1
|
Foran D, Antoniades C, Akoumianakis I. Emerging Roles for Sphingolipids in Cardiometabolic Disease: A Rational Therapeutic Target? Nutrients 2024; 16:3296. [PMID: 39408263 PMCID: PMC11478599 DOI: 10.3390/nu16193296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality. New research elucidates increasingly complex relationships between cardiac and metabolic health, giving rise to new possible therapeutic targets. Sphingolipids are a heterogeneous class of bioactive lipids with critical roles in normal human physiology. They have also been shown to play both protective and deleterious roles in the pathogenesis of cardiovascular disease. Ceramides are implicated in dysregulating insulin signalling, vascular endothelial function, inflammation, oxidative stress, and lipoprotein aggregation, thereby promoting atherosclerosis and vascular disease. Ceramides also advance myocardial disease by enhancing pathological cardiac remodelling and cardiomyocyte death. Glucosylceramides similarly contribute to insulin resistance and vascular inflammation, thus playing a role in atherogenesis and cardiometabolic dysfunction. Sphingosing-1-phosphate, on the other hand, may ameliorate some of the pathological functions of ceramide by protecting endothelial barrier integrity and promoting cell survival. Sphingosine-1-phosphate is, however, implicated in the development of cardiac fibrosis. This review will explore the roles of sphingolipids in vascular, cardiac, and metabolic pathologies and will evaluate the therapeutic potential in targeting sphingolipids with the aim of prevention and reversal of cardiovascular disease in order to improve long-term cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.F.); (C.A.)
| |
Collapse
|
2
|
Kumar S, Singh A, Pandey P, Khopade A, Sawant KK. Application of sphingolipid-based nanocarriers in drug delivery: an overview. Ther Deliv 2024; 15:619-637. [PMID: 39072358 PMCID: PMC11412150 DOI: 10.1080/20415990.2024.2377066] [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: 12/01/2023] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
Sphingolipids (SL) are well recognized for their cell signaling through extracellular and intracellular pathways. Based on chemistry different types of SL are biosynthesized in mammalian cells and have specific function in cellular activity. SL has an ampiphilic structure with have hydrophobic body attached to the polar head enables their use as a drug delivery agent in the form of nanocarriers. SL-based liposomes can improve the solubility of lipophilic drugs through host and drug complexes and are more stable than conventional liposomal formulations. Preclinical studies of SL nanocarriers are reported on topical delivery, oral delivery, ocular delivery, chemotherapeutic delivery, cardiovascular delivery and Alzheimer's disease. The commercial challenges and patents related to SL nanoformulations are highlighted in this article.
Collapse
Affiliation(s)
- Samarth Kumar
- Formulation Research & Development-Non-Orals, Sun Pharmaceutical Industries Ltd, Vadodara, 390012, Gujarat, India
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390001, India
| | - Ajit Singh
- Formulation Research & Development-Non-Orals, Sun Pharmaceutical Industries Ltd, Vadodara, 390012, Gujarat, India
| | - Prachi Pandey
- Krishna School of Pharmacy & Research, KPGU, Vadodara, Gujarat, 391243, India
| | - Ajay Khopade
- Formulation Research & Development-Non-Orals, Sun Pharmaceutical Industries Ltd, Vadodara, 390012, Gujarat, India
| | - Krutika K Sawant
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390001, India
| |
Collapse
|
3
|
Mu G, Cao X, Shao L, Shen H, Guo X, Gao Y, Su C, Fan H, Yu Y, Shen Z. Progress and perspectives of metabolic biomarkers in human aortic dissection. Metabolomics 2024; 20:76. [PMID: 39002042 DOI: 10.1007/s11306-024-02140-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Aortic dissection (AD) significantly threated human cardiovascular health, extensive clinical-scientific research programs have been executed to uncover the pathogenesis and prevention. Unfortunately, no specific biomarker was identified for the causality or development of human AD. AIM OF REVIEW Metabolomics, a high-throughput technique capable of quantitatively detecting metabolites, holds considerable promise in discovering specific biomarkers and unraveling the underlying pathways involved. Aiming to provide a metabolite prediction in human AD, we collected the metabolomics data from 2003 to 2023, and diligently scrutinized with the online system MetaboAnalyst 6.0. KEY SCIENTIFIC CONCEPTS OF REVIEW Based on the data obtained, we have concluded the metabolic dynamics were highly correlated with human AD. Such metabolites (choline, serine and uridine) were frequently involved in the AD. Besides, the pathways, including amino acids metabolism and lipids metabolism, were also dysregulated in the disease. Due to the current limitation of metabolism analysis, the integrative omics data including genomics, transcriptomics, and proteomics were required for developing the specific biomarker for AD.
Collapse
Affiliation(s)
- Gaohang Mu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xiangyu Cao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Han Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xingyou Guo
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
- Department of Vascular Surgery, Suqian First Hospital, Suqian, 223800, Jiangsu, China
| | - Yamei Gao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Chengkai Su
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hongyou Fan
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - You Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China.
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China.
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
4
|
Zhang Y, Yu B, Qi Q, Azarbarzin A, Chen H, Shah NA, Ramos AR, Zee PC, Cai J, Daviglus ML, Boerwinkle E, Kaplan R, Liu PY, Redline S, Sofer T. Metabolomic profiles of sleep-disordered breathing are associated with hypertension and diabetes mellitus development. Nat Commun 2024; 15:1845. [PMID: 38418471 PMCID: PMC10902315 DOI: 10.1038/s41467-024-46019-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 02/12/2024] [Indexed: 03/01/2024] Open
Abstract
Sleep-disordered breathing (SDB) is a prevalent disorder characterized by recurrent episodic upper airway obstruction. Using data from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL), we apply principal component analysis (PCA) to seven SDB-related measures. We estimate the associations of the top two SDB PCs with serum levels of 617 metabolites, in both single-metabolite analysis, and a joint penalized regression analysis. The discovery analysis includes 3299 individuals, with validation in a separate dataset of 1522 individuals. Five metabolite associations with SDB PCs are discovered and replicated. SDB PC1, characterized by frequent respiratory events common in older and male adults, is associated with pregnanolone and progesterone-related sulfated metabolites. SDB PC2, characterized by short respiratory event length and self-reported restless sleep, enriched in young adults, is associated with sphingomyelins. Metabolite risk scores (MRSs), representing metabolite signatures associated with the two SDB PCs, are associated with 6-year incident hypertension and diabetes. These MRSs have the potential to serve as biomarkers for SDB, guiding risk stratification and treatment decisions.
Collapse
Affiliation(s)
- Ying Zhang
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Bing Yu
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Ali Azarbarzin
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, 02115, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Neomi A Shah
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alberto R Ramos
- Sleep Medicine Program, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Phyllis C Zee
- Division of Sleep Medicine, Department of Neurology, Northwestern University, Chicago, IL, 60611, USA
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Martha L Daviglus
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60612, USA
| | - Eric Boerwinkle
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Peter Y Liu
- The Institute for Translational Genomics and Population Sciences, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, 02115, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, 02115, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Ya'ar Bar S, Pintel N, Abd Alghne H, Khattib H, Avni D. The therapeutic potential of sphingolipids for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1224743. [PMID: 37608809 PMCID: PMC10440740 DOI: 10.3389/fcvm.2023.1224743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide and Inflammation plays a critical role in the development of CVD. Despite considerable progress in understanding the underlying mechanisms and various treatment options available, significant gaps in therapy necessitate the identification of novel therapeutic targets. Sphingolipids are a family of lipids that have gained attention in recent years as important players in CVDs and the inflammatory processes that underlie their development. As preclinical studies have shown that targeting sphingolipids can modulate inflammation and ameliorate CVDs, targeting sphingolipids has emerged as a promising therapeutic strategy. This review discusses the current understanding of sphingolipids' involvement in inflammation and cardiovascular diseases, the existing therapeutic approaches and gaps in therapy, and explores the potential of sphingolipids-based drugs as a future avenue for CVD treatment.
Collapse
Affiliation(s)
- Sapir Ya'ar Bar
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Noam Pintel
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Hesen Abd Alghne
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| | - Hamdan Khattib
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Department of Gastroenterology and Hepatology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| |
Collapse
|
6
|
Sofer T, Zhang Y, Yu B, Qi Q, Azarbarzin A, Chen H, Shah N, Ramos A, Zee P, Cai J, Daviglus M, Boerwinkle E, Kaplan R, Liu P, Redline S. Metabolomic Profiles of Sleep-Disordered Breathing are Associated with Hypertension and Diabetes Mellitus Development: the HCHS/SOL. RESEARCH SQUARE 2023:rs.3.rs-3171622. [PMID: 37503089 PMCID: PMC10371150 DOI: 10.21203/rs.3.rs-3171622/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Sleep-disordered breathing (SDB) is a prevalent disorder characterized by recurrent episodic upper airway obstruction. In a dataset from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL), we applied principal component analysis (PCA) on seven measures characterizing SDB-associated respiratory events. We estimated the association of the top two SDB PCs with serum levels of 617 metabolites, in both single-metabolite analysis, and a joint, penalized regression analysis using the least absolute shrinkage and selection operator (LASSO). Discovery analysis included n = 3,299 HCHS/SOL individuals; associations were validated in a separate dataset of n = 1,522 HCHS/SOL individuals. Seven metabolite associations with SDB PCs were discovered and replicated. Metabolite risk scores (MRSs) developed based on LASSO association results and representing metabolite signatures associated with the two SDB PCs were associated with 6-year incident hypertension and incident diabetes. MRSs have the potential to serve as biomarkers for SDB, guiding risk stratification and treatment decisions.
Collapse
Affiliation(s)
| | | | | | - Qibin Qi
- Albert Einstein College of Medicine
| | | | - Han Chen
- The University of Texas Health Science Center at Houston
| | | | | | - Phyllis Zee
- Northwestern University Feinberg School of Medicine
| | | | | | | | | | - Peter Liu
- Lundquist Institute at Harbor-UCLA Medical Center
| | | |
Collapse
|
7
|
Analysis of Intestinal Metabolites in SR-B1 Knockout Mice via Ultra-Performance Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020610. [PMID: 36677669 PMCID: PMC9866485 DOI: 10.3390/molecules28020610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Scavenger receptor class B type 1 (SR-B1), a multiligand membrane receptor, is expressed in a gradient along the gastrocolic axis. SR-B1 deficiency enhances lymphocyte proliferation and elevates inflammatory cytokine production in macrophages. However, whether SR-B1 affects intestinal metabolites is unclear. In this study, we detected metabolite changes in the intestinal tissue of SR-B1-/- mice, including amino acids and neurotransmitters, by ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF/MS) and HPLC. We found that SR-B1-/- mice exhibited changes in intestinal lipid metabolites and metabolic pathways, including the glycerophospholipid, sphingolipid, linoleic acid, taurine, and hypotaurine metabolic pathways. SR-B1 deficiency influenced the contents of amino acids and neurotransmitters in all parts of the intestine; the contents of leucine (LEU), phenylalanine (PHE), tryptophan (TRP), and tyrosine (TYR) were affected in all parts of the intestine; and the contents of 3,4-dihydroxyphenylacetic acid (DOPAC) and dopamine (DA) were significantly decreased in both the colon and rectum. In summary, SR-B1 deficiency regulated intestinal lipids, amino acids, and neurotransmitter metabolism in mice.
