1
|
Duan Y, Li Q, Wu J, Zhou C, Liu X, Yue J, Chen X, Liu J, Zhang Q, Zhang Y, Zhang L. A detrimental role of endothelial S1PR2 in cardiac ischemia-reperfusion injury via modulating mitochondrial dysfunction, NLRP3 inflammasome activation, and pyroptosis. Redox Biol 2024; 75:103244. [PMID: 38909407 PMCID: PMC11254837 DOI: 10.1016/j.redox.2024.103244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024] Open
Abstract
Sphingosine 1-phosphate (S1P), a bioactive lipid molecule, exerts multifaceted effects on cardiovascular functions via S1P receptors, but its effects on cardiac I/R injury are not fully understood. Plasma lipidomics analysis by mass spectrometry revealed that sphingosine lipids, including sphingosine 1-phosphate (S1P), were significantly down-regulated following cardiac I/R injury in mice. The reduced S1P levels were also observed in the plasma of coronary heart disease (CHD) patients after percutaneous coronary intervention (PCI) compared with those without PCI. We found that S1P exerted a cardioprotective effect via endothelial cell (EC)-S1PR1, whereas EC-S1PR2 displayed a detrimental effect on cardiac I/R. Our data showed that EC-specific S1pr2 loss-of-function significantly lessened inflammatory responses and diminished cardiac I/R injury, while EC-specific S1pr2 gain-of-function aggravated cardiac I/R injury. Mechanistically, EC-S1PR2 initiated excessive mitochondrial fission and elevated ROS production via RHO/ROCK1/DRP1 pathway, leading to NLRP3 inflammasome activation and subsequent cell pyroptosis, thereby exacerbating inflammation and I/R injuries. Furthermore, RGD-peptide magnetic nanoparticles packaging S1pr2-siRNA to specifically knockdown S1PR2 in endothelial cells significantly ameliorated cardiac I/R injury. Taken together, our investigations demonstrate that EC-S1PR2 induces excessive mitochondrial fission, which results in NLRP3 inflammasome activation and subsequently triggers cell pyroptosis, ultimately exacerbating inflammatory responses and aggravating heart injuries following I/R.
Collapse
Affiliation(s)
- Yunhao Duan
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Qinyu Li
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, 219 Miao Pu Road, Shanghai, 200135, China
| | - Jinjin Wu
- Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Caixia Zhou
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xiuxiang Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jinnan Yue
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xiaoli Chen
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jie Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Qi Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Yuzhen Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Lin Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China; Clinical Center for Heart Disease Research, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
2
|
Wu Y, Xing L, Lu L, Liu S, Zhao D, Lin L, Wang S, Li C, Pan Y. Alterations in the Salivary Microbiome and Metabolism in Patients With Carotid Atherosclerosis from Rural Northeast China. J Am Heart Assoc 2024; 13:e034014. [PMID: 39082416 DOI: 10.1161/jaha.123.034014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/25/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Periodontitis and atherosclerosis are both chronic inflammatory diseases with a high prevalence. Increasing evidence supports the independent association between severe periodontitis and atherosclerotic cardiovascular disease, in which oral microorganisms may play an important role. We aimed to evaluate the characteristic changes of salivary microbiome and metabolome in patients with carotid atherosclerosis (CAS) and periodontitis. METHODS AND RESULTS The subjects were obtained from a cross-sectional study that included 1933 participants aged 40 years or older from rural northeast China. The study enrolled 48 subjects with CAS and 48 controls without CAS matched by sex, age, body mass index, and prevalence of hypertension, diabetes, and dyslipidemia. We performed full-length 16S rDNA gene sequencing and untargeted metabolomics of saliva samples from 96 subjects. We found that CAS was closely associated with an increased abundance of Streptococcus, Lactobacillus, and Cutibacterium. Furthermore, patients with CAS had higher prevalence of severe periodontitis than the control group. Notably, periodontal pathogens such as Tannerella and Anaeroglobus were not only associated with periodontitis but also enriched in patients with CAS, whereas periodontal health-associated Neisseria was more abundant in those without CAS. We also identified 2 lipid metabolism pathways, including glycerophospholipid and sphingolipid metabolism, as associated with CAS. The levels of trimethylamine N-oxide and inflammatory mediator leukotriene D4 were significantly higher in patients with CAS, whereas the levels of carnosine were significantly lower, than those in controls. Additionally, serum levels of inflammatory marker high-sensitivity C-reactive protein were significantly increased in CAS and positively correlated with the abundance of Anaeroglobus and leukotriene D4 in saliva. CONCLUSIONS Our study suggests that characteristic changes in salivary microbiota and metabolites are closely related to CAS, and periodontitis and associated microorganisms may be involved in the initiation and progression of CAS.
Collapse
Affiliation(s)
- Yahong Wu
- Department of Periodontology, School and Hospital of Stomatology China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases Shenyang China
| | - Liying Xing
- Institute of Chronic Disease Liaoning Provincial Center for Disease Control and Prevention Shenyang China
| | - Lijie Lu
- Department of Periodontology, School and Hospital of Stomatology China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases Shenyang China
| | - Shuang Liu
- Department of Cardiovascular Ultrasound The Fourth Hospital of China Medical University Shenyang China
| | - Dan Zhao
- Department of Implant Dentistry, Beijing Stomatological Hospital Capital Medical University Beijing China
| | - Li Lin
- Department of Periodontology, School and Hospital of Stomatology China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases Shenyang China
| | - Songlin Wang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital Capital Medical University Beijing China
| | - Chen Li
- Department of Periodontology, School and Hospital of Stomatology China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases Shenyang China
| | - Yaping Pan
- Department of Periodontology, School and Hospital of Stomatology China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases Shenyang China
| |
Collapse
|
3
|
Bhat OM, Mir RA, Nehvi IB, Wani NA, Dar AH, Zargar MA. Emerging role of sphingolipids and extracellular vesicles in development and therapeutics of cardiovascular diseases. IJC HEART & VASCULATURE 2024; 53:101469. [PMID: 39139609 PMCID: PMC11320467 DOI: 10.1016/j.ijcha.2024.101469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024]
Abstract
Sphingolipids are eighteen carbon alcohol lipids synthesized from non-sphingolipid precursors in the endoplasmic reticulum (ER). The sphingolipids serve as precursors for a vast range of moieties found in our cells that play a critical role in various cellular processes, including cell division, senescence, migration, differentiation, apoptosis, pyroptosis, autophagy, nutrition intake, metabolism, and protein synthesis. In CVDs, different subclasses of sphingolipids and other derived molecules such as sphingomyelin (SM), ceramides (CERs), and sphingosine-1-phosphate (S1P) are directly related to diabetic cardiomyopathy, dilated cardiomyopathy, myocarditis, ischemic heart disease (IHD), hypertension, and atherogenesis. Several genome-wide association studies showed an association between genetic variations in sphingolipid pathway genes and the risk of CVDs. The sphingolipid pathway plays an important role in the biogenesis and secretion of exosomes. Small extracellular vesicles (sEVs)/ exosomes have recently been found as possible indicators for the onset of CVDs, linking various cellular signaling pathways that contribute to the disease progression. Important features of EVs like biocompatibility, and crossing of biological barriers can improve the pharmacokinetics of drugs and will be exploited to develop next-generation drug delivery systems. In this review, we have comprehensively discussed the role of sphingolipids, and sphingolipid metabolites in the development of CVDs. In addition, concise deliberations were laid to discuss the role of sEVs/exosomes in regulating the pathophysiological processes of CVDs and the exosomes as therapeutic targets.
Collapse
Affiliation(s)
- Owais Mohmad Bhat
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Rakeeb Ahmad Mir
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | | | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Abid Hamid Dar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - M Afzal Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| |
Collapse
|
4
|
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
|
5
|
Hammad SM, Lopes-Virella MF. Circulating Sphingolipids in Insulin Resistance, Diabetes and Associated Complications. Int J Mol Sci 2023; 24:14015. [PMID: 37762318 PMCID: PMC10531201 DOI: 10.3390/ijms241814015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Sphingolipids play an important role in the development of diabetes, both type 1 and type 2 diabetes, as well as in the development of both micro- and macro-vascular complications. Several reviews have been published concerning the role of sphingolipids in diabetes but most of the emphasis has been on the possible mechanisms by which sphingolipids, mainly ceramides, contribute to the development of diabetes. Research on circulating levels of the different classes of sphingolipids in serum and in lipoproteins and their importance as biomarkers to predict not only the development of diabetes but also of its complications has only recently emerged and it is still in its infancy. This review summarizes the previously published literature concerning sphingolipid-mediated mechanisms involved in the development of diabetes and its complications, focusing on how circulating plasma sphingolipid levels and the relative content carried by the different lipoproteins may impact their role as possible biomarkers both in the development of diabetes and mainly in the development of diabetic complications. Further studies in this field may open new therapeutic avenues to prevent or arrest/reduce both the development of diabetes and progression of its complications.
Collapse
Affiliation(s)
- Samar M. Hammad
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Maria F. Lopes-Virella
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC 29425, USA
| |
Collapse
|
6
|
Guo J, Feng J, Qu H, Xu H, Zhou H. Potential Drug Targets for Ceramide Metabolism in Cardiovascular Disease. J Cardiovasc Dev Dis 2022; 9:434. [PMID: 36547431 PMCID: PMC9782850 DOI: 10.3390/jcdd9120434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease poses a significant threat to the quality of human life. Metabolic abnormalities caused by excessive caloric intake have been shown to lead to the development of cardiovascular diseases. Ceramides are structural molecules found in biological membranes; they are crucial for cell survival and lipid metabolism, as they maintain barrier function and membrane fluidity. Increasing evidence has demonstrated that ceramide has a strong correlation with cardiovascular disease progression. Nevertheless, it remains a challenge to develop sphingolipids as therapeutic targets to improve the prognosis of cardiovascular diseases. In this review, we summarize the three synthesis pathways of ceramide and other intermediates that are important in ceramide metabolism. Furthermore, mechanistic studies and therapeutic strategies, including clinical drugs and bioactive molecules based on these intermediates, are discussed.
