1
|
Shao C, Wu S, Liu T, Zheng M, Zhi X, Miao J, Hu L, Ge L. Exploring the association between plasma lipids changes and embolization recurrence following intravenous thrombolysis with alteplase in acute ischemic stroke based on lipidomics. J Stroke Cerebrovasc Dis 2025; 34:108310. [PMID: 40209964 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Alteplase is a widely used thrombolytic therapy for acute ischemic stroke (AIS). However, the efficacy of intravenous thrombolysis (IVT) with alteplase varies due to individual factors, and some patients are prone to embolization recurrence (ER) after IVT. The mechanisms underlying these variations are not fully understood. METHODS In this study, we analyzed changes in plasma lipid profiles of AIS patients treated with alteplase based on lipidomics, focusing on the association between plasma lipids changes and embolization recurrence. RESULTS Our findings revealed significant changes in several lipid species in patients who experienced ER, including triglycerides (TG), diglycerides (DG), phosphatidylcholine (PC), and sphingomyelin (SM). Notably, most TG and DG levels in ER group were significantly upregulated, PC and SM levels were downregulated. However, most PC was upregulated 24 h after IVT treatment, while DG, SM, TG remained downregulated. These differential lipid metabolites are mainly involved in lipid metabolism, immune response inflammation-related, and insulin resistance signaling pathways. CONCLUSION This study demonstrates that lipid alterations, particularly in TG, DG, PC, and SM, may be associated with ER after alteplase treatment. However, further studies are needed to elucidate the exact mechanisms through which these lipids influence the onset and recovery of AIS.
Collapse
Affiliation(s)
- Chuanfeng Shao
- College of pharmacy, Wenzhou Medical University, Wenzhou, 325035, China; Department of pharmacy, The Third People's Hospital of Yueqing, Yueqing, 325600, China
| | - Saizhen Wu
- Department of Neurology, Clinical Medicine, Intensive Care and Clinical Laboratory of the Affiliated Yueqing People's Hospital, Yueqing, 325600, China
| | - Tingting Liu
- Department of Neurology, Clinical Medicine, Intensive Care and Clinical Laboratory of the Affiliated Yueqing People's Hospital, Yueqing, 325600, China
| | - Ming Zheng
- Department of Neurology, Clinical Medicine, Intensive Care and Clinical Laboratory of the Affiliated Yueqing People's Hospital, Yueqing, 325600, China
| | - Xiaoyang Zhi
- Department of Neurology, Clinical Medicine, Intensive Care and Clinical Laboratory of the Affiliated Yueqing People's Hospital, Yueqing, 325600, China
| | - Jinli Miao
- The Yangtze River Delta Biological Medicine Research and Development Center of Zhejiang Province, Yangtze Delta Region Institution of Tsinghua University, Hangzhou, Zhejiang, 314006, China
| | - Lufeng Hu
- College of pharmacy, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Lingli Ge
- Department of pharmacy, The People's Hospital of Lucheng, Wenzhou, 325000, China.
| |
Collapse
|
2
|
Nieddu G, Formato M, Lepedda AJ. Deciphering Acute Coronary Syndromes Pathobiology Through Proteomics. J Cardiovasc Dev Dis 2025; 12:188. [PMID: 40422959 DOI: 10.3390/jcdd12050188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
Acute coronary syndrome (ACS) refers to a spectrum of conditions characterized by a sudden decrease in blood flow to the heart. This includes unstable angina, the mildest form, as well as non-ST- and ST-segment elevation myocardial infarction. The primary cause of ACS is typically the rupture or erosion of an atherosclerotic plaque in a coronary artery, resulting in the formation of a blood clot that can, partially or completely, block the blood flow to the heart muscle. The ongoing discovery and comprehension of emerging biomarkers for atherosclerosis could enhance our capacity to predict future events, particularly when integrated alongside traditional risk factors in assessing overall risk profiles. With advancements in proteomic technologies, large-scale approaches have been increasingly instrumental in unraveling pathways implicated in atherosclerotic degeneration and identifying novel circulating markers, which may serve as early diagnostic indicators or targets for innovative therapies. Over recent decades, numerous matrices including plasma, urine, microparticles, lipoproteins, atherosclerotic plaque extracts and secretomes, as well as thrombi, have been examined to address these questions. Furthermore, proteomics has been applied to various experimental models of atherosclerosis to deepen our understanding of the mechanisms underlying atherogenesis. This review offers a critical overview of the past two decades of untargeted omics research focused on identifying circulating and tissue biomarkers relevant to ACS.
Collapse
Affiliation(s)
- Gabriele Nieddu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Marilena Formato
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | | |
Collapse
|
3
|
Bookmeyer CHM, Correig FX, Masana L, Magni P, Yanes Ó, Vinaixa M. Advancing atherosclerosis research: The Power of lipid imaging with MALDI-MSI. Atherosclerosis 2025; 403:119130. [PMID: 40059002 DOI: 10.1016/j.atherosclerosis.2025.119130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/20/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease that is one of the leading causes of mortality globally. It is characterized by the formation of atheromatous plaques in the intima layer of larger arteries. The (fibro-)fatty plaques usually develop asymptomatically within the vessel until a serious event such as myocardial infarction or stroke occurs. Lipids play a pivotal role in disease progression, but while the causal role of cholesterol is beyond doubt, the distribution of numerous other lipids within the heterogeneous layers of atherosclerotic plaques, and their biological function remain unclear. A deeper understanding of the pathophysiological progression of the disease for prognostics, diagnostics, treatment, and prevention is of great need. Mass spectrometry imaging (MSI), in particular with matrix-assisted laser desorption/ionization (MALDI) offers an unprecedented untargeted characterization of the physiological microenvironment, unraveling the spatial distribution of numerous biochemical compounds. MALDI-MSI offers an advantageous balance of sample preparation, chemical sensitivity, and spatial resolution, and thus has been established as a key technology in modern biomedical analysis. This review focuses on the analysis of lipids in atherosclerotic lesions with MALDI-MSI, for which the past years showed major developments in the spatial characterization of lipids and their interaction within atherosclerotic plaques. We will cover main contributions with a focus on the recent decade, elaborate possibilities, limitations, main findings, and recent developments from sample handling to instrumentation, and estimate current challenges and potentials of MALDI-MSI with respect to a clinical application.
Collapse
Affiliation(s)
- Christoph H M Bookmeyer
- Universitat Rovira i Virgili, Department of Electronic Engineering, Metabolomics Interdisciplinary Laboratory, Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain.
| | - F Xavier Correig
- Universitat Rovira i Virgili, Department of Electronic Engineering, Metabolomics Interdisciplinary Laboratory, Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Luis Masana
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; Universitat Rovira i Virgili, Research Unit on Lipids and Atherosclerosis, Reus, Spain
| | - Paolo Magni
- Dept. of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Italy
| | - Óscar Yanes
- Universitat Rovira i Virgili, Department of Electronic Engineering, Metabolomics Interdisciplinary Laboratory, Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Maria Vinaixa
- Universitat Rovira i Virgili, Department of Electronic Engineering, Metabolomics Interdisciplinary Laboratory, Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.
| |
Collapse
|
4
|
Wan J, Sun Z, Feng X, Zhou P, Macho MTN, Jiao Z, Cao H, Zhang C, Lin R, Zhang X, Fan M, Zhang N, Zhang J, Liu H, Li J, Guan S. Spatial omics strategies for investigating human carotid atherosclerotic disease. Clin Transl Med 2025; 15:e70277. [PMID: 40156163 PMCID: PMC11953060 DOI: 10.1002/ctm2.70277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/01/2025] Open
Abstract
Atherosclerosis is a chronic inflammatory condition of the arteries, marked by the development of plaques within the arterial intima. The rupture of unstable plaques can lead to thrombosis, downstream vessel occlusion and serious clinical events. The composition of atherosclerotic plaques is complex and highly heterogeneous, posing challenges for their study. The current pathology and histological subtype classification of plaques may fail to fully encompass the microscopic molecular components in the tissue, the disease progress in various stages of atherosclerosis and the potential mechanism of plaque rupture. However, spatial mapping of the heterogeneity in plaque tissue components can enhance our understanding of these lesions. Despite the considerable progress made by traditional omics in the field of disease research, and its status as an indispensable technology, there remain inherent limitations in the investigation of minute molecular. In recent years, spatial omics techniques have advanced significantly, enabling the visualisation and analysis of specific components within plaques that may serve as causal targets associated with disease progression. The effective application of spatial omics in both research and clinical settings represents a promising area for further exploration. This review focuses on the recent advancements and findings related to spatial omics in the study of extracranial carotid atherosclerotic cerebrovascular disease. Spatial omics analysis of atherosclerotic plaques can facilitate the detection of biomarkers with diagnostic significance or potential relevance to disease, offering new methods and insights into the diagnosis of atherosclerosis and its complications.