Collapse
|
8
|
Constantinescu V, Haase R, Akgün K, Ziemssen T. S1P receptor modulators and the cardiovascular autonomic nervous system in multiple sclerosis: a narrative review. Ther Adv Neurol Disord 2022; 15:17562864221133163. [PMID: 36437849 PMCID: PMC9685213 DOI: 10.1177/17562864221133163] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/29/2022] [Indexed: 01/21/2024] Open
Abstract
UNLABELLED Sphingosine 1-phosphate (S1P) receptor (S1PR) modulators have a complex mechanism of action, which are among the most efficient therapeutic options in multiple sclerosis (MS) and represent a promising approach for other immune-mediated diseases. The S1P signaling pathway involves the activation of five extracellular S1PR subtypes (S1PR1-S1PR5) that are ubiquitous and have a wide range of effects. Besides the immunomodulatory beneficial outcome in MS, S1P signaling regulates the cardiovascular function via S1PR1-S1PR3 subtypes, which reside on cardiac myocytes, endothelial, and vascular smooth muscle cells. In our review, we describe the mechanisms and clinical effects of S1PR modulators on the cardiovascular system. In the past, mostly short-term effects of S1PR modulators on the cardiovascular system have been studied, while data on long-term effects still need to be investigated. Immediate effects detected after treatment initiation are due to parasympathetic overactivation. In contrast, long-term effects may arise from a shift of the autonomic regulation toward sympathetic predominance along with S1PR1 downregulation. A mild increase in blood pressure has been reported in long-term studies, as well as decreased baroreflex sensitivity. In most studies, sustained hypertension was found to represent a significant adverse event related to treatment. The shift in the autonomic control and blood pressure values could not be just a consequence of disease progression but also related to S1PR modulation. Reduced cardiac autonomic activation and decreased heart rate variability during the long-term treatment with S1PR modulators may increase the risk for subsequent cardiac events. For second-generation S1PR modulators, this observation has to be confirmed in further studies with longer follow-ups. The periodic surveillance of cardiovascular function and detection of any cardiac autonomic dysfunction can help predict cardiac outcomes not only after the first dose but also throughout treatment. PLAIN LANGUAGE SUMMARY What is the cardiovascular effect of S1P receptor modulator therapy in multiple sclerosis? Sphingosine 1-phosphate (S1P) receptor (S1PR) modulators are among the most efficient therapies for multiple sclerosis. As small molecules, they are not only acting on the immune but on cardiovascular and nervous systems as well. Short-term effects of S1PR modulators on the cardiovascular system have already been extensively described, while long-term effects are less known. Our review describes the mechanisms of action and the short- and long-term effects of these therapeutic agents on the cardiovascular system in different clinical trials. We systematically reviewed the literature that had been published by January 2022. One hundred seven articles were initially identified by title and abstract using targeted keywords, and thirty-nine articles with relevance to cardiovascular effects of S1PR therapy in multiple sclerosis patients were thereafter considered, including their references for further accurate clarification. Studies on fingolimod, the first S1PR modulator approved for treating multiple sclerosis, primarily support the safety profile of this therapeutic class. The second-generation therapeutic agents along with a different treatment initiation approach helped mitigate several of the cardiovascular adverse effects that had previously been observed at the start of treatment. The heart rate may decrease when initiating S1PR modulators and, less commonly, the atrioventricular conduction may be prolonged, requiring cardiac monitoring for the first 6 h of medication. Continuous therapy with S1PR modulators can increase blood pressure values; therefore, the presence of arterial hypertension should be checked during long-term treatment. Periodic surveillance of the cardiovascular and autonomic functions can help predict cardiac outcomes and prevent possible adverse events in S1PR modulators treatment. Further studies with longer follow-ups are needed, especially for the second-generation of S1PR modulators, to confirm the safety profile of this therapeutic class.
Collapse
Affiliation(s)
- Victor Constantinescu
- Department of Neurology, Center of Clinical
Neuroscience, University Hospital Carl Gustav Carus, Dresden University of
Technology, Dresden, Germany
| | - Rocco Haase
- Department of Neurology, Center of Clinical
Neuroscience, University Hospital Carl Gustav Carus, Dresden University of
Technology, Dresden, Germany
| | - Katja Akgün
- Department of Neurology, Center of Clinical
Neuroscience, University Hospital Carl Gustav Carus, Dresden University of
Technology, Dresden, Germany
| | - Tjalf Ziemssen
- Department of Neurology, Center of Clinical
Neuroscience, University Hospital Carl Gustav Carus, Dresden University of
Technology, Fetscherstrasse 74, D-01307 Dresden, Germany
| |
Collapse
|
9
|
Constantinescu V, Akgün K, Ziemssen T. Current status and new developments in sphingosine-1-phosphate receptor antagonism: fingolimod and more. Expert Opin Drug Metab Toxicol 2022; 18:675-693. [PMID: 36260948 DOI: 10.1080/17425255.2022.2138330] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Fingolimod was the first oral disease-modifying treatment approved for relapsing-remitting multiple sclerosis (MS) that serves as a sphingosine-1-phosphate receptor (S1PR) agonist. The efficacy is primarily mediated by S1PR subtype 1 activation, leading to agonist-induced down-modulation of receptor expression and further functional antagonism, blocking the egression of auto-aggressive lymphocytes from the lymph nodes in the peripheral compartment. The role of S1P signaling in the regulation of other pathways in human organisms through different S1PR subtypes has received much attention due to its immune-modulatory function and its significance for the regeneration of the central nervous system (CNS). The more selective second-generation S1PR modulators have improved safety and tolerability profiles. AREAS COVERED This review has been carried out based on current data on S1PR modulators, emphasizing the benefits of recent advances in this emergent class of immunomodulatory treatment for MS. EXPERT OPINION Ongoing clinical research suggests that S1PR modulators represent an alternative to first-line therapies in selected cases of MS. A better understanding of the relevance of selective S1PR pathways and the ambition to optimize selective modulation has improved the safety and tolerability of S1PR modulators in MS therapy and opened new perspectives for the treatment of other diseases.
Collapse
Affiliation(s)
- Victor Constantinescu
- Center of Clinical Neuroscience, University Hospital, Fetscher Str. 74, 01307 Dresden, Germany
| | - Katja Akgün
- Center of Clinical Neuroscience, University Hospital, Fetscher Str. 74, 01307 Dresden, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, University Hospital, Fetscher Str. 74, 01307 Dresden, Germany
| |
Collapse
|
10
|
Molecular Characterization of Plasma HDL, LDL, and VLDL Lipids Cargos from Atherosclerotic Patients with Advanced Carotid Lesions: A Preliminary Report. Int J Mol Sci 2022; 23:ijms232012449. [PMID: 36293312 PMCID: PMC9604033 DOI: 10.3390/ijms232012449] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/26/2022] Open
Abstract
Carotid atherosclerosis represents a relevant healthcare problem, since unstable plaques are responsible for approximately 15% of neurologic events, namely transient ischemic attack and stroke. Although statins treatment has proven effective in reducing LDL-cholesterol and the onset of acute clinical events, a residual risk may persist suggesting the need for the detection of reliable molecular markers useful for the identification of patients at higher risk regardless of optimal medical therapy. In this regard, several lines of evidence show a relationship among specific biologically active plasma lipids, atherosclerosis, and acute clinical events. We performed a Selected Reaction Monitoring-based High Performance Liquid Chromatography-tandem Mass Spectrometry (SRM-based HPLC-MS/MS) analysis on plasma HDL, LDL, and VLDL fractions purified, by isopycnic salt gradient ultracentrifugation, from twenty-eight patients undergoing carotid endarterectomy, having either a “hard” or a “soft” plaque, with the aim of characterizing the specific lipidomic patterns associated with features of carotid plaque instability. One hundred and thirty lipid species encompassing different lipid (sub)classes were monitored. Supervised multivariate analysis showed that lipids belonging to phosphatidylethanolamine (PE), sphingomyelin (SM), and diacylglycerol (DG) classes mostly contribute to discrimination within each lipoprotein fraction according to the plaque typology. Differential analysis evidenced a significant dysregulation of LDL PE (38:6), SM (32:1), and SM (32:2) between the two groups of patients (adj. p-value threshold = 0.05 and log2FC ≥ |0.58|). Using this approach, some LDL-associated markers of plaque vulnerability have been identified, in line with the current knowledge of the key roles of these phospholipids in lipoprotein metabolism and cardiovascular disease. This proof-of-concept study reports promising results, showing that lipoprotein lipidomics may present a valuable approach for identifying new biomarkers of potential clinical relevance.
Collapse
|
11
|
Lee H, Jang SY, Jung Y, Kwon O, Hwang GS. Lipidomic profiling analysis of human plasma from subjects with hypercholesterolemia to evaluate the intake of yellow yeast rice fermented by Aspergillus terreus DSMK01. Food Funct 2022; 13:7629-7637. [PMID: 35734953 DOI: 10.1039/d1fo04010c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Yellow yeast rice (YYR) is a Korean functional food fermented with Aspergillus terreus and contains monacolin K, a cholesterol-lowering ingredient. However, the effects of YYR on lipid metabolism alterations have not been reported until now. In this study, we performed a mass spectrometry-based lipidomic analysis of plasma samples from subjects (31 from the YYR group and 27 from the placebo group) with LDL-C higher than 130 mg dL-1 to investigate the effects of the intake of YYR. Lipidomic profiling showed that the levels of sphingomyelin (SM) were significantly decreased in the YYR intake group compared with the placebo group. The SM level in the YYR intake group showed a significant association with the ApoB/ApoA1 ratio (p = 0.004, r = 0.503), an indicator of the effect of lipid-lowering therapy. This study suggests that global lipidomic profiling could be used to identify changes in lipid metabolism induced by YYR intake and provide information that these lipid changes are associated with improved hypercholesterolemia.
Collapse
Affiliation(s)
- Heeyeon Lee
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea. .,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seo Young Jang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea. .,Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngae Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea.
| | - Oran Kwon
- Department of Nutritional Science and Food Management, Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea. .,Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
12
|
Song Y, Cai C, Song Y, Sun X, Liu B, Xue P, Zhu M, Chai W, Wang Y, Wang C, Li M. A Comprehensive Review of Lipidomics and Its Application to Assess Food Obtained from Farm Animals. Food Sci Anim Resour 2022; 42:1-17. [PMID: 35028570 PMCID: PMC8728500 DOI: 10.5851/kosfa.2021.e59] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 12/04/2022] Open
Abstract
Lipids are one of the major macronutrients essential for adequate growth and
maintenance of human health. Their structure is not only complex but also
diverse, which makes systematic and holistic analyses challenging; consequently,
little is known regarding the relationship between phenotype and mechanism of
action. In recent years, rapid advancements have been made in the fields of
lipidomics and bioinformatics. In comparison with traditional approaches, mass
spectrometry-based lipidomics can rapidly identify as well as quantify
>1,000 lipid species at the same time, facilitating comprehensive, robust
analyses of lipids in tissues, cells, and body fluids. Accordingly, lipidomics
is now being widely applied in various fields, particularly food and nutrition
science. In this review, we discuss lipid classification, extraction techniques,
and detection and analysis using lipidomics. We also cover how lipidomics is
being used to assess food obtained from livestock and poultry. The information
included herein should serve as a reference to determine how to characterize
lipids in animal food samples, enhancing our understanding of the application of
lipidomics in the field in animal husbandry.
Collapse
Affiliation(s)
- Yinghua Song
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Changyun Cai
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Yingzi Song
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Xue Sun
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Baoxiu Liu
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Peng Xue
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Mingxia Zhu
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Wenqiong Chai
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Yonghui Wang
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Changfa Wang
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| | - Mengmeng Li
- College of Agronomy, Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252059, China
| |
Collapse
|
13
|
Luo D, Guo Z, Zhao X, Wu L, Liu X, Zhang Y, Zhang Y, Deng Z, Qu X, Cui S, Wan S. Novel 5-fluorouracil sensitizers for colorectal cancer therapy: Design and synthesis of S1P receptor 2 (S1PR2) antagonists. Eur J Med Chem 2022; 227:113923. [PMID: 34688013 DOI: 10.1016/j.ejmech.2021.113923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/21/2022]
Abstract
Sphingosine-1-phosphate receptor 2 (S1PR2) has been identified as a brand-new GPCR target for designing antagonists to reverse 5-FU resistance. We herein report the structural optimization and structure-activity relationship of JTE-013 derivatives as S1PR2 antagonists. Compound 9d was the most potent S1PR2 antagonist (KD = 34.8 nM) among developed compounds. Here, compound 9d could significantly inhibit the expression of dihydropyrimidine dehydrogenase (DPD) to reverse 5-FU-resistance in HCT116DPD and SW620/5-FU cells. Further mechanism studies demonstrated that compound 9d not only inhibited S1PR2 but also affected the transcription of S1PR2. In addition, compound 9d also showed acceptable selectivity to normal cells (NCM460). Importantly, compound 9d with suitable pharmacokinetic properties could significantly reverse 5-FU-resistance in the HCT116DPD and SW620/5-FU xenograft models without obvious toxicity, in which the inhibition rates of 5-FU were increased from 23.97% to 65.29% and 27.23% to 60.81%, respectively. Further immunohistochemistry and western blotting analysis also demonstrated that compound 9d significantly decreases the expression of DPD in tumor and liver tissues. These results indicated that compound 9d is a promising lead compound to reverse 5-FU-resistance for colorectal cancer therapy.
