1
|
Gorman BL, Lukowski JK. Spatial Metabolomics and Lipidomics in Kidney Disease. Semin Nephrol 2025:151582. [PMID: 40234137 DOI: 10.1016/j.semnephrol.2025.151582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Kidney disease is a global health issue that affects over 850 million people, and early detection is key to preventing severe disease and complications. Kidney diseases are associated with complex and dysregulation of lipid metabolism. Spatial metabolomics through mass spectrometry imaging (MSI) enables spatial mapping of the lipids in tissue and includes a variety of techniques that can be used to image lipids. In the kidney, MSI studies often seek to resolve individual functional tissue units such as glomeruli and proximal tubules. Several different MSI techniques, such as matrix-assisted laser desorption/ionization (MALDI) and desorption electrospray ionization (DESI), have been used to characterize lipids and small molecules in chronic kidney disease, acute kidney injury, genetic kidney disease, and cancer. In this review we provide several examples of how spatial metabolomics data can provide critical information concerning the localization of changes in various disease states. Additionally, when combined with pathology, transcriptomics, or proteomics, the metabolomic changes can illuminate underlying mechanisms and provide new clinical insights into disease mechanisms. Semin Nephrol 36:x-xx © 20xx Elsevier Inc. All rights reserved.
Collapse
Affiliation(s)
| | - Jessica K Lukowski
- Mass Spectrometry Imaging Lead, Mass Spectrometry Technology Access Center at the McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO
| |
Collapse
|
2
|
Sun F, Li H, Sun D, Fu S, Gu L, Shao X, Wang Q, Dong X, Duan B, Xing F, Wu J, Xiao M, Zhao F, Han JDJ, Liu Q, Fan X, Li C, Wang C, Shi T. Single-cell omics: experimental workflow, data analyses and applications. SCIENCE CHINA. LIFE SCIENCES 2025; 68:5-102. [PMID: 39060615 DOI: 10.1007/s11427-023-2561-0] [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: 12/07/2023] [Accepted: 04/18/2024] [Indexed: 07/28/2024]
Abstract
Cells are the fundamental units of biological systems and exhibit unique development trajectories and molecular features. Our exploration of how the genomes orchestrate the formation and maintenance of each cell, and control the cellular phenotypes of various organismsis, is both captivating and intricate. Since the inception of the first single-cell RNA technology, technologies related to single-cell sequencing have experienced rapid advancements in recent years. These technologies have expanded horizontally to include single-cell genome, epigenome, proteome, and metabolome, while vertically, they have progressed to integrate multiple omics data and incorporate additional information such as spatial scRNA-seq and CRISPR screening. Single-cell omics represent a groundbreaking advancement in the biomedical field, offering profound insights into the understanding of complex diseases, including cancers. Here, we comprehensively summarize recent advances in single-cell omics technologies, with a specific focus on the methodology section. This overview aims to guide researchers in selecting appropriate methods for single-cell sequencing and related data analysis.
Collapse
Affiliation(s)
- Fengying Sun
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Haoyan Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dongqing Sun
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shaliu Fu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Lei Gu
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Shao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Qinqin Wang
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bin Duan
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Feiyang Xing
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Minmin Xiao
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China.
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Chen Li
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Tieliu Shi
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
3
|
Prentice BM. Imaging with mass spectrometry: Which ionization technique is best? JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5016. [PMID: 38625003 DOI: 10.1002/jms.5016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 04/17/2024]
Abstract
The use of mass spectrometry (MS) to acquire molecular images of biological tissues and other substrates has developed into an indispensable analytical tool over the past 25 years. Imaging mass spectrometry technologies are widely used today to study the in situ spatial distributions for a variety of analytes. Early MS images were acquired using secondary ion mass spectrometry and matrix-assisted laser desorption/ionization. Researchers have also designed and developed other ionization techniques in recent years to probe surfaces and generate MS images, including desorption electrospray ionization (DESI), nanoDESI, laser ablation electrospray ionization, and infrared matrix-assisted laser desorption electrospray ionization. Investigators now have a plethora of ionization techniques to select from when performing imaging mass spectrometry experiments. This brief perspective will highlight the utility and relative figures of merit of these techniques within the context of their use in imaging mass spectrometry.
Collapse
Affiliation(s)
- Boone M Prentice
- Department of Chemistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
4
|
Randolph CE, Manchanda P, Arora H, Iyer S, Saklani P, Beveridge C, Chopra G. Mass Spectrometry-based Single-Cell Lipidomics: Advancements, Challenges, and the Path Forward. Trends Analyt Chem 2023; 169:117350. [PMID: 40255629 PMCID: PMC12007889 DOI: 10.1016/j.trac.2023.117350] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
In the past decade, lipidomics, now recognized as standalone subdiscipline of metabolomics, has gained considerable attention. Due to its sensitivity and unparalleled versatility, mass spectrometry (MS) has emerged as the tool of choice for lipid identification and detection. Traditional MS-based lipidomics are performed on bulk cell samples. While informative, these bulk-scale cellular lipidome measurements mask cellular heterogeneity across seemingly homogeneous populations of cells. Unfortunately, single cell lipidomics methodology and analyses are considerably behind genomics, transcriptomics, and proteomics. Therefore, the cell-to-cell heterogeneity and related function remains largely unexplored for lipidomics. Herein, we review recent advances in MS-based single cell lipidomics. We also explore the root causes for the slow development of single-cell lipidomics techniques. We aim to provide insights on the pivotal knowledge gaps that have been neglected, prohibiting the propulsion of the single-cell lipidomics field forward, while also providing our perspective towards future methodologies that can pave a path forward.
Collapse
Affiliation(s)
| | - Palak Manchanda
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Harshit Arora
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sanjay Iyer
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Pooja Saklani
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Connor Beveridge
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Drug Discovery, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA
- Purdue Institute for Inflammation, Immunology and Infectious Disease, West Lafayette, IN 47907, USA
- Purdue Center for Cancer Research, West Lafayette, IN 47907, USA
- Purdue University Integrative Data Science Initiative, West Lafayette, IN 47907, USA
| |
Collapse
|
5
|
Ferrari A, Whang E, Xiao X, Kennelly JP, Romartinez-Alonso B, Mack JJ, Weston T, Chen K, Kim Y, Tol MJ, Bideyan L, Nguyen A, Gao Y, Cui L, Bedard AH, Sandhu J, Lee SD, Fairall L, Williams KJ, Song W, Munguia P, Russell RA, Martin MG, Jung ME, Jiang H, Schwabe JW, Young SG, Tontonoz P. Aster-dependent nonvesicular transport facilitates dietary cholesterol uptake. Science 2023; 382:eadf0966. [PMID: 37943936 PMCID: PMC11073449 DOI: 10.1126/science.adf0966] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 09/27/2023] [Indexed: 11/12/2023]
Abstract
Intestinal absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1) assists in the initial step of dietary cholesterol uptake, but how cholesterol moves downstream of NPC1L1 is unknown. We show that Aster-B and Aster-C are critical for nonvesicular cholesterol movement in enterocytes. Loss of NPC1L1 diminishes accessible plasma membrane (PM) cholesterol and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate PM cholesterol and show endoplasmic reticulum cholesterol depletion. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, the Aster pathway can be targeted with a small-molecule inhibitor to manipulate cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption.