Collapse
Affiliation(s)
- Jiaying Guo
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
| | - Jiling Feng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, No. 1200, Cailun Road, Shanghai 201203, China
| | - Huiyan Qu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
| | - Hongxi Xu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, No. 1200, Cailun Road, Shanghai 201203, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
7
|
Borodzicz-Jażdżyk S, Jażdżyk P, Łysik W, Cudnoch-Jȩdrzejewska A, Czarzasta K. Sphingolipid metabolism and signaling in cardiovascular diseases. Front Cardiovasc Med 2022; 9:915961. [PMID: 36119733 PMCID: PMC9471951 DOI: 10.3389/fcvm.2022.915961] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/05/2022] [Indexed: 01/10/2023] Open
Abstract
Sphingolipids are a structural component of the cell membrane, derived from sphingosine, an amino alcohol. Its sphingoid base undergoes various types of enzymatic transformations that lead to the formation of biologically active compounds, which play a crucial role in the essential pathways of cellular signaling, proliferation, maturation, and death. The constantly growing number of experimental and clinical studies emphasizes the pivotal role of sphingolipids in the pathophysiology of cardiovascular diseases, including, in particular, ischemic heart disease, hypertension, heart failure, and stroke. It has also been proven that altering the sphingolipid metabolism has cardioprotective properties in cardiac pathologies, including myocardial infarction. Recent studies suggest that selected sphingolipids may serve as valuable biomarkers useful in the prognosis of cardiovascular disorders in clinical practice. This review aims to provide an overview of the current knowledge of sphingolipid metabolism and signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Sonia Borodzicz-Jażdżyk
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Jażdżyk
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, Warsaw, Poland
| | - Wojciech Łysik
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jȩdrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Czarzasta
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Spampinato SF, Sortino MA, Salomone S. Sphingosine-1-phosphate and Sphingosine-1-phosphate receptors in the cardiovascular system: pharmacology and clinical implications. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:95-139. [PMID: 35659378 DOI: 10.1016/bs.apha.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a lipid that binds and activates five distinct receptor subtypes, S1P1, S1P2, S1P3, S1P4, S1P5, widely expressed in different cells, tissues and organs. In the cardiovascular system these receptors have been extensively studied, but no drug acting on them has been approved so far for treating cardiovascular diseases. In contrast, a number of S1P receptor agonists are approved as immunomodulators, mainly for multiple sclerosis, because of their action on lymphocyte trafficking. This chapter summarizes the available information on S1P receptors in the cardiovascular system and discusses their potential for treating cardiovascular conditions and/or their role on the clinical pharmacology of drugs so far approved for non-cardiovascular conditions. Basic research has recently produced data useful to understand the molecular pharmacology of S1P and S1P receptors, regarding biased agonism, S1P storage, release and vehiculation and chaperoning by lipoproteins, paracrine actions, intracellular non-receptorial S1P actions. On the other hand, the approval of fingolimod and newer generation S1P receptor ligands as immunomodulators, provides information on a number of clinical observations on the impact of these drugs on cardiovascular system which need to be integrated with preclinical data. S1P receptors are potential targets for prevention and treatment of major cardiovascular conditions, including hypertension, myocardial infarction, heart failure and stroke.
Collapse
Affiliation(s)
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy.
| |
Collapse
|
9
|
Luo L, Xue Q, Qi Y, Zeng L, Liang S. Therapeutic effects of different polar fractions of hawthorn extract on blood stasis model rats revealed by liquid chromatography-mass spectrometry metabolomics. J Sep Sci 2021; 44:4005-4016. [PMID: 34490993 DOI: 10.1002/jssc.202100569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022]
Abstract
Hawthorn, a commonly used traditional Chinese medicine, has been suggested to have therapeutic effects on cardiovascular disease. However, effective fractions of hawthorn extract in the treatment of cardiovascular disease, together with possible therapeutic mechanisms, remain unclear. This study aimed to investigate the effects of four different polar fractions of hawthorn extract on blood stasis model rats, and explore the possible metabolic mechanisms by using a liquid chromatography-mass spectrometry metabolomics approach. Evaluation of hemorheology and fibrinogen showed that n-butanol and ethyl acetate fractions of hawthorn extract had significant therapeutic effects on blood stasis model rats. Furthermore, metabolomics analysis showed that n-butanol and ethyl acetate fractions of hawthorn extract could reverse imbalanced biomarkers in plasma and urine of blood stasis model rats. Additionally, metabolic pathway analysis revealed that plasma biomarkers were responsible for several important pathways, including d-glutamine and d-glutamate metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, alanine, aspartate, and glutamate metabolism, sphingolipid metabolism, and arginine biosynthesis. Meanwhile, urine biomarkers were responsible for some important pathways, including phenylalanine metabolism, tyrosine metabolism, and lysine degradation. This study demonstrated that n-butanol and ethyl acetate fractions of hawthorn extract had significant therapeutic effects on blood stasis model rats, and the underlying mechanisms involved multiple metabolic pathways.
Collapse
Affiliation(s)
- Lan Luo
- College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administrationof TCM, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Qi Xue
- College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administrationof TCM, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Yue Qi
- College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administrationof TCM, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Lu Zeng
- College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administrationof TCM, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Shengwang Liang
- College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administrationof TCM, Guangdong Pharmaceutical University, Guangzhou, P. R. China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| |
Collapse
|
10
|
B Gowda S, Gowda D, Kain V, Chiba H, Hui SP, Chalfant CE, Parcha V, Arora P, Halade GV. Sphingosine-1-phosphate interactions in the spleen and heart reflect extent of cardiac repair in mice and failing human hearts. Am J Physiol Heart Circ Physiol 2021; 321:H599-H611. [PMID: 34415189 DOI: 10.1152/ajpheart.00314.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive mediator in inflammation. Dysregulated S1P is demonstrated as a cause of heart failure (HF). However, the time-dependent and integrative role of S1P interaction with receptors in HF is unclear after myocardial infarction (MI). In this study, the sphingolipid mediators were quantified in ischemic human hearts. We also measured the time kinetics of these mediators post-MI in murine spleen and heart as an integrative approach to understand the interaction of S1P and respective S1P receptors in the transition of acute (AHF) to chronic HF (CHF). Risk-free 8-12 wk male C57BL/6 mice were subjected to MI surgery, and MI was confirmed by echocardiography and histology. Mass spectrometry was used to quantify sphingolipids in plasma, infarcted heart, spleen of mice, and ischemic and healthy human heart. The physiological cardiac repair was observed in mice with a notable increase of S1P quantity (pmol/g) in the heart and spleen significantly reduced in patients with ischemic HF. The circulating murine S1P levels were increased during AHF and CHF despite lowered substrate in CHF. The S1PR1 receptor expression was observed to coincide with the respective S1P quantity in mice and human hearts. Furthermore, selective S1P1 agonist limited inflammatory markers CCL2 and TNF-α and accelerated reparative markers ARG-1 and YM-1 in macrophages in the presence of Kdo2-Lipid A (KLA; potent inflammatory stimulant). This report demonstrated the importance of S1P/S1PR1 signaling in physiological inflammation during cardiac repair in mice. Alteration in these axes may serve as the signs of pathological remodeling in patients with ischemia.NEW & NOTEWORTHY Previous studies indicate that sphingosine-1-phosphate (S1P) has some role in cardiovascular disease. This study adds quantitative and integrative systems-based approaches that are necessary for discovery and bedside translation. Here, we quantitated sphinganine, sphingosine, sphingosine-1-phosphate (S1P) in mice and human cardiac pathobiology. Interorgan S1P quantity and respective systems-based receptor activation suggest cardiac repair after myocardial infarction. Thus, S1P serves as a therapeutic target for cardiac protection in clinical translation.