Collapse
Affiliation(s)
- Dongdong Luo
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003, Qingdao, China
| | - Zhikun Guo
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China
| | - Xuecui Zhao
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003, Qingdao, China
| | - Lijuan Wu
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003, Qingdao, China
| | - Xiaochun Liu
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003, Qingdao, China
| | - Yingzhi Zhang
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China
| | - Yuhang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Zirong Deng
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003, Qingdao, China
| | - Xianjun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Shuxiang Cui
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China.
| | - Shengbiao Wan
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003, Qingdao, China.
| |
Collapse
|
14
|
Ermini L, Farrell A, Alahari S, Ausman J, Park C, Sallais J, Melland-Smith M, Porter T, Edson M, Nevo O, Litvack M, Post M, Caniggia I. Ceramide-Induced Lysosomal Biogenesis and Exocytosis in Early-Onset Preeclampsia Promotes Exosomal Release of SMPD1 Causing Endothelial Dysfunction. Front Cell Dev Biol 2021; 9:652651. [PMID: 34017832 PMCID: PMC8130675 DOI: 10.3389/fcell.2021.652651] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Aberrant ceramide build-up in preeclampsia, a serious disorder of pregnancy, causes exuberant autophagy-mediated trophoblast cell death. The significance of ceramide accumulation for lysosomal biogenesis in preeclampsia is unknown. Here we report that lysosome formation is markedly increased in trophoblast cells of early-onset preeclamptic placentae, in particular in syncytiotrophoblasts. This is accompanied by augmented levels of transcription factor EB (TFEB). In vitro and in vivo experiments demonstrate that ceramide increases TFEB expression and nuclear translocation and induces lysosomal formation and exocytosis. Further, we show that TFEB directly regulates the expression of lysosomal sphingomyelin phosphodiesterase (L-SMPD1) that degrades sphingomyelin to ceramide. In early-onset preeclampsia, ceramide-induced lysosomal exocytosis carries L-SMPD1 to the apical membrane of the syncytial epithelium, resulting in ceramide accumulation in lipid rafts and release of active L-SMPD1 via ceramide-enriched exosomes into the maternal circulation. The SMPD1-containing exosomes promote endothelial activation and impair endothelial tubule formation in vitro. Both exosome-induced processes are attenuated by SMPD1 inhibitors. These findings suggest that ceramide-induced lysosomal biogenesis and exocytosis in preeclamptic placentae contributes to maternal endothelial dysfunction, characteristic of this pathology.
Collapse
Affiliation(s)
- Leonardo Ermini
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Abby Farrell
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Sruthi Alahari
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Jonathan Ausman
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Chanho Park
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Julien Sallais
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Megan Melland-Smith
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Tyler Porter
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Michael Edson
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Ori Nevo
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Michael Litvack
- Translational Medicine Program, Peter Gilgan Center, The Hospital for Sick Children, Toronto, ON, Canada
| | - Martin Post
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Translational Medicine Program, Peter Gilgan Center, The Hospital for Sick Children, Toronto, ON, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Al‐Sari N, Schmidt S, Suvitaival T, Kim M, Trošt K, Ranjan AG, Christensen MB, Overgaard AJ, Pociot F, Nørgaard K, Legido‐Quigley C. Changes in the lipidome in type 1 diabetes following low carbohydrate diet: Post-hoc analysis of a randomized crossover trial. Endocrinol Diabetes Metab 2021; 4:e00213. [PMID: 33855215 PMCID: PMC8029500 DOI: 10.1002/edm2.213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/14/2020] [Indexed: 01/14/2023] Open
Abstract
Aims Lipid metabolism might be compromised in type 1 diabetes, and the understanding of lipid physiology is critically important. This study aimed to compare the change in plasma lipid concentrations during carbohydrate dietary changes in individuals with type 1 diabetes and identify links to early-stage dyslipidaemia. We hypothesized that (1) the lipidomic profiles after ingesting low or high carbohydrate diet for 12 weeks would be different; and (2) specific annotated lipid species could have significant associations with metabolic outcomes. Methods Ten adults with type 1 diabetes (mean ± SD: age 43.6 ± 13.8 years, diabetes duration 24.5 ± 13.4 years, BMI 24.9 ± 2.1 kg/m2, HbA1c 57.6 ± 2.6 mmol/mol) using insulin pumps participated in a randomized 2-period crossover study with a 12-week intervention period of low carbohydrate diet (< 100 g carbohydrates/day) or high carbohydrate diet (> 250 g carbohydrates/day), respectively, separated by a 12-week washout period. A large-scale non-targeted lipidomics was performed with mass spectrometry in fasting plasma samples obtained before and after each diet intervention. Longitudinal lipid levels were analysed using linear mixed-effects models. Results In total, 289 lipid species were identified from 14 major lipid classes. Comparing the two diets, 11 lipid species belonging to sphingomyelins, phosphatidylcholines and LPC(O-16:0) were changed. All the 11 lipid species were significantly elevated during low carbohydrate diet. Two lipid species were most differentiated between diets, namely SM(d36:1) (β ± SE: 1.44 ± 0.28, FDR = 0.010) and PC(P-36:4)/PC(O-36:5) (β ± SE: 1.34 ± 0.25, FDR = 0.009) species. Polyunsaturated PC(35:4) was inversely associated with BMI and positively associated with HDL cholesterol (p < .001). Conclusion Lipidome-wide outcome analysis of a randomized crossover trial of individuals with type 1 diabetes following a low carbohydrate diet showed an increase in sphingomyelins and phosphatidylcholines which are thought to reduce dyslipidaemia. The polyunsaturated phosphatidylcholine 35:4 was inversely associated with BMI and positively associated with HDL cholesterol (p < .001). Results from this study warrant for more investigation on the long-term effect of single lipid species in type 1 diabetes.
Collapse
Affiliation(s)
| | - Signe Schmidt
- Steno Diabetes Center CopenhagenGentofteDenmark
- Danish Diabetes AcademyOdenseDenmark
- Department of EndocrinologyCopenhagen University Hospital HvidovreHvidovreDenmark
| | | | - Min Kim
- Steno Diabetes Center CopenhagenGentofteDenmark
| | - Kajetan Trošt
- Steno Diabetes Center CopenhagenGentofteDenmark
- Present address:
Novo Nordisk foundation Center for Basic Metabolic ResearchKøbenhavn NDenmark
| | - Ajenthen G. Ranjan
- Steno Diabetes Center CopenhagenGentofteDenmark
- Danish Diabetes AcademyOdenseDenmark
- Department of EndocrinologyCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Merete B. Christensen
- Steno Diabetes Center CopenhagenGentofteDenmark
- Department of EndocrinologyCopenhagen University Hospital HvidovreHvidovreDenmark
| | | | - Flemming Pociot
- Steno Diabetes Center CopenhagenGentofteDenmark
- Department of Clinical MedicineUniversity of CopenhagenKøbenhavnDenmark
| | - Kirsten Nørgaard
- Steno Diabetes Center CopenhagenGentofteDenmark
- Department of EndocrinologyCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Cristina Legido‐Quigley
- Steno Diabetes Center CopenhagenGentofteDenmark
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| |
Collapse
|
16
|
Liu J, Yuan J, Zhao J, Zhang L, Wang Q, Wang G. Serum metabolomic patterns in young patients with ischemic stroke: a case study. Metabolomics 2021; 17:24. [PMID: 33554271 DOI: 10.1007/s11306-021-01774-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/22/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ischemic stroke is one of the leading causes of death and adult disability. The incidence of ischemic stroke continues to rise in young adults. This study aimed to provide a comprehensive evaluation of metabolic changes and explore possible mechanisms in young ischemic stroke patients without common risk factors. METHODS This study investigated serum metabolomics in 50 young patients with newly suffered ischemic stroke and 50 age-, sex-, and body mass index-matched healthy controls. Liquid chromatography coupled with a Waters Xevo TQ-S mass spectrometer with an electrospray ionization (ESI) source was used to analyze amino acid or bile acid, and free fatty acid or lipid was analyzed by liquid chromatography coupled with a Qtrap5500 mass spectrometer with an ESI source. The metabolomic data were analyzed by performing a multivariate statistical analysis. RESULTS A total of 197 metabolites, including amino acids, bile acids, free fatty acids, and lipids, were identified in all participants. Multivariate models showed significant differences in serum metabolomic patterns between young patients with ischemic stroke and healthy controls. The stroke patients had increased L-methionine, homocysteine, glutamine, uric acid, GCDCA, and PE (18:0/20:4, 16:0/22:5), and decreased levels of L-citrulline, taurine, PC (16:2/22:6, 16:2/20:5, 15:0/18:2), and SM (d18:1/23:0, d20:0/19:1, d18:1/22:0, d16:0/26:1, d16:0/18:0, d16:0/22:1, d18:1/19:1, d16:0/17:1, d16:1/24:1, d18:1/19:0). Based on the identified metabolites, the metabolic pathways of arginine biosynthesis, glycerophospholipid metabolism, and taurine and hypotaurine metabolism were significantly enriched in the young patients with ischemic stroke. CONCLUSIONS Serum metabolomic patterns were significantly different between young patients with ischemic stroke and healthy controls. Our study is beneficial in providing a further view into the pathophysiology of young patients with ischemic stroke.
Collapse
Affiliation(s)
- Jia Liu
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Junliang Yuan
- Department of Neurology, Peking University Sixth Hospital, Beijing, 100191, China
| | - Jingwei Zhao
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Lin Zhang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Qiu Wang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
17
|
Chorell E, Olsson T, Jansson JH, Wennberg P. Lysophospholipids as Predictive Markers of ST-Elevation Myocardial Infarction (STEMI) and Non-ST-Elevation Myocardial Infarction (NSTEMI). Metabolites 2020; 11:metabo11010025. [PMID: 33396480 PMCID: PMC7823877 DOI: 10.3390/metabo11010025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
The present study explored patterns of circulating metabolites and proteins that can predict future risk for ST-elevation myocardial infarction (STEMI) and non-ST-elevation myocardial infarction (NSTEMI). We conducted a prospective nested case-control study in northern Sweden in individuals who developed STEMI (N = 50) and NSTEMI (N = 50) within 5 years and individually matched controls (N = 100). Fasted plasma samples were subjected to multiplatform mass spectrometry-based metabolomics and multiplex protein analyses. Multivariate analyses were used to elucidate infarction-specific metabolite and protein risk profiles associated with future incident STEMI and NSTEMI. We found that altered lysophosphatidylcholine (LPC) to lysophosphatidylethanolamine (LPE) ratio predicted STEMI and NSTEMI events in different ways. In STEMI, lysophospholipids (mainly LPEs) were lower, whereas in NSTEMI, lysophospholipids (mainly LPEs) were higher. We found a similar response for all detected lysophospholipids but significant alterations only for those containing linoleic acid (C18:2, p < 0.05). Patients with STEMI had higher secretoglobin family 3A member 2 and tartrate-resistant acid phosphate type 5 and lower platelet-derived growth factor subunit A, which are proteins associated with atherosclerosis severity and plaque development mediated via altered phospholipid metabolism. In contrast, patients with NSTEMI had higher levels of proteins associated with inflammation and macrophage activation, including interleukin 6, C-reactive protein, chemerin, and cathepsin X and D. The STEMI risk marker profile includes factors closely related to the development of unstable plaque, including a higher LPC:LPE ratio, whereas NSTEMI is characterized by a lower LPC:LPE ratio and increased inflammation.