Collapse
Affiliation(s)
- Alessandra Ferrari
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Emily Whang
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Pediatric Gastroenterology, Hepatology, and Nutrition, David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Xu Xiao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - John P. Kennelly
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | | | - Julia J. Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
| | - Thomas Weston
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Kai Chen
- Department of Chemistry, The University of Hong Kong, Hong Kong, 999077, China
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley WA 6009, Australia
| | - Youngjae Kim
- Department of Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Marcus J. Tol
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Lara Bideyan
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Alexander Nguyen
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Yajing Gao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Liujuan Cui
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Alexander H. Bedard
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Jaspreet Sandhu
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephen D. Lee
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Louise Fairall
- Institute for Structural and Chemical Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Kevin J. Williams
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
- UCLA Lipidomics Core, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wenxin Song
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Priscilla Munguia
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Robert A. Russell
- National Deuteration Facility, Australian Nuclear Science and Technology Organisation, Lucas Heights, Australia
| | - Martin G. Martin
- Pediatric Gastroenterology, Hepatology, and Nutrition, David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Michael E. Jung
- Department of Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Hong Kong, 999077, China
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley WA 6009, Australia
| | - John W.R. Schwabe
- Institute for Structural and Chemical Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Stephen G. Young
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| |
Collapse
|
6
|
Song W, Beigneux AP, Weston TA, Chen K, Yang Y, Nguyen LP, Guagliardo P, Jung H, Tran AP, Tu Y, Tran C, Birrane G, Miyashita K, Nakajima K, Murakami M, Tontonoz P, Jiang H, Ploug M, Fong LG, Young SG. The lipoprotein lipase that is shuttled into capillaries by GPIHBP1 enters the glycocalyx where it mediates lipoprotein processing. Proc Natl Acad Sci U S A 2023; 120:e2313825120. [PMID: 37871217 PMCID: PMC10623010 DOI: 10.1073/pnas.2313825120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 09/19/2023] [Indexed: 10/25/2023] Open
Abstract
Lipoprotein lipase (LPL), the enzyme that carries out the lipolytic processing of triglyceride-rich lipoproteins (TRLs), is synthesized by adipocytes and myocytes and secreted into the interstitial spaces. The LPL is then bound by GPIHBP1, a GPI-anchored protein of endothelial cells (ECs), and transported across ECs to the capillary lumen. The assumption has been that the LPL that is moved into capillaries remains attached to GPIHBP1 and that GPIHBP1 serves as a platform for TRL processing. In the current studies, we examined the validity of that assumption. We found that an LPL-specific monoclonal antibody (mAb), 88B8, which lacks the ability to detect GPIHBP1-bound LPL, binds avidly to LPL within capillaries. We further demonstrated, by confocal microscopy, immunogold electron microscopy, and nanoscale secondary ion mass spectrometry analyses, that the LPL detected by mAb 88B8 is located within the EC glycocalyx, distant from the GPIHBP1 on the EC plasma membrane. The LPL within the glycocalyx mediates the margination of TRLs along capillaries and is active in TRL processing, resulting in the delivery of lipoprotein-derived lipids to immediately adjacent parenchymal cells. Thus, the LPL that GPIHBP1 transports into capillaries can detach and move into the EC glycocalyx, where it functions in the intravascular processing of TRLs.
Collapse
Affiliation(s)
- Wenxin Song
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Anne P. Beigneux
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Thomas A. Weston
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Kai Chen
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
- School of Molecular Sciences, The University of Western Australia, Perth6009, Australia
| | - Ye Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Le Phuong Nguyen
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Paul Guagliardo
- Centre for Microscopy Characterisation and Analysis, The University of Western Australia, Perth6009, Australia
| | - Hyesoo Jung
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Anh P. Tran
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Yiping Tu
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Caitlyn Tran
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Gabriel Birrane
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Boston, MA02215
| | - Kazuya Miyashita
- Department of Clinical Laboratory Medicine, Gunma University School of Medicine, Maebashi371-8511, Japan
| | - Katsuyuki Nakajima
- Department of Clinical Laboratory Medicine, Gunma University School of Medicine, Maebashi371-8511, Japan
| | - Masami Murakami
- Department of Clinical Laboratory Medicine, Gunma University School of Medicine, Maebashi371-8511, Japan
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA90095
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Michael Ploug
- Finsen Laboratory, Copenhagen University Hospital-Rigshospitalet, Copenhagen NDK–2200, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
| | - Loren G. Fong
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Stephen G. Young
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| |
Collapse
|
7
|
Ferrari A, Whang E, Xiao X, Kennelly JP, Romartinez-Alonso B, Mack JJ, Weston T, Chen K, Kim Y, Tol MJ, Bideyan L, Nguyen A, Gao Y, Cui L, Bedard AH, Sandhu J, Lee SD, Fairall L, Williams KJ, Song W, Munguia P, Russell RA, Martin MG, Jung ME, Jiang H, Schwabe JWR, Young SG, Tontonoz P. Aster-dependent non-vesicular transport facilitates dietary cholesterol uptake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.548168. [PMID: 37503112 PMCID: PMC10369906 DOI: 10.1101/2023.07.07.548168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Intestinal cholesterol absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1), the target of the drug ezetimibe (EZ), assists in the initial step of dietary cholesterol uptake. However, how cholesterol moves downstream of NPC1L1 is unknown. Here we show that Aster-B and Aster-C are critical for non-vesicular cholesterol movement in enterocytes, bridging NPC1L1 at the plasma membrane (PM) and ACAT2 in the endoplasmic reticulum (ER). Loss of NPC1L1 diminishes accessible PM cholesterol in enterocytes and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate cholesterol at the PM and display evidence of ER cholesterol depletion, including decreased cholesterol ester stores and activation of the SREBP-2 transcriptional pathway. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, we show that the Aster pathway can be targeted with a small molecule inhibitor to manipulate dietary cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption. One-Sentence Summary Identification of a targetable pathway for regulation of dietary cholesterol absorption.
Collapse
|
8
|
Schwaiger-Haber M, Stancliffe E, Anbukumar DS, Sells B, Yi J, Cho K, Adkins-Travis K, Chheda MG, Shriver LP, Patti GJ. Using mass spectrometry imaging to map fluxes quantitatively in the tumor ecosystem. Nat Commun 2023; 14:2876. [PMID: 37208361 PMCID: PMC10199024 DOI: 10.1038/s41467-023-38403-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 04/26/2023] [Indexed: 05/21/2023] Open
Abstract
Tumors are comprised of a multitude of cell types spanning different microenvironments. Mass spectrometry imaging (MSI) has the potential to identify metabolic patterns within the tumor ecosystem and surrounding tissues, but conventional workflows have not yet fully integrated the breadth of experimental techniques in metabolomics. Here, we combine MSI, stable isotope labeling, and a spatial variant of Isotopologue Spectral Analysis to map distributions of metabolite abundances, nutrient contributions, and metabolic turnover fluxes across the brains of mice harboring GL261 glioma, a widely used model for glioblastoma. When integrated with MSI, the combination of ion mobility, desorption electrospray ionization, and matrix assisted laser desorption ionization reveals alterations in multiple anabolic pathways. De novo fatty acid synthesis flux is increased by approximately 3-fold in glioma relative to surrounding healthy tissue. Fatty acid elongation flux is elevated even higher at 8-fold relative to surrounding healthy tissue and highlights the importance of elongase activity in glioma.
Collapse
Affiliation(s)
- Michaela Schwaiger-Haber
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Ethan Stancliffe
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Dhanalakshmi S Anbukumar
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Blake Sells
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Jia Yi
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Kevin Cho
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Kayla Adkins-Travis
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Milan G Chheda
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Leah P Shriver
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA.
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
9
|
Chen K, Song W, Russell R, Ferrari A, Darwish T, Tontonoz P, Young SG, Jiang H. NanoSIMS imaging of lipid absorption by intestinal enterocytes. J Lipid Res 2022; 63:100290. [PMID: 36179912 PMCID: PMC9672438 DOI: 10.1016/j.jlr.2022.100290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 10/14/2022] Open
Affiliation(s)
- Kai Chen
- Department of Chemistry, The University of Hong Kong, Hong Kong, China,School of Molecular Sciences, The University of Western Australia, Crawley, Australia
| | - Wenxin Song
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Robert Russell
- National Deuteration Facility, ANSTO, Lucas Heights, Australia
| | - Alessandra Ferrari
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Tamim Darwish
- National Deuteration Facility, ANSTO, Lucas Heights, Australia
| | - Peter Tontonoz
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Stephen G. Young
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Hong Kong, China; School of Molecular Sciences, The University of Western Australia, Crawley, Australia.
| |
Collapse
|
10
|
Young SG, Song W, Yang Y, Birrane G, Jiang H, Beigneux AP, Ploug M, Fong LG. A protein of capillary endothelial cells, GPIHBP1, is crucial for plasma triglyceride metabolism. Proc Natl Acad Sci U S A 2022; 119:e2211136119. [PMID: 36037340 PMCID: PMC9457329 DOI: 10.1073/pnas.2211136119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
GPIHBP1, a protein of capillary endothelial cells (ECs), is a crucial partner for lipoprotein lipase (LPL) in the lipolytic processing of triglyceride-rich lipoproteins. GPIHBP1, which contains a three-fingered cysteine-rich LU (Ly6/uPAR) domain and an intrinsically disordered acidic domain (AD), captures LPL from within the interstitial spaces (where it is secreted by parenchymal cells) and shuttles it across ECs to the capillary lumen. Without GPIHBP1, LPL remains stranded within the interstitial spaces, causing severe hypertriglyceridemia (chylomicronemia). Biophysical studies revealed that GPIHBP1 stabilizes LPL structure and preserves LPL activity. That discovery was the key to crystallizing the GPIHBP1-LPL complex. The crystal structure revealed that GPIHBP1's LU domain binds, largely by hydrophobic contacts, to LPL's C-terminal lipid-binding domain and that the AD is positioned to project across and interact, by electrostatic forces, with a large basic patch spanning LPL's lipid-binding and catalytic domains. We uncovered three functions for GPIHBP1's AD. First, it accelerates the kinetics of LPL binding. Second, it preserves LPL activity by inhibiting unfolding of LPL's catalytic domain. Third, by sheathing LPL's basic patch, the AD makes it possible for LPL to move across ECs to the capillary lumen. Without the AD, GPIHBP1-bound LPL is trapped by persistent interactions between LPL and negatively charged heparan sulfate proteoglycans (HSPGs) on the abluminal surface of ECs. The AD interrupts the HSPG interactions, freeing LPL-GPIHBP1 complexes to move across ECs to the capillary lumen. GPIHBP1 is medically important; GPIHBP1 mutations cause lifelong chylomicronemia, and GPIHBP1 autoantibodies cause some acquired cases of chylomicronemia.