Collapse
Affiliation(s)
| | - Divyavani Gowda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Hitoshi Chiba
- Department of Nutrition, Sapporo University of Health Sciences, Sapporo, Japan
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, Florida.,Research Service, James A. Haley Veterans' Hospital, Tampa, Florida
| | - Vibhu Parcha
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Pankaj Arora
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
11
|
Montero-Bullon JF, Aveiro SS, Melo T, Martins-Marques T, Lopes D, Neves B, Girão H, Rosário M Domingues M, Domingues P. Cardiac phospholipidome is altered during ischemia and reperfusion in an ex vivo rat model. Biochem Biophys Rep 2021; 27:101037. [PMID: 34169155 PMCID: PMC8207217 DOI: 10.1016/j.bbrep.2021.101037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/17/2022] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of death, morbidity, and health costs worldwide. In AMI, a sudden blockage of blood flow causes myocardial ischemia and cell death. Reperfusion after ischemia has paradoxical effects and may exacerbate the myocardial injury, a process known as ischemic reperfusion injury. In this work we evaluated the lipidome of isolated rat hearts, maintained in controlled perfusion (CT), undergoing global ischemia (ISC) or ischemia followed by reperfusion (IR). 153 polar lipid levels were significantly different between conditions. 48 features had q < 0.001 and included 8 phosphatidylcholines and 4 lysophospholipids, which were lower in ISC compared to CT, and even lower in the IR group, suggesting that IR induces more profound changes than ISC. We observed that the levels of 16 alkyl acyl phospholipids were significantly altered during ISC and IR. Overall, these data indicate that myocardial lipid remodelling and possibly damage occurs to a greater extent during reperfusion. The adaptation of cardiac lipidome during ISC and IR described is consistent with the presence of oxidative damage and may reflect the impact of AMI on the lipidome at the cellular level and provide new insights into the role of lipids in the pathophysiology of acute myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Javier-Fernando Montero-Bullon
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
| | - Susana S. Aveiro
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
| | - Tânia Melo
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
| | - Tânia Martins-Marques
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Diana Lopes
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
| | - Bruna Neves
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
| | - Henrique Girão
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - M. Rosário M Domingues
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193, Aveiro, Portugal
| |
Collapse
|
12
|
Müller J, Bertsch T, Volke J, Schmid A, Klingbeil R, Metodiev Y, Karaca B, Kim SH, Lindner S, Schupp T, Kittel M, Poschet G, Akin I, Behnes M. Narrative review of metabolomics in cardiovascular disease. J Thorac Dis 2021; 13:2532-2550. [PMID: 34012599 PMCID: PMC8107570 DOI: 10.21037/jtd-21-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are accompanied by disorders in the cardiac metabolism. Furthermore, comorbidities often associated with cardiovascular disease can alter systemic and myocardial metabolism contributing to worsening of cardiac performance and health status. Biomarkers such as natriuretic peptides or troponins already support diagnosis, prognosis and treatment of patients with cardiovascular diseases and are represented in international guidelines. However, as cardiovascular diseases affect various pathophysiological pathways, a single biomarker approach cannot be regarded as ideal to reveal optimal clinical application. Emerging metabolomics technology allows the measurement of hundreds of metabolites in biological fluids or biopsies and thus to characterize each patient by its own metabolic fingerprint, improving our understanding of complex diseases, significantly altering the management of cardiovascular diseases and possibly personalizing medicine. This review outlines current knowledge, perspectives as well as limitations of metabolomics for diagnosis, prognosis and treatment of cardiovascular diseases such as heart failure, atherosclerosis, ischemic and non-ischemic cardiomyopathy. Furthermore, an ongoing research project tackling current inconsistencies as well as clinical applications of metabolomics will be discussed. Taken together, the application of metabolomics will enable us to gain more insights into pathophysiological interactions of metabolites and disease states as well as improving therapies of patients with cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Julian Müller
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Bertsch
- Institute of Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremburg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Justus Volke
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alexander Schmid
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rebecca Klingbeil
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Yulian Metodiev
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bican Karaca
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Seung-Hyun Kim
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Simon Lindner
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Tobias Schupp
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Maximilian Kittel
- Institute for Clinical Chemistry, Faculty of Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Behnes
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
13
|
Magaye RR, Savira F, Hua Y, Xiong X, Huang L, Reid C, Flynn BL, Kaye D, Liew D, Wang BH. Attenuating PI3K/Akt- mTOR pathway reduces dihydrosphingosine 1 phosphate mediated collagen synthesis and hypertrophy in primary cardiac cells. Int J Biochem Cell Biol 2021; 134:105952. [PMID: 33609744 DOI: 10.1016/j.biocel.2021.105952] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/15/2022]
Abstract
Cardiac fibrosis and myocyte hypertrophy play contributory roles in the progression of diseases such as heart Failure (HF) through what is collectively termed cardiac remodelling. The phosphoinositide 3- kinase (PI3K), protein kinase B (Akt) and mammalian target for rapamycin (mTOR) signalling pathway (PI3K/Akt- mTOR) is an important pathway in protein synthesis, cell growth, cell proliferation, and lipid metabolism. The sphingolipid, dihydrosphingosine 1 phosphate (dhS1P) has been shown to bind to high density lipids in plasma. Unlike its analog, spingosine 1 phosphate (S1P), the role of dhS1P in cardiac fibrosis is still being deciphered. This study was conducted to investigate the effect of dhS1P on PI3K/Akt signalling in primary cardiac fibroblasts and myocytes. Our findings demonstrate that inhibiting PI3K reduced collagen synthesis in neonatal cardiac fibroblasts (NCFs), and hypertrophy in neonatal cardiac myocytes (NCMs) induced by dhS1P, in vitro. Reduced activation of the PI3K/Akt- mTOR signalling pathway led to impaired translation of fibrotic proteins such as collagen 1 (Coll1) and transforming growth factor β (TGFβ) and inhibited the transcription and translation of tissue inhibitor of matrix metalloproteinase 1 (TIMP1). PI3K inhibition also affected the gene expression of S1P receptors and enzymes such as the dihydroceramide delta 4 desaturase (DEGS1) and sphingosine kinase 1 (SK1) in the de novo sphingolipid pathway. While in myocytes, PI3K inhibition reduced myocyte hypertrophy induced by dhS1P by reducing skeletal muscle α- actin (αSKA) mRNA expression, and protein translation due to increased glycogen synthase kinase 3β (GSK3β) mRNA expression. Our findings show a relationship between the PI3K/Akt- mTOR signalling cascade and exogenous dhS1P induced collagen synthesis and myocyte hypertrophy in primary neonatal cardiac cells.
Collapse
Affiliation(s)
- Ruth R Magaye
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Feby Savira
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Xiong
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; Shanghai Institute of Heart Failure, Research Centre for Translational Medicine, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai 200120, China
| | - Li Huang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Christopher Reid
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; School of Public Health School, Curtin University, Perth, Australia
| | - Bernard L Flynn
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - David Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Bing H Wang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia.
| |
Collapse
|
14
|
Nitzsche A, Poittevin M, Benarab A, Bonnin P, Faraco G, Uchida H, Favre J, Garcia-Bonilla L, Garcia MCL, Léger PL, Thérond P, Mathivet T, Autret G, Baudrie V, Couty L, Kono M, Chevallier A, Niazi H, Tharaux PL, Chun J, Schwab SR, Eichmann A, Tavitian B, Proia RL, Charriaut-Marlangue C, Sanchez T, Kubis N, Henrion D, Iadecola C, Hla T, Camerer E. Endothelial S1P 1 Signaling Counteracts Infarct Expansion in Ischemic Stroke. Circ Res 2021; 128:363-382. [PMID: 33301355 PMCID: PMC7874503 DOI: 10.1161/circresaha.120.316711] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Cerebrovascular function is critical for brain health, and endogenous vascular protective pathways may provide therapeutic targets for neurological disorders. S1P (Sphingosine 1-phosphate) signaling coordinates vascular functions in other organs, and S1P1 (S1P receptor-1) modulators including fingolimod show promise for the treatment of ischemic and hemorrhagic stroke. However, S1P1 also coordinates lymphocyte trafficking, and lymphocytes are currently viewed as the principal therapeutic target for S1P1 modulation in stroke. OBJECTIVE To address roles and mechanisms of engagement of endothelial cell S1P1 in the naive and ischemic brain and its potential as a target for cerebrovascular therapy. METHODS AND RESULTS Using spatial modulation of S1P provision and signaling, we demonstrate a critical vascular protective role for endothelial S1P1 in the mouse brain. With an S1P1 signaling reporter, we reveal that abluminal polarization shields S1P1 from circulating endogenous and synthetic ligands after maturation of the blood-neural barrier, restricting homeostatic signaling to a subset of arteriolar endothelial cells. S1P1 signaling sustains hallmark endothelial functions in the naive brain and expands during ischemia by engagement of cell-autonomous S1P provision. Disrupting this pathway by endothelial cell-selective deficiency in S1P production, export, or the S1P1 receptor substantially exacerbates brain injury in permanent and transient models of ischemic stroke. By contrast, profound lymphopenia induced by loss of lymphocyte S1P1 provides modest protection only in the context of reperfusion. In the ischemic brain, endothelial cell S1P1 supports blood-brain barrier function, microvascular patency, and the rerouting of blood to hypoperfused brain tissue through collateral anastomoses. Boosting these functions by supplemental pharmacological engagement of the endothelial receptor pool with a blood-brain barrier penetrating S1P1-selective agonist can further reduce cortical infarct expansion in a therapeutically relevant time frame and independent of reperfusion. CONCLUSIONS This study provides genetic evidence to support a pivotal role for the endothelium in maintaining perfusion and microvascular patency in the ischemic penumbra that is coordinated by S1P signaling and can be harnessed for neuroprotection with blood-brain barrier-penetrating S1P1 agonists.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/pathology
- Blood-Brain Barrier/physiopathology
- Cerebral Arteries/drug effects
- Cerebral Arteries/metabolism
- Cerebral Arteries/pathology
- Cerebral Arteries/physiopathology
- Cerebrovascular Circulation
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Female
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Infarction, Middle Cerebral Artery/prevention & control
- Ischemic Attack, Transient/metabolism
- Ischemic Attack, Transient/pathology
- Ischemic Attack, Transient/physiopathology
- Ischemic Attack, Transient/prevention & control
- Ischemic Stroke/metabolism
- Ischemic Stroke/pathology
- Ischemic Stroke/physiopathology
- Ischemic Stroke/prevention & control
- Lysophospholipids/metabolism
- Male
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Microcirculation
- Neuroprotective Agents/pharmacology
- Signal Transduction
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Sphingosine-1-Phosphate Receptors/agonists
- Sphingosine-1-Phosphate Receptors/genetics
- Sphingosine-1-Phosphate Receptors/metabolism
- Vascular Patency
- Mice
Collapse
Affiliation(s)
- Anja Nitzsche