Collapse
Affiliation(s)
- Jiaxin Wan
- Department of Neurointerventionthe First Affiliated Hospital of Zhengzhou, UniversityHenan Provincial Neurointerventional Engineering Research CenterZhengzhouHenanChina
| | - Zhi Sun
- Department of Pharmacythe First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. Henan Engineering Research Center of Clinical Mass Spectrometry for Precision MedicineZhengzhouHenan ProvinceChina
| | - Xueqiong Feng
- Department of Pharmacythe First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. Henan Engineering Research Center of Clinical Mass Spectrometry for Precision MedicineZhengzhouHenan ProvinceChina
| | - Peipei Zhou
- Department of Pharmacythe First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. Henan Engineering Research Center of Clinical Mass Spectrometry for Precision MedicineZhengzhouHenan ProvinceChina
| | - Mateus T. N. Macho
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhouyang Jiao
- Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hui Cao
- Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chuang Zhang
- Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Rijin Lin
- Department of Neurointerventionthe First Affiliated Hospital of Zhengzhou, UniversityHenan Provincial Neurointerventional Engineering Research CenterZhengzhouHenanChina
| | - Xiaowen Zhang
- Department of Neurointerventionthe First Affiliated Hospital of Zhengzhou, UniversityHenan Provincial Neurointerventional Engineering Research CenterZhengzhouHenanChina
| | - Mengyan Fan
- Department of Neurointerventionthe First Affiliated Hospital of Zhengzhou, UniversityHenan Provincial Neurointerventional Engineering Research CenterZhengzhouHenanChina
| | - Nan Zhang
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou, UniversityZhengzhouChina
| | - Jiamei Zhang
- Department of Neurointerventionthe First Affiliated Hospital of Zhengzhou, UniversityHenan Provincial Neurointerventional Engineering Research CenterZhengzhouHenanChina
| | - Huixiang Liu
- Department of Neurointerventionthe First Affiliated Hospital of Zhengzhou, UniversityHenan Provincial Neurointerventional Engineering Research CenterZhengzhouHenanChina
| | - Jing Li
- Department of Neurointerventionthe First Affiliated Hospital of Zhengzhou, UniversityHenan Provincial Neurointerventional Engineering Research CenterZhengzhouHenanChina
| | - Sheng Guan
- Department of Neurointerventionthe First Affiliated Hospital of Zhengzhou, UniversityHenan Provincial Neurointerventional Engineering Research CenterZhengzhouHenanChina
| |
Collapse
|
5
|
Saba L, Cau R, Vergallo R, Kooi ME, Staub D, Faa G, Congiu T, Ntaios G, Wasserman BA, Benson J, Nardi V, Kawakami R, Lanzino G, Virmani R, Libby P. Carotid artery atherosclerosis: mechanisms of instability and clinical implications. Eur Heart J 2025; 46:904-921. [PMID: 39791527 DOI: 10.1093/eurheartj/ehae933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/25/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular disease remains a prominent cause of disability and premature death worldwide. Within this spectrum, carotid artery atherosclerosis is a complex and multifaceted condition, and a prominent precursor of acute ischaemic stroke and other cardiovascular events. The intricate interplay among inflammation, oxidative stress, endothelial dysfunction, lipid metabolism, and immune responses participates in the development of lesions, leading to luminal stenosis and potential plaque instability. Even non-stenotic plaques can precipitate a sudden cerebrovascular event, regardless of the degree of luminal encroachment. In this context, carotid imaging modalities have proved their efficacy in providing in vivo characterization of plaque features, contributing substantially to patient risk stratification and clinical management. This review emphasizes the importance of identifying high-risk individuals by use of current imaging modalities, biomarkers, and risk stratification tools. Such approaches inform early intervention and the implementation of personalized therapeutic strategies, ultimately enhancing patient outcomes in the realm of cardiovascular disease management.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, University of Cagliari, Via Università, 40, 09124 Cagliari, Italy
| | - Riccardo Cau
- Department of Radiology, University of Cagliari, Via Università, 40, 09124 Cagliari, Italy
| | - Rocco Vergallo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - M Eline Kooi
- Department of Radiology and Nuclear Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Daniel Staub
- Vascular Medicine/Angiology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Gavino Faa
- Department of Pathology, University of Cagliari, Cagliari, Italy
| | - Terenzio Congiu
- Department of Pathology, University of Cagliari, Cagliari, Italy
| | - George Ntaios
- Department of Internal Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Larissa 41132, Greece
| | - Bruce A Wasserman
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, MD, USA
| | - John Benson
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Valentina Nardi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Inc., Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Ding M, Zheng L, Hua X, Chen M, Zhong Q, Huang T, Li P, Yang H. Simultaneous metabolomics and lipidomics analysis based on 4in1 online analysis system reveal metabolic signatures in atherosclerotic mice. Talanta 2025; 283:127109. [PMID: 39481347 DOI: 10.1016/j.talanta.2024.127109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Developing efficient and comprehensive analysis methods for metabolomics and lipidomics in the biological tissues and body fluids is essential for understanding the disease mechanisms. Although various two-dimensional liquid chromatography-mass spectrometry (2D-LC-MS) methods have been proposed to expand metabolite coverage, achieving higher efficiency in integrated metabolomics and lipidomics studies remains a technical challenge. In this work, a novel 4in1 online analysis system with excellent reproducibility and mass accuracy was constructed for metabolomics and lipidomics study in various biological samples from atherosclerotic mice. This system enabled the simultaneous detection in both positive and negative ion modes with extensive polarity separation in a single analytical run. Using the 4in1 online analysis system, we identified distinct but complementary metabolic signatures associated with atherosclerosis in different biological samples. Specifically, a total of 230 and 170 differential metabolites or lipids were detected in mice plasma samples and aortic tissue samples, respectively, including glycerophospholipids, sphingolipids, fatty acyls, glycerolipids, carboxylic acids, and pyrimidine nucleosides. Additionally, atherosclerosis-related metabolic pathways involved in biosynthesis of unsaturated fatty acids, sphingolipid metabolism, cholesterol metabolism, glycerophospholipid metabolism, and choline metabolism further revealed. These findings demonstrate that the novel 4in1 online analysis system is a faithful, stable and powerful tool for comprehensive metabolomics and lipidomics studies in complex biological matrices.
Collapse
Affiliation(s)
- Meng Ding
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing, 211198, China
| | - Luwei Zheng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing, 211198, China
| | - Xiaolin Hua
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing, 211198, China
| | - Mengxuan Chen
- Analytical Applications Center, Shimadzu (China) Co., Ltd., Shanghai, 200233, China
| | - Qisheng Zhong
- Analytical Applications Center, Shimadzu (China) Co., Ltd., Shanghai, 200233, China
| | - Taohong Huang
- Analytical Applications Center, Shimadzu (China) Co., Ltd., Shanghai, 200233, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing, 211198, China.
| | - Hua Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing, 211198, China.
| |
Collapse
|
7
|
Slijkhuis N, Towers M, Claude E, van Soest G. MALDI versus DESI mass spectrometry imaging of lipids in atherosclerotic plaque. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e9927. [PMID: 39435741 DOI: 10.1002/rcm.9927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024]
Abstract
Mass spectrometry imaging (MSI) is a powerful tool for detecting lipids in tissue sections, with matrix-assisted laser desorption/ionization (MALDI) and desorption electrospray ionization (DESI) as its key ionization techniques. In this study, we examine how MALDI compares with state-of-the-art DESI ionization in identifying lipids in heterogeneous samples, specifically atherosclerotic plaques. Carotid plaques (n = 4) from patients undergoing endarterectomy were snap-frozen, stored at -80°C, and then sectioned for MSI analysis and H&E staining. Measurements were conducted using a SYNAPT XS mass spectrometer in positive ion mode, employing MALDI with a 2,5-dihydroxybenzoic acid (DHB) matrix and DESI with a methanol: water (98:2) (v/v) solvent. Our comparison covered spectral profiles, sensitivity, and image quality generated by these two techniques. We found that both MALDI and DESI are highly suitable techniques for detecting a wide range of lipids in atherosclerotic plaque sections. DESI-MSI exhibited higher ion counts for most lipid classes than MALDI-MSI and provided sharper images. MALDI detected larger amounts of ceramide and hexosylceramide species, possibly due to its efficient generation of dehydrated ions. In contrast, DESI showed greater peak intensities of cholesteryl ester and triacylglyceride species than MALDI, consistent with reduced fragmentation. These findings establish the relative merits of DESI and MALDI and demonstrate their complementarity as techniques for lipid research in MSI.
Collapse
Affiliation(s)
- Nuria Slijkhuis
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Gijs van Soest
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
- Department of Precision and Microsystems Engineering, Delft University of Technology, Delft, The Netherlands
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Ntshangase S, Khan S, Bezuidenhout L, Gazárková T, Kaczynski J, Sellers S, Rattray NJ, Newby DE, Hadoke PW, Andrew R. Spatial lipidomic profiles of atherosclerotic plaques: A mass spectrometry imaging study. Talanta 2025; 282:126954. [PMID: 39423636 DOI: 10.1016/j.talanta.2024.126954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
Lipids contribute to atherosclerotic cardiovascular disease but their roles are not fully understood. Spatial lipid composition of atherosclerotic plaques was compared between species focusing on aortic plaques from New Zealand White rabbits and carotid plaques from humans (n = 3), using matrix-assisted laser desorption/ionization mass spectrometry imaging. Histologically discriminant lipids within plaque features (neointima and media in rabbits, and lipid-necrotic core and fibrous cap/tissue in humans) included sphingomyelins, phosphatidylcholines, and cholesteryl esters. There were 67 differential lipids between rabbit plaque features and 199 differential lipids in human, each with variable importance in projection score ≥1.0 and p < 0.05. The lipid profile of plaques in the rabbit model closely mimicked that of human plaques and two key pathways (impact value ≥ 0.1), sphingolipid and glycerophospholipid metabolism, were disrupted by atherosclerosis in both species. Thus, mass spectrometry imaging of spatial biomarkers offers valuable insights into atherosclerosis.
Collapse
Affiliation(s)
- Sphamandla Ntshangase
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Shazia Khan
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Louise Bezuidenhout
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Taťána Gazárková
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Jakub Kaczynski
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Stephanie Sellers
- Centre for Heart Lung Innovation, St Paul's Hospital and University of British Columbia, Vancouver, Canada
| | - Nicholas Jw Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - David E Newby
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Patrick Wf Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Ruth Andrew
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
9
|
Sun M, Otsuka Y, Okada M, Shimma S, Toyoda M. Probe oscillation control in tapping-mode scanning probe electrospray ionization for stabilization of mass spectrometry imaging. Analyst 2024; 149:4011-4019. [PMID: 38953117 DOI: 10.1039/d4an00712c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Mass spectrometry imaging (MSI) is used for visualizing the distribution of components in solid samples, such as biological tissues, and requires a technique to ionize the components from local areas of the sample. Tapping-mode scanning probe electrospray ionization (t-SPESI) uses an oscillating capillary probe to extract components from a local area of a sample with a small volume of solvent and to perform electrospray ionization of those components at high speed. MSI can be conducted by scanning the sample surface with a capillary probe. To ensure stable extraction and ionization for MSI, the probe oscillation during measurements must be understood. In this study, we examined the changes in oscillation amplitude and phase due to the interaction between the oscillating probe and the brain tissue section when the probe tip was dynamically brought close to the sample surface. The changes in the probe oscillation depended on the oscillation frequency and polarity of the bias voltage applied to the solvent because an electrostatic force shifted the frequency of the probe oscillation. These findings suggest that controlling the probe oscillation frequency is important for stabilizing MSI by t-SPESI.
Collapse
Affiliation(s)
- Mengze Sun
- Department of Physics, Graduate School of Science, Osaka University, Japan.
| | - Yoichi Otsuka
- Department of Physics, Graduate School of Science, Osaka University, Japan.
- Forefront Research Center, Graduate School of Science, Osaka University, Japan
| | - Maki Okada
- Department of Physics, Graduate School of Science, Osaka University, Japan.
| | - Shuichi Shimma
- Department of Bioengineering, Graduate School of Engineering, Osaka University, Japan
| | - Michisato Toyoda
- Department of Physics, Graduate School of Science, Osaka University, Japan.