Collapse
Affiliation(s)
- Stephen G. Young
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Wenxin Song
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Ye Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Gabriel Birrane
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Anne P. Beigneux
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen 2200N, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Loren G. Fong
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| |
Collapse
|
11
|
Mellinger AL, Muddiman DC, Gamcsik MP. Highlighting Functional Mass Spectrometry Imaging Methods in Bioanalysis. J Proteome Res 2022; 21:1800-1807. [PMID: 35749637 DOI: 10.1021/acs.jproteome.2c00220] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Most mass spectrometry imaging (MSI) methods provide a molecular map of tissue content but little information on tissue function. Mapping tissue function is possible using several well-known examples of "functional imaging" such as positron emission tomography and functional magnetic resonance imaging that can provide the spatial distribution of time-dependent biological processes. These functional imaging methods represent the net output of molecular networks influenced by local tissue environments that are difficult to predict from molecular/cellular content alone. However, for decades, MSI methods have also been demonstrated to provide functional imaging data on a variety of biological processes. In fact, MSI exceeds some of the classic functional imaging methods, demonstrating the ability to provide functional data from the nanoscale (subcellular) to whole tissue or organ level. This Perspective highlights several examples of how different MSI ionization and detection technologies can provide unprecedented detailed spatial maps of time-dependent biological processes, namely, nucleic acid synthesis, lipid metabolism, bioenergetics, and protein metabolism. By classifying various MSI methods under the umbrella of "functional MSI", we hope to draw attention to both the unique capabilities and accessibility with the aim of expanding this underappreciated field to include new approaches and applications.
Collapse
Affiliation(s)
- Allyson L Mellinger
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - David C Muddiman
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States.,Molecular Education, Technology and Research Innovation Center (METRIC), North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Michael P Gamcsik
- UNC/NCSU Joint Department of Biomedical Engineering, Raleigh, North Carolina 27695, United States
| |
Collapse
|
12
|
Zhu X, Xu T, Peng C, Wu S. Advances in MALDI Mass Spectrometry Imaging Single Cell and Tissues. Front Chem 2022; 9:782432. [PMID: 35186891 PMCID: PMC8850921 DOI: 10.3389/fchem.2021.782432] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Compared with conventional optical microscopy techniques, mass spectrometry imaging (MSI) or imaging mass spectrometry (IMS) is a powerful, label-free analytical technique, which can sensitively and simultaneously detect, quantify, and map hundreds of biomolecules, such as peptides, proteins, lipid, and other organic compounds in cells and tissues. So far, although several soft ionization techniques, such as desorption electrospray ionization (DESI) and secondary ion mass spectrometry (SIMS) have been used for imaging biomolecules, matrix-assisted laser desorption/ionization (MALDI) is still the most widespread MSI scanning method. Here, we aim to provide a comprehensive review of MALDI-MSI with an emphasis on its advances of the instrumentation, methods, application, and future directions in single cell and biological tissues.
Collapse
Affiliation(s)
- Xiaoping Zhu
- Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China
- Research Center of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Tianyi Xu
- Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China
- Research Center of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Chen Peng
- Research Center of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Shihua Wu
- Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China
- Research Center of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou, China
- *Correspondence: Shihua Wu, ; Shihua Wu,
| |
Collapse
|
13
|
Trans-endothelial trafficking of metabolic substrates and its importance in cardio-metabolic disease. Biochem Soc Trans 2021; 49:507-517. [PMID: 33616631 DOI: 10.1042/bst20200991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 11/17/2022]
Abstract
The endothelium acts as a gatekeeper, controlling the movement of biomolecules between the circulation and underlying tissues. Although conditions of metabolic stress are traditionally considered as causes of endothelial dysfunction, a principal driver of cardiovascular disease, accumulating evidence suggests that endothelial cells are also active players in maintaining local metabolic homeostasis, in part, through regulating the supply of metabolic substrates, including lipids and glucose, to energy-demanding organs. Therefore, endothelial dysfunction, in terms of altered trans-endothelial trafficking of these substrates, may in fact be an early contributor towards the establishment of metabolic dysfunction and subsequent cardiovascular disease. Understanding the molecular mechanisms that underpin substrate trafficking through the endothelium represents an important area within the vascular and metabolism fields that may offer an opportunity for identifying novel therapeutic targets. This mini-review summarises the emerging mechanisms regulating the trafficking of lipids and glucose through the endothelial barrier and how this may impact on the development of cardio-metabolic disease.
Collapse
|
14
|
Applications of stable isotopes in MALDI imaging: current approaches and an eye on the future. Anal Bioanal Chem 2021; 413:2637-2653. [PMID: 33532914 DOI: 10.1007/s00216-021-03189-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/30/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
Matrix-assisted laser desorption/ionisation-imaging mass spectrometry (MALDI-IMS) is now an established imaging modality with particular utility in the study of biological, biomedical and pathological processes. In the first instance, the use of stable isotopically labelled (SIL) compounds in MALDI-IMS has addressed technical barriers to increase the accuracy and versatility of this technique. This has undoubtedly enhanced our ability to interpret the two-dimensional ion intensity distributions produced from biological tissue sections. Furthermore, studies using delivery of SIL compounds to live tissues have begun to decipher cell, tissue and inter-tissue metabolism while maintaining spatial resolution. Here, we review both the technical and biological applications of SIL compounds in MALDI-IMS, before using the uptake and metabolism of glucose in bovine ocular lens tissue to illustrate the current limitations of SIL compound use in MALDI-IMS. Finally, we highlight recent instrumentation advances that may further enhance our ability to use SIL compounds in MALDI-IMS to understand biological and pathological processes. Graphical Abstract.
Collapse
|
15
|
He C, Migawa MT, Chen K, Weston TA, Tanowitz M, Song W, Guagliardo P, Iyer KS, Bennett CF, Fong LG, Seth PP, Young SG, Jiang H. High-resolution visualization and quantification of nucleic acid-based therapeutics in cells and tissues using Nanoscale secondary ion mass spectrometry (NanoSIMS). Nucleic Acids Res 2021; 49:1-14. [PMID: 33275144 PMCID: PMC7797060 DOI: 10.1093/nar/gkaa1112] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/27/2020] [Accepted: 11/04/2020] [Indexed: 01/19/2023] Open
Abstract
Nucleic acid therapeutics (NATs) have proven useful in promoting the degradation of specific transcripts, modifying gene expression, and regulating mRNA splicing. In each situation, efficient delivery of nucleic acids to cells, tissues and intracellular compartments is crucial—both for optimizing efficacy and reducing side effects. Despite successes in NATs, our understanding of their cellular uptake and distribution in tissues is limited. Current methods have yielded insights into distribution of NATs within cells and tissues, but the sensitivity and resolution of these approaches are limited. Here, we show that nanoscale secondary ion mass spectrometry (NanoSIMS) imaging can be used to define the distribution of 5-bromo-2′-deoxythymidine (5-BrdT) modified antisense oligonucleotides (ASO) in cells and tissues with high sensitivity and spatial resolution. This approach makes it possible to define ASO uptake and distribution in different subcellular compartments and to quantify the impact of targeting ligands designed to promote ASO uptake by cells. Our studies showed that phosphorothioate ASOs are associated with filopodia and the inner nuclear membrane in cultured cells, and also revealed substantial cellular and subcellular heterogeneity of ASO uptake in mouse tissues. NanoSIMS imaging represents a significant advance in visualizing uptake and distribution of NATs; this approach will be useful in optimizing efficacy and delivery of NATs for treating human disease.