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| | - Marine Poittevin
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
- Institut des Vaisseaux et du Sang, Hôpital Lariboisière
| | - Ammar Benarab
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| | - Philippe Bonnin
- Université de Paris, INSERM U965 and Physiologie Clinique - Explorations-Fonctionnelles, AP-HP, Hôpital Lariboisière
| | - Giuseppe Faraco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York
| | - Hiroki Uchida
- Center for Vascular Biology, Weill Cornell Medical College, Cornell University, New York
| | - Julie Favre
- MITOVASC Institute, CARFI Facility, CNRS UMR 6015, INSERM U1083, Angers University
| | - Lidia Garcia-Bonilla
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York
| | - Manuela C. L. Garcia
- MITOVASC Institute, CARFI Facility, CNRS UMR 6015, INSERM U1083, Angers University
| | - Pierre-Louis Léger
- Institut des Vaisseaux et du Sang, Hôpital Lariboisière
- INSERM U1141, Hôpital Robert Debré
| | - Patrice Thérond
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Biochimie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France; Université Paris-Sud
- UFR de Pharmacie, EA 4529, Châtenay-Malabry, France
| | - Thomas Mathivet
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| | - Gwennhael Autret
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| | | | - Ludovic Couty
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| | - Mari Kono
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Institutes of Health, Bethesda, MD, USA
| | - Aline Chevallier
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| | - Hira Niazi
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| | | | - Jerold Chun
- Neuroscience Drug Discovery, Sanford Burnham Prebys Medical Discovery Institute, La Jolla
| | - Susan R. Schwab
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York
| | - Anne Eichmann
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| | | | - Richard L. Proia
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Institutes of Health, Bethesda, MD, USA
| | | | - Teresa Sanchez
- Center for Vascular Biology, Weill Cornell Medical College, Cornell University, New York
| | - Nathalie Kubis
- Université de Paris, INSERM U965 and Physiologie Clinique - Explorations-Fonctionnelles, AP-HP, Hôpital Lariboisière
- Université de Paris, INSERM U1148, Hôpital Bichat, Paris, France
| | - Daniel Henrion
- MITOVASC Institute, CARFI Facility, CNRS UMR 6015, INSERM U1083, Angers University
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital
| | - Eric Camerer
- Université de Paris, Paris Cardiovascular Research Centre, INSERM
| |
Collapse
|
15
|
Li L, Dai W, Li W, Zhang Y, Wu Y, Guan C, Zhang A, Huang H, Li Y. Integrated Network Pharmacology and Metabonomics to Reveal the Myocardial Protection Effect of Huang-Lian-Jie-Du-Tang on Myocardial Ischemia. Front Pharmacol 2021; 11:589175. [PMID: 33613277 PMCID: PMC7890363 DOI: 10.3389/fphar.2020.589175] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Myocardial ischemia (MI) is one of the most common cardiovascular diseases with high incidence and mortality. Huang-Lian-Jie-Du-Tang (HLJDT) is a classic traditional Chinese prescription to clear “heat” and “poison”. In this study, we used a deliberate strategy integrating the methods of network pharmacology, pharmacodynamics, and metabonomics to investigate the molecular mechanism and potential targets of HLJDT in the treatment of MI. Firstly, by a network pharmacology approach, a global view of the potential compound-target-pathway network based on network pharmacology was constructed to provide a preliminary understanding of bioactive compounds and related targets of HLJDT for elucidating its molecular mechanisms in MI. Subsequently, in vivo efficacy of HLJDT was validated in a rat model. Meanwhile, the corresponding metabonomic profiles were used to explore differentially induced metabolic markers thus providing the metabolic mechanism of HLJDT in treating MI. The results demonstrated the myocardial protection effect of HLJDT on ischemia by a multicomponent-multitarget mode. This study highlights the reliability and effectiveness of a network pharmacology-based approach that identifies and validates the complex of natural compounds in HLJDT for illustrating the mechanism for the treatment of MI.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weixing Dai
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Kowloon, China
| | - Wenting Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yumao Zhang
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yanqin Wu
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chenfeng Guan
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Anye Zhang
- Department of Gastroenterology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hui Huang
- Department of Cardiovascular, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuzhen Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
16
|
Knapp P, Bodnar L, Błachnio-Zabielska A, Reszeć J, Świderska M, Chabowski A. Blood bioactive sphingolipids in patients with advanced serous epithelial ovarian cancer - mass spectrometry analysis. Arch Med Sci 2021; 17:53-61. [PMID: 33488856 PMCID: PMC7811313 DOI: 10.5114/aoms.2018.76996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/12/2017] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Due to the lack of highly specific and sensitive methods for diagnosing ovarian cancer at advanced stages (according to the International Federation of Gynecology and Obstetrics (FIGO) classification stage III-IV), new noninvasive biomarkers are urgently needed. This study aims to investigate how the levels of plasma bioactive sphingolipids (ceramides, sphingosine-1-phosphate, sphingosine and sphinganine) are altered in serum, erythrocytes and platelets of patients with advanced serous ovarian cancer. MATERIAL AND METHODS A total of 135 patients with advanced serous ovarian cancer and 159 women with normal ovarian morphology were enrolled. Plasma levels of sphingosine, sphingosine-1-phosphate, sphinganine, ceramide C14:0-Cer, C16:0-Cer, C18:1-Cer, C18:0-Cer, C20:0-Cer, C22:0-Cer, C24:1-Cer and C24:0-Cer were assessed by LC/MS/MS. RESULTS Plasma concentrations of C16-Cer, C18:1-Cer and C18-Cer were significantly higher in the advanced ovarian cancer group than in the control group (1.5-fold, p = 0.021; 1.8-fold, p = 0.036 and 1.5-fold, p = 0.031, respectively). Plasma concentration of C18:1-Cer was significantly higher in erythrocytes of women with advanced serous cancer compared to the control group (p = 0.027). Plasma C16-Cer and C18:1-Cer levels and erythrocyte C18:1-Cer levels were able to distinguish patients with moderate/severe serous ovarian cancer from patients with mild ovarian cancer (AUC: 0.86, 0.898, 0.795, respectively). Plasma concentrations of C16, C18.1 and C18 significantly correlated with FIGO staging (p = 0.001, p = 0.024 and p = 0.005), and grading (p = 0.021, p = 0.021 and p = 0.033). CONCLUSIONS Plasma concentrations of C16, C18.1 and C18 correlated with the progression of ovarian cancer (FIGO staging and grading). Plasma levels of C16-Cer and C18:1-Cer and erythrocyte C18:1-Cer levels could be used to distinguish patients with advanced serous ovarian cancer.
Collapse
Affiliation(s)
- Paweł Knapp
- Department of Gynecology and Gynecologic Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Lubomir Bodnar
- Department of Clinical Oncology, Military Institute of Medicine in Warsaw, Warsaw, Poland
| | | | - Joanna Reszeć
- Department of Clinical Pathomorphology, Medical University of Bialystok, Białystok, Poland
| | - Magdalena Świderska
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
17
|
Lock MC, Tellam RL, Darby JRT, Soo JY, Brooks DA, Seed M, Selvanayagam JB, Morrison JL. Identification of Novel miRNAs Involved in Cardiac Repair Following Infarction in Fetal and Adolescent Sheep Hearts. Front Physiol 2020; 11:614. [PMID: 32587529 PMCID: PMC7298149 DOI: 10.3389/fphys.2020.00614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/15/2020] [Indexed: 01/14/2023] Open
Abstract
Aims Animal models have been used to show that there are critical molecular mechanisms that can be activated to induce myocardial repair at specific times in development. For example, specific miRNAs are critical for regulating the response to myocardial infarction (MI) and improving the response to injury. Manipulating these miRNAs in small animal models provides beneficial effects post-MI; however it is not known if these miRNAs are regulated similarly in large mammals. Studying a large animal where the timing of heart development in relation to birth is similar to humans may provide insights to better understand the capacity to repair a developing mammalian heart and its application to the adult heart. Methods We used a sheep model of MI that included permanent ligation of the left anterior descending (LAD) coronary artery. Surgery was performed on fetuses (at 105 days gestation when all cardiomyocytes are mononucleated and proliferative) and adolescent sheep (at 6 months of age when all cardiomyocytes contribute to heart growth by hypertrophy). A microarray was utilized to determine the expression of known miRNAs within the damaged and undamaged tissue regions in fetal and adolescent hearts after MI. Results 73 miRNAs were up-regulated and 58 miRNAs were down-regulated significantly within the fetal infarct compared to remote cardiac samples. From adolescent hearts 69 non-redundant miRNAs were up-regulated and 63 miRNAs were down-regulated significantly in the infarct area compared to remote samples. Opposite differential expression profiles of 10 miRNAs within tissue regions (Infarct area, Border zone and Remote area of the left ventricle) occurred between the fetuses and adolescent sheep. These included miR-558 and miR-1538, which when suppressed using LNA anti-miRNAs in cell culture, increased cardiomyoblast proliferation. Conclusion There were significant differences in miRNA responses in fetal and adolescent sheep hearts following a MI, suggesting that the modulation of novel miRNA expression may have therapeutic potential, by promoting proliferation or repair in a damaged heart.
Collapse
Affiliation(s)
- Mitchell C Lock
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| | - Doug A Brooks
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia.,Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Mike Seed
- Division of Cardiology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Joseph B Selvanayagam
- Cardiac Imaging Research, Department of Heart Health, South Australian Health & Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
18
|
Iessi E, Marconi M, Manganelli V, Sorice M, Malorni W, Garofalo T, Matarrese P. On the role of sphingolipids in cell survival and death. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:149-195. [PMID: 32247579 DOI: 10.1016/bs.ircmb.2020.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sphingolipids, universal components of biological membranes of all eukaryotic organisms, from yeasts to mammals, in addition of playing a structural role, also play an important part of signal transduction pathways. They participate or, also, ignite several fundamental subcellular signaling processes but, more in general, they directly contribute to key biological activities such as cell motility, growth, senescence, differentiation as well as cell fate, i.e., survival or death. The sphingolipid metabolic pathway displays an intricate network of reactions that result in the formation of multiple sphingolipids, including ceramide, and sphingosine-1-phosphate. Different sphingolipids, that have key roles in determining cell fate, can induce opposite effects: as a general rule, sphingosine-1-phosphate promotes cell survival and differentiation, whereas ceramide is known to induce apoptosis. Furthermore, together with cholesterol, sphingolipids also represent the basic lipid component of lipid rafts, cholesterol- and sphingolipid-enriched membrane microdomains directly involved in cell death and survival processes. In this review, we briefly describe the characteristics of sphingolipids and lipid membrane microdomains. In particular, we will consider the involvement of various sphingolipids per se and of lipid rafts in apoptotic pathway, both intrinsic and extrinsic, in nonapoptotic cell death, in autophagy, and in cell differentiation. In addition, their roles in the most common physiological and pathological contexts either as pathogenetic elements or as biomarkers of diseases will be considered. We would also hint how the manipulation of sphingolipid metabolism could represent a potential therapeutic target to be investigated and functionally validated especially for those diseases for which therapeutic options are limited or ineffective.
