
- Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM); Shah Alam Selangor Darul Ehsan 40450 Malaysia
| | - Siong Meng Lim
- Faculty of Pharmacy; University Teknologi MARA (UiTM); Bandar Puncak Alam Selangor Darul Ehsan 42300 Malaysia
- Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM); Shah Alam Selangor Darul Ehsan 40450 Malaysia
| | - Fei Tieng Lim
- Faculty of Pharmacy; University Teknologi MARA (UiTM); Bandar Puncak Alam Selangor Darul Ehsan 42300 Malaysia
- Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM); Shah Alam Selangor Darul Ehsan 40450 Malaysia
| | - Rakesh K. Mishra
- Faculty of Pharmacy; University Teknologi MARA (UiTM); Bandar Puncak Alam Selangor Darul Ehsan 42300 Malaysia
- Brain Degeneration and Therapeutics Group; Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM); Shah Alam Selangor Darul Ehsan 40450 Malaysia
| | - Kalavathy Ramasamy
- Faculty of Pharmacy; University Teknologi MARA (UiTM); Bandar Puncak Alam Selangor Darul Ehsan 42300 Malaysia
- Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM); Shah Alam Selangor Darul Ehsan 40450 Malaysia
| |
Collapse
|
31
|
Solanko KA, Modzel M, Solanko LM, Wüstner D. Fluorescent Sterols and Cholesteryl Esters as Probes for Intracellular Cholesterol Transport. Lipid Insights 2016; 8:95-114. [PMID: 27330304 PMCID: PMC4902042 DOI: 10.4137/lpi.s31617] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 12/20/2022] Open
Abstract
Cholesterol transport between cellular organelles comprised vesicular trafficking and nonvesicular exchange; these processes are often studied by quantitative fluorescence microscopy. A major challenge for using this approach is producing analogs of cholesterol with suitable brightness and structural and chemical properties comparable with those of cholesterol. This review surveys currently used fluorescent sterols with respect to their behavior in model membranes, their photophysical properties, as well as their transport and metabolism in cells. In the first part, several intrinsically fluorescent sterols, such as dehydroergosterol or cholestatrienol, are discussed. These polyene sterols (P-sterols) contain three conjugated double bonds in the steroid ring system, giving them slight fluorescence in ultraviolet light. We discuss the properties of P-sterols relative to cholesterol, outline their chemical synthesis, and explain how to image them in living cells and organisms. In particular, we show that P-sterol esters inserted into low-density lipoprotein can be tracked in the fibroblasts of Niemann–Pick disease using high-resolution deconvolution microscopy. We also describe fluorophore-tagged cholesterol probes, such as BODIPY-, NBD-, Dansyl-, or Pyrene-tagged cholesterol, and eventual esters of these analogs. Finally, we survey the latest developments in the synthesis and use of alkyne cholesterol analogs to be labeled with fluorophores by click chemistry and discuss the potential of all approaches for future applications.
Collapse
Affiliation(s)
- Katarzyna A Solanko
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Maciej Modzel
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Lukasz M Solanko
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| |
Collapse
|
32
|
Kishimoto T, Ishitsuka R, Kobayashi T. Detectors for evaluating the cellular landscape of sphingomyelin- and cholesterol-rich membrane domains. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:812-829. [PMID: 26993577 DOI: 10.1016/j.bbalip.2016.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/09/2016] [Accepted: 03/09/2016] [Indexed: 12/11/2022]
Abstract
Although sphingomyelin and cholesterol are major lipids of mammalian cells, the detailed distribution of these lipids in cellular membranes remains still obscure. However, the recent development of protein probes that specifically bind sphingomyelin and/or cholesterol provides new information about the landscape of the lipid domains that are enriched with sphingomyelin or cholesterol or both. Here, we critically summarize the tools to study distribution and dynamics of sphingomyelin and cholesterol. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
Affiliation(s)
| | - Reiko Ishitsuka
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan; INSERM U1060, Université Lyon 1, Villeurbanne 69621, France.