Collapse
Affiliation(s)
- Cuiwen He
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | - Kai Chen
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia
| | - Thomas A Weston
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | - Wenxin Song
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Paul Guagliardo
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia
| | - K Swaminathan Iyer
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia
| | | | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Punit P Seth
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, CA 90095, USA.,Department of Human Genetics, University of California, Los Angeles, CA 90095, USA
| | - Haibo Jiang
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia.,Department of Chemistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Ellis SR, Hall E, Panchal M, Flinders B, Madsen J, Koster G, Heeren RMA, Clark HW, Postle AD. Mass spectrometry imaging of phosphatidylcholine metabolism in lungs administered with therapeutic surfactants and isotopic tracers. J Lipid Res 2021; 62:100023. [PMID: 33453219 PMCID: PMC7961103 DOI: 10.1016/j.jlr.2021.100023] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/18/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Mass spectrometry imaging (MSI) visualizes molecular distributions throughout tissues but is blind to dynamic metabolic processes. Here, MSI with high mass resolution together with multiple stable isotope labeling provided spatial analyses of phosphatidylcholine (PC) metabolism in mouse lungs. Dysregulated surfactant metabolism is central to many respiratory diseases. Metabolism and turnover of therapeutic pulmonary surfactants were imaged from distributions of intact and metabolic products of an added tracer, universally 13C-labeled dipalmitoyl PC (U13C-DPPC). The parenchymal distributions of newly synthesized PC species were also imaged from incorporations of methyl-D9-choline. This dual labeling strategy demonstrated both lack of inhibition of endogenous PC synthesis by exogenous surfactant and location of acyl chain remodeling processes acting on the U13C-DPPC-labeled surfactant, leading to formation of polyunsaturated PC lipids. This ability to visualize discrete metabolic events will greatly enhance our understanding of lipid metabolism in diverse tissues and has potential application to both clinical and experimental studies.
Collapse
Affiliation(s)
- Shane R Ellis
- Division of Imaging Mass Spectrometry, Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Maastricht, The Netherlands; Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia; Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia.
| | - Emily Hall
- Academic Unit of Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Madhuriben Panchal
- Academic Unit of Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; National Institute for Health Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Bryn Flinders
- Division of Imaging Mass Spectrometry, Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Maastricht, The Netherlands
| | - Jens Madsen
- Elizabeth Garrett Anderson Institute for Women's Health, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Grielof Koster
- Academic Unit of Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; National Institute for Health Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Ron M A Heeren
- Division of Imaging Mass Spectrometry, Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Maastricht, The Netherlands
| | - Howard W Clark
- Elizabeth Garrett Anderson Institute for Women's Health, Faculty of Population Health Sciences, University College London, London, United Kingdom; National Institute for Health Biomedical Research Centre, University College London Hospital Biomedical Research Centre, London, United Kingdom
| | - Anthony D Postle
- Academic Unit of Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; National Institute for Health Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom.
| |
Collapse
|
17
|
Salado-Leza D, Porcel E, Yang X, Štefančíková L, Bolsa-Ferruz M, Savina F, Dragoe D, Guerquin-Kern JL, Wu TD, Hirayama R, Remita H, Lacombe S. Green One-Step Synthesis of Medical Nanoagents for Advanced Radiation Therapy. Nanotechnol Sci Appl 2020; 13:61-76. [PMID: 32848371 PMCID: PMC7426062 DOI: 10.2147/nsa.s257392] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/30/2020] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Metal-based nanoparticles (M-NPs) have attracted great attention in nanomedicine due to their capacity to amplify and improve the tumor targeting of medical beams. However, their simple, efficient, high-yield and reproducible production remains a challenge. Currently, M-NPs are mainly synthesized by chemical methods or radiolysis using toxic reactants. The waste of time, loss of material and potential environmental hazards are major limitations. MATERIALS AND METHODS This work proposes a simple, fast and green strategy to synthesize small, non-toxic and stable NPs in water with a 100% production rate. Ionizing radiation is used to simultaneously synthesize and sterilize the containing NPs solutions. The synthesis of platinum nanoparticles (Pt NPs) coated with biocompatible poly(ethylene glycol) ligands (PEG) is presented as proof of concept. The physicochemical properties of NPs were studied by complementary specialized techniques. Their toxicity and radio-enhancing properties were evaluated in a cancerous in vitro model. Using plasmid nanoprobes, we investigated the elementary mechanisms underpinning radio-enhancement. RESULTS AND DISCUSSION Pt NPs showed nearly spherical-like shapes and an average hydrodynamic diameter of 9 nm. NPs are zero-valent platinum successfully coated with PEG. They were found non-toxic and have the singular property of amplifying cell killing induced by γ-rays (14%) and even more, the effects of carbon ions (44%) used in particle therapy. They induce nanosized-molecular damage, which is a major finding to potentially implement this protocol in treatment planning simulations. CONCLUSION This new eco-friendly, fast and simple proposed method opens a new era of engineering water-soluble biocompatible NPs and boosts the development of NP-aided radiation therapies.
Collapse
Affiliation(s)
- Daniela Salado-Leza
- Université Paris Saclay, CNRS UMR 8214, Institut des Sciences Moléculaires d’Orsay, 91405 Orsay, France
- Cátedra CONACyT, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, 78210 San Luis Potosí, Mexico
| | - Erika Porcel
- Université Paris Saclay, CNRS UMR 8214, Institut des Sciences Moléculaires d’Orsay, 91405 Orsay, France
| | - Xiaomin Yang
- Université Paris Saclay, CNRS UMR 8214, Institut des Sciences Moléculaires d’Orsay, 91405 Orsay, France
| | - Lenka Štefančíková
- Université Paris Saclay, CNRS UMR 8214, Institut des Sciences Moléculaires d’Orsay, 91405 Orsay, France
| | - Marta Bolsa-Ferruz
- Université Paris Saclay, CNRS UMR 8214, Institut des Sciences Moléculaires d’Orsay, 91405 Orsay, France
| | - Farah Savina
- Université Paris Saclay, CNRS UMR 8214, Institut des Sciences Moléculaires d’Orsay, 91405 Orsay, France
| | - Diana Dragoe
- Université Paris Saclay, CNRS UMR 8182, Institut de Chimie Moléculaire et des Matériaux d’Orsay, 91405 Orsay, France
| | - Jean-Luc Guerquin-Kern
- Paris-Saclay University, Multimodal Imaging Center (UMS 2016/US 43) CNRS, INSERM, Institut Curie, 91405 Orsay, France
| | - Ting-Di Wu
- Paris-Saclay University, Multimodal Imaging Center (UMS 2016/US 43) CNRS, INSERM, Institut Curie, 91405 Orsay, France
| | - Ryoichi Hirayama
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 263-8555 Chiba, Japan
| | - Hynd Remita
- Université Paris Saclay, CNRS UMR 8000, Institut de Chimie Physique, 91405 Orsay, France
| | - Sandrine Lacombe
- Université Paris Saclay, CNRS UMR 8214, Institut des Sciences Moléculaires d’Orsay, 91405 Orsay, France
| |
Collapse
|
18
|
He C, Song W, Weston TA, Tran C, Kurtz I, Zuckerman JE, Guagliardo P, Miner JH, Ivanov SV, Bougoure J, Hudson BG, Colon S, Voziyan PA, Bhave G, Fong LG, Young SG, Jiang H. Peroxidasin-mediated bromine enrichment of basement membranes. Proc Natl Acad Sci U S A 2020; 117:15827-15836. [PMID: 32571911 PMCID: PMC7354931 DOI: 10.1073/pnas.2007749117] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Bromine and peroxidasin (an extracellular peroxidase) are essential for generating sulfilimine cross-links between a methionine and a hydroxylysine within collagen IV, a basement membrane protein. The sulfilimine cross-links increase the structural integrity of basement membranes. The formation of sulfilimine cross-links depends on the ability of peroxidasin to use bromide and hydrogen peroxide substrates to produce hypobromous acid (HOBr). Once a sulfilimine cross-link is created, bromide is released into the extracellular space and becomes available for reutilization. Whether the HOBr generated by peroxidasin is used very selectively for creating sulfilimine cross-links or whether it also causes oxidative damage to bystander molecules (e.g., generating bromotyrosine residues in basement membrane proteins) is unclear. To examine this issue, we used nanoscale secondary ion mass spectrometry (NanoSIMS) imaging to define the distribution of bromine in mammalian tissues. We observed striking enrichment of bromine (79Br, 81Br) in basement membranes of normal human and mouse kidneys. In peroxidasin knockout mice, bromine enrichment of basement membranes of kidneys was reduced by ∼85%. Proteomic studies revealed bromination of tyrosine-1485 in the NC1 domain of α2 collagen IV from kidneys of wild-type mice; the same tyrosine was brominated in collagen IV from human kidney. Bromination of tyrosine-1485 was reduced by >90% in kidneys of peroxidasin knockout mice. Thus, in addition to promoting sulfilimine cross-links in collagen IV, peroxidasin can also brominate a bystander tyrosine. Also, the fact that bromine enrichment is largely confined to basement membranes implies that peroxidasin activity is largely restricted to basement membranes in mammalian tissues.