Collapse
Affiliation(s)
- Elisabetta Iessi
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Matteo Marconi
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | | | - Maurizio Sorice
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Walter Malorni
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy; Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Tina Garofalo
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
19
|
Gaggini M, Sabatino L, Vassalle C. Conventional and innovative methods to assess oxidative stress biomarkers in the clinical cardiovascular setting. Biotechniques 2020; 68:223-231. [PMID: 32066251 DOI: 10.2144/btn-2019-0138] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Oxidative stress has a pivotal and widely described role in the onset and progression of atherosclerotic plaque and cardiovascular disease. Many oxidative stress-related biomarkers can be measured in biological samples; however, there are still many aspects that limit the adoption of oxidative stress assessment in clinical laboratory practice. Here, we report an overview of the different sources and main common oxidative stress biomarkers relevant for cardiovascular physiopathology, describing in detail a recently proposed lipidomic assay for ceramide assessment, as a promising future development in oxidative stress evaluation among the many available redox-related tests.
Collapse
|
20
|
Kovilakath A, Cowart LA. Sphingolipid Mediators of Myocardial Pathology. J Lipid Atheroscler 2020; 9:23-49. [PMID: 32821720 PMCID: PMC7379069 DOI: 10.12997/jla.2020.9.1.23] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/25/2019] [Accepted: 10/09/2019] [Indexed: 12/15/2022] Open
Abstract
Cardiomyopathy is the leading cause of mortality worldwide. While the causes of cardiomyopathy continue to be elucidated, current evidence suggests that aberrant bioactive lipid signaling plays a crucial role as a component of cardiac pathophysiology. Sphingolipids have been implicated in the pathophysiology of cardiovascular disease, as they regulate numerous cellular processes that occur in primary and secondary cardiomyopathies. Experimental evidence gathered over the last few decades from both in vitro and in vivo model systems indicates that inhibitors of sphingolipid synthesis attenuate a variety of cardiomyopathic symptoms. In this review, we focus on various cardiomyopathies in which sphingolipids have been implicated and the potential therapeutic benefits that could be gained by targeting sphingolipid metabolism.
Collapse
Affiliation(s)
- Anna Kovilakath
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - L. Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
- Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, VA, USA
| |
Collapse
|
21
|
Kovilakath A, Jamil M, Cowart LA. Sphingolipids in the Heart: From Cradle to Grave. Front Endocrinol (Lausanne) 2020; 11:652. [PMID: 33042014 PMCID: PMC7522163 DOI: 10.3389/fendo.2020.00652] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/11/2020] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular diseases are the leading cause of mortality worldwide and this has largely been driven by the increase in metabolic disease in recent decades. Metabolic disease alters metabolism, distribution, and profiles of sphingolipids in multiple organs and tissues; as such, sphingolipid metabolism and signaling have been vigorously studied as contributors to metabolic pathophysiology in various pathological outcomes of obesity, including cardiovascular disease. Much experimental evidence suggests that targeting sphingolipid metabolism may be advantageous in the context of cardiometabolic disease. The heart, however, is a structurally and functionally complex organ where bioactive sphingolipids have been shown not only to mediate pathological processes, but also to contribute to essential functions in cardiogenesis and cardiac function. Additionally, some sphingolipids are protective in the context of ischemia/reperfusion injury. In addition to mechanistic contributions, untargeted lipidomics approaches used in recent years have identified some specific circulating sphingolipids as novel biomarkers in the context of cardiovascular disease. In this review, we summarize recent literature on both deleterious and beneficial contributions of sphingolipids to cardiogenesis and myocardial function as well as recent identification of novel sphingolipid biomarkers for cardiovascular disease risk prediction and diagnosis.
Collapse
Affiliation(s)
- Anna Kovilakath
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Maryam Jamil
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Lauren Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, VA, United States
- *Correspondence: Lauren Ashley Cowart
| |
Collapse
|
22
|
Alessenko AV, Zateyshchikov DA, Lebedev AТ, Kurochkin IN. Participation of Sphingolipids in the Pathogenesis of Atherosclerosis. ACTA ACUST UNITED AC 2019; 59:77-87. [DOI: 10.18087/cardio.2019.8.10270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/08/2019] [Indexed: 11/18/2022]
Affiliation(s)
| | - D. A. Zateyshchikov
- City Clinical Hospital № 51; Central State Medical Academy of Department of Presidential Affairs
| | | | | |
Collapse
|
23
|
Fuentes E, Moore-Carrasco R, de Andrade Paes AM, Trostchansky A. Role of Platelet Activation and Oxidative Stress in the Evolution of Myocardial Infarction. J Cardiovasc Pharmacol Ther 2019; 24:509-520. [DOI: 10.1177/1074248419861437] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myocardial infarction, commonly known as heart attack, evolves from the rupture of unstable atherosclerotic plaques to coronary thrombosis and myocardial ischemia–reperfusion injury. A body of evidence supports a close relationship between the alterations following an ischemia–reperfusion injury-induced oxidative stress and platelet activity. Through their critical role in thrombogenesis and inflammatory responses, platelets are fully (totally) implicated from atherothrombotic plaque formation to myocardial infarction onset and expansion. However, mere platelet aggregation prevention does not offer full protection, suggesting that other antiplatelet therapy mechanisms may also be involved. Thus, the present review discusses the integrative role of platelets, oxidative stress, and antiplatelet therapy in triggering myocardial infarction pathophysiology.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca, Chile
| | - Rodrigo Moore-Carrasco
- Departamento de Bioquímica Clínica e Inmunohematología, Facultad de Ciencias de la Salud, Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Talca, Chile
| | - Antonio Marcus de Andrade Paes
- Laboratory of Experimental Physiology, Health Sciences Graduate Program and Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Andres Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
24
|
Metabolic Disturbances Identified in Plasma Samples from ST-Segment Elevation Myocardial Infarction Patients. DISEASE MARKERS 2019; 2019:7676189. [PMID: 31354891 PMCID: PMC6636502 DOI: 10.1155/2019/7676189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/07/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022]
Abstract
ST-segment elevation myocardial infarction (STEMI) is the most severe form of myocardial infarction (MI) and the main contributor to morbidity and mortality caused by MI worldwide. Frequently, STEMI is caused by complete and persistent occlusion of a coronary artery by a blood clot, which promotes heart damage. STEMI impairment triggers changes in gene transcription, protein expression, and metabolite concentrations, which grants a biosignature to the heart dysfunction. There is a major interest in identifying novel biomarkers that could improve the diagnosis of STEMI. In this study, the phenotypic characterization of STEMI patients (n = 15) and healthy individuals (n = 19) was performed, using a target metabolomics approach. Plasma samples were analyzed by UPLC-MS/MS (ultra-high-performance liquid chromatography-tandem mass spectrometry) and FIA-MS (MS-based flow injection analysis). The goal was to identify novel plasma biomarkers and metabolic signatures underlying STEMI. Concentrations of phosphatidylcholines, lysophosphatidylcholines, sphingomyelins, and biogenic amines were altered in STEMI patients in relation to healthy subjects. Also, after multivariate analysis, it was possible to identify alterations in the glycerophospholipids, alpha-linolenic acid, and sphingolipid metabolisms in STEMI patients.
Collapse
|
25
|
Alessenko AV, Lebedev AT, Kurochkin IN. The Role of Sphingolipids in Cardiovascular Pathologies. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2019. [DOI: 10.1134/s1990750819020021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Alessenko AV, Lebedev АТ, Kurochkin IN. [The role of sphingolipids in cardiovascular pathologies]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 64:487-495. [PMID: 30632976 DOI: 10.18097/pbmc20186406487] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cardiovascular diseases (CVD) remain the leading cause of death in industrialized countries. One of the most significant risk factors for atherosclerosis is hypercholesterolemia. Its diagnostics is based on routine lipid profile analysis, including the determination of total cholesterol, low and high density lipoprotein cholesterol, and triglycerides. However in recent years, much attention has been paid to the crosstalk between the metabolic pathways of the cholesterol and sphingolipids biosynthesis. Sphingolipids are a group of lipids, containing a molecule of aliphatic alcohol sphingosine. These include sphingomyelins, cerebrosides, gangliosides and ceramides, sphingosines, and sphingosine-1-phosphate (S-1-P). It has been found that catabolism of sphingolipids is associated with catabolism of cholesterol. However, the exact mechanism of this interaction is still unknown. Particular attention as CVD inducer attracts ceramide (Cer). Lipoprotein aggregates isolated from atherosclerotic pluques are enriched with Cer. The level of Cer and sphingosine increases after ischemia reperfusion of the heart, in the infarction zone and in the blood, and also in hypertension. S-1-P exhibits pronounced cardioprotective properties. Its content sharply decreases with ischemia and myocardial infarction. S-1-P presents predominantly in HDL, and influences their multiple functions. Increased levels of Cer and sphingosine and decreased levels of S-1-P formed in the course of coronary heart disease can be an important factor in the development of atherosclerosis. It is proposed to use determination of sphingolipids in blood plasma as markers for early diagnosis of cardiac ischemia and for hypertension in humans. There are intensive studies aimed at correction of metabolism S-1-P. The most successful drugs are those that use S-1-P receptors as a targets, since all of its actions are receptor-mediated.
Collapse
Affiliation(s)
- A V Alessenko
- Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | | | - I N Kurochkin
- Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
27
|
Magaye RR, Savira F, Hua Y, Kelly DJ, Reid C, Flynn B, Liew D, Wang BH. The role of dihydrosphingolipids in disease. Cell Mol Life Sci 2019; 76:1107-1134. [PMID: 30523364 PMCID: PMC11105797 DOI: 10.1007/s00018-018-2984-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/06/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022]
Abstract
Dihydrosphingolipids refer to sphingolipids early in the biosynthetic pathway that do not contain a C4-trans-double bond in the sphingoid backbone: 3-ketosphinganine (3-ketoSph), dihydrosphingosine (dhSph), dihydrosphingosine-1-phosphate (dhS1P) and dihydroceramide (dhCer). Recent advances in research related to sphingolipid biochemistry have shed light on the importance of sphingolipids in terms of cellular signalling in health and disease. However, dihydrosphingolipids have received less attention and research is lacking especially in terms of their molecular mechanisms of action. This is despite studies implicating them in the pathophysiology of disease, for example dhCer in predicting type 2 diabetes in obese individuals, dhS1P in cardiovascular diseases and dhSph in hepato-renal toxicity. This review gives a comprehensive summary of research in the last 10-15 years on the dihydrosphingolipids, 3-ketoSph, dhSph, dhS1P and dhCer, and their relevant roles in different diseases. It also highlights gaps in research that could be of future interest.