- Forefront Research Center, Graduate School of Science, Osaka University, Japan
| |
Collapse
|
10
|
Malý M, Kučerka O, Bechyňská K, Kočí K, Mandys V, Hajšlová J, Kosek V. Plasma lipidome differences in patients with and without significant carotid plaque. Vascul Pharmacol 2024; 155:107377. [PMID: 38705432 DOI: 10.1016/j.vph.2024.107377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Atherosclerosis is a major cause of ischemic stroke, and early detection of advanced atherosclerosis in the carotid artery is important for reducing morbidity and mortality. What is even more important is not only detection of atherosclerosis but early determination whether the patients are at high risk of an event with adverse effects as the size of the plaque does not necessarily reflect its potential to trigger such events. AIM We studied whether plasma lipidomics profile can be used as a diagnostic tool for stratification of stable or unstable plaques without the need of removing the carotid plaque. METHODS This study used liquid chromatography high-resolution tandem mass spectrometry lipidomics to characterize lipid profiles in patients' plasma and found that patients with significant and complicated (vulnerable) atherosclerotic plaque had distinct lipid profiles compared to those with insignificant plaques. RESULTS The lipid classes that were most predictive of vulnerable plaque were lysophosphoethanolamines, fatty acyl esters of hydroxy fatty acids, free fatty acids, plasmalogens, and triacylglycerols. Most of these compounds were found decreased in plasma of patients with unstable plaques which enabled sufficient performance of a statistical model used for patient stratification. CONCLUSIONS Plasma lipidomes measured by liquid chromatography-mass spectrometry show differences in patients with stable and unstable carotid plaques, therefore these compounds could potentially be used as biomarkers for unstable plaque in future clinical diagnosis.
Collapse
Affiliation(s)
- Martin Malý
- Department of Medicine, First Faculty of Medicine, Charles University in Prague and the Military University Hospital, Prague 16902, Czech Republic
| | - Ondřej Kučerka
- Department of Medicine, First Faculty of Medicine, Charles University in Prague and the Military University Hospital, Prague 16902, Czech Republic; Department of Military Internal Medicine and Military Hygiene, Faculty of Military Health Sciences, University of Defence, Hradec Kralove 50002, Czech Republic
| | - Kamila Bechyňská
- University of Chemistry and Technology, Department of Food Chemistry and Analysis, Technická 3, Prague 6 166 28, Czech Republic
| | - Karolína Kočí
- Department of Medicine, First Faculty of Medicine, Charles University in Prague and the Military University Hospital, Prague 16902, Czech Republic
| | - Václav Mandys
- Department of Pathology, Third Faculty of Medicine, Charles University and the University Hospital Kralovske Vinohrady, Prague 100 00, Czech Republic
| | - Jana Hajšlová
- University of Chemistry and Technology, Department of Food Chemistry and Analysis, Technická 3, Prague 6 166 28, Czech Republic
| | - Vít Kosek
- University of Chemistry and Technology, Department of Food Chemistry and Analysis, Technická 3, Prague 6 166 28, Czech Republic.
| |
Collapse
|
11
|
Che Y, Ren J, Zhao H, Yang Y, Chen Z. Orosomucoid 2 as a biomarker of carotid artery atherosclerosis plaque vulnerability through its generation of reactive oxygen species and lipid accumulation in vascular smooth muscle cells. Biochem Biophys Res Commun 2024; 705:149736. [PMID: 38447392 DOI: 10.1016/j.bbrc.2024.149736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Orosomucoid (ORM) has been reported as a biomarker of carotid atherosclerosis, but the role of ORM 2, a subtype of ORM, in carotid atherosclerotic plaque formation and the underlying mechanism have not been established. METHODS Plasma was collected from patients with carotid artery stenosis (CAS) and healthy participants and assessed using mass spectrometry coupled with isobaric tags for relative and absolute quantification (iTRAQ) technology to identify differentially expressed proteins. The key proteins and related pathways were identified via western blotting, immunohistochemistry, and polymerase chain reaction of carotid artery plaque tissues and in vitro experiments involving vascular smooth muscle cells (VSMCs). RESULTS We screened 33 differentially expressed proteins out of 535 proteins in the plasma. Seventeen proteins showed increased expressions in the CAS groups relative to the healthy groups, while 16 proteins showed decreased expressions during iTRAQ and bioinformatic analysis. The reactive oxygen species metabolic process was the most common enrichment pathway identified by Gene Ontology analysis, while ORM2, PRDX2, GPX3, HP, HBB, ANXA5, PFN1, CFL1, and S100A11 were key proteins identified by STRING and MCODE analysis. ORM2 showed increased expression in patients with CAS plaques, and ORM2 was accumulated in smooth muscle cells. Oleic acid increased the lipid accumulation and ORM2 and PRDX6 expressions in the VSMCs. The recombinant-ORM2 also increased the lipid accumulation and reactive oxygen species (ROS) in the VSMCs. The expressions of ORM2 and PRDX-6 were correlated, and MJ33 (an inhibitor of PRDX6-PLA2) decreased ROS production and lipid accumulation in VSMCs. CONCLUSION ORM2 may be a biomarker for CAS; it induced lipid accumulation and ROS production in VSMCs during atherosclerosis plaque formation. However, the relationships between ORM2 and PRDX-6 underlying lipid accumulation-induced plaque vulnerability require further research.
Collapse
Affiliation(s)
- Yuan Che
- Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Jinrui Ren
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Haoyang Zhao
- Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Yaoguo Yang
- Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Zhong Chen
- Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Wu X, Zhang H. Omics Approaches Unveiling the Biology of Human Atherosclerotic Plaques. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:482-498. [PMID: 38280419 PMCID: PMC10988765 DOI: 10.1016/j.ajpath.2023.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall, characterized by the buildup of plaques with the accumulation and transformation of lipids, immune cells, vascular smooth muscle cells, and necrotic cell debris. Plaques with collagen-poor thin fibrous caps infiltrated by macrophages and lymphocytes are considered unstable because they are at the greatest risk of rupture and clinical events. However, the current histologic definition of plaque types may not fully capture the complex molecular nature of atherosclerotic plaque biology and the underlying mechanisms contributing to plaque progression, rupture, and erosion. The advances in omics technologies have changed the understanding of atherosclerosis plaque biology, offering new possibilities to improve risk prediction and discover novel therapeutic targets. Genomic studies have shed light on the genetic predisposition to atherosclerosis, and integrative genomic analyses expedite the translation of genomic discoveries. Transcriptomic, proteomic, metabolomic, and lipidomic studies have refined the understanding of the molecular signature of atherosclerotic plaques, aiding in data-driven hypothesis generation for mechanistic studies and offering new prospects for biomarker discovery. Furthermore, advancements in single-cell technologies and emerging spatial analysis techniques have unveiled the heterogeneity and plasticity of plaque cells. This review discusses key omics-based discoveries that have advanced the understanding of human atherosclerotic plaque biology, focusing on insights derived from omics profiling of human atherosclerotic vascular specimens.
Collapse
Affiliation(s)
- Xun Wu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
13
|
Dzobo KE, Cupido AJ, Mol BM, Stiekema LC, Versloot M, Winkelmeijer M, Peter J, Pennekamp AM, Havik SR, Vaz FM, van Weeghel M, Prange KH, Levels JH, de Winther MP, Tsimikas S, Groen AK, Stroes ES, de Kleijn DP, Kroon J. Diacylglycerols and Lysophosphatidic Acid, Enriched on Lipoprotein(a), Contribute to Monocyte Inflammation. Arterioscler Thromb Vasc Biol 2024; 44:720-740. [PMID: 38269588 PMCID: PMC10880937 DOI: 10.1161/atvbaha.123.319937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Oxidized phospholipids play a key role in the atherogenic potential of lipoprotein(a) (Lp[a]); however, Lp(a) is a complex particle that warrants research into additional proinflammatory mediators. We hypothesized that additional Lp(a)-associated lipids contribute to the atherogenicity of Lp(a). METHODS Untargeted lipidomics was performed on plasma and isolated lipoprotein fractions. The atherogenicity of the observed Lp(a)-associated lipids was tested ex vivo in primary human monocytes by RNA sequencing, ELISA, Western blot, and transendothelial migratory assays. Using immunofluorescence staining and single-cell RNA sequencing, the phenotype of macrophages was investigated in human atherosclerotic lesions. RESULTS Compared with healthy individuals with low/normal Lp(a) levels (median, 7 mg/dL [18 nmol/L]; n=13), individuals with elevated Lp(a) levels (median, 87 mg/dL [218 nmol/L]; n=12) demonstrated an increase in lipid species, particularly diacylglycerols (DGs) and lysophosphatidic acid (LPA). DG and the LPA precursor lysophosphatidylcholine were enriched in the Lp(a) fraction. Ex vivo stimulation with DG(40:6) demonstrated a significant upregulation in proinflammatory pathways related to leukocyte migration, chemotaxis, NF-κB (nuclear factor kappa B) signaling, and cytokine production. Functional assessment showed a dose-dependent increase in the secretion of IL (interleukin)-6, IL-8, and IL-1β after DG(40:6) and DG(38:4) stimulation, which was, in part, mediated via the NLRP3 (NOD [nucleotide-binding oligomerization domain]-like receptor family pyrin domain containing 3) inflammasome. Conversely, LPA-stimulated monocytes did not exhibit an inflammatory phenotype. Furthermore, activation of monocytes by DGs and LPA increased their transendothelial migratory capacity. Human atherosclerotic plaques from patients with high Lp(a) levels demonstrated colocalization of Lp(a) with M1 macrophages, and an enrichment of CD68+IL-18+TLR4+ (toll-like receptor) TREM2+ (triggering receptor expressed on myeloid cells) resident macrophages and CD68+CASP1+ (caspase) IL-1B+SELL+ (selectin L) inflammatory macrophages compared with patients with low Lp(a). Finally, potent Lp(a)-lowering treatment (pelacarsen) resulted in a reduction in specific circulating DG lipid subspecies in patients with cardiovascular disease with elevated Lp(a) levels (median, 82 mg/dL [205 nmol/L]). CONCLUSIONS Lp(a)-associated DGs and LPA have a potential role in Lp(a)-induced monocyte inflammation by increasing cytokine secretion and monocyte transendothelial migration. This DG-induced inflammation is, in part, NLRP3 inflammasome dependent.
Collapse
Affiliation(s)
- Kim E. Dzobo
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
| | - Arjen J. Cupido
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Barend M. Mol
- Department of Vascular Surgery, University Medical Centre Utrecht, the Netherlands (B.M.M., D.P.V.d.K.)
| | - Lotte C.A. Stiekema
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Miranda Versloot
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
| | - Maaike Winkelmeijer
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Jorge Peter
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Anne-Marije Pennekamp
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Stefan R. Havik
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Frédéric M. Vaz
- Core Facility Metabolomics (F.M.V., M.v.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Michel van Weeghel
- Core Facility Metabolomics (F.M.V., M.v.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Koen H.M. Prange
- Department of Medical Biochemistry, Amsterdam Infection and Immunity (K.H.M.P., M.P.J.d.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Johannes H.M. Levels
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam Infection and Immunity (K.H.M.P., M.P.J.d.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla (S.T.)
| | - Albert K. Groen
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Erik S.G. Stroes
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Dominique P.V. de Kleijn
- Department of Vascular Surgery, University Medical Centre Utrecht, the Netherlands (B.M.M., D.P.V.d.K.)
| | - Jeffrey Kroon
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
- Laboratory of Angiogenesis and Vascular Metabolism, Flanders Institute for Biotechnology (VIB)-KU Leuven Center for Cancer Biology, VIB, Belgium (J.K.)