Collapse
Affiliation(s)
- Cuiwen He
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Wenxin Song
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Thomas A Weston
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Caitlyn Tran
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Ira Kurtz
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Jonathan E Zuckerman
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
| | - Paul Guagliardo
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, 6009 Perth, Australia
| | - Jeffrey H Miner
- Division of Nephrology, Washington University School of Medicine, St. Louis, MO 63110
| | - Sergey V Ivanov
- Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37212
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jeremy Bougoure
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, 6009 Perth, Australia
| | - Billy G Hudson
- Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37212
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232
| | - Selene Colon
- Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37212
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37212
| | - Paul A Voziyan
- Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37212
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Gautam Bhave
- Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37212
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37212
- Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, CA 90095;
- Department of Human Genetics, University of California, Los Angeles, CA 90095
| | - Haibo Jiang
- School of Molecular Sciences, University of Western Australia, 6009 Perth, Australia;
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
19
|
Cao G, Li K, Guo J, Lu M, Hong Y, Cai Z. Mass Spectrometry for Analysis of Changes during Food Storage and Processing. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6956-6966. [PMID: 32516537 DOI: 10.1021/acs.jafc.0c02587] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Many physicochemical changes occur during food storage and processing, such as rancidity, hydrolysis, oxidation, and aging, which may alter the taste, flavor, and texture of food products and pose risks to public health. Analysis of these changes has become of great interest to many researchers. Mass spectrometry is a promising technique for the study of food and nutrition domains as a result of its excellent ability in molecular profiling, food authentication, and marker detection. In this review, we summarized recent advances in mass spectrometry techniques and their applications in food storage and processing. Furthermore, current technical challenges associated with these methodologies were discussed.
Collapse
Affiliation(s)
- Guodong Cao
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong Special Administrative Region of the People's Republic of China
| | - Kun Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong Special Administrative Region of the People's Republic of China
- State Key Laboratory of Cotton Biology, Key Laboratory of Plant Stress Biology, School of Life Sciences, Henan University, Kaifeng, Henan 475004, People's Republic of China
| | - Jinggong Guo
- State Key Laboratory of Cotton Biology, Key Laboratory of Plant Stress Biology, School of Life Sciences, Henan University, Kaifeng, Henan 475004, People's Republic of China
| | - Minghua Lu
- State Key Laboratory of Cotton Biology, Key Laboratory of Plant Stress Biology, School of Life Sciences, Henan University, Kaifeng, Henan 475004, People's Republic of China
| | - Yanjun Hong
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong Special Administrative Region of the People's Republic of China
- HKBU Institute of Research and Continuing Education, Shenzhen, Guangdong 518057, People's Republic of China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong Special Administrative Region of the People's Republic of China
| |
Collapse
|
20
|
Jiang H, He C, Fong LG, Young SG. The fatty acids from LPL-mediated processing of triglyceride-rich lipoproteins are taken up rapidly by cardiomyocytes. J Lipid Res 2020; 61:815. [PMID: 32241805 PMCID: PMC7269765 DOI: 10.1194/jlr.ilr120000783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Haibo Jiang
- School of Molecular Sciences, University of Western Australia, 6009 Perth, WA, Australia
| | - Cuiwen He
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
Lin Y, Hao J, Miao Z, Zhang J, Yang W. NanoSIMS image enhancement by reducing random noise using low‐rank method. SURF INTERFACE ANAL 2020. [DOI: 10.1002/sia.6736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Yi Lin
- Key Laboratory of Earth and Planetary PhysicsInstitute of Geology and Geophysics, Chinese Academy of Sciences Beijing China
- Innovation Academy for Earth Science, Chinese Academy of Sciences Beijing China
- College of Earth and Planetary SciencesUniversity of Chinese Academy of Sciences Beijing China
| | - Jialong Hao
- Key Laboratory of Earth and Planetary PhysicsInstitute of Geology and Geophysics, Chinese Academy of Sciences Beijing China
- Innovation Academy for Earth Science, Chinese Academy of Sciences Beijing China
| | - Zhongzheng Miao
- Key Laboratory of Earth and Planetary PhysicsInstitute of Geology and Geophysics, Chinese Academy of Sciences Beijing China
- Innovation Academy for Earth Science, Chinese Academy of Sciences Beijing China
- College of Earth and Planetary SciencesUniversity of Chinese Academy of Sciences Beijing China
| | - Jinhai Zhang
- Key Laboratory of Earth and Planetary PhysicsInstitute of Geology and Geophysics, Chinese Academy of Sciences Beijing China
- Innovation Academy for Earth Science, Chinese Academy of Sciences Beijing China
| | - Wei Yang
- Key Laboratory of Earth and Planetary PhysicsInstitute of Geology and Geophysics, Chinese Academy of Sciences Beijing China
- Innovation Academy for Earth Science, Chinese Academy of Sciences Beijing China
| |
Collapse
|
22
|
Young SG. Images in Lipid Research. J Lipid Res 2020; 61:589-590. [PMID: 32132143 DOI: 10.1194/jlr.e120000736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
23
|
Ramanome technology platform for label-free screening and sorting of microbial cell factories at single-cell resolution. Biotechnol Adv 2019; 37:107388. [DOI: 10.1016/j.biotechadv.2019.04.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 01/09/2023]
|
24
|
Goldberg IJ, Reue K, Abumrad NA, Bickel PE, Cohen S, Fisher EA, Galis ZS, Granneman JG, Lewandowski ED, Murphy R, Olive M, Schaffer JE, Schwartz-Longacre L, Shulman GI, Walther TC, Chen J. Deciphering the Role of Lipid Droplets in Cardiovascular Disease: A Report From the 2017 National Heart, Lung, and Blood Institute Workshop. Circulation 2019; 138:305-315. [PMID: 30012703 DOI: 10.1161/circulationaha.118.033704] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lipid droplets (LDs) are distinct and dynamic organelles that affect the health of cells and organs. Much progress has been made in understanding how these structures are formed, how they interact with other cellular organelles, how they are used for storage of triacylglycerol in adipose tissue, and how they regulate lipolysis. Our understanding of the biology of LDs in the heart and vascular tissue is relatively primitive in comparison with LDs in adipose tissue and liver. The National Heart, Lung, and Blood Institute convened a working group to discuss how LDs affect cardiovascular diseases. The goal of the working group was to examine the current state of knowledge on the cell biology of LDs, including current methods to study them in cells and organs and reflect on how LDs influence the development and progression of cardiovascular diseases. This review summarizes the working group discussion and recommendations on research areas ripe for future investigation that will likely improve our understanding of atherosclerosis and heart function.
Collapse
Affiliation(s)
| | - Karen Reue
- University of California, Los Angeles (K.R.)
| | | | - Perry E Bickel
- University of Texas Southwestern Medical Center, Dallas (P.E.B.)
| | - Sarah Cohen
- University of North Carolina at Chapel Hill (S.C.)
| | | | - Zorina S Galis
- National Institutes of Health/National, Heart, Lung, and Blood Institute, Bethesda, MD (Z.S.G., M.O., L.S.-L., J.C.)
| | | | | | | | - Michelle Olive
- National Institutes of Health/National, Heart, Lung, and Blood Institute, Bethesda, MD (Z.S.G., M.O., L.S.-L., J.C.)
| | | | - Lisa Schwartz-Longacre
- National Institutes of Health/National, Heart, Lung, and Blood Institute, Bethesda, MD (Z.S.G., M.O., L.S.-L., J.C.)
| | - Gerald I Shulman
- Yale University, Howard Hughes Medical Institute, New Haven, CT (G.I.S.)
| | - Tobias C Walther
- Harvard University, Howard Hughes Medical Institute, Boston, MA (T.C.W.)
| | - Jue Chen
- National Institutes of Health/National, Heart, Lung, and Blood Institute, Bethesda, MD (Z.S.G., M.O., L.S.-L., J.C.).
| |
Collapse
|
25
|
Hu X, Matsumoto K, Jung RS, Weston TA, Heizer PJ, He C, Sandoval NP, Allan CM, Tu Y, Vinters HV, Liau LM, Ellison RM, Morales JE, Baufeld LJ, Bayley NA, He L, Betsholtz C, Beigneux AP, Nathanson DA, Gerhardt H, Young SG, Fong LG, Jiang H. GPIHBP1 expression in gliomas promotes utilization of lipoprotein-derived nutrients. eLife 2019; 8:e47178. [PMID: 31169500 PMCID: PMC6594755 DOI: 10.7554/elife.47178] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/05/2019] [Indexed: 12/25/2022] Open
Abstract
GPIHBP1, a GPI-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) within the subendothelial spaces and shuttles it to the capillary lumen. GPIHBP1-bound LPL is essential for the margination of triglyceride-rich lipoproteins (TRLs) along capillaries, allowing the lipolytic processing of TRLs to proceed. In peripheral tissues, the intravascular processing of TRLs by the GPIHBP1-LPL complex is crucial for the generation of lipid nutrients for adjacent parenchymal cells. GPIHBP1 is absent from the capillaries of the brain, which uses glucose for fuel; however, GPIHBP1 is expressed in the capillaries of mouse and human gliomas. Importantly, the GPIHBP1 in glioma capillaries captures locally produced LPL. We use NanoSIMS imaging to show that TRLs marginate along glioma capillaries and that there is uptake of TRL-derived lipid nutrients by surrounding glioma cells. Thus, GPIHBP1 expression in gliomas facilitates TRL processing and provides a source of lipid nutrients for glioma cells.