Collapse
Affiliation(s)
- Ruth R Magaye
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Feby Savira
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Darren J Kelly
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - Christopher Reid
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Bernard Flynn
- Australian Translational Medicinal Chemistry Facility, Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Bing H Wang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.
| |
Collapse
|
28
|
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: 36] [Impact Index Per Article: 6.0] [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
|
29
|
Meeusen JW, Donato LJ, Bryant SC, Baudhuin LM, Berger PB, Jaffe AS. Plasma Ceramides. Arterioscler Thromb Vasc Biol 2018; 38:1933-1939. [DOI: 10.1161/atvbaha.118.311199] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Ceramides are sphingolipids involved with cellular signaling. Synthesis of ceramides occurs in all tissues. Ceramides accumulate within tissues and the blood plasma during metabolic dysfunction, dyslipidemia, and inflammation. Elevations of ceramides are predictive of cardiovascular mortality. We sought to verify the utility of plasma concentrations of 4 ceramides: N-palmitoyl-sphingosine [Cer(16:0)], N-stearoyl-sphingosine [Cer(18:0)], N-nervonoyl-sphingosine [Cer(24:1)], and N-lignoceroyl-sphingosine [Cer(24:0)] in predicting major adverse cardiovascular events in a diverse patient population referred for coronary angiography.
Approach and Results—
Plasma ceramides were measured in 495 participants before nonurgent coronary angiography. Coronary artery disease, defined as >50% stenosis in ≥1 coronary artery, was identified 265 (54%) cases. Ceramides were not significantly associated with coronary artery disease. Patients were followed for a combined primary end point of myocardial infarction, percutaneous intervention, coronary artery bypass, stroke, or death within 4 years. Ceramides were significantly predictive of outcomes after adjusting for age, sex, body mass index, hypertension, smoking, LDL (low-density lipoprotein) cholesterol, HDL (high-density lipoprotein) cholesterol, triglycerides, serum glucose, and family history of coronary artery disease. The fully adjusted per SD hazard ratios (95% confidence interval) were 1.50 (1.16–1.93) for Cer(16:0), 1.42 (1.11–1.83) for Cer(18:0), 1.43 (1.08–1.89) for Cer(24:1), and 1.58 (1.22–2.04) for the ceramide risk score.
Conclusions—
Elevated plasma concentrations of ceramides are independently associated with major adverse cardiovascular events in patients with and without coronary artery disease.
Collapse
Affiliation(s)
- Jeffrey W. Meeusen
- From the Department of Laboratory Medicine and Pathology (J.W.M., L.J.D., L.M.B., A.S.J.)
| | - Leslie J. Donato
- From the Department of Laboratory Medicine and Pathology (J.W.M., L.J.D., L.M.B., A.S.J.)
| | | | - Linnea M. Baudhuin
- From the Department of Laboratory Medicine and Pathology (J.W.M., L.J.D., L.M.B., A.S.J.)
| | - Peter B. Berger
- Department of Cardiology (P.B.B., A.S.J.), Mayo Clinic, Rochester, MN
| | - Allan S. Jaffe
- From the Department of Laboratory Medicine and Pathology (J.W.M., L.J.D., L.M.B., A.S.J.)
- Department of Cardiology (P.B.B., A.S.J.), Mayo Clinic, Rochester, MN
| |
Collapse
|
30
|
The significance of diminished sTWEAK and P-selectin content in platelets of patients with pulmonary arterial hypertension. Cytokine 2018; 107:52-58. [DOI: 10.1016/j.cyto.2017.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 11/21/2017] [Accepted: 11/25/2017] [Indexed: 01/18/2023]
|
31
|
Yücel G, Behnes M, Barth C, Wenke A, Sartorius B, Mashayekhi K, Yazdani B, Bertsch T, Rusnak J, Saleh A, Hoffmann U, Fastner C, Lang S, Zhou X, Sattler K, Borggrefe M, Akin I. Percutaneous Closure of Left Atrial Appendage significantly affects Lipidome Metabolism. Sci Rep 2018; 8:5894. [PMID: 29650978 PMCID: PMC5897373 DOI: 10.1038/s41598-018-23935-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 03/23/2018] [Indexed: 11/09/2022] Open
Abstract
Patients with non-valvular atrial fibrillation (AF) and a high risk for oral anticoagulation can be treated by percutaneous implantation of left atrial appendage occlusion devices (LAAC) to reduce the risk of cardio-embolic stroke. This study evaluates whether LAAC may influence lipid metabolism, which has never been investigated before. Patients with successful LAAC were included consecutively. Venous peripheral blood samples of patients were collected immediately before (T0, baseline) and 6 months after (T1, mid-term) LAAC. A targeted metabolomics approach based on electrospray ionization liquid chromatography-mass spectrometry (ESI-LC-MS/MS) and MS/MS measurements was performed. A total of 34 lipids revealed a significant change from baseline to mid-term follow-up after successful LAAC. Subgroup analysis revealed confounding influence by gender, age, diabetes mellitus type II, body mass index, left ventricular ejection fraction, creatinine and NT-proBNP. After multivariable adjustment within logistic regression models, these 34 lipids were still significantly altered after LAAC. Successful percutaneous LAAC may affect lipid metabolism and thereby may potentially affect pro-atherogenic and cardio-toxic effects.
Collapse
Affiliation(s)
- G Yücel
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - M Behnes
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.
| | - C Barth
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - A Wenke
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - B Sartorius
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - K Mashayekhi
- Clinic for Cardiology and Angiology II, Universitäts-Herzzentrum Freiburg - Bad Krozingen, Bad Krozingen, Germany
| | - B Yazdani
- Fifth Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - T Bertsch
- Institute of Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, General Hospital Nuremberg and Paracelsus Medical University, Nuremberg, Germany
| | - J Rusnak
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - A Saleh
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - U Hoffmann
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - C Fastner
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - S Lang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - X Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - K Sattler
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - M Borggrefe
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - I Akin
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| |
Collapse
|
32
|
Kazimierczyk R, Blaszczak P, Jasiewicz M, Knapp M, Ptaszynska-Kopczynska K, Sobkowicz B, Waszkiewicz E, Grzywna R, Musial WJ, Kaminski KA. Increased platelet content of SDF-1alpha is associated with worse prognosis in patients with pulmonary prterial hypertension. Platelets 2018; 30:445-451. [PMID: 29617176 DOI: 10.1080/09537104.2018.1457780] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Inflammatory processes and platelet activity play an important role in the pathophysiology of pulmonary arterial hypertension (PAH). Enhanced IL-6 signaling and higher concentration of stromal-derived factor alpha (SDF-1) have been previously shown to be linked with prognosis in PAH. We hypothesized that platelets of PAH patients have higher content of IL-6 and SDF-1 and thus are involved in disease progression. We enrolled into study 22 PAH patients and 18 healthy controls. Patients with PAH presented significantly higher plasma concentrations and platelet contents of IL-6, sIL-6R, and SDF-1 than healthy subjects (platelet content normalized to protein concentration: IL-6 (0.85*10-10 [0.29 - 1.37] vs. 0.45*10-10 [0.19-0.65], sIL-6R 1.54*10-7 [1.32-2.21] vs. 1.14*10-7 [1.01-1.28] and SDF-1 (2.72*10-7 [1.85-3.23] vs. 1.70*10-7 [1.43-2.60], all p < 0.05). Patients with disease progression (death, WHO class worsening, or therapy escalation, n = 10) had a significantly higher platelet SDF-1/total platelet protein ratio (3.68*10-7 [2.45-4.62] vs. 1.69*10-7 [1.04-2.28], p = 0.001), with no significant differences between plasma levels. Kaplan-Meier analysis revealed that patients with higher platelet SDF-1/total platelet protein ratio had more frequently deterioration of PAH in the follow-up (15.24 ± 4.26 months, log-rank test, p = 0.01). Concentrations of IL-6, sIL-6 receptor and SDF-1 in plasma and platelets are elevated in PAH patients. Higher content of SDF-1 in platelets is associated with poorer prognosis. Our study, despite of limitation due to small number of enrolled patients, suggests that activated platelets may be an important source of cytokines at the site of endothelial injury, but their exact role in the pathogenesis of PAH requires further investigation.
Collapse
Affiliation(s)
| | - Piotr Blaszczak
- b Department of Cardiology , Cardinal Wyszynski Hospital , Lublin , Poland
| | - Małgorzata Jasiewicz
- a Department of Cardiology , Medical University of Bialystok , Bialystok , Poland
| | - Małgorzata Knapp
- a Department of Cardiology , Medical University of Bialystok , Bialystok , Poland
| | | | - Bozena Sobkowicz
- a Department of Cardiology , Medical University of Bialystok , Bialystok , Poland
| | - Ewa Waszkiewicz
- a Department of Cardiology , Medical University of Bialystok , Bialystok , Poland
| | - Ryszard Grzywna
- b Department of Cardiology , Cardinal Wyszynski Hospital , Lublin , Poland
| | - Wlodzimierz J Musial
- a Department of Cardiology , Medical University of Bialystok , Bialystok , Poland
| | - Karol A Kaminski
- a Department of Cardiology , Medical University of Bialystok , Bialystok , Poland.,c Department of Population Medicine and Civilization Diseases Prevention , Medical University of Bialystok , Bialystok , Poland
| |
Collapse
|
33
|
Pang HQ, Yue SJ, Tang YP, Chen YY, Tan YJ, Cao YJ, Shi XQ, Zhou GS, Kang A, Huang SL, Shi YJ, Sun J, Tang ZS, Duan JA. Integrated Metabolomics and Network Pharmacology Approach to Explain Possible Action Mechanisms of Xin-Sheng-Hua Granule for Treating Anemia. Front Pharmacol 2018; 9:165. [PMID: 29551975 PMCID: PMC5840524 DOI: 10.3389/fphar.2018.00165] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/14/2018] [Indexed: 11/13/2022] Open
Abstract
As a well-known traditional Chinese medicine (TCM) prescription, Xin-Sheng-Hua Granule (XSHG) has been applied in China for more than 30 years to treat postpartum diseases, especially anemia. However, underlying therapeutic mechanisms of XSHG for anemia were still unclear. In this study, plasma metabolomics profiling with UHPLC-QTOF/MS and multivariate data method was firstly analyzed to discover the potential regulation mechanisms of XSHG on anemia rats induced by bleeding from the orbit. Afterward, the compound-target-pathway network of XSHG was constructed by the use of network pharmacology, thus anemia-relevant signaling pathways were dissected. Finally, the crucial targets in the shared pathways of metabolomics and network pharmacology were experimentally validated by ELISA and Western Blot analysis. The results showed that XSHG could exert excellent effects on anemia probably through regulating coenzyme A biosynthesis, sphingolipids metabolism and HIF-1α pathways, which was reflected by the increased levels of EPOR, F2, COASY, as well as the reduced protein expression of HIF-1α, SPHK1, and S1PR1. Our work successfully explained the polypharmcological mechanisms underlying the efficiency of XSHG on treating anemia, and meanwhile, it probed into the potential treatment strategies for anemia from TCM prescription.