Collapse
Affiliation(s)
- Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, SE-901 87 Umeå, Sweden;
- Correspondence: ; Tel.: +46-(0)90-785-1326
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Umeå University, SE-901 87 Umeå, Sweden;
| | - Jan-Håkan Jansson
- Research Unit Skellefteå, Department of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Patrik Wennberg
- Department of Public Health and Clinical Medicine, Family Medicine, Medicine, Umeå University, 90187 Umeå, Sweden;
| |
Collapse
|
18
|
Jozefczuk E, Guzik TJ, Siedlinski M. Significance of sphingosine-1-phosphate in cardiovascular physiology and pathology. Pharmacol Res 2020; 156:104793. [PMID: 32278039 DOI: 10.1016/j.phrs.2020.104793] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 02/07/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a signaling lipid, synthetized by sphingosine kinases (SPHK1 and SPHK2), that affects cardiovascular function in various ways. S1P signaling is complex, particularly since its molecular action is reliant on the differential expression of its receptors (S1PR1, S1PR2, S1PR3, S1PR4, S1PR5) within various tissues. Significance of this sphingolipid is manifested early in vertebrate development as certain defects in S1P signaling result in embryonic lethality due to defective vasculo- or cardiogenesis. Similar in the mature organism, S1P orchestrates both physiological and pathological processes occurring in the heart and vasculature of higher eukaryotes. S1P regulates cell fate, vascular tone, endothelial function and integrity as well as lymphocyte trafficking, thus disbalance in its production and signaling has been linked with development of such pathologies as arterial hypertension, atherosclerosis, endothelial dysfunction and aberrant angiogenesis. Number of signaling mechanisms are critical - from endothelial nitric oxide synthase through STAT3, MAPK and Akt pathways to HDL particles involved in redox and inflammatory balance. Moreover, S1P controls both acute cardiac responses (cardiac inotropy and chronotropy), as well as chronic processes (such as apoptosis and hypertrophy), hence numerous studies demonstrate significance of S1P in the pathogenesis of hypertrophic/fibrotic heart disease, myocardial infarction and heart failure. This review presents current knowledge concerning the role of S1P in the cardiovascular system, as well as potential therapeutic approaches to target S1P signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- E Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - T J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland; Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - M Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland; Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
19
|
Elevated plasma ceramide levels in post-menopausal women: a cross-sectional study. Aging (Albany NY) 2020; 11:73-88. [PMID: 30620722 PMCID: PMC6339790 DOI: 10.18632/aging.101719] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/06/2018] [Indexed: 12/22/2022]
Abstract
Circulating ceramide levels are abnormally elevated in age-dependent pathologies such as cardiovascular diseases, obesity and Alzheimer's disease. Nevertheless, the potential impact of age on plasma ceramide levels has not yet been systematically examined. In the present study, we quantified a focused panel of plasma ceramides and dihydroceramides in a cohort of 164 subjects (84 women) 19 to 80 years of age. After adjusting for potential confounders, multivariable linear regression analysis revealed a positive association between age and ceramide (d18:1/24:0) (β (SE) = 5.67 (2.38); p = .0198) and ceramide (d18:1/24:1) (β (SE) = 2.88 (.61); p < .0001) in women, and between age and ceramide (d18:1/24:1) in men (β (SE) = 1.86 (.77); p = .0179). In women of all ages, but not men, plasma ceramide (d18:1/24:1) was negatively correlated with plasma estradiol (r = -0.294; p = .007). Finally, in vitro experiments in human cancer cells expressing estrogen receptors showed that incubation with estradiol (10 nM, 24 h) significantly decreased ceramide accumulation. Together, the results suggest that aging is associated with an increase in circulating ceramide levels, which in post-menopausal women is at least partially associated with lower estradiol levels.
Collapse
|
20
|
Józefczuk E, Nosalski R, Saju B, Crespo E, Szczepaniak P, Guzik TJ, Siedlinski M. Cardiovascular Effects of Pharmacological Targeting of Sphingosine Kinase 1. Hypertension 2020; 75:383-392. [PMID: 31838904 PMCID: PMC7055939 DOI: 10.1161/hypertensionaha.119.13450] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/11/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023]
Abstract
High blood pressure is a risk factor for cardiovascular diseases. Ang II (angiotensin II), a key pro-hypertensive hormone, mediates target organ consequences such as endothelial dysfunction and cardiac hypertrophy. S1P (sphingosine-1-phosphate), produced by Sphk1 (sphingosine kinase 1), plays a pivotal role in the pathogenesis of hypertension and downstream organ damage, as it controls vascular tone and regulates cardiac remodeling. Accordingly, we aimed to examine if pharmacological inhibition of Sphk1 using selective inhibitor PF543 can represent a useful vasoprotective and cardioprotective anti-hypertensive strategy in vivo. PF543 was administered intraperitoneally throughout a 14-day Ang II-infusion in C57BL6/J male mice. Pharmacological inhibition of Sphk1 improved endothelial function of arteries of hypertensive mice that could be mediated via decrease in eNOS (endothelial nitric oxide synthase) phosphorylation at T495. This effect was independent of blood pressure. Importantly, PF543 also reduced cardiac hypertrophy (heart to body weight ratio, 5.6±0.2 versus 6.4±0.1 versus 5.9±0.2 mg/g; P<0.05 for Sham, Ang II+placebo, and Ang II+PF543-treated mice, respectively). Mass spectrometry revealed that PF543 elevated cardiac sphingosine, that is, Sphk1 substrate, content in vivo. Mechanistically, RNA-Seq indicated a decreased expression of cardiac genes involved in actin/integrin organization, S1pr1 signaling, and tissue remodeling. Indeed, downregulation of Rock1 (Rho-associated coiled-coil containing protein kinase 1), Stat3 (signal transducer and activator of transcription 3), PKC (protein kinase C), and ERK1/2 (extracellular signal-regulated kinases 1/2) level/phosphorylation by PF543 was observed. In summary, pharmacological inhibition of Sphk1 partially protects against Ang II-induced cardiac hypertrophy and endothelial dysfunction. Therefore, it may represent a promising target for harnessing residual cardiovascular risk in hypertension.
Collapse
Affiliation(s)
- Ewelina Józefczuk
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| | - Ryszard Nosalski
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Blessy Saju
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Eva Crespo
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Piotr Szczepaniak
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| | - Tomasz Jan Guzik
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Mateusz Siedlinski
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| |
Collapse
|
21
|
Mielke MM, Syrjanen JA, Bui HH, Petersen RC, Knopman DS, Jack CR, Graff-Radford J, Vemuri P. Elevated Plasma Ceramides Are Associated With Higher White Matter Hyperintensity Volume-Brief Report. Arterioscler Thromb Vasc Biol 2019; 39:2431-2436. [PMID: 31510790 PMCID: PMC6812619 DOI: 10.1161/atvbaha.119.313099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/03/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Sphingolipids, including S1P (sphingosine-1-phosphate) and ceramides, have been associated with vascular tone, blood pressure regulation, cardiovascular outcomes, and mortality. However, the relationship between plasma sphingolipids and cerebrovascular disease has not been examined. We aimed to assess the cross-sectional association between plasma sphingolipids and white matter hyperintensity (WMH) volume, which is a marker of cerebrovascular disease. Approach and Results: We included 588 participants (302 men and 286 women), aged 60 to 93, enrolled in the population-based Mayo Clinic Study of Aging who had MRI and plasma sphingolipids at the same study visit. Fasting plasma was obtained, and ceramides and S1P were assayed using liquid chromatography-electrospray ionization tandem mass spectrometry. Fluid-attenuated inversion recovery was used to measure WMH volume, defined as percent total intracranial volume. We used linear regression to cross-sectionally examine the relationships between plasma sphingolipids and WMH; both were log-transformed. In multivariable analyses adjusting for age, sex, and hypertension, higher levels of ceramide C16:0 (b [95% CI]=0.24 [0.02-0.45]) and the ceramide ratios C16:0_24:0 (b [95% CI]=0.30 [0.12-0.48]) and C24:1_24:0 (b [95% CI]=0.24 [0.07-0.41]) were associated with a higher WMH volume. A higher ceramide score was also associated with higher WMH volume (b [95% CI]=0.03 (0.01-0.04]). We did not observe any association between S1P and WMH volume. CONCLUSIONS Higher plasma ceramide C16:0 and 2 specific ceramide ratios (C16:0_24:0 and C24:1_24:0) are associated with greater WMH volumes, independent of hypertension, suggesting their utility for measurement of cerebrovascular disease.
Collapse
Affiliation(s)
- Michelle M Mielke
- From the Departments of Health Sciences Research (M.M.M., J.A.S., R.C.P.), Mayo Clinic Rochester, MN
- Neurology (M.M.M., R.C.P., D.S.K., J.G.-R.), Mayo Clinic Rochester, MN
| | - Jeremy A Syrjanen
- From the Departments of Health Sciences Research (M.M.M., J.A.S., R.C.P.), Mayo Clinic Rochester, MN
| | - Hai H Bui
- Eli Lilly and Company, Indianapolis, IN (H.H.B.)
| | - Ronald C Petersen
- From the Departments of Health Sciences Research (M.M.M., J.A.S., R.C.P.), Mayo Clinic Rochester, MN
- Neurology (M.M.M., R.C.P., D.S.K., J.G.-R.), Mayo Clinic Rochester, MN
| | - David S Knopman
- Neurology (M.M.M., R.C.P., D.S.K., J.G.-R.), Mayo Clinic Rochester, MN
| | | | | | | |
Collapse
|
22
|
Hernández-Coronado CG, Guzmán A, Castillo-Juárez H, Zamora-Gutiérrez D, Rosales-Torres AM. Sphingosine-1-phosphate (S1P) in ovarian physiology and disease. ANNALES D'ENDOCRINOLOGIE 2019; 80:263-272. [DOI: 10.1016/j.ando.2019.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/06/2019] [Accepted: 06/20/2019] [Indexed: 12/15/2022]
|
23
|
Alganga H, Almabrouk TAM, Katwan OJ, Daly CJ, Pyne S, Pyne NJ, Kennedy S. Short Periods of Hypoxia Upregulate Sphingosine Kinase 1 and Increase Vasodilation of Arteries to Sphingosine 1-Phosphate (S1P) via S1P 3. J Pharmacol Exp Ther 2019; 371:63-74. [PMID: 31371480 DOI: 10.1124/jpet.119.257931] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
Sphingosine kinase [(SK), isoforms SK1 and SK2] catalyzes the formation of the bioactive lipid, sphingosine 1-phosphate (S1P). This can be exported from cells and bind to S1P receptors to modulate vascular function. We investigated the effect of short-term hypoxia on SK1 expression and the response of arteries to S1P. SK1 expression in rat aortic and coronary artery endothelial cells was studied using immunofluorescence and confocal microscopy. Responses of rat aortic rings were studied using wire myography and reversible hypoxia induced by bubbling myography chambers with 95% N2:5% CO2 Inhibitors were added 30 minutes before induction of hypoxia. S1P induced endothelium-dependent vasodilation via activation of S1P3 receptors and generation of nitric oxide. Hypoxia significantly increased relaxation to S1P and this was attenuated by (2R)-1-[[(4-[[3-methyl-5-[(phenylsulfonyl)methyl] phenoxy]methyl]phenyl]methyl]-2-pyrrolidinemethanol [(PF-543), SK1 inhibitor] but not (R)-FTY720 methyl ether [(ROMe), SK2 inhibitor]. Hypoxia also increased vessel contractility to the thromboxane mimetic, 9,11-dideoxy-11α,9α-epoxymethanoprostaglandin F2α, which was further increased by PF-543 and ROMe. Hypoxia upregulated SK1 expression in aortic and coronary artery endothelial cells and this was blocked by PF-543 and 2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole [(SKi), SK1/2 inhibitor]. The effects of PF-543 and SKi were associated with increased proteasomal/lysosomal degradation of SK1. A short period of hypoxia increases the expression of SK1, which may generate S1P to oppose vessel contraction. Under hypoxic conditions, upregulation of SK1 is likely to lead to increased export of S1P from the cell and vasodilation via activation of endothelial S1P3 receptors. These data have significance for perfusion of tissue during episodes of ischemia.