Collapse
Affiliation(s)
- Xuchen Hu
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Ken Matsumoto
- VIB-KU Leuven Center for Cancer Biology (CCB)LeuvenBelgium
| | - Rachel S Jung
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Thomas A Weston
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Patrick J Heizer
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Cuiwen He
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Norma P Sandoval
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Christopher M Allan
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Yiping Tu
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Rochelle M Ellison
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Jazmin E Morales
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Lynn J Baufeld
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Nicholas A Bayley
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsalaSweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsalaSweden
- Integrated Cardio Metabolic Centre (ICMC)Karolinska InstitutetHuddingeSweden
| | - Anne P Beigneux
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Holger Gerhardt
- VIB-KU Leuven Center for Cancer Biology (CCB)LeuvenBelgium
- Max Delbrück Center for Molecular MedicineBerlinGermany
| | - Stephen G Young
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Loren G Fong
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Haibo Jiang
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- School of Molecular SciencesUniversity of Western AustraliaPerthAustralia
| |
Collapse
|
26
|
Abstract
Imaging techniques greatly facilitate the comprehensive knowledge of biological systems. Although imaging methodology for biomacromolecules such as protein and nucleic acids has been long established, microscopic techniques and contrast mechanisms are relatively limited for small biomolecules, which are equally important participants in biological processes. Recent developments in Raman imaging, including both microscopy and tailored vibrational tags, have created exciting opportunities for noninvasive imaging of small biomolecules in living cells, tissues, and organisms. Here, we summarize the principle and workflow of small-biomolecule imaging by Raman microscopy. Then, we review recent efforts in imaging, for example, lipids, metabolites, and drugs. The unique advantage of Raman imaging has been manifested in a variety of applications that have provided novel biological insights.
Collapse
Affiliation(s)
- Yihui Shen
- Department of Chemistry, Columbia University, New York, NY 10027, USA;
| | - Fanghao Hu
- Department of Chemistry, Columbia University, New York, NY 10027, USA;
| | - Wei Min
- Department of Chemistry, Columbia University, New York, NY 10027, USA;
| |
Collapse
|
27
|
He C, Hu X, Weston TA, Jung RS, Heizer P, Tu Y, Ellison R, Matsumoto K, Gerhardt H, Tontonoz P, Fong LG, Young SG, Jiang H. NanoSIMS imaging reveals unexpected heterogeneity in nutrient uptake by brown adipocytes. Biochem Biophys Res Commun 2018; 504:899-902. [PMID: 30224066 DOI: 10.1016/j.bbrc.2018.09.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/08/2018] [Indexed: 11/16/2022]
Abstract
Heterogeneity in the metabolic properties of adipocytes in white adipose tissue has been well documented. We sought to investigate metabolic heterogeneity in adipocytes of brown adipose tissue (BAT), focusing on heterogeneity in nutrient uptake. To explore the possibility of metabolic heterogeneity in brown adipocytes, we used nanoscale secondary ion mass spectrometry (NanoSIMS) to quantify uptake of lipids in adipocytes interscapular BAT and perivascular adipose tissue (PVAT) after an intravenous injection of triglyceride-rich lipoproteins (TRLs) containing [2H]triglycerides (2H-TRLs). The uptake of deuterated lipids into brown adipocytes was quantified by NanoSIMS. We also examined 13C enrichment in brown adipocytes after administering [13C]glucose or 13C-labeled mixed fatty acids by gastric gavage. The uptake of 2H-TRLs-derived lipids into brown adipocytes was heterogeneous, with 2H enrichment in adjacent adipocytes varying by more than fourfold. We also observed substantial heterogeneity in 13C enrichment in adjacent brown adipocytes after administering [13C]glucose or [13C]fatty acids by gastric gavage. The uptake of nutrients by adjacent brown adipocytes within a single depot is variable, suggesting that there is heterogeneity in the metabolic properties of brown adipocytes.
Collapse
Affiliation(s)
- Cuiwen He
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Xuchen Hu
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Thomas A Weston
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Rachel S Jung
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Patrick Heizer
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Yiping Tu
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Rochelle Ellison
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Ken Matsumoto
- VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Holger Gerhardt
- VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Peter Tontonoz
- Departments of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Loren G Fong
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Stephen G Young
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA; Departments of Human Genetics, University of California, Los Angeles, CA, 90095, USA.
| | - Haibo Jiang
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA; School of Molecular Sciences and Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth, 6009, Australia.
| |
Collapse
|
28
|
Laguerre A, Schultz C. Novel lipid tools and probes for biological investigations. Curr Opin Cell Biol 2018; 53:97-104. [PMID: 30015291 DOI: 10.1016/j.ceb.2018.06.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/12/2018] [Accepted: 06/29/2018] [Indexed: 12/21/2022]
Abstract
We present the latest advances in lipid tool development for studying cellular membrane trafficking and metabolism. We focus on chemical modifications that are introduced to natural lipid structures. The new functionalities are used to follow and interfere with lipid dynamics in intact cells.
Collapse
Affiliation(s)
- Aurélien Laguerre
- Dept. of Physiology & Pharmacology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Carsten Schultz
- Dept. of Physiology & Pharmacology, Oregon Health and Science University (OHSU), Portland, OR, USA; European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Unit, 69117 Heidelberg, Germany.
| |
Collapse
|
29
|
He C, Weston TA, Jung RS, Heizer P, Larsson M, Hu X, Allan CM, Tontonoz P, Reue K, Beigneux AP, Ploug M, Holme A, Kilburn M, Guagliardo P, Ford DA, Fong LG, Young SG, Jiang H. NanoSIMS Analysis of Intravascular Lipolysis and Lipid Movement across Capillaries and into Cardiomyocytes. Cell Metab 2018; 27:1055-1066.e3. [PMID: 29719224 PMCID: PMC5945212 DOI: 10.1016/j.cmet.2018.03.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/16/2018] [Accepted: 03/24/2018] [Indexed: 10/17/2022]
Abstract
The processing of triglyceride-rich lipoproteins (TRLs) in capillaries provides lipids for vital tissues, but our understanding of TRL metabolism is limited, in part because TRL processing and lipid movement have never been visualized. To investigate the movement of TRL-derived lipids in the heart, mice were given an injection of [2H]triglyceride-enriched TRLs, and the movement of 2H-labeled lipids across capillaries and into cardiomyocytes was examined by NanoSIMS. TRL processing and lipid movement in tissues were extremely rapid. Within 30 s, TRL-derived lipids appeared in the subendothelial spaces and in the lipid droplets and mitochondria of cardiomyocytes. Enrichment of 2H in capillary endothelial cells was not greater than in cardiomyocytes, implying that endothelial cells may not be a control point for lipid movement into cardiomyocytes. Remarkably, a deficiency of the putative fatty acid transport protein CD36, which is expressed highly in capillary endothelial cells, did not impede entry of TRL-derived lipids into cardiomyocytes.
Collapse
Affiliation(s)
- Cuiwen He
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas A Weston
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rachel S Jung
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Patrick Heizer
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mikael Larsson
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xuchen Hu
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christopher M Allan
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Karen Reue
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Anne P Beigneux
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Holme
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia
| | - Matthew Kilburn
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia
| | - Paul Guagliardo
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Center for Cardiovascular Research, Saint Louis University, St. Louis, MO 63104, USA
| | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Haibo Jiang
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia.
| |
Collapse
|
30
|
Jiang H, Passarelli MK, Munro PMG, Kilburn MR, West A, Dollery CT, Gilmore IS, Rakowska PD. High-resolution sub-cellular imaging by correlative NanoSIMS and electron microscopy of amiodarone internalisation by lung macrophages as evidence for drug-induced phospholipidosis. Chem Commun (Camb) 2018; 53:1506-1509. [PMID: 28085162 DOI: 10.1039/c6cc08549k] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Correlative NanoSIMS and EM imaging of amiodarone-treated macrophages shows the internalisation of the drug at a sub-cellular level and reveals its accumulation within the lysosomes, providing direct evidence for amiodarone-induced phospholipidosis. Chemical fixation using tannic acid effectively seals cellular membranes aiding intracellular retention of diffusible drugs.