Collapse
Affiliation(s)
- Han-Qing Pang
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shi-Jun Yue
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu-Ping Tang
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan-Yan Chen
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Ya-Jie Tan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu-Jie Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xu-Qin Shi
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Gui-Sheng Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - An Kang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Ya-Jun Shi
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Sun
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zhi-Shu Tang
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Camerer E. Endotoxemia rocks sphingolipid metabolism at the blood-brain barrier: An Editorial Highlight for 'Alteration of sphingolipid metabolism as a putative mechanism underlying LPS-induced BBB disruption' on page 172. J Neurochem 2017; 144:115-117. [PMID: 29285771 DOI: 10.1111/jnc.14246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/18/2017] [Indexed: 11/28/2022]
Abstract
In this issue of the Journal of Neurochemistry, Vutukuri et al. evaluate the impact of endotoxemia-induced encephalopathy on the sphingosine-1-phosphate (S1P) signaling pathway at the blood-brain barrier (BBB). Four hours after intraperitoneal administration of lipopolysaccharides (LPS, 4 mg/kg) to mice, they first demonstrate BBB dysfunction and then evaluate changes in sphingolipid metabolites in serum, isolated brain microvessels (MBMV), and whole brain. In parallel, they investigate the fate of indicated S1P generating and metabolizing enzymes and S1P receptors in brain and MBMV. S1P levels decreased in serum and brain and a similar tendency was observed in MBMV. Sphk2 expression was strongly reduced in MBMV together with an up-regulation of lipid phosphate and S1P phosphatases, resulting in a net decrease in S1P levels despite a compensatory increase in Sphk1 expression. The implications of disturbed sphingolipid metabolism for the pathogenesis of septic encephalopathy will depend on the net impact of these changes on S1P receptor signaling at the BBB and the importance of the S1P pathway in regulating vascular homeostasis in this context.
Collapse
Affiliation(s)
- Eric Camerer
- INSERM U970, Paris Cardiovascular Research Center, Paris, France.,Université Paris-Descartes, Paris, France
| |
Collapse
|
35
|
Knapp P, Bodnar L, Błachnio-Zabielska A, Świderska M, Chabowski A. Plasma and ovarian tissue sphingolipids profiling in patients with advanced ovarian cancer. Gynecol Oncol 2017; 147:139-144. [PMID: 28800942 DOI: 10.1016/j.ygyno.2017.07.143] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/27/2017] [Accepted: 07/30/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE The role of lipids in carcinogenesis through induction of abnormal cell lines in the human body is currently undisputable. Based on the literature, bioactive sphingolipids play an essential role in the development and progression of cancer and are involved in the metastatic process. The aim of this study was to determine the concentration of selected sphingolipids in patients with advanced ovarian cancer (AOC, FIGO III/IV, high grade ovarian cancer). METHODS Seventy-four patients with ovarian cancer were enrolled. Plasma concentrations of C16-Cer, C18:1-Cer and C18-Cer were assessed by LC/MS/MS. The content of tissue sphingolipids was measured using a UHPLC/MS/MS. RESULTS Plasma concentration of 3 ceramides: C16-Cer, C18:1-Cer and C18-Cer was significantly elevated in women with advanced ovarian cancer compared to control group (P=0.031; 0.022; 0.020; respectively). There were increases in concentration of 5 ceramides: C16-Cer, C18:1-Cer, C18-Cer, C24:1-Cer, C24-Cer (P=0.025; 0.049; 0.032; 0.005; 0.013, respectively) and S1P (P=0.004) in ovarian tissue of women with advanced ovarian cancer compared to healthy individuals. Importantly, significantly higher risk of ovarian cancer when the plasma concentration of C16-Cer>311.88ng/100μl (AUC: 0.76, P=0.0261); C18:1-Cer>4.75ng/100μl (AUC: 0.77, P=0.0160) and C18-Cer>100.76ng/100μl (AUC:0.77, P=0.0136) was noticed. CONCLUSIONS Bioactive sphingolipids play an essential role in the development and progression of cancer and they also take part in the process of metastasizing. This study suggests that some sphingolipids can be used as potential biomarkers of advanced ovarian cancer and that they can play an important role in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Paweł Knapp
- Department of Gynecology and Gynecologic Oncology, Medical University of Bialystok, 24a Sklodowskiej-Curie Str., 15-276 Bialystok, Poland.
| | - Lubomir Bodnar
- Department of Clinical Oncology, Military Institute of Medicine in Warsaw, 128 Szaserow Str., 04-141 Warsaw, Poland.
| | - Agnieszka Błachnio-Zabielska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 2c Mickiewicza Str., 15-222 Bialystok, Poland; Department of Physiology, Medical University of Bialystok, 2c Mickiewicza Str., 15-222 Bialystok, Poland.
| | - Magdalena Świderska
- Department of Physiology, Medical University of Bialystok, 2c Mickiewicza Str., 15-222 Bialystok, Poland.
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 2c Mickiewicza Str., 15-222 Bialystok, Poland.
| |
Collapse
|
36
|
Iqbal J, Walsh MT, Hammad SM, Hussain MM. Sphingolipids and Lipoproteins in Health and Metabolic Disorders. Trends Endocrinol Metab 2017; 28:506-518. [PMID: 28462811 PMCID: PMC5474131 DOI: 10.1016/j.tem.2017.03.005] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/09/2017] [Accepted: 03/28/2017] [Indexed: 12/28/2022]
Abstract
Sphingolipids are structurally and functionally diverse molecules with significant physiologic functions and are found associated with cellular membranes and plasma lipoproteins. The cellular and plasma concentrations of sphingolipids are altered in several metabolic disorders and may serve as prognostic and diagnostic markers. Here we discuss various sphingolipid transport mechanisms and highlight how changes in cellular and plasma sphingolipid levels contribute to cardiovascular disease, obesity, diabetes, insulin resistance, and nonalcoholic fatty liver disease (NAFLD). Understanding of the mechanisms involved in intracellular transport, secretion, and extracellular transport may provide novel information that might be amenable to therapeutic targeting for the treatment of various metabolic disorders.
Collapse
Affiliation(s)
- Jahangir Iqbal
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, NY 11203, USA; King Abdullah International Medical Research Center, MNGHA, Al Ahsa 31982, Saudi Arabia
| | - Meghan T Walsh
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, NY 11203, USA
| | - Samar M Hammad
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - M Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, NY 11203, USA; VA New York Harbor Healthcare System, Brooklyn, New York, NY 11209; Center for Diabetes and Obesity Research, NYU Winthrop Hospital, Mineola, NY 11501, USA.
| |
Collapse
|
37
|
QGQS Granule in SHR Serum Metabonomics Study Based on Tools of UPLC-Q-TOF and Renin-Angiotensin-Aldosterone System Form Protein Profilin-1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:4854720. [PMID: 28367224 PMCID: PMC5358465 DOI: 10.1155/2017/4854720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 01/15/2017] [Accepted: 01/24/2017] [Indexed: 01/09/2023]
Abstract
QGQS granule is effective for the therapeutic of hypertension in clinic. The aim of this research is to observe the antihypertension effect of QGQS granule on SHR and explain the mechanism of its lowering blood pressure. 30 SHR were selected as model group, captopril group, and QGQS group, 10 WKYr were used as control group, and RBP were measured on tail artery consciously. And all the serum sample analysis was carried out on UPLC-TOF-MS system to determine endogenous metabolites and to find the metabonomics pathways. Meanwhile, ELISA kits for the determination pharmacological indexes of PRA, AngI, AngII, and ALD were used for pathway confirmatory; WB for determination of profilin-1 protein expression was conducted for Ang II pathway analysis as well. It is demonstrated that QGQS granule has an excellent therapeutic effect on antihypertension, which exerts effect mainly on metabonomics pathway by regulating glycerophospholipid, sphingolipid, and arachidonic acid metabolism, and it could inhibit the overexpression of the profilin-1 protein. We can come to a conclusion that RAAS should be responsible mainly for the metabonomics pathway of QGQS granule on antihypertension, and it plays a very important role in protein of profilin-1 inhibition.