Collapse
Affiliation(s)
- H Alganga
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - T A M Almabrouk
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - O J Katwan
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - C J Daly
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - S Pyne
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - N J Pyne
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - S Kennedy
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| |
Collapse
|
24
|
Zhou X, Wang R, Zhang T, Liu F, Zhang W, Wang G, Gu G, Han Q, Xu D, Yao C, Guo D, Fu W, Qi Y, Wang L. Identification of Lysophosphatidylcholines and Sphingolipids as Potential Biomarkers for Acute Aortic Dissection via Serum Metabolomics. Eur J Vasc Endovasc Surg 2018; 57:434-441. [PMID: 30087010 DOI: 10.1016/j.ejvs.2018.07.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/03/2018] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Acute aortic dissection (AAD) is a severe clinical emergency with a high mortality, and is easily misdiagnosed in its early stage. This study aimed at discovering serum metabolomic markers with the potential to diagnose AAD and distinguish between two subtypes of AAD. METHODS Thirty-five patients with AAD, including 20 with Stanford type A and 15 with Stanford type B were enrolled in this study, together with 20 healthy controls. All patients with AAD were admitted within 72 h of onset. Serum metabolomics profiles were determined by ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry and the data were analysed by principal component analysis and partial least squares discriminant analysis. RESULTS A total of 17 metabolites differing between the control and AAD groups were finally screened and identified as lysophosphatidylcholines (LPC) and sphingolipids including sphinganine, phytosphingosine, sphingomyelin, and ceramide. Compared with those in the healthy control group, LPC levels were significantly lower in both the Stanford type A and type B AAD groups. Interestingly, sphingolipids, including sphinganine, phytosphingosine, and ceramide, were remarkably reduced in the Stanford type A AAD group, but not in the Stanford type B AAD group. Subgroup analysis showed that the changes in LPC and sphingolipid levels were unrelated to hypertension or gender. CONCLUSIONS The present results indicate that LPCs and sphingolipids are significantly altered in patients with AAD, and several sphingolipids, such as sphinganine, phytosphingosine, and ceramide, were dramatically decreased in patients with Stanford type A AAD. A combination of these two families of metabolites could serve as a potential biomarker for the diagnosis of AAD and distinguishing between Stanford type A and Stanford type B.
Collapse
Affiliation(s)
- Xiushi Zhou
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Vascular Surgery Institute of Fudan University, Shanghai, China
| | - Renping Wang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tian Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Fei Liu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Vascular Surgery Institute of Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Vascular Surgery Institute of Fudan University, Shanghai, China
| | - Guili Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Vascular Surgery Institute of Fudan University, Shanghai, China
| | - Guorong Gu
- Department of Emergency, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qinqi Han
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Demin Xu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenling Yao
- Department of Emergency, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Daqiao Guo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Vascular Surgery Institute of Fudan University, Shanghai, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Vascular Surgery Institute of Fudan University, Shanghai, China; Department of Vascular Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Yunpeng Qi
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Vascular Surgery Institute of Fudan University, Shanghai, China; Department of Vascular Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China.
| |
Collapse
|
25
|
Mitsnefes MM, Fitzpatrick J, Sozio SM, Jaar BG, Estrella MM, Monroy-Trujillo JM, Zhang W, Setchell K, Parekh RS. Plasma glucosylceramides and cardiovascular risk in incident hemodialysis patients. J Clin Lipidol 2018; 12:1513-1522.e4. [PMID: 30143433 DOI: 10.1016/j.jacl.2018.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/20/2018] [Accepted: 07/22/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Recent population-based studies identified plasma sphingolipids as independent predictors of increased cardiovascular disease (CVD) morbidity and mortality. Understanding the impact of sphingolipids on CVD outcomes in patients on dialysis, who suffer from higher risk of these conditions, is important for risk assessment and treatment. OBJECTIVE To measure plasma sphingolipid levels and determine their associations with CVD in adults initiating maintenance hemodialysis. METHODS To evaluate associations of plasma sphingolipids with intermediate cardiovascular outcomes (hypertension, left ventricular hypertrophy, and decreased ejection fraction), cardiovascular mortality, and all-cause mortality in patients with end-stage renal disease, we measured plasma levels of ceramides, glucosylceramides, and lactosylceramides from the family of sphingolipids in 368 incident hemodialysis patients enrolled in the Predictors of Arrhythmic and Cardiovascular Risk in End-Stage Renal Disease study. RESULTS Glucosylceramide C16GC (per 1 log μM increase) was associated with higher odds of having uncontrolled hypertension (odds ratio [OR]: 1.34; 95% confidential interval [CI]: 1.01-1.76), left ventricular hypertrophy (OR: 1.53; 95% CI: 1.11-2.13), and reduced ejection fraction (OR: 1.05; 95% CI: 1.00-1.11) in fully adjusted models. During a median 2.5 years of follow-up, there were 78 deaths from all causes, of which 33 were from CVD. Mortality was higher among those in the highest tertile of C16GC for all causes (HR: 1.81; 95% CI: 1.02-3.22) and CVD (HR: 2.63, 95% CI: 1.08-6.55). CONCLUSIONS These results suggest that abnormal glycosphingolipid metabolism might contribute to increased CVD risk in end-stage renal disease.
Collapse
Affiliation(s)
- Mark M Mitsnefes
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Jessica Fitzpatrick
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephen M Sozio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Baltimore, MD, USA
| | - Bernard G Jaar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Baltimore, MD, USA; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Nephrology Center of Maryland, Baltimore, MD, USA
| | - Michelle M Estrella
- Kidney Health Research Collaborative, Department of Medicine, University of California, San Francisco and Department of Medicine, San Francisco VA Medical Center, San Francisco, CA, USA
| | | | - Wujuan Zhang
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kenneth Setchell
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rulan S Parekh
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Division of Nephrology, Department of Pediatrics and Medicine, The Hospital for Sick Children, University Health Network and University of Toronto, Ontario, Canada
| |
Collapse
|
26
|
Meng Y, Du Z, Li Y, Wang L, Gao P, Gao X, Li C, Zhao M, Jiang Y, Tu P, Guo X. Integration of Metabolomics With Pharmacodynamics to Elucidate the Anti-myocardial Ischemia Effects of Combination of Notoginseng Total Saponins and Safflower Total Flavonoids. Front Pharmacol 2018; 9:667. [PMID: 29988484 PMCID: PMC6026671 DOI: 10.3389/fphar.2018.00667] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 06/04/2018] [Indexed: 01/20/2023] Open
Abstract
Notoginseng (Sanqi), the roots and rhizomes of Panax notoginseng and safflower, the flowers of Carthamus tinctorius, are widely used traditional Chinese medicines (TCMs) for the treatment of cardiovascular diseases. Positive evidences have fueled growing acceptance for cardioprotective effects of the combination of the notoginseng total saponins and safflower total flavonoids (CNS) against myocardial ischemia (MI). However, the underlying cardioprotective mechanisms of CNS are still obscured. Metabolomics is a comprehensive tool for investigating biological mechanisms of disease, monitoring therapeutic outcomes, and advancing drug discovery and development. Herein, we investigated the cardioprotective effects of CNS on the isoproterenol (ISO)-induced MI rats by using plasma and urine metabolomics based on ultra-performance liquid chromatography coupled with quadrupole-time of flight mass spectrometry (UPLC-Q-TOF/MS) and multiple pharmacodynamics approaches. The results showed that pretreatment with CNS could attenuate the cardiac injury resulting from ISO, as evidenced by decreasing the myocardial infarct size, converting the echocardiographic, histopathological, and plasma biochemical abnormalities, and reversing the perturbations of plasma and urine metabolic profiles, particularly for the 55.0 mg/kg dosage group. In addition, 44 metabolites were identified as the potential MI biomarkers, mainly including a range of free fatty acids (FFAs), sphingolipids, and glycerophospholipids. CNS pretreatment group may robustly ameliorate these potential MI-related biomarkers. The accumulation of LysoPCs and FFAs, caused by PLA2, may activate NF-κB pathway and increase proinflammatory cytokines. However, our results showed that CNS at 55.0 mg/kg dosage could maximally attenuate the NF-κB signaling pathway, depress the expressions of TNF-α, IL-6, IL-1β, and PLA2. The results suggested that the anti-inflammatory property of CNS may contribute to its cardioprotection against MI. Our results demonstrate that the integrating of metabolomics with pharmacodynamics provides a reasonable approach for understanding the therapeutic effects of TCMs and CNS provide a potential candidate for prevention and treatment of MI.
Collapse
Affiliation(s)
- Yuqing Meng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhiyong Du
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yan Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Lichao Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Peng Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoyan Gao
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Mingbo Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoyu Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
27
|
Anupriya MG, Singh S, Hulyalkar NV, Sreekumar E. Sphingolipid signaling modulates trans-endothelial cell permeability in dengue virus infected HMEC-1 cells. Prostaglandins Other Lipid Mediat 2018; 136:44-54. [PMID: 29733947 DOI: 10.1016/j.prostaglandins.2018.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/01/2018] [Accepted: 05/03/2018] [Indexed: 12/07/2022]
Abstract
Dengue has emerged as a major mosquito-borne disease in the tropics and subtropics. In severe dengue, enhanced microvascular endothelial permeability leads to plasma leakage. Direct dengue virus (DENV) infection in human microvascular endothelial cells (HMEC-1) can enhance trans-endothelial leakage. Using a microarray-based analysis, we identified modulation of key endothelial cell signaling pathways in DENV-infected HMEC-1 cells. One among them was the sphingolipid pathway that regulates vascular barrier function. Sphingosine-1-phosphate receptor 2 (S1PR2) and S1PR5 showed significant up-regulation in the microarray data. In DENV-infected cells, the kinetics of S1PR2 transcript expression and enhanced in vitro trans-endothelial permeability showed a correlation. We also observed an internalization and cytoplasmic translocation of VE-Cadherin, a component of adherens junctions (AJ), upon infection indicating AJ disassembly. Further, inhibition of S1PR2 signaling by a specific pharmacological inhibitor prevented translocation of VE-Cadherin, thus helping AJ maintenance, and abrogated DENV-induced trans-endothelial leakage. Our results show that sphingolipid signaling, especially that involving S1PR2, plays a critical role in vascular leakage in dengue.
Collapse
Affiliation(s)
- M G Anupriya
- Molecular Virology laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud P.O., Thiruvananthapuram, 695014, Kerala, India; Research Scholar, University of Kerala, India
| | - Sneha Singh
- Molecular Virology laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud P.O., Thiruvananthapuram, 695014, Kerala, India; Research Scholar, University of Kerala, India
| | - Neha Vijay Hulyalkar
- Molecular Virology laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud P.O., Thiruvananthapuram, 695014, Kerala, India
| | - Easwaran Sreekumar
- Molecular Virology laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud P.O., Thiruvananthapuram, 695014, Kerala, India.
| |
Collapse
|
28
|
Morris TG, Borland SJ, Clarke CJ, Wilson C, Hannun YA, Ohanian V, Canfield AE, Ohanian J. Sphingosine 1-phosphate activation of ERM contributes to vascular calcification. J Lipid Res 2017; 59:69-78. [PMID: 29167409 DOI: 10.1194/jlr.m079731] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/11/2017] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification is the deposition of mineral in the artery wall by vascular smooth muscle cells (VSMCs) in response to pathological stimuli. The process is similar to bone formation and is an independent risk factor for cardiovascular disease. Given that ceramide and sphingosine 1-phosphate (S1P) are involved in cardiovascular pathophysiology and biomineralization, their role in VSMC matrix mineralization was investigated. During phosphate-induced VSMC mineralization, endogenous S1P levels increased accompanied by increased sphingosine kinase (SK) activity and increased mRNA expression of SK1 and SK2. Consistent with this, mineralization was increased by exogenous S1P, but decreased by C2-ceramide. Mechanistically, exogenous S1P stimulated ezrin-radixin-moesin (ERM) phosphorylation in VSMCs and ERM phosphorylation was increased concomitantly with endogenous S1P during mineralization. Moreover, inhibition of acid sphingomyelinase and ceramidase with desipramine prevented increased S1P levels, ERM activation, and mineralization. Finally, pharmacological inhibition of ERM phosphorylation with NSC663894 decreased mineralization induced by phosphate and exogenous S1P. Although further studies will be needed to verify these findings in vivo, this study defines a novel role for the SK-S1P-ERM pathways in phosphate-induced VSMC matrix mineralization and shows that blocking these pathways with pharmacological inhibitors reduces mineralization. These results may inform new therapeutic approaches to inhibit or delay vascular calcification.