Collapse
Affiliation(s)
- Haibo Jiang
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Melissa K Passarelli
- National Centre of Excellence in Mass Spectrometry Imaging (NiCE-MSI), National Physical Laboratory, Hampton Road, Teddington, Middlesex TW11 0LW, UK.
| | - Peter M G Munro
- Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Matt R Kilburn
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Andrew West
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Colin T Dollery
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Ian S Gilmore
- National Centre of Excellence in Mass Spectrometry Imaging (NiCE-MSI), National Physical Laboratory, Hampton Road, Teddington, Middlesex TW11 0LW, UK.
| | - Paulina D Rakowska
- National Centre of Excellence in Mass Spectrometry Imaging (NiCE-MSI), National Physical Laboratory, Hampton Road, Teddington, Middlesex TW11 0LW, UK.
| |
Collapse
|
31
|
Zhang L, Vertes A. Einzelzell‐Massenspektrometrie zur Untersuchung zellulärer Heterogenität. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201709719] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Linwen Zhang
- Department of Chemistry The George Washington University Washington DC 20052 USA
| | - Akos Vertes
- Department of Chemistry The George Washington University Washington DC 20052 USA
| |
Collapse
|
32
|
Zhang L, Vertes A. Single‐Cell Mass Spectrometry Approaches to Explore Cellular Heterogeneity. Angew Chem Int Ed Engl 2018; 57:4466-4477. [DOI: 10.1002/anie.201709719] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/27/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Linwen Zhang
- Department of Chemistry The George Washington University Washington DC 20052 USA
| | - Akos Vertes
- Department of Chemistry The George Washington University Washington DC 20052 USA
| |
Collapse
|
33
|
Welte MA, Gould AP. Lipid droplet functions beyond energy storage. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1260-1272. [PMID: 28735096 PMCID: PMC5595650 DOI: 10.1016/j.bbalip.2017.07.006] [Citation(s) in RCA: 353] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/17/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Lipid droplets are cytoplasmic organelles that store neutral lipids and are critically important for energy metabolism. Their function in energy storage is firmly established and increasingly well characterized. However, emerging evidence indicates that lipid droplets also play important and diverse roles in the cellular handling of lipids and proteins that may not be directly related to energy homeostasis. Lipid handling roles of droplets include the storage of hydrophobic vitamin and signaling precursors, and the management of endoplasmic reticulum and oxidative stress. Roles of lipid droplets in protein handling encompass functions in the maturation, storage, and turnover of cellular and viral polypeptides. Other potential roles of lipid droplets may be connected with their intracellular motility and, in some cases, their nuclear localization. This diversity highlights that lipid droplets are very adaptable organelles, performing different functions in different biological contexts. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Michael A Welte
- Department of Biology, University of Rochester, Rochester, NY, United States.
| | | |
Collapse
|
34
|
High-resolution imaging and quantification of plasma membrane cholesterol by NanoSIMS. Proc Natl Acad Sci U S A 2017; 114:2000-2005. [PMID: 28167768 DOI: 10.1073/pnas.1621432114] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cholesterol is a crucial lipid within the plasma membrane of mammalian cells. Recent biochemical studies showed that one pool of cholesterol in the plasma membrane is "accessible" to binding by a modified version of the cytolysin perfringolysin O (PFO*), whereas another pool is sequestered by sphingomyelin and cannot be bound by PFO* unless the sphingomyelin is destroyed with sphingomyelinase (SMase). Thus far, it has been unclear whether PFO* and related cholesterol-binding proteins bind uniformly to the plasma membrane or bind preferentially to specific domains or morphologic features on the plasma membrane. Here, we used nanoscale secondary ion mass spectrometry (NanoSIMS) imaging, in combination with 15N-labeled cholesterol-binding proteins (PFO* and ALO-D4, a modified anthrolysin O), to generate high-resolution images of cholesterol distribution in the plasma membrane of Chinese hamster ovary (CHO) cells. The NanoSIMS images revealed preferential binding of PFO* and ALO-D4 to microvilli on the plasma membrane; lower amounts of binding were detectable in regions of the plasma membrane lacking microvilli. The binding of ALO-D4 to the plasma membrane was virtually eliminated when cholesterol stores were depleted with methyl-β-cyclodextrin. When cells were treated with SMase, the binding of ALO-D4 to cells increased, largely due to increased binding to microvilli. Remarkably, lysenin (a sphingomyelin-binding protein) also bound preferentially to microvilli. Thus, high-resolution images of lipid-binding proteins on CHO cells can be acquired with NanoSIMS imaging. These images demonstrate that accessible cholesterol, as judged by PFO* or ALO-D4 binding, is not evenly distributed over the entire plasma membrane but instead is highly enriched on microvilli.
Collapse
|
35
|
He C, Fong LG, Young SG, Jiang H. NanoSIMS imaging: an approach for visualizing and quantifying lipids in cells and tissues. J Investig Med 2016; 65:669-672. [PMID: 27793974 DOI: 10.1136/jim-2016-000239] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2016] [Indexed: 12/22/2022]
Abstract
Over the past few decades, several approaches have been used to image lipids in cells and tissues, but most have limited spatial resolution and sensitivity. Here, we discuss a relatively new approach, nanoscale secondary ion mass spectrometry imaging, that makes it possible to visualize lipids in cells and tissues in a quantitative fashion and with high spatial resolution and high sensitivity.
Collapse
Affiliation(s)
- Cuiwen He
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Loren G Fong
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Stephen G Young
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA.,Departments of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Haibo Jiang
- Centre for Microscopy, Characterisation, and Analysis, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
36
|
NanoSIMS chemical imaging combined with correlative microscopy for biological sample analysis. Curr Opin Biotechnol 2016; 41:130-135. [PMID: 27506876 DOI: 10.1016/j.copbio.2016.06.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/17/2016] [Accepted: 06/23/2016] [Indexed: 12/18/2022]
Abstract
Nano-scale Secondary Ion Mass Spectrometry (NanoSIMS) is one of the most powerful in situ elemental and isotopic analysis techniques available to biologists. The combination of stable isotope probing with NanoSIMS (nanoSIP) has opened up new avenues for biological studies over the past decade. However, due to limitations inherent with any analytical methodology, additional information from correlative techniques is usually required to address real biological questions. Here we review recent developments in correlative analysis applied to complex biological systems: first, high-resolution tracking of molecules (e.g. peptides, lipids) by correlation with electron microscopy and atomic force microscopy; second, identification of a specific microbial taxon with fluorescence in situ hybridization and quantification of its metabolic capacities; and, third, molecular specific imaging with new probes.
Collapse
|
37
|
Fong LG, Young SG, Beigneux AP, Bensadoun A, Oberer M, Jiang H, Ploug M. GPIHBP1 and Plasma Triglyceride Metabolism. Trends Endocrinol Metab 2016; 27:455-469. [PMID: 27185325 PMCID: PMC4927088 DOI: 10.1016/j.tem.2016.04.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 10/21/2022]
Abstract
GPIHBP1, a GPI-anchored protein in capillary endothelial cells, is crucial for the lipolytic processing of triglyceride-rich lipoproteins (TRLs). GPIHBP1 shuttles lipoprotein lipase (LPL) to its site of action in the capillary lumen and is essential for the margination of TRLs along capillaries - such that lipolytic processing can proceed. GPIHBP1 also reduces the unfolding of the LPL catalytic domain, thereby stabilizing LPL catalytic activity. Many different GPIHBP1 mutations have been identified in patients with severe hypertriglyceridemia (chylomicronemia), the majority of which interfere with folding of the protein and abolish its capacity to bind and transport LPL. The discovery of GPIHBP1 has substantially revised our understanding of intravascular triglyceride metabolism but has also raised many new questions for future research.