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute, Belgium (J.K.)
| |
Collapse
|
14
|
Cao J, Martin-Lorenzo M, van Kuijk K, Wieland EB, Gijbels MJ, Claes BSR, Heredero A, Aldamiz-Echevarria G, Heeren RMA, Goossens P, Sluimer JC, Balluff B, Alvarez-Llamas G. Spatial Metabolomics Identifies LPC(18:0) and LPA(18:1) in Advanced Atheroma With Translation to Plasma for Cardiovascular Risk Estimation. Arterioscler Thromb Vasc Biol 2024; 44:741-754. [PMID: 38299357 DOI: 10.1161/atvbaha.123.320278] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/16/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND The metabolic alterations occurring within the arterial architecture during atherosclerosis development remain poorly understood, let alone those particular to each arterial tunica. We aimed first to identify, in a spatially resolved manner, the specific metabolic changes in plaque, media, adventitia, and cardiac tissue between control and atherosclerotic murine aortas. Second, we assessed their translatability to human tissue and plasma for cardiovascular risk estimation. METHODS In this observational study, mass spectrometry imaging (MSI) was applied to identify region-specific metabolic differences between atherosclerotic (n=11) and control (n=11) aortas from low-density lipoprotein receptor-deficient mice, via histology-guided virtual microdissection. Early and advanced plaques were compared within the same atherosclerotic animals. Progression metabolites were further analyzed by MSI in 9 human atherosclerotic carotids and by targeted mass spectrometry in human plasma from subjects with elective coronary artery bypass grafting (cardiovascular risk group, n=27) and a control group (n=27). RESULTS MSI identified 362 local metabolic alterations in atherosclerotic mice (log2 fold-change ≥1.5; P≤0.05). The lipid composition of cardiac tissue is altered during atherosclerosis development and presents a generalized accumulation of glycerophospholipids, except for lysolipids. Lysolipids (among other glycerophospholipids) were found at elevated levels in all 3 arterial layers of atherosclerotic aortas. LPC(18:0) (lysophosphatidylcholine; P=0.024) and LPA(18:1) (lysophosphatidic acid; P=0.025) were found to be significantly elevated in advanced plaques as compared with mouse-matched early plaques. Higher levels of both lipid species were also observed in fibrosis-rich areas of advanced- versus early-stage human samples. They were found to be significantly reduced in human plasma from subjects with elective coronary artery bypass grafting (P<0.001 and P=0.031, respectively), with LPC(18:0) showing significant association with cardiovascular risk (odds ratio, 0.479 [95% CI, 0.225-0.883]; P=0.032) and diagnostic potential (area under the curve, 0.778 [95% CI, 0.638-0.917]). CONCLUSIONS An altered phospholipid metabolism occurs in atherosclerosis, affecting both the aorta and the adjacent heart tissue. Plaque-progression lipids LPC(18:0) and LPA(18:1), as identified by MSI on tissue, reflect cardiovascular risk in human plasma.
Collapse
Affiliation(s)
- Jianhua Cao
- Maastricht MultiModal Molecular Imaging institute, M4i, Maastricht University, the Netherlands (J.C., B.S.R.C., R.M.A.H., B.B.)
| | - Marta Martin-Lorenzo
- Immunology Department, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain (M.M.-L., G.A.-L.)
| | - Kim van Kuijk
- Department of Pathology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, the Netherlands (K.v.K., E.B.W., M.J.G., P.G., J.C.S.)
| | - Elias B Wieland
- Department of Pathology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, the Netherlands (K.v.K., E.B.W., M.J.G., P.G., J.C.S.)
| | - Marion J Gijbels
- Department of Pathology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, the Netherlands (K.v.K., E.B.W., M.J.G., P.G., J.C.S.)
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, the Netherlands (M.J.G.)
| | - Britt S R Claes
- Maastricht MultiModal Molecular Imaging institute, M4i, Maastricht University, the Netherlands (J.C., B.S.R.C., R.M.A.H., B.B.)
| | - Angeles Heredero
- Cardiac Surgery Service, Fundación Jiménez Díaz University Hospital-UAM, Madrid, Spain (A.H., G.A.-E.)
| | | | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging institute, M4i, Maastricht University, the Netherlands (J.C., B.S.R.C., R.M.A.H., B.B.)
| | - Pieter Goossens
- Department of Pathology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, the Netherlands (K.v.K., E.B.W., M.J.G., P.G., J.C.S.)
| | - Judith C Sluimer
- Department of Pathology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, the Netherlands (K.v.K., E.B.W., M.J.G., P.G., J.C.S.)
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.C.S.)
| | - Benjamin Balluff
- Maastricht MultiModal Molecular Imaging institute, M4i, Maastricht University, the Netherlands (J.C., B.S.R.C., R.M.A.H., B.B.)
| | - Gloria Alvarez-Llamas
- Immunology Department, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain (M.M.-L., G.A.-L.)
- RICORS2040, IIS-Fundación Jiménez Díaz, Madrid, Spain (G.A.-L.)
- Biochemistry and Molecular Biology Department, Complutense University, Madrid, Spain (G.A.-L.)
| |
Collapse
|
15
|
Gerhardtova I, Jankech T, Majerova P, Piestansky J, Olesova D, Kovac A, Jampilek J. Recent Analytical Methodologies in Lipid Analysis. Int J Mol Sci 2024; 25:2249. [PMID: 38396926 PMCID: PMC10889185 DOI: 10.3390/ijms25042249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/09/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Lipids represent a large group of biomolecules that are responsible for various functions in organisms. Diseases such as diabetes, chronic inflammation, neurological disorders, or neurodegenerative and cardiovascular diseases can be caused by lipid imbalance. Due to the different stereochemical properties and composition of fatty acyl groups of molecules in most lipid classes, quantification of lipids and development of lipidomic analytical techniques are problematic. Identification of different lipid species from complex matrices is difficult, and therefore individual analytical steps, which include extraction, separation, and detection of lipids, must be chosen properly. This review critically documents recent strategies for lipid analysis from sample pretreatment to instrumental analysis and data interpretation published in the last five years (2019 to 2023). The advantages and disadvantages of various extraction methods are covered. The instrumental analysis step comprises methods for lipid identification and quantification. Mass spectrometry (MS) is the most used technique in lipid analysis, which can be performed by direct infusion MS approach or in combination with suitable separation techniques such as liquid chromatography or gas chromatography. Special attention is also given to the correct evaluation and interpretation of the data obtained from the lipid analyses. Only accurate, precise, robust and reliable analytical strategies are able to bring complex and useful lipidomic information, which may contribute to clarification of some diseases at the molecular level, and may be used as putative biomarkers and/or therapeutic targets.
Collapse
Affiliation(s)
- Ivana Gerhardtova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovakia
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, SK-842 15 Bratislava, Slovakia
| | - Timotej Jankech
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovakia
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, SK-842 15 Bratislava, Slovakia
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovakia
| | - Juraj Piestansky
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia
| | - Dominika Olesova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 05 Bratislava, Slovakia
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovakia
- Department of Pharmacology and Toxicology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 68/73, SK-041 81 Kosice, Slovakia
| | - Josef Jampilek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovakia
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, SK-842 15 Bratislava, Slovakia
| |
Collapse
|
16
|
Slijkhuis N, Razzi F, Korteland SA, Heijs B, van Gaalen K, Duncker DJ, van der Steen AFW, van Steijn V, van Beusekom HMM, van Soest G. Spatial lipidomics of coronary atherosclerotic plaque development in a familial hypercholesterolemia swine model. J Lipid Res 2024; 65:100504. [PMID: 38246237 PMCID: PMC10879031 DOI: 10.1016/j.jlr.2024.100504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Coronary atherosclerosis is caused by plaque build-up, with lipids playing a pivotal role in its progression. However, lipid composition and distribution within coronary atherosclerosis remain unknown. This study aims to characterize lipids and investigate differences in lipid composition across disease stages to aid in the understanding of disease progression. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) was used to visualize lipid distributions in coronary artery sections (n = 17) from hypercholesterolemic swine. We performed histology on consecutive sections to classify the artery segments and to investigate colocalization between lipids and histological regions of interest in advanced plaque, including necrotic core and inflammatory cells. Segments were classified as healthy (n = 6), mild (n = 6), and advanced disease (n = 5) artery segments. Multivariate data analysis was employed to find differences in lipid composition between the segment types, and the lipids' spatial distribution was investigated using non-negative matrix factorization (NMF). Through this process, MALDI-MSI detected 473 lipid-related features. NMF clustering described three components in positive ionization mode: triacylglycerides (TAG), phosphatidylcholines (PC), and cholesterol species. In negative ionization mode, two components were identified: one driven by phosphatidylinositol(PI)(38:4), and one driven by ceramide-phosphoethanolamine(36:1). Multivariate data analysis showed the association between advanced disease and specific lipid signatures like PC(O-40:5) and cholesterylester(CE)(18:2). Ether-linked phospholipids and LysoPC species were found to colocalize with necrotic core, and mostly CE, ceramide, and PI species colocalized with inflammatory cells. This study, therefore, uncovers distinct lipid signatures correlated with plaque development and their colocalization with necrotic core and inflammatory cells, enhancing our understanding of coronary atherosclerosis progression.
Collapse
Affiliation(s)
- Nuria Slijkhuis
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Francesca Razzi
- Department of Experimental Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands; Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands
| | - Suze-Anne Korteland
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Bram Heijs
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kim van Gaalen
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Dirk J Duncker
- Department of Experimental Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Antonius F W van der Steen
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands; Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Department of Imaging Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Volkert van Steijn
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands
| | - Heleen M M van Beusekom
- Department of Experimental Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Gijs van Soest
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, The Netherlands; Department of Precision and Microsystems Engineering, Delft University of Technology, Delft, The Netherlands; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
17
|
Walczak-Skierska J, Monedeiro F, Maślak E, Złoch M. Lipidomics Characterization of the Microbiome in People with Diabetic Foot Infection Using MALDI-TOF MS. Anal Chem 2023; 95:16251-16262. [PMID: 37877781 PMCID: PMC10633811 DOI: 10.1021/acs.analchem.3c03071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/11/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
Lipidomic profiling has emerged as a powerful tool for the comprehensive characterization of bacterial species, particularly in the context of clinical diagnostics. Utilizing matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS), this study aims to elucidate the lipidomic landscapes of bacterial strains isolated from diabetic foot infections (DFI). Our analysis successfully identified a diverse array of lipids in the cellular membranes of both Gram-positive and Gram-negative bacteria, revealing a total of 108 unique fatty acid combinations. Specifically, we identified 26 LPG, 33 LPE, 43 PE, 114 PG, 89 TAG, and 120 CLP in Gram-positive bacteria and 10 LPG, 14 LPE, 124 PE, 37 PG, 13 TAG, and 22 CLP in Gram-negative strains. Key fatty acids, such as palmitic acid, palmitoleic acid, stearic acid, and oleic acid, were prominently featured. Univariate analysis further highlighted distinct lipidomic signatures among the bacterial strains, revealing elevated levels of phosphatidylethanolamine (PE) and phosphatidylglycerol (PG) in Gram-negative bacteria associated with DFI. In contrast, Gram-positive strains demonstrated increased or uniquely fluctuating levels of triglyceride (TAG) and cardiolipin (CLP). These findings not only underscore the utility of MALDI-TOF MS in bacterial lipidomics but also provide valuable insights into the lipidomic adaptations of bacteria in diabetic foot infections, thereby laying the groundwork for future studies aimed at constructing microbial lipid libraries for enhanced bacterial identification.