Collapse
Affiliation(s)
- Thomas G Morris
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Samantha J Borland
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Christopher J Clarke
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY
| | - Claire Wilson
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Yusuf A Hannun
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY
| | - Vasken Ohanian
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Ann E Canfield
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Jacqueline Ohanian
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
Vehoff J, Haegele-Link S, Humm A, Kaegi G, Mueller SK, Sauter R, Tettenborn BE, Hundsberger T. Heart rate variability decreases after 3 months of sustained treatment with fingolimod. J Neurol 2017; 264:2313-2317. [PMID: 28993873 DOI: 10.1007/s00415-017-8636-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/19/2022]
Abstract
The objective is to prospectively investigate short- and mid-term changes of heart rate variability (HRV) in patients with relapsing-remitting multiple sclerosis (RRMS), being started on fingolimod. In this prospective clinical trial, patient (n = 33) with RRMS starting treatment with fingolimod underwent a time-domain-based analysis of HRV (breathing at rest, deep breath, and in response to the Valsalva maneuver) shortly before, 4.5 h and 3 months after first intake. Blood pressure changes after the Valsalva maneuver were used as a marker of the sympathetic noradrenergic system. We used a non-invasive continuous beat-to-beat heart rate and blood pressure monitoring. In addition, the Fatigue Severity Scale and the refined and abbreviated Composite Autonomic Symptom Score were applied. Significant changes in HRV in RRMS patients, following treatment with fingolimod, were detected. After an initial increase in HRV, measured 4.5 h after the first intake of fingolimod, a substantial decrease in HRV occurred within 3 months on continuous treatment. There is a growing body of evidence for short-term cardiovascular side effects in continuous treatment with fingolimod, driven by the ANS. The mechanisms and the clinical relevance of the observed changes in HRV need further evaluation, especially in longer and larger prospective studies.
Collapse
Affiliation(s)
- Jochen Vehoff
- Department of Neurology, Cantonal Hospital, St. Gallen, Switzerland.
| | | | - Andrea Humm
- Department of Neurology, Cantonal Hospital, Fribourg, Switzerland
| | - Georg Kaegi
- Department of Neurology, Cantonal Hospital, St. Gallen, Switzerland
| | | | - Rafael Sauter
- Clinical Trials Unit, Cantonal Hospital, St. Gallen, Switzerland
| | | | | |
Collapse
|
30
|
Belliston S, Sundararajan J, Hammond N, Newell K, Lynch S. Reversible cerebral vasoconstriction syndrome in association with fingolimod use. Int J Neurosci 2016; 127:831-834. [DOI: 10.1080/00207454.2016.1257991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
| | | | | | - Kathy Newell
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS, United States
| | | |
Collapse
|
31
|
Bolli MH, Lescop C, Birker M, de Kanter R, Hess P, Kohl C, Nayler O, Rey M, Sieber P, Velker J, Weller T, Steiner B. Novel S1P1 receptor agonists – Part 5: From amino-to alkoxy-pyridines. Eur J Med Chem 2016; 115:326-41. [DOI: 10.1016/j.ejmech.2016.03.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 03/07/2016] [Accepted: 03/09/2016] [Indexed: 12/15/2022]
|
32
|
Nakahashi A, C. Siddegowda AK, Hammam MAS, Gowda SGB, Murai Y, Monde K. Stereochemical Study of Sphingosine by Vibrational Circular Dichroism. Org Lett 2016; 18:2327-30. [DOI: 10.1021/acs.orglett.6b00477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Atsufumi Nakahashi
- Faculty
of Advanced Life
Science, Frontier Research Center for the Post-Genome Science and
Technology, Hokkaido University, Kita 21, Nishi 11, Sapporo 001-0021, Japan
| | - Ananda Kumar C. Siddegowda
- Faculty
of Advanced Life
Science, Frontier Research Center for the Post-Genome Science and
Technology, Hokkaido University, Kita 21, Nishi 11, Sapporo 001-0021, Japan
| | - Mostafa A. S. Hammam
- Faculty
of Advanced Life
Science, Frontier Research Center for the Post-Genome Science and
Technology, Hokkaido University, Kita 21, Nishi 11, Sapporo 001-0021, Japan
| | - Siddabasave Gowda B. Gowda
- Faculty
of Advanced Life
Science, Frontier Research Center for the Post-Genome Science and
Technology, Hokkaido University, Kita 21, Nishi 11, Sapporo 001-0021, Japan
| | - Yuta Murai
- Faculty
of Advanced Life
Science, Frontier Research Center for the Post-Genome Science and
Technology, Hokkaido University, Kita 21, Nishi 11, Sapporo 001-0021, Japan
| | - Kenji Monde
- Faculty
of Advanced Life
Science, Frontier Research Center for the Post-Genome Science and
Technology, Hokkaido University, Kita 21, Nishi 11, Sapporo 001-0021, Japan
| |
Collapse
|
33
|
Zhang Y, Huang Y, Cantalupo A, Azevedo PS, Siragusa M, Bielawski J, Giordano FJ, Di Lorenzo A. Endothelial Nogo-B regulates sphingolipid biosynthesis to promote pathological cardiac hypertrophy during chronic pressure overload. JCI Insight 2016; 1. [PMID: 27158676 DOI: 10.1172/jci.insight.85484] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We recently discovered that endothelial Nogo-B, a membrane protein of the ER, regulates vascular function by inhibiting the rate-limiting enzyme, serine palmitoyltransferase (SPT), in de novo sphingolipid biosynthesis. Here, we show that endothelium-derived sphingolipids, particularly sphingosine-1-phosphate (S1P), protect the heart from inflammation, fibrosis, and dysfunction following pressure overload and that Nogo-B regulates this paracrine process. SPT activity is upregulated in banded hearts in vivo as well as in TNF-α-activated endothelium in vitro, and loss of Nogo removes the brake on SPT, increasing local S1P production. Hence, mice lacking Nogo-B, systemically or specifically in the endothelium, are resistant to the onset of pathological cardiac hypertrophy. Furthermore, pharmacological inhibition of SPT with myriocin restores permeability, inflammation, and heart dysfunction in Nogo-A/B-deficient mice to WT levels, whereas SEW2871, an S1P1 receptor agonist, prevents myocardial permeability, inflammation, and dysfunction in WT banded mice. Our study identifies a critical role of endothelial sphingolipid biosynthesis and its regulation by Nogo-B in the development of pathological cardiac hypertrophy and proposes a potential therapeutic target for the attenuation or reversal of this clinical condition.
Collapse
Affiliation(s)
- Yi Zhang
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Yan Huang
- Section of Cardiovascular Medicine, Department of Internal Medicine, and Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anna Cantalupo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Paula S Azevedo
- Department of Internal Medicine, Botucatu Medical School, University of Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Mauro Siragusa
- Center for Molecular Medicine, Institute for Vascular Signalling, Goethe University Frankfurt, Frankfurt, Germany
| | - Jacek Bielawski
- Lipidomics Mass Spectrometry Facility, Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Frank J Giordano
- Section of Cardiovascular Medicine, Department of Internal Medicine, and Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Annarita Di Lorenzo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| |
Collapse
|
34
|
Tipping WJ, Lee M, Serrels A, Brunton VG, Hulme AN. Stimulated Raman scattering microscopy: an emerging tool for drug discovery. Chem Soc Rev 2016; 45:2075-89. [PMID: 26839248 PMCID: PMC4839273 DOI: 10.1039/c5cs00693g] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Indexed: 12/26/2022]
Abstract
Optical microscopy techniques have emerged as a cornerstone of biomedical research, capable of probing the cellular functions of a vast range of substrates, whilst being minimally invasive to the cells or tissues of interest. Incorporating biological imaging into the early stages of the drug discovery process can provide invaluable information about drug activity within complex disease models. Spontaneous Raman spectroscopy has been widely used as a platform for the study of cells and their components based on chemical composition; but slow acquisition rates, poor resolution and a lack of sensitivity have hampered further development. A new generation of stimulated Raman techniques is emerging which allows the imaging of cells, tissues and organisms at faster acquisition speeds, and with greater resolution and sensitivity than previously possible. This review focuses on the development of stimulated Raman scattering (SRS), and covers the use of bioorthogonal tags to enhance sample detection, and recent applications of both spontaneous Raman and SRS as novel imaging platforms to facilitate the drug discovery process.
Collapse
Affiliation(s)
- W. J. Tipping
- EaStCHEM School of Chemistry , The University of Edinburgh , Joseph Black Building , David Brewster Road , Edinburgh , EH9 3FJ , UK .
- Edinburgh Cancer Research Centre , Institute of Genetics and Molecular Medicine , The University of Edinburgh , Crewe Road South , Edinburgh , EH4 2XR , UK
| | - M. Lee
- Edinburgh Cancer Research Centre , Institute of Genetics and Molecular Medicine , The University of Edinburgh , Crewe Road South , Edinburgh , EH4 2XR , UK
| | - A. Serrels
- Edinburgh Cancer Research Centre , Institute of Genetics and Molecular Medicine , The University of Edinburgh , Crewe Road South , Edinburgh , EH4 2XR , UK
| | - V. G. Brunton
- Edinburgh Cancer Research Centre , Institute of Genetics and Molecular Medicine , The University of Edinburgh , Crewe Road South , Edinburgh , EH4 2XR , UK
| | - A. N. Hulme
- EaStCHEM School of Chemistry , The University of Edinburgh , Joseph Black Building , David Brewster Road , Edinburgh , EH9 3FJ , UK .
| |
Collapse
|
35
|
Aguiar C, Batista S, Pacheco R. Cardiovascular effects of fingolimod: Relevance, detection and approach. Rev Port Cardiol 2015; 34:279-85. [PMID: 25843307 DOI: 10.1016/j.repc.2014.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/15/2014] [Indexed: 01/10/2023] Open
Abstract
Fingolimod, a structural analogue of sphingosine, is the first oral treatment available for multiple sclerosis. The presence of sphingosine-1-phosphate receptors in the sinus and atrioventricular nodes, myocardial cells, endothelial cells and arterial smooth muscle cells is responsible for fingolimod's cardiovascular effects. We provide a comprehensive review of the mechanisms of these effects and characterize their clinical relevance.