Collapse
Affiliation(s)
- Loren G Fong
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Stephen G Young
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Anne P Beigneux
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - André Bensadoun
- Division of Nutritional Science, Cornell University, Ithaca, NY 14853, USA
| | - Monika Oberer
- Institute of Molecular Biosciences, University of Graz and BioTechMed, Graz, Austria
| | - Haibo Jiang
- Centre for Microscopy, Characterisation, and Analysis, The University of Western Australia
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, 2200 Copenhagen N, Denmark; Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 220 Copenhagen N, Denmark.
| |
Collapse
|
38
|
Three-dimensional imaging of cholesterol and sphingolipids within a Madin-Darby canine kidney cell. Biointerphases 2016; 11:02A309. [PMID: 26746168 DOI: 10.1116/1.4939681] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Metabolic stable isotope incorporation and secondary ion mass spectrometry (SIMS) depth profiling performed on a Cameca NanoSIMS 50 were used to image the (18)O-cholesterol and (15)N-sphingolipid distributions within a portion of a Madin-Darby canine kidney (MDCK) cell. Three-dimensional representations of the component-specific isotope distributions show clearly defined regions of (18)O-cholesterol and (15)N-sphingolipid enrichment that seem to be separate subcellular compartments. The low levels of nitrogen-containing secondary ions detected at the (18)O-enriched regions suggest that these (18)O-cholesterol-rich structures may be lipid droplets, which have a core consisting of cholesterol esters and triacylglycerides.
Collapse
|
39
|
Dong Y, Guella G, Franceschi P. Impact of tissue surface properties on the desorption electrospray ionization imaging of organic acids in grapevine stem. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2016; 30:711-718. [PMID: 26864524 DOI: 10.1002/rcm.7495] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/10/2015] [Accepted: 12/20/2015] [Indexed: 06/05/2023]
Abstract
RATIONALE Desorption electrospray ionization (DESI) imaging is a fast analytical technique used to assess spatially resolved biological processes over unmodified sample surfaces. Although DESI profiling experiments have demonstrated that the properties of the sample surface significantly affect the outcomes of DESI analyses, the potential implications of these phenomena in imaging applications have not yet been explored extensively. METHODS The distribution of endogenous and exogenous organic acids in pith and out pith region of grapevine stems was investigated by using DESI imaging, ion chromatography and direct infusion methods. Several common normalization strategies to account for the surface effect, including TIC normalization, addition of the internal standard in the spray solvent and deposition of the standard over the sample surface, were critically evaluated. RESULTS DESI imaging results show that, in our case, the measured distributions of these small organic acids are not consistent with their 'true' localizations within the tissues. Furthermore, our results indicate that the common normalization strategies are not able to completely compensate for the observed surface effect. CONCLUSIONS Variations in the tissue surface properties across the tissue sample can greatly affect the semi-quantitative detection of organic acids. Attention should be paid when interpreting DESI imaging results and an independent analytical validation step is important in untargeted DESI imaging investigations.
Collapse
Affiliation(s)
- Yonghui Dong
- Biostatistics and Data Management, Research and Innovation Centre - Fondazione Edmund Mach, S. Michele all'Adige, (TN), Italy
| | - Graziano Guella
- Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, Povo, (TN), Italy
| | - Pietro Franceschi
- Biostatistics and Data Management, Research and Innovation Centre - Fondazione Edmund Mach, S. Michele all'Adige, (TN), Italy
| |
Collapse
|
40
|
Pascual F, Coleman RA. Fuel availability and fate in cardiac metabolism: A tale of two substrates. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1425-33. [PMID: 26993579 DOI: 10.1016/j.bbalip.2016.03.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/12/2022]
Abstract
The heart's extraordinary metabolic flexibility allows it to adapt to normal changes in physiology in order to preserve its function. Alterations in the metabolic profile of the heart have also been attributed to pathological conditions such as ischemia and hypertrophy; however, research during the past decade has established that cardiac metabolic adaptations can precede the onset of pathologies. It is therefore critical to understand how changes in cardiac substrate availability and use trigger events that ultimately result in heart dysfunction. This review examines the mechanisms by which the heart obtains fuels from the circulation or from mobilization of intracellular stores. We next describe experimental models that exhibit either an increase in glucose use or a decrease in FA oxidation, and how these aberrant conditions affect cardiac metabolism and function. Finally, we highlight the importance of alternative, relatively under-investigated strategies for the treatment of heart failure. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.
Collapse
Affiliation(s)
- Florencia Pascual
- Department of Nutrition, University of North Carolina at Chapel Hill, 27599, USA.
| | - Rosalind A Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, 27599, USA.
| |
Collapse
|
41
|
Kohnhorst CL, Schmitt DL, Sundaram A, An S. Subcellular functions of proteins under fluorescence single-cell microscopy. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1864:77-84. [PMID: 26025769 PMCID: PMC5679394 DOI: 10.1016/j.bbapap.2015.05.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 05/08/2015] [Accepted: 05/18/2015] [Indexed: 11/25/2022]
Abstract
A cell is a highly organized, dynamic, and intricate biological entity orchestrated by a myriad of proteins and their self-assemblies. Because a protein's actions depend on its coordination in both space and time, our curiosity about protein functions has extended from the test tube into the intracellular space of the cell. Accordingly, modern technological developments and advances in enzymology have been geared towards analyzing protein functions within intact single cells. We discuss here how fluorescence single-cell microscopy has been employed to identify subcellular locations of proteins, detect reversible protein-protein interactions, and measure protein activity and kinetics in living cells. Considering that fluorescence single-cell microscopy has been only recently recognized as a primary technique in enzymology, its potentials and outcomes in studying intracellular protein functions are projected to be immensely useful and enlightening. We anticipate that this review would inspire many investigators to study their proteins of interest beyond the conventional boundary of specific disciplines. This article is part of a Special Issue entitled: Physiological Enzymology and Protein Functions.
Collapse
Affiliation(s)
- Casey L Kohnhorst
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Danielle L Schmitt
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Anand Sundaram
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Songon An
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
42
|
Daemen S, van Zandvoort MAMJ, Parekh SH, Hesselink MKC. Microscopy tools for the investigation of intracellular lipid storage and dynamics. Mol Metab 2015; 5:153-163. [PMID: 26977387 PMCID: PMC4770264 DOI: 10.1016/j.molmet.2015.12.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 12/19/2015] [Accepted: 12/27/2015] [Indexed: 12/01/2022] Open
Abstract
Background Excess storage of lipids in ectopic tissues, such as skeletal muscle, liver, and heart, seems to associate closely with metabolic abnormalities and cardiac disease. Intracellular lipid storage occurs in lipid droplets, which have gained attention as active organelles in cellular metabolism. Recent developments in high-resolution microscopy and microscopic spectroscopy have opened up new avenues to examine the physiology and biochemistry of intracellular lipids. Scope of review The aim of this review is to give an overview of recent technical advances in microscopy, and its application for the visualization, identification, and quantification of intracellular lipids, with special focus to lipid droplets. In addition, we attempt to summarize the probes currently available for the visualization of lipids. Major conclusions The continuous development of lipid probes in combination with the rapid development of microscopic techniques can provide new insights in the role and dynamics of intracellular lipids. Moreover, in situ identification of intracellular lipids is now possible and promises to add a new dimensionality to analysis of lipid biochemistry, and its relation to (patho)physiology.
Collapse
Key Words
- BODIPY, Boron-dipyrromethene
- CARS, coherent anti-stokes Raman scattering
- CLEM, correlative light electron microscopy
- CLSM, confocal laser scanning microscopy
- DIC, differential interference microscopy
- FA, fatty acid
- FIB-SEM, focused ion beam scanning electron microscopy
- FLIP, fluorescence loss in photobleaching
- FRAP, fluorescent recovery after photobleaching
- FRET, fluorescence resonance energy transfer
- Fluorescent lipid probes
- GFP, green fluorescent protein
- HCV, hepatitis C virus
- LD, lipid droplet
- Lipid droplets
- Live-cell imaging
- Metabolic disease
- NBD, nitro-benzoxadiazolyl
- PALM, photoactivation localization microscopy
- SBEM, serial block face scanning electron microscopy
- SIMS, Secondary Ion Mass Spectrometry
- SRS, Stimulated Raman Scattering
- STED, stimulated emission depletion
- STORM, stochastic optical reconstruction microscopy
- Super-resolution
- TAG, triacylglycerol
- TEM, transmission electron microscopy
- TOF-SIMS, time-of-flight SIMS
- TPLSM, two-photon laser scanning microscopy
- Vibrational microscopy
Collapse
Affiliation(s)
- Sabine Daemen
- Department of Human Movement Sciences and Human Biology, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.
| | - Marc A M J van Zandvoort
- Department of Genetics and Molecular Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands; Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.
| | - Sapun H Parekh
- Department of Molecular Spectroscopy, Max Planck Institute for Polymer Research, Mainz, Germany.
| | - Matthijs K C Hesselink
- Department of Human Movement Sciences and Human Biology, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|