| |
Collapse
|
33
|
Fujimoto T, Parmryd I. Interleaflet Coupling, Pinning, and Leaflet Asymmetry-Major Players in Plasma Membrane Nanodomain Formation. Front Cell Dev Biol 2016. [PMID: 28119914 DOI: 10.3389/fcell.2016.0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
The plasma membrane has a highly asymmetric distribution of lipids and contains dynamic nanodomains many of which are liquid entities surrounded by a second, slightly different, liquid environment. Contributing to the dynamics is a continuous repartitioning of components between the two types of liquids and transient links between lipids and proteins, both to extracellular matrix and cytoplasmic components, that temporarily pin membrane constituents. This make plasma membrane nanodomains exceptionally challenging to study and much of what is known about membrane domains has been deduced from studies on model membranes at equilibrium. However, living cells are by definition not at equilibrium and lipids are distributed asymmetrically with inositol phospholipids, phosphatidylethanolamines and phosphatidylserines confined mostly to the inner leaflet and glyco- and sphingolipids to the outer leaflet. Moreover, each phospholipid group encompasses a wealth of species with different acyl chain combinations whose lateral distribution is heterogeneous. It is becoming increasingly clear that asymmetry and pinning play important roles in plasma membrane nanodomain formation and coupling between the two lipid monolayers. How asymmetry, pinning, and interdigitation contribute to the plasma membrane organization is only beginning to be unraveled and here we discuss their roles and interdependence.
Collapse
Affiliation(s)
- Toyoshi Fujimoto
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Ingela Parmryd
- Science for Life Laboratory, Medical Cell Biology, Uppsala University Uppsala, Sweden
| |
Collapse
|
34
|
Sezgin E, Can FB, Schneider F, Clausen MP, Galiani S, Stanly TA, Waithe D, Colaco A, Honigmann A, Wüstner D, Platt F, Eggeling C. A comparative study on fluorescent cholesterol analogs as versatile cellular reporters. J Lipid Res 2015; 57:299-309. [PMID: 26701325 DOI: 10.1194/jlr.m065326] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Indexed: 12/12/2022] Open
Abstract
Cholesterol (Chol) is a crucial component of cellular membranes, but knowledge of its intracellular dynamics is scarce. Thus, it is of utmost interest to develop tools for visualization of Chol organization and dynamics in cells and tissues. For this purpose, many studies make use of fluorescently labeled Chol analogs. Unfortunately, the introduction of the label may influence the characteristics of the analog, such as its localization, interaction, and trafficking in cells; hence, it is important to get knowledge of such bias. In this report, we compared different fluorescent lipid analogs for their performance in cellular assays: 1) plasma membrane incorporation, specifically the preference for more ordered membrane environments in phase-separated giant unilamellar vesicles and giant plasma membrane vesicles; 2) cellular trafficking, specifically subcellular localization in Niemann-Pick type C disease cells; and 3) applicability in fluorescence correlation spectroscopy (FCS)-based and super-resolution stimulated emission depletion-FCS-based measurements of membrane diffusion dynamics. The analogs exhibited strong differences, with some indicating positive performance in the membrane-based experiments and others in the intracellular trafficking assay. However, none showed positive performance in all assays. Our results constitute a concise guide for the careful use of fluorescent Chol analogs in visualizing cellular Chol dynamics.