Collapse
Affiliation(s)
- Carlos Aguiar
- Cardiology Department, Hospital Santa Cruz, CHLO, Carnaxide, Portugal.
| | - Sónia Batista
- Multiple Sclerosis Outpatient Clinic, Coimbra University, Coimbra, Portugal
| | - Ricardo Pacheco
- Medical Department, Novartis Farma S.A., Porto Salvo, Portugal
| |
Collapse
|
36
|
Aguiar C, Batista S, Pacheco R. Cardiovascular effects of fingolimod: Relevance, detection and approach. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2015. [DOI: 10.1016/j.repce.2014.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
37
|
Fenger M, Linneberg A, Jeppesen J. Network-based analysis of the sphingolipid metabolism in hypertension. Front Genet 2015; 6:84. [PMID: 25788903 PMCID: PMC4349157 DOI: 10.3389/fgene.2015.00084] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/17/2015] [Indexed: 01/11/2023] Open
Abstract
Common diseases like essential hypertension or diabetes mellitus are complex as they are polygenic in nature, such that each genetic variation only has a small influence on the disease. Genes operates in integrated networks providing the blue-print for all biological processes and conditional of the complex genotype determines the state and dynamics of any trait, which may be modified to various extent by non-genetic factors. Thus, diseases are heterogenous ensembles of conditions with a common endpoint. Numerous studies have been performed to define genes of importance for a trait or disease, but only a few genes with small effect have been identified. The major reasons for this modest progress is the unresolved heterogeneity of the regulation of blood pressure and the shortcomings of the prevailing monogenic approach to capture genetic effects in a polygenic condition. Here, a two-step procedure is presented in which physiological heterogeneity is disentangled and genetic effects are analyzed by variance decomposition of genetic interactions and by an information theoretical approach including 162 single nucleotide polymorphisms (SNP) in 84 genes in the sphingolipid metabolism and related networks in blood pressure regulation. As expected, almost no genetic main effects were detected. In contrast, two-gene interactions established the entire sphingolipid metabolic and related genetic network to be highly involved in the regulation of blood pressure. The pattern of interaction clearly revealed that epistasis does not necessarily reflects the topology of the metabolic pathways i.e., the flow of metabolites. Rather, the enzymes and proteins are integrated in complex cellular substructures where communication flows between the components of the networks, which may be composite in structure. The heritabilities for diastolic and systolic blood pressure were estimated to be 0.63 and 0.01, which may in fact be the maximum heritabilities of these traits. This procedure provide a platform for studying and capturing the genetic networks of any polygenic trait, condition, or disease.
Collapse
Affiliation(s)
- Mogens Fenger
- Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, Denmark
| | | | - Jørgen Jeppesen
- Department of Cardiology, Glostrup University Hospital Glostrup, Denmark
| |
Collapse
|
38
|
Ogle ME, Sefcik LS, Awojoodu AO, Chiappa NF, Lynch K, Peirce-Cottler S, Botchwey EA. Engineering in vivo gradients of sphingosine-1-phosphate receptor ligands for localized microvascular remodeling and inflammatory cell positioning. Acta Biomater 2014; 10:4704-4714. [PMID: 25128750 PMCID: PMC4529737 DOI: 10.1016/j.actbio.2014.08.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/30/2014] [Accepted: 08/06/2014] [Indexed: 12/29/2022]
Abstract
Biomaterial-mediated controlled release of soluble signaling molecules is a tissue engineering approach to spatially control processes of inflammation, microvascular remodeling and host cell recruitment, and to generate biochemical gradients in vivo. Lipid mediators, such as sphingosine 1-phosphate (S1P), are recognized for their essential roles in spatial guidance, signaling and highly regulated endogenous gradients. S1P and pharmacological analogs such as FTY720 are therapeutically attractive targets for their critical roles in the trafficking of cells between blood and tissue spaces, both physiologically and pathophysiologically. However, the interaction of locally delivered sphingolipids with the complex metabolic networks controlling the flux of lipid species in inflamed tissue has yet to be elucidated. In this study, complementary in vitro and in vivo approaches are investigated to identify relationships between polymer composition, drug release kinetics, S1P metabolic activity, signaling gradients and spatial positioning of circulating cells around poly(lactic-co-glycolic acid) biomaterials. Results demonstrate that biomaterial-based gradients of S1P are short-lived in the tissue due to degradation by S1P lyase, an enzyme that irreversibly degrades intracellular S1P. On the other hand, in vivo gradients of the more stable compound, FTY720, enhance microvascular remodeling by selectively recruiting an anti-inflammatory subset of monocytes (S1P3(high)) to the biomaterial. Results highlight the need to better understand the endogenous balance of lipid import/export machinery and lipid kinase/phosphatase activity in order to design biomaterial products that spatially control the innate immune environment to maximize regenerative potential.
Collapse
Affiliation(s)
- Molly E. Ogle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Lauren S. Sefcik
- Department of Chemical & Biomolecular Engineering, Lafayette College, 740 High Street, Easton, PA 18042
| | - Anthony O. Awojoodu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Nathan F. Chiappa
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Kevin Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903
| | - Shayn Peirce-Cottler
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903
| | - Edward A. Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
39
|
Ross JS, Russo SB, Chavis GC, Cowart LA. Sphingolipid regulators of cellular dysfunction in Type 2 diabetes mellitus: a systems overview. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/clp.14.37] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
40
|
Fernández-Pisonero I, López J, Onecha E, Dueñas AI, Maeso P, Crespo MS, Román JAS, García-Rodríguez C. Synergy between sphingosine 1-phosphate and lipopolysaccharide signaling promotes an inflammatory, angiogenic and osteogenic response in human aortic valve interstitial cells. PLoS One 2014; 9:e109081. [PMID: 25275309 PMCID: PMC4183546 DOI: 10.1371/journal.pone.0109081] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 09/08/2014] [Indexed: 12/20/2022] Open
Abstract
Given that the bioactive lipid sphingosine 1-phosphate is involved in cardiovascular pathophysiology, and since lipid accumulation and inflammation are hallmarks of calcific aortic stenosis, the role of sphingosine 1-phosphate on the pro-inflammatory/pro-osteogenic pathways in human interstitial cells from aortic and pulmonary valves was investigated. Real-time PCR showed sphingosine 1-phosphate receptor expression in aortic valve interstitial cells. Exposure of cells to sphingosine 1-phosphate induced pro-inflammatory responses characterized by interleukin-6, interleukin-8, and cyclooxygenase-2 up-regulations, as observed by ELISA and Western blot. Strikingly, cell treatment with sphingosine 1-phosphate plus lipopolysaccharide resulted in the synergistic induction of cyclooxygenase-2, and intercellular adhesion molecule 1, as well as the secretion of prostaglandin E2, the soluble form of the intercellular adhesion molecule 1, and the pro-angiogenic factor vascular endothelial growth factor-A. Remarkably, the synergistic effect was significantly higher in aortic valve interstitial cells from stenotic than control valves, and was drastically lower in cells from pulmonary valves, which rarely undergo stenosis. siRNA and pharmacological analysis revealed the involvement of sphingosine 1-phosphate receptors 1/3 and Toll-like receptor-4, and downstream signaling through p38/MAPK, protein kinase C, and NF-κB. As regards pro-osteogenic pathways, sphingosine 1-phosphate induced calcium deposition and the expression of the calcification markers bone morphogenetic protein-2 and alkaline phosphatase, and enhanced the effect of lipopolysaccharide, an effect that was partially blocked by inhibition of sphingosine 1-phosphate receptors 3/2 signaling. In conclusion, the interplay between sphingosine 1-phosphate receptors and Toll-like receptor 4 signaling leads to a cooperative up-regulation of inflammatory, angiogenic, and osteogenic pathways in aortic valve interstitial cells that seems relevant to the pathogenesis of aortic stenosis and may allow the inception of new therapeutic approaches.
Collapse
Affiliation(s)
| | - Javier López
- Cardiology Department, Instituto Ciencias del Corazón (ICICOR) Hospital Clínico Universitario, Valladolid, Spain
| | - Esther Onecha
- Instituto de Biología y Genética Molecular (CSIC-Universidad Valladolid), Valladolid, Spain
| | - Ana I. Dueñas
- Research Unit, Hospital Clínico Universitario, Valladolid, Spain
| | - Patricia Maeso
- Instituto de Biología y Genética Molecular (CSIC-Universidad Valladolid), Valladolid, Spain
| | - Mariano Sánchez Crespo
- Instituto de Biología y Genética Molecular (CSIC-Universidad Valladolid), Valladolid, Spain
| | - José Alberto San Román
- Cardiology Department, Instituto Ciencias del Corazón (ICICOR) Hospital Clínico Universitario, Valladolid, Spain
| | - Carmen García-Rodríguez
- Instituto de Biología y Genética Molecular (CSIC-Universidad Valladolid), Valladolid, Spain
- * E-mail:
| |
Collapse
|
41
|
Qi Y, Jiang C, Tanaka N, Krausz KW, Brocker CN, Fang ZZ, Bredell BX, Shah YM, Gonzalez FJ. PPARα-dependent exacerbation of experimental colitis by the hypolipidemic drug fenofibrate. Am J Physiol Gastrointest Liver Physiol 2014; 307:G564-G573. [PMID: 25035112 PMCID: PMC4154119 DOI: 10.1152/ajpgi.00153.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/14/2014] [Indexed: 02/07/2023]
Abstract
Fibrates, such as fenofibrate, are peroxisome proliferator-activated receptor-α (PPARα) agonists and have been used for several decades as hypolipidemic agents in the clinic. However, contradictory observations exist on the role of fibrates in host response to acute inflammation, with unclear mechanisms. The role of PPARα in colitis was assessed using fenofibrate and Ppara-null mice. Wild-type or Ppara-null mice were subjected to acute colitis under three distinct protocols, dextran sulfate sodium, trinitrobenzenesulfonic acid, and Salmonella Typhi. Serum and colon lipidomics were analyzed to characterize the metabolic profiles by ultra-performance liquid chromatography-coupled with electrospray ionization quadrupole time-of-flight mass spectrometry. Messenger RNAs of PPARα target genes and genes involved in inflammation were determined by qunatitative PCR analysis. Fenofibrate treatment exacerbated inflammation and tissue injury in acute colitis, and this was dependent on PPARα activation. Lipidomics analysis revealed that bioactive sphingolipids, including sphingomyelins (SM) and ceramides, were significantly increased in the colitis group compared with the control group; this was further potentiated following fenofibrate treatment. In the colon, fenofibrate did not reduce the markedly increased expression of mRNA encoding TNFα found in the acute colitis model, while it decreased hydrolysis and increased synthesis of SM, upregulated RIPK3-dependent necrosis, and elevated mitochondrial fatty acid β-oxidation, which were possibly related to the exacerbated colitis.
Collapse
Affiliation(s)
- Yunpeng Qi
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Changtao Jiang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Naoki Tanaka
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Chad N Brocker
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Zhong-Ze Fang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Bryce X Bredell
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
42
|
Thomas K, Schrötter H, Halank M, Ziemssen T. Fingolimod in a patient with heart failure on the background of pulmonary arterial hypertension and coronary artery disease. BMC Neurol 2014; 14:126. [PMID: 24906818 PMCID: PMC4055797 DOI: 10.1186/1471-2377-14-126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/03/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fingolimod is the first oral immunomodulatory therapy approved for highly active relapsing remitting multiple sclerosis. Based on the distribution pattern of fingolimod interacting sphingosine-1-phosphat receptors in organism including immune system and cardiovascular system clinical monitoring of patients and evaluation of adverse events are recommended. Despite extensive data on cardiovascular safety, experience with fingolimod in patients with concomitant cardiological disease, especially within the pulmonary circulation, is rare. CASE PRESENTATION We report the case of a 46-year-old woman presented with relapsing remitting multiple sclerosis and severe idiopathic pulmonary arterial hypertension. Fingolimod was initiated because of disease activity of multiple sclerosis with two relapses and gadolinium-enhancing lesions in MRI. The patient demonstrated stable disease course of idiopathic pulmonary arterial hypertension when fingolimod was started. Fingolimod therapy did not alter or even worsen the pulmonary or cardiovascular conditions during first dose application as well as follow up of nine months. CONCLUSION In this report, we present the first case of fingolimod treatment in a patient with highly active multiple sclerosis and severe idiopathic pulmonary arterial hypertension. We suggest an interdisciplinary approach with detailed cardiopulmonary monitoring for safety in such patients.