Collapse
Affiliation(s)
- Justyna Walczak-Skierska
- Centre
for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str., 87-100 Toruń, Poland
- Chair
of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus a Copernicus University in Toruń, Gagarina 7 Str., 87-100 Toruń, Poland
| | - Fernanda Monedeiro
- Centre
for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str., 87-100 Toruń, Poland
| | - Ewelina Maślak
- Centre
for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str., 87-100 Toruń, Poland
| | - Michał Złoch
- Centre
for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str., 87-100 Toruń, Poland
- Chair
of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus a Copernicus University in Toruń, Gagarina 7 Str., 87-100 Toruń, Poland
| |
Collapse
|
18
|
Slijkhuis N, Towers M, Mirzaian M, Korteland SA, Heijs B, van Gaalen K, Nieuwenhuizen I, Nigg A, van der Heiden K, de Rijke YB, van der Lugt A, Sijbrands EJG, Claude E, van Soest G. Identifying lipid traces of atherogenic mechanisms in human carotid plaque. Atherosclerosis 2023; 385:117340. [PMID: 37913561 DOI: 10.1016/j.atherosclerosis.2023.117340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND AND AIMS Lipids play an important role in atherosclerotic plaque development and are interesting candidate predictive biomarkers. However, the link between circulating lipids, accumulating lipids in the vessel wall, and plaque destabilization processes in humans remains largely unknown. This study aims to provide new insights into the role of lipids in atherosclerosis using lipidomics and mass spectrometry imaging to investigate lipid signatures in advanced human carotid plaque and plasma samples. METHODS We used lipidomics and desorption electrospray ionization mass spectrometry imaging (DESI-MSI) to investigate lipid signatures of advanced human carotid plaque and plasma obtained from patients who underwent carotid endarterectomy (n = 14 out of 17 whose plaque samples were analyzed by DESI-MSI). Multivariate data analysis and unsupervised clustering were applied to identify lipids that were the most discriminative species between different patterns in plaque and plasma. These patterns were interpreted by quantitative comparison with conventional histology. RESULTS Lipidomics detected more than 300 lipid species in plasma and plaque, with markedly different relative abundances. DESI-MSI visualized the spatial distribution of 611 lipid-related m/z features in plaques, of which 330 m/z features could be assigned based on exact mass, comparison to the lipidomic data, and high mass resolution MSI. Matching spatial lipid patterns to histological areas of interest revealed several molecular species that were colocalized with pertinent disease processes in plaque including specific sphingomyelin and ceramide species with calcification, phospholipids and free fatty acids with inflammation, and triacylglycerols and phosphatidylinositols with fibrin-rich areas. CONCLUSIONS By comparing lipid species in plaque and plasma, we identified those circulating species that were also prominently present in plaque. Quantitative comparison of lipid spectral patterns with histology revealed the presence of specific lipid species in destabilized plaque areas, corroborating previous in vitro and animal studies.
Collapse
Affiliation(s)
- Nuria Slijkhuis
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Mark Towers
- Waters Corporation, Wilmslow, United Kingdom
| | - Mina Mirzaian
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Suze-Anne Korteland
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Bram Heijs
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Kim van Gaalen
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Ingeborg Nieuwenhuizen
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Alex Nigg
- Optical Imaging Center, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Kim van der Heiden
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | | | - Gijs van Soest
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, the Netherlands.
| |
Collapse
|
19
|
Greco F, Pardini LF, Botto A, McDonnell LA. Low-melting point agarose as embedding medium for MALDI mass spectrometry imaging and laser-capture microdissection-based proteomics. Sci Rep 2023; 13:18678. [PMID: 37907539 PMCID: PMC10618491 DOI: 10.1038/s41598-023-45799-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/24/2023] [Indexed: 11/02/2023] Open
Abstract
The combination of MALDI mass spectrometry imaging, laser-capture microdissection, and quantitative proteomics allows the identification and characterization of molecularly distinct tissue compartments. Such workflows are typically performed using consecutive tissue sections, and so reliable sectioning and mounting of high-quality tissue sections is a prerequisite of such investigations. Embedding media facilitate the sectioning process but can introduce contaminants which may adversely affect either the mass spectrometry imaging or proteomics analyses. Seven low-temperature embedding media were tested in terms of embedding temperature and cutting performance. The two media that provided the best results (5% gelatin and 2% low-melting point agarose) were compared with non-embedded tissue by both MALDI mass spectrometry imaging of lipids and laser-capture microdissection followed by bottom-up proteomics. Two out of the seven tested media (5% gelatin and 2% low-melting point agarose) provided the best performances on terms of mechanical properties. These media allowed for low-temperature embedding and for the collection of high-quality consecutive sections. Comparisons with non-embedded tissues revealed that both embedding media had no discernable effect on proteomics analysis; 5% gelatin showed a light ion suppression effect in the MALDI mass spectrometry imaging experiments, 2% agarose performed similarly to the non-embedded tissue. 2% low-melting point agarose is proposed for tissue embedding in experiments involving MALDI mass spectrometry imaging of lipids and laser-capture microdissection, proteomics of consecutive tissue sections.
Collapse
Affiliation(s)
- Francesco Greco
- Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme (PI), Italy
| | - Luca Fidia Pardini
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme (PI), Italy
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Asia Botto
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme (PI), Italy
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | | |
Collapse
|
20
|
Nieddu G, Formato M, Lepedda AJ. Searching for Atherosclerosis Biomarkers by Proteomics: A Focus on Lesion Pathogenesis and Vulnerability. Int J Mol Sci 2023; 24:15175. [PMID: 37894856 PMCID: PMC10607641 DOI: 10.3390/ijms242015175] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Plaque rupture and thrombosis are the most important clinical complications in the pathogenesis of stroke, coronary arteries, and peripheral vascular diseases. The identification of early biomarkers of plaque presence and susceptibility to ulceration could be of primary importance in preventing such life-threatening events. With the improvement of proteomic tools, large-scale technologies have been proven valuable in attempting to unravel pathways of atherosclerotic degeneration and identifying new circulating markers to be utilized either as early diagnostic traits or as targets for new drug therapies. To address these issues, different matrices of human origin, such as vascular cells, arterial tissues, plasma, and urine, have been investigated. Besides, proteomics was also applied to experimental atherosclerosis in order to unveil significant insights into the mechanisms influencing atherogenesis. This narrative review provides an overview of the last twenty years of omics applications to the study of atherogenesis and lesion vulnerability, with particular emphasis on lipoproteomics and vascular tissue proteomics. Major issues of tissue analyses, such as plaque complexity, sampling, availability, choice of proper controls, and lipoproteins purification, will be raised, and future directions will be addressed.
Collapse
Affiliation(s)
| | | | - Antonio Junior Lepedda
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (G.N.); (M.F.); Antonio Junior Lepedda (A.J.L.)
| |
Collapse
|
21
|
Seeley EH, Liu Z, Yuan S, Stroope C, Cockerham E, Rashdan NA, Delgadillo L, Finney AC, Kumar D, Das S, Razani B, Liu W, Traylor J, Orr AW, Rom O, Pattillo CB, Yurdagul A. Spatially Resolved Metabolites in Stable and Unstable Human Atherosclerotic Plaques Identified by Mass Spectrometry Imaging. Arterioscler Thromb Vasc Biol 2023; 43:1626-1635. [PMID: 37381983 PMCID: PMC10527524 DOI: 10.1161/atvbaha.122.318684] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Impairments in carbohydrate, lipid, and amino acid metabolism drive features of plaque instability. However, where these impairments occur within the atheroma remains largely unknown. Therefore, we sought to characterize the spatial distribution of metabolites within stable and unstable atherosclerosis in both the fibrous cap and necrotic core. METHODS Atherosclerotic tissue specimens from 9 unmatched individuals were scored based on the Stary classification scale and subdivided into stable and unstable atheromas. After performing mass spectrometry imaging on these samples, we identified over 850 metabolite-related peaks. Using MetaboScape, METASPACE, and Human Metabolome Database, we confidently annotated 170 of these metabolites and found over 60 of these were different between stable and unstable atheromas. We then integrated these results with an RNA-sequencing data set comparing stable and unstable human atherosclerosis. RESULTS Upon integrating our mass spectrometry imaging results with the RNA-sequencing data set, we discovered that pathways related to lipid metabolism and long-chain fatty acids were enriched in stable plaques, whereas reactive oxygen species, aromatic amino acid, and tryptophan metabolism were increased in unstable plaques. In addition, acylcarnitines and acylglycines were increased in stable plaques whereas tryptophan metabolites were enriched in unstable plaques. Evaluating spatial differences in stable plaques revealed lactic acid in the necrotic core, whereas pyruvic acid was elevated in the fibrous cap. In unstable plaques, 5-hydroxyindoleacetic acid was enriched in the fibrous cap. CONCLUSIONS Our work here represents the first step to defining an atlas of metabolic pathways involved in plaque destabilization in human atherosclerosis. We anticipate this will be a valuable resource and open new avenues of research in cardiovascular disease.
Collapse
Affiliation(s)
- Erin H. Seeley
- Department of Chemistry, University of Texas at Austin, TX, USA
| | - Zhipeng Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, IN, USA
| | - Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA, USA
| | - Chad Stroope
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Elizabeth Cockerham
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Nabil A Rashdan
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Luisa Delgadillo
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Dhananjay Kumar
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences and Department of Pharmacology, Wayne State University, MI, USA
| | - James Traylor
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - A Wayne Orr
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| |
Collapse
|
22
|
Chong SY, Wang X, van Bloois L, Huang C, Syeda NS, Zhang S, Ting HJ, Nair V, Lin Y, Lou CKL, Benetti AA, Yu X, Lim NJY, Tan MS, Lim HY, Lim SY, Thiam CH, Looi WD, Zharkova O, Chew NWS, Ng CH, Bonney GK, Muthiah M, Chen X, Pastorin G, Richards AM, Angeli V, Storm G, Wang JW. Injectable liposomal docosahexaenoic acid alleviates atherosclerosis progression and enhances plaque stability. J Control Release 2023; 360:344-364. [PMID: 37406819 DOI: 10.1016/j.jconrel.2023.06.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 06/12/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory vascular disease that is characterized by the accumulation of lipids and immune cells in plaques built up inside artery walls. Docosahexaenoic acid (DHA, 22:6n-3), an omega-3 polyunsaturated fatty acid (PUFA), which exerts anti-inflammatory and antioxidant properties, has long been purported to be of therapeutic benefit to atherosclerosis patients. However, large clinical trials have yielded inconsistent data, likely due to variations in the formulation, dosage, and bioavailability of DHA following oral intake. To fully exploit its potential therapeutic effects, we have developed an injectable liposomal DHA formulation intended for intravenous administration as a plaque-targeted nanomedicine. The liposomal formulation protects DHA against chemical degradation and increases its local concentration within atherosclerotic lesions. Mechanistically, DHA liposomes are readily phagocytosed by activated macrophages, exert potent anti-inflammatory and antioxidant effects, and inhibit foam cell formation. Upon intravenous administration, DHA liposomes accumulate preferentially in atherosclerotic lesional macrophages and promote polarization of macrophages towards an anti-inflammatory M2 phenotype, resulting in attenuation of atherosclerosis progression in both ApoE-/- and Ldlr-/- experimental models. Plaque composition analysis demonstrates that liposomal DHA inhibits macrophage infiltration, reduces lipid deposition, and increases collagen content, thus improving the stability of atherosclerotic plaques against rupture. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) further reveals that DHA liposomes can partly restore the complex lipid profile of the plaques to that of early-stage plaques. In conclusion, DHA liposomes offer a promising approach for applying DHA to stabilize atherosclerotic plaques and attenuate atherosclerosis progression, thereby preventing atherosclerosis-related cardiovascular events.