Collapse
Affiliation(s)
- Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX39DS Oxford, United Kingdom
| | - Fatma Betul Can
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX39DS Oxford, United Kingdom
| | - Falk Schneider
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX39DS Oxford, United Kingdom
| | - Mathias P Clausen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX39DS Oxford, United Kingdom MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, 5230 Odense M, Denmark
| | - Silvia Galiani
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX39DS Oxford, United Kingdom
| | - Tess A Stanly
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX39DS Oxford, United Kingdom
| | - Dominic Waithe
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX39DS Oxford, United Kingdom
| | - Alexandria Colaco
- Department of Pharmacology, University of Oxford, OX13QT Oxford, United Kingdom
| | - Alf Honigmann
- Max Planck Institute of Cell Biology and Genetics, 01307 Dresden, Germany
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Frances Platt
- Department of Pharmacology, University of Oxford, OX13QT Oxford, United Kingdom
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX39DS Oxford, United Kingdom
| |
Collapse
|
35
|
Weismann CM, Ferreira J, Keeler AM, Su Q, Qui L, Shaffer SA, Xu Z, Gao G, Sena-Esteves M. Systemic AAV9 gene transfer in adult GM1 gangliosidosis mice reduces lysosomal storage in CNS and extends lifespan. Hum Mol Genet 2015; 24:4353-64. [PMID: 25964428 DOI: 10.1093/hmg/ddv168] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 05/05/2015] [Indexed: 02/06/2023] Open
Abstract
GM1 gangliosidosis (GM1) is an autosomal recessive lysosomal storage disease where GLB1 gene mutations result in a reduction or absence of lysosomal acid β-galactosidase (βgal) activity. βgal deficiency leads to accumulation of GM1-ganglioside in the central nervous system (CNS). GM1 is characterized by progressive neurological decline resulting in generalized paralysis, extreme emaciation and death. In this study, we assessed the therapeutic efficacy of an adeno-associated virus (AAV) 9-mβgal vector infused systemically in adult GM1 mice (βGal(-/-)) at 1 × 10(11) or 3 × 10(11) vector genomes (vg). Biochemical analysis of AAV9-treated GM1 mice showed high βGal activity in liver and serum. Moderate βGal levels throughout CNS resulted in a 36-76% reduction in GM1-ganglioside content in the brain and 75-86% in the spinal cord. Histological analyses of the CNS of animals treated with 3 × 10(11) vg dose revealed increased presence of βgal and clearance of lysosomal storage throughout cortex, hippocampus, brainstem and spinal cord. Storage reduction in these regions was accompanied by a marked decrease in astrogliosis. AAV9 treatment resulted in improved performance in multiple tests of motor function and behavior. Also the majority of GM1 mice in the 3 × 10(11) vg cohort retained ambulation and rearing despite reaching the humane endpoint due to weight loss. Importantly, the median survival of AAV9 treatment groups (316-576 days) was significantly increased over controls (250-264 days). This study shows that moderate widespread expression of βgal in the CNS of GM1 gangliosidosis mice is sufficient to achieve significant biochemical impact with phenotypic amelioration and extension in lifespan.
Collapse
Affiliation(s)
| | | | | | | | | | - Scott A Shaffer
- Biochemistry and Molecular Pharmacology and Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
36
|
Vovk I, Gerčar N, Simonovska B, Sok M. Chromatographic determination of total cholesterol in human lung healthy and cancer tissues of the same patient. JPC-J PLANAR CHROMAT 2015. [DOI: 10.1556/jpc.28.2015.2.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
37
|
Akgoc Z, Sena-Esteves M, Martin DR, Han X, d'Azzo A, Seyfried TN. Bis(monoacylglycero)phosphate: a secondary storage lipid in the gangliosidoses. J Lipid Res 2015; 56:1006-13. [PMID: 25795792 DOI: 10.1194/jlr.m057851] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Indexed: 01/24/2023] Open
Abstract
Bis(monoacylglycero)phosphate (BMP) is a negatively charged glycerophospholipid with an unusual sn-1;sn-1' structural configuration. BMP is primarily enriched in endosomal/lysosomal membranes. BMP is thought to play a role in glycosphingolipid degradation and cholesterol transport. Elevated BMP levels have been found in many lysosomal storage diseases (LSDs), suggesting an association with lysosomal storage material. The gangliosidoses are a group of neurodegenerative LSDs involving the accumulation of either GM1 or GM2 gangliosides resulting from inherited deficiencies in β-galactosidase or β-hexosaminidase, respectively. Little information is available on BMP levels in gangliosidosis brain tissue. Our results showed that the content of BMP in brain was significantly greater in humans and in animals (mice, cats, American black bears) with either GM1 or GM2 ganglioside storage diseases, than in brains of normal subjects. The storage of BMP and ganglioside GM2 in brain were reduced similarly following adeno-associated viral-mediated gene therapy in Sandhoff disease mice. We also found that C22:6, C18:0, and C18:1 were the predominant BMP fatty acid species in gangliosidosis brains. The results show that BMP accumulates as a secondary storage material in the brain of a broad range of mammals with gangliosidoses.