Collapse
Affiliation(s)
| | | | | | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, Neurological University Clinic Dresden, University Clinic Carl Gustav Carus Dresden, University of Technology Dresden, Fetscherstr, 74, 01307 Dresden, Germany.
| |
Collapse
|
43
|
Ohanian J, Liao A, Forman SP, Ohanian V. Age-related remodeling of small arteries is accompanied by increased sphingomyelinase activity and accumulation of long-chain ceramides. Physiol Rep 2014; 2:2/5/e12015. [PMID: 24872355 PMCID: PMC4098743 DOI: 10.14814/phy2.12015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The structure and function of large arteries alters with age leading to increased risk of cardiovascular disease. Age‐related large artery remodeling and arteriosclerosis is associated with increased collagen deposition, inflammation, and endothelial dysfunction. Bioactive sphingolipids are known to regulate these processes, and are also involved in aging and cellular senescence. However, less is known about age‐associated alterations in small artery morphology and function or whether changes in arterial sphingolipids occur in aging. We show that mesenteric small arteries from old sheep have increased lumen diameter and media thickness without a change in media to lumen ratio, indicative of outward hypertrophic remodeling. This remodeling occurred without overt changes in blood pressure or pulse pressure indicating it was a consequence of aging per se. There was no age‐associated change in mechanical properties of the arteries despite an increase in total collagen content and deposition of collagen in a thickened intima layer in arteries from old animals. Analysis of the sphingolipid profile showed an increase in long‐chain ceramide (C14–C20), but no change in the levels of sphingosine or sphingosine‐1‐phosphate in arteries from old compared to young animals. This was accompanied by a parallel increase in acid and neutral sphingomyelinase activity in old arteries compared to young. This study demonstrates remodeling of small arteries during aging that is accompanied by accumulation of long‐chain ceramides. This suggests that sphingolipids may be important mediators of vascular aging. In this study, we have investigated remodeling of small arteries in a large animal model of aging, the sheep. We show that there is age‐related formation of neointima and increased collagen deposition that is accompanied by changes in sphingolipid metabolism resulting in ceramide accumulation in the tissues. These are the first data implicating sphingolipids as important mediators of vascular aging in small arteries. Given that aging is a major risk factor for cardiovascular disease, our study opens a new area for further research into the mechanisms that underlie vascular remodeling in aging.
Collapse
Affiliation(s)
- Jacqueline Ohanian
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Aiyin Liao
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Simon P Forman
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Vasken Ohanian
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| |
Collapse
|
44
|
Qi Y, Jiang C, Cheng J, Krausz KW, Li T, Ferrell JM, Gonzalez FJ, Chiang JYL. Bile acid signaling in lipid metabolism: metabolomic and lipidomic analysis of lipid and bile acid markers linked to anti-obesity and anti-diabetes in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:19-29. [PMID: 24796972 DOI: 10.1016/j.bbalip.2014.04.008] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/17/2014] [Accepted: 04/28/2014] [Indexed: 12/11/2022]
Abstract
Bile acid synthesis is the major pathway for catabolism of cholesterol. Cholesterol 7α-hydroxylase (CYP7A1) is the rate-limiting enzyme in the bile acid biosynthetic pathway in the liver and plays an important role in regulating lipid, glucose and energy metabolism. Transgenic mice overexpressing CYP7A1 (CYP7A1-tg mice) were resistant to high-fat diet (HFD)-induced obesity, fatty liver, and diabetes. However the mechanism of resistance to HFD-induced obesity of CYP7A1-tg mice has not been determined. In this study, metabolomic and lipidomic profiles of CYP7A1-tg mice were analyzed to explore the metabolic alterations in CYP7A1-tg mice that govern the protection against obesity and insulin resistance by using ultra-performance liquid chromatography-coupled with electrospray ionization quadrupole time-of-flight mass spectrometry combined with multivariate analyses. Lipidomics analysis identified seven lipid markers including lysophosphatidylcholines, phosphatidylcholines, sphingomyelins and ceramides that were significantly decreased in serum of HFD-fed CYP7A1-tg mice. Metabolomics analysis identified 13 metabolites in bile acid synthesis including taurochenodeoxycholic acid, taurodeoxycholic acid, tauroursodeoxycholic acid, taurocholic acid, and tauro-β-muricholic acid (T-β-MCA) that differed between CYP7A1-tg and wild-type mice. Notably, T-β-MCA, an antagonist of the farnesoid X receptor (FXR) was significantly increased in intestine of CYP7A1-tg mice. This study suggests that reducing 12α-hydroxylated bile acids and increasing intestinal T-β-MCA may reduce high fat diet-induced increase of phospholipids, sphingomyelins and ceramides, and ameliorate diabetes and obesity. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.
Collapse
Affiliation(s)
- Yunpeng Qi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Changtao Jiang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jie Cheng
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tiangang Li
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Jessica M Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John Y L Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA.
| |
Collapse
|
45
|
Sphingosine-1-phosphate receptor 3 mediates sphingosine-1-phosphate induced release of weibel-palade bodies from endothelial cells. PLoS One 2014; 9:e91346. [PMID: 24632890 PMCID: PMC3954688 DOI: 10.1371/journal.pone.0091346] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/10/2014] [Indexed: 12/28/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is an agonist for five distinct G-protein coupled receptors, that is released by platelets, mast cells, erythrocytes and endothelial cells. S1P promotes endothelial cell barrier function and induces release of endothelial cell-specific storage-organelles designated Weibel-Palade bodies (WPBs). S1P-mediated enhancement of endothelial cell barrier function is dependent on S1P receptor 1 (S1PR1) mediated signaling events that result in the activation of the small GTPase Rac1. Recently, we have reported that Rac1 regulates epinephrine-induced WPB exocytosis following its activation by phosphatidylinositol-3,4,5-triphosphate-dependent Rac exchange factor 1 (PREX1). S1P has also been described to induce WPB exocytosis. Here, we confirm that S1P induces release of WPBs using von Willebrand factor (VWF) as a marker. Using siRNA mediated knockdown of gene expression we show that S1PR1 is not involved in S1P-mediated release of WPBs. In contrast depletion of the S1PR3 greatly reduced S1P-induced release of VWF. S1P-mediated enhancement of endothelial barrier function was not affected by S1PR3-depletion whereas it was greatly impaired in cells lacking S1PR1. The Rho kinase inhibitor Y27632 completely abrogated S1P-mediated release of VWF. Also, the calcium chelator BAPTA-AM significantly reduced S1P-induced release of VWF. Our findings indicate that S1P-induced release of haemostatic, inflammatory and angiogenic components stored within WPBs depends on the S1PR3.
Collapse
|
46
|
Vihervaara T, Suoniemi M, Laaksonen R. Lipidomics in drug discovery. Drug Discov Today 2014; 19:164-70. [DOI: 10.1016/j.drudis.2013.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 08/23/2013] [Accepted: 09/10/2013] [Indexed: 12/19/2022]
|
47
|
High density lipoproteins and ischemia reperfusion injury: the therapeutic potential of HDL to modulate cell survival pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 824:19-26. [PMID: 25038990 DOI: 10.1007/978-3-319-07320-0_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The clinical importance of high density lipoproteins has grown in recent years with demonstrations of their impact on diverse pathological mechanisms implicated not only in vascular disease, but also in other physiological systems. This is related to the multiple functions associated with high-density lipoproteins (HDL), notably their ability to limit oxidant and inflammatory processes, which are common to different disease states. A second feature of particular clinical relevance is the possibility of synthesising a simplified form of HDL that exhibits some of the functions of the mature lipoprotein. The therapeutic potential of synthetic HDL is already under clinical scrutiny. To illustrate these points, the present chapter will discuss the role of HDL in limiting damage to the heart consequent to myocardial ischemia. It will review molecular survival pathways stimulated by HDL to combat oxidative stress and the potential of synthetic HDL to activate such pathways.
Collapse
|
48
|
Bolli MH, Abele S, Birker M, Bravo R, Bur D, de Kanter R, Kohl C, Grimont J, Hess P, Lescop C, Mathys B, Müller C, Nayler O, Rey M, Scherz M, Schmidt G, Seifert J, Steiner B, Velker J, Weller T. Novel S1P(1) receptor agonists--part 3: from thiophenes to pyridines. J Med Chem 2013; 57:110-30. [PMID: 24367923 DOI: 10.1021/jm4014696] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In preceding communications we summarized our medicinal chemistry efforts leading to the identification of potent, selective, and orally active S1P1 agonists such as the thiophene derivative 1. As a continuation of these efforts, we replaced the thiophene in 1 by a 2-, 3-, or 4-pyridine and obtained less lipophilic, potent, and selective S1P1 agonists (e.g., 2) efficiently reducing blood lymphocyte count in the rat. Structural features influencing the compounds' receptor affinity profile and pharmacokinetics are discussed. In addition, the ability to penetrate brain tissue has been studied for several compounds. As a typical example for these pyridine based S1P1 agonists, compound 53 showed EC50 values of 0.6 and 352 nM for the S1P1 and S1P3 receptor, respectively, displayed favorable PK properties, and penetrated well into brain tissue. In the rat, compound 53 maximally reduced the blood lymphocyte count for at least 24 h after oral dosing of 3 mg/kg.
Collapse
Affiliation(s)
- Martin H Bolli
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd. , Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mikłosz A, Łukaszuk B, Baranowski M, Górski J, Chabowski A. Effects of inhibition of serine palmitoyltransferase (SPT) and sphingosine kinase 1 (SphK1) on palmitate induced insulin resistance in L6 myotubes. PLoS One 2013; 8:e85547. [PMID: 24376889 PMCID: PMC3871603 DOI: 10.1371/journal.pone.0085547] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 12/04/2013] [Indexed: 11/18/2022] Open
Abstract
Background The objective of this study was to examine the effects of short (2 h) and prolonged (18 h) inhibition of serine palmitoyltransferase (SPT) and sphingosine kinase 1 (SphK1) on palmitate (PA) induced insulin resistance in L6 myotubes. Methods L6 myotubes were treated simultaneously with either PA and myriocin (SPT inhibitor) or PA and Ski II (SphK1inhibitor) for different time periods (2 h and 18 h). Insulin stimulated glucose uptake was measured using radioactive isotope. Expression of insulin signaling proteins was determined using Western blot analyses. Intracellular sphingolipids content [sphinganine (SFA), ceramide (CER), sphingosine (SFO), sphingosine-1-phosphate (S1P)] were estimated by HPLC. Results Our results revealed that both short and prolonged time of inhibition of SPT by myriocin was sufficient to prevent ceramide accumulation and simultaneously reverse palmitate induced inhibition of insulin-stimulated glucose transport. In contrast, prolonged inhibition of SphK1 intensified the effect of PA on insulin-stimulated glucose uptake and attenuated further the activity of insulin signaling proteins (pGSK3β/GSK3β ratio) in L6 myotubes. These effects were related to the accumulation of sphingosine in palmitate treated myotubes. Conclusion Myriocin is more effective in restoration of palmitate induced insulin resistance in L6 myocytes, despite of the time of SPT inhibition, comparing to SKII (a specific SphK1 inhibitor). Observed changes in insulin signaling proteins were related to the content of specific sphingolipids, namely to the reduction of ceramide. Interestingly, inactivation of SphK1 augmented the effect of PA induced insulin resistance in L6 myotubes, which was associated with further inhibition of insulin stimulated PKB and GSK3β phosphorylation, glucose uptake and the accumulation of sphingosine.
Collapse
Affiliation(s)
- Agnieszka Mikłosz
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
- * E-mail:
| | - Bartłomiej Łukaszuk
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Marcin Baranowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Jan Górski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
50
|
Bolli MH, Velker J, Müller C, Mathys B, Birker M, Bravo R, Bur D, de Kanter R, Hess P, Kohl C, Lehmann D, Meyer S, Nayler O, Rey M, Scherz M, Steiner B. Novel S1P1 Receptor Agonists - Part 2: From Bicyclo[3.1.0]hexane-Fused Thiophenes to Isobutyl Substituted Thiophenes. J Med Chem 2013; 57:78-97. [DOI: 10.1021/jm401456d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Martin H. Bolli
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Jörg Velker
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Claus Müller
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Boris Mathys
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Magdalena Birker
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Roberto Bravo
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Daniel Bur
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Ruben de Kanter
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Patrick Hess
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Christopher Kohl
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - David Lehmann
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Solange Meyer
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Oliver Nayler
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Markus Rey
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Michael Scherz
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Beat Steiner
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| |
Collapse
|