Collapse
Affiliation(s)
- Suet Yen Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Louis van Bloois
- Department of Pharmaceutics, Faculty of Science, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Chenyuan Huang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Nilofer Sayed Syeda
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Sitong Zhang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Hui Jun Ting
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Vaarsha Nair
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Yuanzhe Lin
- Department of Biomedical Engineering, National University of Singapore, 117583 Singapore, Singapore
| | - Charles Kang Liang Lou
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Ayca Altay Benetti
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543 Singapore, Singapore
| | - Xiaodong Yu
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Nicole Jia Ying Lim
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Michelle Siying Tan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Sheau Yng Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Chung Hwee Thiam
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Wen Donq Looi
- Bruker Daltonics, Bruker Singapore Pte. Ltd., 138671 Singapore, Singapore
| | - Olga Zharkova
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Nicholas W S Chew
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Department of Cardiology, National University Heart Centre, National University Hospital, 119074 Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Glenn Kunnath Bonney
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, National University Hospital, 119074 Singapore, Singapore
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, 119074 Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, 119074 Singapore, Singapore
| | - Xiaoyuan Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 119074 Singapore, Singapore; Departments of Chemical and Biomolecular Engineering, and Biomedical Engineering, Faculty of Engineering, National University of Singapore, 117575 Singapore, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543 Singapore, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Veronique Angeli
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Pharmaceutics, Faculty of Science, Utrecht University, 3584 CG Utrecht, the Netherlands; Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, the Netherlands.
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Physiology, National University of Singapore, 117593 Singapore, Singapore.
| |
Collapse
|
23
|
Nie L, Xiang X, Wen C, Zhang F, Xia Y, Wang Y, Mao L. The Role of Lipid Subcomponents in the Development of Atherosclerotic Plaques. Rev Cardiovasc Med 2023; 24:139. [PMID: 39076730 PMCID: PMC11273020 DOI: 10.31083/j.rcm2405139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/17/2022] [Accepted: 01/06/2023] [Indexed: 07/31/2024] Open
Abstract
Atherosclerosis (AS) is a long-standing cardiovascular and cerebrovascular disease. Its occurrence and development are related to the pathophysiology of lipids including cholesteryl ester (CE), cholesterol, triacylglycerol (TG), and phospholipid (PL). In this review, we focus on the roles and possible mechanisms of different lipid subcomponents in the process of AS, and provide new ideas for the prevention, diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Lei Nie
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Xuying Xiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Cheng Wen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Feng Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Yuanpeng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Yong Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| |
Collapse
|
24
|
Abstract
Over the last decade, immunometabolism has emerged as a novel interdisciplinary field of research and yielded significant fundamental insights into the regulation of immune responses. Multiple classical approaches to interrogate immunometabolism, including bulk metabolic profiling and analysis of metabolic regulator expression, paved the way to appreciating the physiological complexity of immunometabolic regulation in vivo. Studying immunometabolism at the systems level raised the need to transition towards the next-generation technology for metabolic profiling and analysis. Spatially resolved metabolic imaging and computational algorithms for multi-modal data integration are new approaches to connecting metabolism and immunity. In this review, we discuss recent studies that highlight the complex physiological interplay between immune responses and metabolism and give an overview of technological developments that bear the promise of capturing this complexity most directly and comprehensively.
Collapse
Affiliation(s)
- Denis A Mogilenko
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
- Current affiliation: Department of Medicine, Department of Pathology, Microbiology, and Immunology, and Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| | - Alexey Sergushichev
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
| |
Collapse
|
25
|
Bonacina F, Di Costanzo A, Genkel V, Kong XY, Kroon J, Stimjanin E, Tsiantoulas D, Grootaert MO. The heterogeneous cellular landscape of atherosclerosis: Implications for future research and therapies. A collaborative review from the EAS young fellows. Atherosclerosis 2023; 372:48-56. [PMID: 37030081 DOI: 10.1016/j.atherosclerosis.2023.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023]
Abstract
Single cell technologies, lineage tracing mouse models and advanced imaging techniques unequivocally improved the resolution of the cellular landscape of atherosclerosis. Although the discovery of the heterogeneous nature of the cellular plaque architecture has undoubtedly improved our understanding of the specific cellular states in atherosclerosis progression, it also adds more complexity to current and future research and will change how we approach future drug development. In this review, we will discuss how the revolution of new single cell technologies allowed us to map the cellular networks in the plaque, but we will also address current (technological) limitations that confine us to identify the cellular drivers of the disease and to pinpoint a specific cell state, cell subset or cell surface antigen as new candidate drug target for atherosclerosis.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Alessia Di Costanzo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Vadim Genkel
- Department of Internal Medicine, South-Ural State Medical University, Chelyabinsk, Russia
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Jeffrey Kroon
- Amsterdam UMC Location University of Amsterdam, Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, VIB, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Belgium
| | - Ena Stimjanin
- Department of Internal Medicine, Cantonal Hospital Zenical, Zenica, Bosnia and Herzegovina
| | | | - Mandy Oj Grootaert
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
26
|
Embracing lipidomics at single-cell resolution: Promises and pitfalls. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
27
|
Chandran M, S S, Abhirami, Chandran A, Jaleel A, Plakkal Ayyappan J. Defining atherosclerotic plaque biology by mass spectrometry-based omics approaches. Mol Omics 2023; 19:6-26. [PMID: 36426765 DOI: 10.1039/d2mo00260d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Atherosclerosis is the principal cause of vascular diseases and one of the leading causes of worldwide death. Even though several insights into its natural course, risk factors and interventions have been identified, it is still an ongoing global pandemic. Since the structure and biochemical composition of the plaques show high heterogeneity, a comprehensive understanding of the intraplaque composition, its microenvironment, and the mechanisms of the progression and instability across different vascular beds at their progression stages is crucial for better risk stratification and treatment modalities. Even though several cell-based studies, animal studies, and extensive multicentric population studies have been conducted concerning cardiovascular diseases for assessing the risk factors and plaque biology, the studies on human clinical samples are very limited. New novel approaches utilize samples from percutaneous coronary interventions, which could possibly gain more access to clinical samples at different stages of the diseases without complex invasive resections. As an emerging technological platform in disease discovery research, mass spectrometry-based omics technologies offer capabilities for a comprehensive understanding of the mechanisms linked to several vascular diseases. Here, we discuss the cellular and molecular processes of atherosclerosis, different mass spectrometry-based omics approaches, and the studies mostly done on clinical samples of atheroma plaque using mass spectrometry-based proteomics, metabolomics and lipidomics approaches.
Collapse
Affiliation(s)
- Mahesh Chandran
- Translational Nanomedicine and Lifestyle Disease Research Laboratory, Department of Biochemistry, University of Kerala, Thiruvananthapuram 695034, Kerala, India. .,Department of Biotechnology, University of Kerala, Thiruvananthapuram 695034, Kerala, India.,Mass Spectrometry and Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695012, India
| | - Sudhina S
- Translational Nanomedicine and Lifestyle Disease Research Laboratory, Department of Biochemistry, University of Kerala, Thiruvananthapuram 695034, Kerala, India.
| | - Abhirami
- Translational Nanomedicine and Lifestyle Disease Research Laboratory, Department of Biochemistry, University of Kerala, Thiruvananthapuram 695034, Kerala, India.
| | - Akash Chandran
- Department of Nanoscience and Nanotechnology, University of Kerala, Kariavattom, Thiruvananthapuram-695581, Kerala, India
| | - Abdul Jaleel
- Mass Spectrometry and Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695012, India
| | - Janeesh Plakkal Ayyappan
- Translational Nanomedicine and Lifestyle Disease Research Laboratory, Department of Biochemistry, University of Kerala, Thiruvananthapuram 695034, Kerala, India. .,Department of Biotechnology, University of Kerala, Thiruvananthapuram 695034, Kerala, India.,Department of Nanoscience and Nanotechnology, University of Kerala, Kariavattom, Thiruvananthapuram-695581, Kerala, India.,Centre for Advanced Cancer Research, Department of Biochemistry, University of Kerala, Thiruvananthapuram 695034, Kerala, India
| |
Collapse
|
28
|
Li W, Luo J, Peng F, Liu R, Bai X, Wang T, Zhang X, Zhu J, Li XY, Wang Z, Liu W, Wang J, Zhang L, Chen X, Xue T, Ding C, Wang C, Jiao L. Spatial metabolomics identifies lipid profiles of human carotid atherosclerosis. Atherosclerosis 2023; 364:20-28. [PMID: 36459728 DOI: 10.1016/j.atherosclerosis.2022.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/31/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS Carotid atherosclerosis is an important cause of ischemic stroke. Lipids play a key role in the progression of atherosclerosis. To date, the spatial lipid profile of carotid atherosclerotic plaques related to histology has not been systematically investigated. METHODS Carotid atherosclerosis samples from 12 patients were obtained and classified into four classical pathological stages (preatheroma, atheroma, fibroatheroma and complicated lesion) by histological staining. Desorption electrospray ionization-mass spectrometry imaging (DESI-MSI) was used to investigate the lipid profile of carotid atherosclerosis, and correlated it with histological information. Bioinformatics technology was used to process MSI data among different pathological stages of atherosclerosis lesions. RESULTS A total of 55 lipids (26 throughout cross-section regions [TCSRs], 13 in lipid-rich regions [LRRs], and 16 in collagen-rich regions [CRRs]) were initially identified in carotid plaque from one patient. Subsequently, 32 of 55 lipids (12 in TCSRs, eight in LRRs, and 12 in CRRs) were further screened in 11 patients. Pathway enrichment analysis showed that multiple metabolic pathways, such as fat digestion and absorption, cholesterol metabolism, lipid and atherosclerosis, were enriched in TCSRs; sphingolipid signaling pathway, necroptosis pathway were enriched in LRRs; and glycerophospholipid metabolism, ether lipid metabolism pathway were mainly enriched in CRRs. CONCLUSIONS This study comprehensively showed the spatial lipid metabolism footprint in human carotid atherosclerotic plaques. The lipid profiles and related metabolism pathways in three regions of plaque with disease progression were different markedly, suggesting that the different metabolic mechanisms in these regions of carotid plaque may be critical in atherosclerosis progression.