Collapse
Affiliation(s)
- Zeynep Akgoc
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | - Miguel Sena-Esteves
- Department of Neurology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605
| | - Douglas R Martin
- Scott-Ritchey Research Center and Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL 36849
| | - Xianlin Han
- Sanford-Burnham Medical Research Institute, Orlando, FL 32827
| | | | | |
Collapse
|
38
|
Lee HJ, Zhang W, Zhang D, Yang Y, Liu B, Barker EL, Buhman KK, Slipchenko LV, Dai M, Cheng JX. Assessing cholesterol storage in live cells and C. elegans by stimulated Raman scattering imaging of phenyl-Diyne cholesterol. Sci Rep 2015; 5:7930. [PMID: 25608867 PMCID: PMC4302291 DOI: 10.1038/srep07930] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/22/2014] [Indexed: 02/07/2023] Open
Abstract
We report a cholesterol imaging method using rationally synthesized phenyl-diyne cholesterol (PhDY-Chol) and stimulated Raman scattering (SRS) microscope. The phenyl-diyne group is biologically inert and provides a Raman scattering cross section that is 88 times larger than the endogenous C = O stretching mode. SRS microscopy offers an imaging speed that is faster than spontaneous Raman microscopy by three orders of magnitude, and a detection sensitivity of 31 μM PhDY-Chol (~1,800 molecules in the excitation volume). Inside living CHO cells, PhDY-Chol mimics the behavior of cholesterol, including membrane incorporation and esterification. In a cellular model of Niemann-Pick type C disease, PhDY-Chol reflects the lysosomal accumulation of cholesterol, and shows relocation to lipid droplets after HPβCD treatment. In live C. elegans, PhDY-Chol mimics cholesterol uptake by intestinal cells and reflects cholesterol storage. Together, our work demonstrates an enabling platform for study of cholesterol storage and trafficking in living cells and vital organisms.
Collapse
Affiliation(s)
- Hyeon Jeong Lee
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Wandi Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Delong Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Yang Yang
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Bin Liu
- National Key Laboratory of Science and Technology on Tunable Laser, Harbin Institute of Technology, Harbin 150080, China
| | - Eric L. Barker
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA
- Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Kimberly K. Buhman
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | | | - Mingji Dai
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Ji-Xin Cheng
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
39
|
Krivoi II. Functional interactions of Na,K-ATPase with molecular environment. Biophysics (Nagoya-shi) 2014. [DOI: 10.1134/s000635091405011x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
40
|
Ariga T. Pathogenic role of ganglioside metabolism in neurodegenerative diseases. J Neurosci Res 2014; 92:1227-42. [PMID: 24903509 DOI: 10.1002/jnr.23411] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 12/13/2022]
Abstract
Ganglioside metabolism is altered in several neurodegenerative diseases, and this may participate in several events related to the pathogenesis of these diseases. Most changes occur in specific areas of the brain and their distinct membrane microdomains or lipid rafts. Antiganglioside antibodies may be involved in dysfunction of the blood-brain barrier and disease progression in these diseases. In lipid rafts, interactions of glycosphingolipids, including ganglioside, with proteins may be responsible for the misfolding events that cause the fibril and/or aggregate processing of disease-specific proteins, such as α-synuclein, in Parkinson's disease, huntingtin protein in Huntington's disease, and copper-zinc superoxide dismutase in amyotrophic lateral sclerosis. Targeting ganglioside metabolism may represent an underexploited opportunity to design novel therapeutic strategies for neurodegeneration in these diseases.