Collapse
Affiliation(s)
- Wei Li
- Department of Stroke Center, Central Hospital Affiliated to Shandong First Medical University, China; Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Fangda Peng
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China
| | - Ruiting Liu
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Xuesong Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Junge Zhu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xu-Ying Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zhanjun Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wubin Liu
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Xianyang Chen
- Zhongguancun Biological and Medical Big Data Center, Beijing, China; BaoFeng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Teng Xue
- BaoFeng Key Laboratory of Genetics and Metabolism, Beijing, China; Zhongyuanborui Key Laborotory of Genetics and Metabolism, Guangdong-Macao In-depth Cooperation Zone in Hengqin, China
| | - Chunguang Ding
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China.
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; National Clinical Research Center for Geriatric Diseases, Beijing, China.
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Beijing, 100053, China; Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
29
|
Busnelli M, Manzini S, Colombo A, Franchi E, Lääperi M, Laaksonen R, Chiesa G. Effect of Diets on Plasma and Aorta Lipidome: A Study in the apoE Knockout Mouse Model. Mol Nutr Food Res 2023; 67:e2200367. [PMID: 36419336 DOI: 10.1002/mnfr.202200367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/20/2022] [Indexed: 11/27/2022]
Abstract
SCOPE Specific lipid molecules circulating in plasma at low concentrations have emerged as biomarkers of atherosclerotic risk. The aim of the present study is that of evaluating, in an athero-prone mouse model, how different diets can affect plasma and aorta lipidome. METHODS AND RESULTS Thirty-six apoE knockout mice are divided in three groups and feed 12 weeks with diets differing for cholesterol and fatty acid content. Atherosclerosis is measured at the aortic sinus and aorta. Lipids are quantified in plasma and aorta with mass spectrometry. The cholesterol content of the diets is the main driver of lipid accumulation in plasma and aorta. The fatty acid composition of the diets affects plasma levels both of essential (linoleic acid) and nonessential (myristic and arachidonic acid) ones. Lipidomics show a comparable distribution, in plasma and aorta, of the main lipid components of oxidized LDL, including cholesteryl esters and lysophosphatidylcholines. Interestingly, lactosylceramide, glucosyl/galactosylceramide, and individual ceramide species are found to accumulate in diseased aortic segments. CONCLUSION Both the cholesterol and fatty acid content of the diets profoundly affect plasma lipidome. Aorta lipidome is likewise affected with the accumulation of specific lipids known as markers of atherosclerosis.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Elsa Franchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | | | - Reijo Laaksonen
- Zora Biosciences Oy, Espoo, 02150, Finland.,Finnish Cardiovascular Research Center, University of Tampere, Tampere, 33520, Finland
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| |
Collapse
|
30
|
Visscher M, Pleitez MA, Van Gaalen K, Nieuwenhuizen-Bakker IM, Ntziachristos V, Van Soest G. Label-free analytic histology of carotid atherosclerosis by mid-infrared optoacoustic microscopy. PHOTOACOUSTICS 2022; 26:100354. [PMID: 35465607 PMCID: PMC9020099 DOI: 10.1016/j.pacs.2022.100354] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 05/20/2023]
Abstract
BACKGROUND AND AIMS Analysis of atherosclerotic plaque composition is a vital tool for unraveling the pathological metabolic processes that contribute to plaque growth. METHODS We visualize the constitution of human carotid plaques by mid-infrared optoacoustic microscopy (MiROM), a method for label-free analytic histology that requires minimal tissue preparation, rapidly yielding large field-of-view en-face images with a resolution of a few micrometers. We imaged endarterectomy specimens (n = 3, 12 sections total) at specific vibrational modes, targeting carbohydrates, lipids and proteins. Additionally, we recorded spectra at selected tissue locations. We identified correlations in the variability in this high-dimensional data set using non-negative matrix factorization (NMF). RESULTS We visualized high-risk plaque features with molecular assignment. Consistent NMF components relate to different dominant tissue constituents, dominated by lipids, proteins, and cholesterol and carbohydrates respectively. CONCLUSIONS These results introduce MiROM as an innovative, stain-free, analytic histology technology for the biochemical characterization of complex human vascular pathology.
Collapse
Affiliation(s)
- Mirjam Visscher
- Department of Cardiology, Erasmus MC University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Miguel A. Pleitez
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging (CBI) and Center for Translational Cancer Research (TranslaTUM), Technische Universität München, München, Germany
- Corresponding author at: Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Kim Van Gaalen
- Department of Cardiology, Erasmus MC University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | - Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging (CBI) and Center for Translational Cancer Research (TranslaTUM), Technische Universität München, München, Germany
| | - Gijs Van Soest
- Department of Cardiology, Erasmus MC University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
- Corresponding author.
| |
Collapse
|
31
|
Velenosi TJ, Ben-Yakov G, Podszun MC, Hercun J, Etzion O, Yang S, Nadal C, Haynes-Williams V, Huang WCA, Gonzalez-Hodar L, Brychta RJ, Takahashi S, Akkaraju V, Krausz KW, Walter M, Cai H, Walter PJ, Muniyappa R, Chen KY, Gonzalez FJ, Rotman Y. Postprandial Plasma Lipidomics Reveal Specific Alteration of Hepatic-derived Diacylglycerols in Nonalcoholic Fatty Liver Disease. Gastroenterology 2022; 162:1990-2003. [PMID: 35283114 PMCID: PMC9117487 DOI: 10.1053/j.gastro.2022.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS Hepatic energy metabolism is a dynamic process modulated by multiple stimuli. In nonalcoholic fatty liver disease (NAFLD), human studies typically focus on the static fasting state. We hypothesized that unique postprandial alterations in hepatic lipid metabolism are present in NAFLD. METHODS In a prospective clinical study, 37 patients with NAFLD and 10 healthy control subjects ingested a standardized liquid meal with pre- and postprandial blood sampling. Postprandial plasma lipid kinetics were characterized at the molecular lipid species level by untargeted lipidomics, cluster analysis, and lipid particle isolation, then confirmed in a mouse model. RESULTS There was a specific increase of multiple plasma diacylglycerol (DAG) species at 4 hours postprandially in patients with NAFLD but not in controls. This was replicated in a nonalcoholic steatohepatitis mouse model, where postprandial DAGs increased in plasma and concomitantly decreased in the liver. The increase in plasma DAGs appears early in the disease course, is dissociated from NAFLD severity and obesity, and correlates with postprandial insulin levels. Immunocapture isolation of very low density lipoprotein in human samples and stable isotope tracer studies in mice revealed that elevated postprandial plasma DAGs reflect hepatic secretion of endogenous, rather than meal-derived lipids. CONCLUSIONS We identified a selective insulin-related increase in hepatic secretion of endogenously derived DAGs after a mixed meal as a unique feature of NAFLD. DAGs are known to be lipotoxic and associated with atherosclerosis. Although it is still unknown whether the increased exposure to hepatic DAGs contributes to extrahepatic manifestations and cardiovascular risk in NAFLD, our study highlights the importance of extending NAFLD research beyond the fasting state.
Collapse
Affiliation(s)
- Thomas J. Velenosi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH)
| | - Gil Ben-Yakov
- Liver & Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH,Liver Diseases Branch, NIDDK, NIH
| | - Maren C. Podszun
- Liver & Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH,Liver Diseases Branch, NIDDK, NIH
| | | | | | | | | | | | | | - Lila Gonzalez-Hodar
- Liver & Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH,Liver Diseases Branch, NIDDK, NIH
| | | | - Shogo Takahashi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH)
| | - Vikas Akkaraju
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH)
| | - Kristopher W. Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH)
| | | | - Hongyi Cai
- Clinical Mass Spectrometry Core, NIDDK, NIH
| | | | | | - Kong Y. Chen
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH)
| | - Yaron Rotman
- Liver and Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland; Liver Diseases Branch, NIDDK, NIH, Bethesda, Maryland.
| |
Collapse
|
32
|
Lee H, Seeger MR, Lippok N, Nadkarni SK, van Soest G, Bouma BE. Nanosecond SRS fiber amplifier for label-free near-infrared photoacoustic microscopy of lipids. PHOTOACOUSTICS 2022; 25:100331. [PMID: 35096525 PMCID: PMC8783138 DOI: 10.1016/j.pacs.2022.100331] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 05/18/2023]
Abstract
Near-infrared photoacoustics receives increasing interest as an intravital modality to sense key biomolecules. One of the most central types of biomolecules of interest are lipids as they constitute essential bio-hallmarks of cardiovascular and metabolic diseases and their in-vivo detection holds insightful information about disease progression and treatment monitoring. However, the full potential of near-infrared photoacoustic for high-resolution and high-sensitivity biomedical studies of lipids has so far not been exploited due a lack of appropriate excitation sources delivering short-pulses at high-repetition-rate, high-pulse-energy, and wavelength around 1200 nm. Here, we demonstrate a custom-built SRS fiber amplifier that provides optical excitations at 1192.8 nm, repetition rates of 200 kHz, pulse durations below 2 ns, and pulse energies beyond 5 μJ. We capitalize on the performance of our excitation source and show near-infrared photoacoustics resolving intrinsic lipid contrast in biomedically relevant specimens ranging from single cells to lipid-rich tissue with subcellular resolution.
Collapse
Affiliation(s)
- Hwidon Lee
- Harvard Medical School, Boston, Massachusetts, MA 02115, USA
- Wellman Center for Photomedicine, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA
| | - Markus R. Seeger
- Harvard Medical School, Boston, Massachusetts, MA 02115, USA
- Wellman Center for Photomedicine, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA
| | - Norman Lippok
- Harvard Medical School, Boston, Massachusetts, MA 02115, USA
- Wellman Center for Photomedicine, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA
| | - Seemantini K. Nadkarni
- Harvard Medical School, Boston, Massachusetts, MA 02115, USA
- Wellman Center for Photomedicine, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA
| | - Gijs van Soest
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 Rotterdam, CA The Netherlands
| | - Brett E. Bouma
- Harvard Medical School, Boston, Massachusetts, MA 02115, USA
- Wellman Center for Photomedicine, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 Rotterdam, CA The Netherlands
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Corresponding author at: Harvard Medical School, Boston, Massachusetts, MA 02115, USA.
| |
Collapse
|
33
|
Engel KM, Prabutzki P, Leopold J, Nimptsch A, Lemmnitzer K, Vos DRN, Hopf C, Schiller J. A new update of MALDI-TOF mass spectrometry in lipid research. Prog Lipid Res 2022; 86:101145. [PMID: 34995672 DOI: 10.1016/j.plipres.2021.101145] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 01/06/2023]
Abstract
Matrix-assisted laser desorption and ionization (MALDI) mass spectrometry (MS) is an indispensable tool in modern lipid research since it is fast, sensitive, tolerates sample impurities and provides spectra without major analyte fragmentation. We will discuss some methodological aspects, the related ion-forming processes and the MALDI MS characteristics of the different lipid classes (with the focus on glycerophospholipids) and the progress, which was achieved during the last ten years. Particular attention will be given to quantitative aspects of MALDI MS since this is widely considered as the most serious drawback of the method. Although the detailed role of the matrix is not yet completely understood, it will be explicitly shown that the careful choice of the matrix is crucial (besides the careful evaluation of the positive and negative ion mass spectra) in order to be able to detect all lipid classes of interest. Two developments will be highlighted: spatially resolved Imaging MS is nowadays well established and the distribution of lipids in tissues merits increasing interest because lipids are readily detectable and represent ubiquitous compounds. It will also be shown that a combination of MALDI MS with thin-layer chromatography (TLC) enables a fast spatially resolved screening of an entire TLC plate which makes the method competitive with LC/MS.
Collapse
Affiliation(s)
- Kathrin M Engel
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - Patricia Prabutzki
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - Jenny Leopold
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - Ariane Nimptsch
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - Katharina Lemmnitzer
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - D R Naomi Vos
- Center for Biomedical Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Strasse 10, D-68163 Mannheim, Germany
| | - Carsten Hopf
- Center for Biomedical Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Strasse 10, D-68163 Mannheim, Germany
| | - Jürgen Schiller
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany.
| |
Collapse
|
34
|
Tanase DM, Gosav EM, Petrov D, Jucan AE, Lacatusu CM, Floria M, Tarniceriu CC, Costea CF, Ciocoiu M, Rezus C. Involvement of Ceramides in Non-Alcoholic Fatty Liver Disease (NAFLD) Atherosclerosis (ATS) Development: Mechanisms and Therapeutic Targets. Diagnostics (Basel) 2021; 11:2053. [PMID: 34829402 PMCID: PMC8621166 DOI: 10.3390/diagnostics11112053] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/26/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and atherosclerosis (ATS) are worldwide known diseases with increased incidence and prevalence. These two are driven and are interconnected by multiple oxidative and metabolic functions such as lipotoxicity. A gamut of evidence suggests that sphingolipids (SL), such as ceramides, account for much of the tissue damage. Although in humans they are proving to be accurate biomarkers of adverse cardiovascular disease outcomes and NAFLD progression, in rodents, pharmacological inhibition or depletion of enzymes driving de novo ceramide synthesis prevents the development of metabolic driven diseases such as diabetes, ATS, and hepatic steatosis. In this narrative review, we discuss the pathways which generate the ceramide synthesis, the potential use of circulating ceramides as novel biomarkers in the development and progression of ATS and related diseases, and their potential use as therapeutic targets in NAFDL-ATS development which can further provide new clues in this field.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.M.G.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.M.G.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Daniela Petrov
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Alina Ecaterina Jucan
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Institute of Gastroenterology and Hepatology, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Cristina Mihaela Lacatusu
- Unit of Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.M.G.); (C.R.)
- Internal Medicine Clinic, Emergency Military Clinical Hospital Iasi, 700483 Iasi, Romania
| | - Claudia Cristina Tarniceriu
- Department of Morpho-Functional Sciences I, Discipline of Anatomy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Claudia Florida Costea
- Department of Ophthalmology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.M.G.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| |
Collapse
|
35
|
Miura Y, Suzuki Y, Kanamaru H, Shiba M, Yasuda R, Toma N, Suzuki H. Higher Non-fasting Serum Triglyceride Preceding the Carotid Stenosis Progression. Neurol Med Chir (Tokyo) 2021; 61:422-432. [PMID: 34078829 PMCID: PMC8280330 DOI: 10.2176/nmc.oa.2020-0430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The present study was conducted to investigate whether non-fasting serum triglyceride (TG) levels can be used to assess a risk for the progression of carotid artery stenosis. This was a single-center retrospective study. Consecutive 96 patients with ≥50% stenosis of at least unilateral cervical internal carotid artery and normal fasting serum low-density lipoprotein cholesterol (LDL-C) levels of ≤140 mg/dL were followed up for at least 1 year (mean, 3.1 years), and clinical variables were compared between patients with and without carotid stenosis progression (≥10% increases in the degree on ultrasonography). Carotid stenosis progression was shown in 21 patients, associated with less frequent treatment with calcium channel blockers (CCBs), higher non-fasting TG and glucose levels. In carotid artery-based analyses including <50% stenosis side, stenosis progression was shown in 23 of 121 arteries except for those with complete occlusion and less than 1-year follow-up period because of carotid artery stenting (CAS) or carotid endarterectomy (CEA). Stenosis progression was more frequently observed in symptomatic and/or radiation-induced lesions, and was also accompanied with less frequent treatment with CCBs, higher non-fasting TG and glucose levels in carotid artery-based analyses. The receiver operating characteristic (ROC) curve analyses revealed that a cutoff value of non-fasting TG to discriminate carotid stenosis progression was 169.5 mg/dL for carotid arteries with the baseline stenosis of <50%, and 154.5mg/dL for those of ≥50%. Non-fasting TG level was an independent risk factor of carotid stenosis progression, and more strict control of non-fasting TG may be necessary for higher degree of carotid artery stenosis.
Collapse
Affiliation(s)
- Yoichi Miura
- Department of Neurosurgery, Mie University Graduate School of Medicine
| | - Yume Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine
| | - Hideki Kanamaru
- Department of Neurosurgery, Mie University Graduate School of Medicine
| | - Masato Shiba
- Department of Neurosurgery, Mie University Graduate School of Medicine
| | - Ryuta Yasuda
- Department of Neurosurgery, Mie University Graduate School of Medicine
| | - Naoki Toma
- Department of Neurosurgery, Mie University Graduate School of Medicine
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine
| |
Collapse
|
36
|
Iskander-Rizk S, Visscher M, Moerman AM, Korteland SA, Van der Heiden K, Van der Steen AF, Van Soest G. Micro Spectroscopic Photoacoustic (μsPA) imaging of advanced carotid atherosclerosis. PHOTOACOUSTICS 2021; 22:100261. [PMID: 33854946 PMCID: PMC8027769 DOI: 10.1016/j.pacs.2021.100261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 05/11/2023]
Abstract
Atherosclerosis is a lipid-driven and an inflammatory disease of the artery walls. The composition of atherosclerotic plaque stratifies the risk of a specific plaque to cause a cardiovascular event. In an optical resolution photoacoustic microscopy setup, of 45 μm resolution, we extracted plaque lipid photoacoustic (PA) spectral signatures of human endarterectomy samples in the range of 1150-1240 nm, using matrix assisted laser desorption ionization mass spectrometry imaging as a reference. We found plaque PA signals to correlate best with sphingomyelins and cholesteryl esters. PA signal spectral variations within the plaque area were compared to reference molecular patterns and absorption spectra of lipid laboratory standards. Variability in the lipid spectroscopic features extracted by principal component analysis of all samples revealed three distinct components with peaks at: 1164, 1188, 1196 and 1210 nm. This result will guide the development of PA-based atherosclerosis disease staging capitalizing on lipidomics of atherosclerotic tissue.
Collapse
Key Words
- Atherosclerosis
- CE, cholesteryl ester
- CEA, carotid endarterectomy
- DG, diacylglycerol
- DHB, 2,5-dihydroxybenzoic acid
- ESI, electrospray ionization
- FTICR, fourier-transform ion cyclotron resonance
- HPLC, high-performance liquid chromatography
- Lipids
- MALDI-MSI, matrix-assisted laser desorption ionization mass spectrometry imaging
- Mass spectrometry imaging
- Microscopy
- NIRS, near-infrared spectroscopy
- PC, phosphatidylcholine
- PCA
- PCA, principal component analysis
- PFA, paraformaldehyde
- SM, sphingomyelin
- Spectroscopy
- TG, triacylglycerol
- WREnS, Waters Research Enabled Software suite
- m/z, mass to charge ratio
- μsPA, Micro Spectroscopic Photoacoustic
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gijs Van Soest
- Corresponding author at: Erasmus Medical Center, Ee-2302, PO Box 2040, 3000 CA, Rotterdam, the Netherlands.
| |
Collapse
|
37
|
Greco F, Quercioli L, Pucci A, Rocchiccioli S, Ferrari M, Recchia FA, McDonnell LA. Mass Spectrometry Imaging as a Tool to Investigate Region Specific Lipid Alterations in Symptomatic Human Carotid Atherosclerotic Plaques. Metabolites 2021; 11:250. [PMID: 33919525 PMCID: PMC8073208 DOI: 10.3390/metabo11040250] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 12/01/2022] Open
Abstract
Atherosclerosis is characterized by fatty plaques in large and medium sized arteries. Their rupture can causes thrombi, occlusions of downstream vessels and adverse clinical events. The investigation of atherosclerotic plaques is made difficult by their highly heterogeneous nature. Here we propose a spatially resolved approach based on matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging to investigate lipids in specific regions of atherosclerotic plaques. The method was applied to a small dataset including symptomatic and asymptomatic human carotid atherosclerosis plaques. Tissue sections of symptomatic and asymptomatic human carotid atherosclerotic plaques were analyzed by MALDI mass spectrometry imaging (MALDI MSI) of lipids, and adjacent sections analyzed by histology and immunofluorescence. These multimodal datasets were used to compare the lipid profiles of specific histopathological regions within the plaque. The lipid profiles of macrophage-rich regions and intimal vascular smooth muscle cells exhibited the largest changes associated with plaque outcome. Macrophage-rich regions from symptomatic lesions were found to be enriched in sphingomyelins, and intimal vascular smooth muscle cells of symptomatic plaques were enriched in cholesterol and cholesteryl esters. The proposed method enabled the MALDI MSI analysis of specific regions of the atherosclerotic lesion, confirming MALDI MSI as a promising tool for the investigation of histologically heterogeneous atherosclerotic plaques.
Collapse
Affiliation(s)
- Francesco Greco
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy; (F.G.); (F.A.R.)
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
| | - Laura Quercioli
- Department of Vascular Surgery, Azienda Ospedaliero Universitaria Pisana, 56124 Pisa, Italy; (L.Q.); (M.F.)
| | - Angela Pucci
- Department of Histopathology, University Hospital, 56124 Pisa, Italy;
| | - Silvia Rocchiccioli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | - Mauro Ferrari
- Department of Vascular Surgery, Azienda Ospedaliero Universitaria Pisana, 56124 Pisa, Italy; (L.Q.); (M.F.)
| | - Fabio A. Recchia
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy; (F.G.); (F.A.R.)
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Liam A. McDonnell
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
| |
Collapse
|