Collapse
Affiliation(s)
- Toshio Ariga
- Institute of Molecular Medicine and Genetics, Institute of Neuroscience, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
41
|
Raina V, Gupta S, Yadav S, Surolia A. Simvastatin induced neurite outgrowth unveils role of cell surface cholesterol and acetyl CoA carboxylase in SH-SY5Y cells. PLoS One 2013; 8:e74547. [PMID: 24040277 PMCID: PMC3770597 DOI: 10.1371/journal.pone.0074547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 08/02/2013] [Indexed: 01/08/2023] Open
Abstract
Statins are known to modulate cell surface cholesterol (CSC) and AMP-activated protein kinase (AMPK) in non-neural cells; however no study demonstrates whether CSC and AMPK may regulate simvastatin induced neuritogenesis (SIN). We found that simvastatin (SIM) maintains CSC as shown by Fillipin III staining, Flotillin-2 protein expression / localization and phosphorylation of various receptor tyrosine kinases (RTKs) in the plasma membrane. Modulation of CSC revealed that SIN is critically dependent on this CSC. Simultaneously, phospho array for mitogen activated protein kinases (MAPKs) revealed PI3K / Akt as intracellular pathway which modulates lipid pathway by inhibiting AMPK activation. Though, SIM led to a transient increase in AMPK phosphorylation followed by a sudden decline; the effect was independent of PI3K. Strikingly, AMPK phosphorylation was regulated by protein phosphatase 2A (PP2A) activity which was enhanced upon SIM treatment as evidenced by increase in threonine phosphorylation. Moreover, it was observed that addition of AMP analogue and PP2A inhibitor inhibited SIN. Bio-composition of neurites shows that lipids form a major part of neurites and AMPK is known to regulate lipid metabolism majorly through acetyl CoA carboxylase (ACC). AMPK activity is negative regulator of ACC activity and we found that phosphorylation of ACC started to decrease after 6 hrs which becomes more pronounced at 12 hrs. Addition of ACC inhibitor showed that SIN is dependent on ACC activity. Simultaneously, addition of Fatty acid synthase (FAS) inhibitor confirmed that endogenous lipid pathway is important for SIN. We further investigated SREBP-1 pathway activation which controls ACC and FAS at transcriptional level. However, SIM did not affect SREBP-1 processing and transcription of its target genes likes ACC1 and FAS. In conclusion, this study highlights a distinct role of CSC and ACC in SIN which might have implication in process of neuronal differentiation induced by other agents.
Collapse
Affiliation(s)
- Varshiesh Raina
- Molecular Sciences Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi, India
| | - Sarika Gupta
- Molecular Sciences Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi, India
- * E-mail: (AS); (SG)
| | - Saurabh Yadav
- Molecular Sciences Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi, India
| | - Avadhesha Surolia
- Molecular Biophysics Unit, Indian Institute of Science, Bangaluru, Karnataka, India
- * E-mail: (AS); (SG)
| |
Collapse
|
42
|
Abstract
A common feature of neurodegenerative diseases is the accumulation of disease-specific, aggregated protein species in the nervous system. Transmissible spongiform encephalopathies are universally fatal neurodegenerative diseases involving the transconformation and aggregation of prion proteins. At the cellular level macroautophagy has been identified as an efficient pathway for the clearance of these toxic protein aggregates. Hence, recent research has focused on the pharmacological manipulation of autophagy as a potential treatment for neurodegenerative diseases. Independent of their effects on the estrogen receptor, tamoxifen and its metabolite 4-hydroxytamoxifen are well known inducers of autophagy. However, we recently reported that the ability of 4-hydroxytamoxifen to clear prion infection is independent of autophagy. In contrast, we provide a model whereby perturbation of cholesterol metabolism, and not autophagy, is the main mechanism whereby 4-hydroxytamoxifen is able to exert its anti-prion effects. Thus, while tamoxifen, a widely available pharmaceutical, may have applications in prion therapy, prions may also represent a special case and may require different pharmacological interventions than other proteinopathies.
Collapse
Affiliation(s)
- Duncan Browman
- Institut Pasteur; Unite ́ de traffic membranaire et pathogenèse; Paris, France
| | | |
Collapse
|
43
|
Echovirus 1 entry into polarized Caco-2 cells depends on dynamin, cholesterol, and cellular factors associated with macropinocytosis. J Virol 2013; 87:8884-95. [PMID: 23740983 DOI: 10.1128/jvi.03415-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Enteroviruses invade their hosts by crossing the intestinal epithelium. We have examined the mechanism by which echovirus 1 (EV1) enters polarized intestinal epithelial cells (Caco-2). Virus binds to VLA-2 on the apical cell surface and moves rapidly to early endosomes. Using inhibitory drugs, dominant negative mutants, and small interfering RNAs (siRNAs) to block specific endocytic pathways, we found that virus entry requires dynamin GTPase and membrane cholesterol but is independent of both clathrin- and caveolin-mediated endocytosis. Instead, infection requires factors commonly associated with macropinocytosis, including amiloride-sensitive Na(+)/H(+) exchange, protein kinase C, and C-terminal-binding protein-1 (CtBP1); furthermore, EV1 accumulates rapidly in intracellular vesicles with dextran, a fluid-phase marker. These results suggest a role for macropinocytosis in the process by which EV1 enters polarized cells to initiate infection.
Collapse
|
44
|
Blattmann P, Schuberth C, Pepperkok R, Runz H. RNAi-based functional profiling of loci from blood lipid genome-wide association studies identifies genes with cholesterol-regulatory function. PLoS Genet 2013; 9:e1003338. [PMID: 23468663 PMCID: PMC3585126 DOI: 10.1371/journal.pgen.1003338] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/07/2013] [Indexed: 01/17/2023] Open
Abstract
Genome-wide association studies (GWAS) are powerful tools to unravel genomic loci associated with common traits and complex human disease. However, GWAS only rarely reveal information on the exact genetic elements and pathogenic events underlying an association. In order to extract functional information from genomic data, strategies for systematic follow-up studies on a phenotypic level are required. Here we address these limitations by applying RNA interference (RNAi) to analyze 133 candidate genes within 56 loci identified by GWAS as associated with blood lipid levels, coronary artery disease, and/or myocardial infarction for a function in regulating cholesterol levels in cells. Knockdown of a surprisingly high number (41%) of trait-associated genes affected low-density lipoprotein (LDL) internalization and/or cellular levels of free cholesterol. Our data further show that individual GWAS loci may contain more than one gene with cholesterol-regulatory functions. Using a set of secondary assays we demonstrate for a number of genes without previously known lipid-regulatory roles (e.g. CXCL12, FAM174A, PAFAH1B1, SEZ6L, TBL2, WDR12) that knockdown correlates with altered LDL–receptor levels and/or that overexpression as GFP–tagged fusion proteins inversely modifies cellular cholesterol levels. By providing strong evidence for disease-relevant functions of lipid trait-associated genes, our study demonstrates that quantitative, cell-based RNAi is a scalable strategy for a systematic, unbiased detection of functional effectors within GWAS loci. Complex traits and diseases are assumed to result from interactions between multiple genes in relevant biological processes. Recent genome-wide association studies have uncovered many novel genomic loci where genes with functional significance are expected. However, functional validation of such genes has thus far remained confined to single gene approaches. Here, we use RNA interference and high-content screening microscopy to profile 133 genes at 56 loci associated with blood lipid traits, cardiovascular disease, and/or myocardial infarction for a function in regulating cellular free cholesterol levels and the efficiency of low-density lipoprotein uptake. Our results suggest that a high number of trait-associated genes have conserved cholesterol-regulatory functions in cells, with several GWAS loci harboring more than one gene of likely functional significance. For a number of genes without previously known lipid-regulatory functions, consequences upon siRNA knockdown positively correlated with cellular levels of LDL receptor, a major determinant of blood LDL levels. Moreover, GFP–tagged fusion proteins of several candidates shifted cellular cholesterol levels to inverse directions than knockdown, and subcellular localization of some candidates was sterol-dependent. Our study generates a valuable resource for prioritization of lipid-trait/CAD/MI-associated genes for future in-depth mechanistic analyses and introduces cell-based RNAi as a scalable and unbiased tool for functional follow-up of GWAS loci.
Collapse
Affiliation(s)
- Peter Blattmann
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christian Schuberth
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany
- * E-mail: (RP); (HR)
| | - Heiko Runz
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
- * E-mail: (RP); (HR)
| |
Collapse
|
45
|
Efremova TN, Chubinskij-Nadezhdin VI, Khaitlina SY, Morachevskaya EA. Assembly of actin filaments induced by sequestration of membrane cholesterol in transformed cells. ACTA ACUST UNITED AC 2012. [DOI: 10.1134/s1990519x12040050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
46
|
Abstract
Cholesterol plays an important role in determining the biophysical properties of biological membranes, and its concentration is tightly controlled by homeostatic processes. The intracellular transport of cholesterol among organelles is a key part of the homeostatic mechanism, but sterol transport processes are not well understood. Fluorescence microscopy is a valuable tool for studying intracellular transport processes, but this method can be challenging for lipid molecules because addition of a fluorophore may alter the properties of the molecule greatly. We discuss the use of fluorescent molecules that can bind to cholesterol to reveal its distribution in cells. We also discuss the use of intrinsically fluorescent sterols that closely mimic cholesterol, as well as some minimally modified fluorophore-labeled sterols. Methods for imaging these sterols by conventional fluorescence microscopy and by multiphoton microscopy are described. Some label-free methods for imaging cholesterol itself are also discussed briefly.
Collapse
|