Collapse
|
38
|
Checa A, Idborg H, Zandian A, Sar DG, Surowiec I, Trygg J, Svenungsson E, Jakobsson PJ, Nilsson P, Gunnarsson I, Wheelock CE. Dysregulations in circulating sphingolipids associate with disease activity indices in female patients with systemic lupus erythematosus: a cross-sectional study. Lupus 2017; 26:1023-1033. [PMID: 28134039 DOI: 10.1177/0961203316686707] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective The objective of this study was to investigate the association of clinical and renal disease activity with circulating sphingolipids in patients with systemic lupus erythematosus. Methods We used liquid chromatography tandem mass spectrometry to measure the levels of 27 sphingolipids in plasma from 107 female systemic lupus erythematosus patients and 23 controls selected using a design of experiment approach. We investigated the associations between sphingolipids and two disease activity indices, the Systemic Lupus Activity Measurement and the Systemic Lupus Erythematosus Disease Activity Index. Damage was scored according to the Systemic Lupus International Collaborating Clinics damage index. Renal activity was evaluated with the British Island Lupus Activity Group index. The effects of immunosuppressive treatment on sphingolipid levels were evaluated before and after treatment in 22 female systemic lupus erythematosus patients with active disease. Results Circulating sphingolipids from the ceramide and hexosylceramide families were increased, and sphingoid bases were decreased, in systemic lupus erythematosus patients compared to controls. The ratio of C16:0-ceramide to sphingosine-1-phosphate was the best discriminator between patients and controls, with an area under the receiver-operating curve of 0.77. The C16:0-ceramide to sphingosine-1-phosphate ratio was associated with ongoing disease activity according to the Systemic Lupus Activity Measurement and the Systemic Lupus Erythematosus Disease Activity Index, but not with accumulated damage according to the Systemic Lupus International Collaborating Clinics Damage Index. Levels of C16:0- and C24:1-hexosylceramides were able to discriminate patients with current versus inactive/no renal involvement. All dysregulated sphingolipids were normalized after immunosuppressive treatment. Conclusion We provide evidence that sphingolipids are dysregulated in systemic lupus erythematosus and associated with disease activity. This study demonstrates the utility of simultaneously targeting multiple components of a pathway to establish disease associations.
Collapse
Affiliation(s)
- A Checa
- 1 Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE- 171 77 Stockholm, Sweden
| | - H Idborg
- 2 Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE- 171 76 Stockholm, Sweden
| | - A Zandian
- 3 Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - D Garcia Sar
- 1 Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE- 171 77 Stockholm, Sweden
| | - I Surowiec
- 4 Computational Life Science Cluster, Department of Chemistry, Umeå University, Umeå, Sweden
| | - J Trygg
- 4 Computational Life Science Cluster, Department of Chemistry, Umeå University, Umeå, Sweden
| | - E Svenungsson
- 2 Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE- 171 76 Stockholm, Sweden
| | - P-J Jakobsson
- 2 Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE- 171 76 Stockholm, Sweden
| | - P Nilsson
- 3 Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - I Gunnarsson
- 2 Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE- 171 76 Stockholm, Sweden
| | - C E Wheelock
- 1 Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE- 171 77 Stockholm, Sweden
| |
Collapse
|
39
|
Ross JS, Russo SB, Chavis GC, Cowart LA. Sphingolipid regulators of cellular dysfunction in Type 2 diabetes mellitus: a systems overview. CLINICAL LIPIDOLOGY 2017; 9:553-569. [PMID: 29643939 PMCID: PMC5891157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Climbing obesity rates have contributed to worldwide increases in obesity-associated diseases, including the metabolic syndrome and Type 2 diabetes mellitus (T2DM). Sphingolipids, an important class of structural and signaling lipids, have emerged as key players in the development and pathogenesis of insulin resistance and T2DM. More specifically, sphingolipids have been demonstrated to play integral roles in lipotoxicity and other aspects of pathogenesis in T2DM, although the cellular mechanisms by which this occurs and by which sphingolipid metabolism is dysregulated in T2DM remain under investigation. This review summarizes current knowledge of sphingolipid metabolism and signaling in key organs and tissues affected by T2DM, including the pancreas, adipose tissue, skeletal muscle, cardiovascular system and liver, and highlights areas that ripe for future investigation.
Collapse
Affiliation(s)
- Jessica S Ross
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425,USA
| | - Sarah B Russo
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425,USA
| | - Georgia C Chavis
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425,USA
| | - Lauren A Cowart
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425,USA
- Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| |
Collapse
|
40
|
Sasset L, Zhang Y, Dunn TM, Di Lorenzo A. Sphingolipid De Novo Biosynthesis: A Rheostat of Cardiovascular Homeostasis. Trends Endocrinol Metab 2016; 27:807-819. [PMID: 27562337 PMCID: PMC5075255 DOI: 10.1016/j.tem.2016.07.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/07/2016] [Accepted: 07/20/2016] [Indexed: 01/01/2023]
Abstract
Sphingolipids (SL) are both fundamental structural components of the eukaryotic membranes and signaling molecules that regulate a variety of biological functions. The highly-bioactive lipids, ceramide and sphingosine-1-phosphate, have emerged as important regulators of cardiovascular function in health and disease. In this review we discuss recent insights into the role of SLs, particularly ceramide and sphingosine-1-phosphate, in the pathophysiology of the cardiovascular system. We also highlight advances into the molecular mechanisms regulating serine palmitoyltransferase, the first and rate-limiting enzyme of de novo SL biosynthesis, with an emphasis on the recently discovered inhibitors of serine palmitoyltransferase, ORMDL and NOGO-B proteins. Understanding the molecular mechanisms regulating this biosynthetic pathway may lead to the development of novel therapeutic approaches for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Linda Sasset
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Yi Zhang
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Teresa M Dunn
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Annarita Di Lorenzo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
41
|
Gazit SL, Mariko B, Thérond P, Decouture B, Xiong Y, Couty L, Bonnin P, Baudrie V, Le Gall SM, Dizier B, Zoghdani N, Ransinan J, Hamilton JR, Gaussem P, Tharaux PL, Chun J, Coughlin SR, Bachelot-Loza C, Hla T, Ho-Tin-Noé B, Camerer E. Platelet and Erythrocyte Sources of S1P Are Redundant for Vascular Development and Homeostasis, but Both Rendered Essential After Plasma S1P Depletion in Anaphylactic Shock. Circ Res 2016; 119:e110-26. [PMID: 27582371 DOI: 10.1161/circresaha.116.308929] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/30/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE Sphingosine-1-phosphate (S1P) signaling is essential for vascular development and postnatal vascular homeostasis. The relative importance of S1P sources sustaining these processes remains unclear. OBJECTIVE To address the level of redundancy in bioactive S1P provision to the developing and mature vasculature. METHODS AND RESULTS S1P production was selectively impaired in mouse platelets, erythrocytes, endothelium, or smooth muscle cells by targeted deletion of genes encoding sphingosine kinases -1 and -2. S1P deficiency impaired aggregation and spreading of washed platelets and profoundly reduced their capacity to promote endothelial barrier function ex vivo. However, and in contrast to recent reports, neither platelets nor any other source of S1P was essential for vascular development, vascular integrity, or hemostasis/thrombosis. Yet rapid and profound depletion of plasma S1P during systemic anaphylaxis rendered both platelet- and erythrocyte-derived S1P essential for survival, with a contribution from blood endothelium observed only in the absence of circulating sources. Recovery was sensitive to aspirin in mice with but not without platelet S1P, suggesting that platelet activation and stimulus-response coupling is needed. S1P deficiency aggravated vasoplegia in this model, arguing a vital role for S1P in maintaining vascular resistance during recovery from circulatory shock. Accordingly, the S1P2 receptor mediated most of the survival benefit of S1P, whereas the endothelial S1P1 receptor was dispensable for survival despite its importance for maintaining vascular integrity. CONCLUSIONS Although source redundancy normally secures essential S1P signaling in developing and mature blood vessels, profound depletion of plasma S1P renders both erythrocyte and platelet S1P pools necessary for recovery and high basal plasma S1P levels protective during anaphylactic shock.
Collapse
Affiliation(s)
- Salomé L Gazit
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Boubacar Mariko
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Patrice Thérond
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Benoit Decouture
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Yuquan Xiong
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Ludovic Couty
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Philippe Bonnin
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Véronique Baudrie
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Sylvain M Le Gall
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Blandine Dizier
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Nesrine Zoghdani
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Jessica Ransinan
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Justin R Hamilton
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Pascale Gaussem
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Pierre-Louis Tharaux
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Jerold Chun
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Shaun R Coughlin
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Christilla Bachelot-Loza
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Timothy Hla
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Benoit Ho-Tin-Noé
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Eric Camerer
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.).
| |
Collapse
|
42
|
Li N, Zhang F. Implication of sphingosin-1-phosphate in cardiovascular regulation. Front Biosci (Landmark Ed) 2016; 21:1296-313. [PMID: 27100508 DOI: 10.2741/4458] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite generated by phosphorylation of sphingosine catalyzed by sphingosine kinase. S1P acts mainly through its high affinity G-protein-coupled receptors and participates in the regulation of multiple systems, including cardiovascular system. It has been shown that S1P signaling is involved in the regulation of cardiac chronotropy and inotropy and contributes to cardioprotection as well as cardiac remodeling; S1P signaling regulates vascular function, such as vascular tone and endothelial barrier, and possesses an anti-atherosclerotic effect; S1P signaling is also implicated in the regulation of blood pressure. Therefore, manipulation of S1P signaling may offer novel therapeutic approaches to cardiovascular diseases. As several S1P receptor modulators and sphingosine kinase inhibitors have been approved or under clinical trials for the treatment of other diseases, it may expedite the test and implementation of these S1P-based drugs in cardiovascular diseases.
Collapse
Affiliation(s)
- Ningjun Li
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA,
| | - Fan Zhang
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
43
|
Błachnio-Zabielska A, Baranowski M, Wójcik B, Górski J. Reduction of ceramide de novo synthesis in solid tissues changes sphingolipid levels in rat plasma, erythrocytes and platelets. Adv Med Sci 2016; 61:72-7. [PMID: 26521206 DOI: 10.1016/j.advms.2015.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 07/03/2015] [Accepted: 09/17/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE De novo sphingolipid synthesis does not occur in plasma, erythrocytes and platelets. The purpose of the study was to examine the effect of inhibition of sphingolipid synthesis in solid tissues on the level of the following bioactive sphingolipids: sphinganine, ceramide, sphingosine and sphingosine 1-phosphate in plasma, erythrocytes and platelets. MATERIAL/METHODS The experiments were carried out on male Wistar rats. Myriocin was used to inhibit serine palmitoyltransferase activity (the enzyme catalyzes the first step of ceramide de novo synthesis) and nicotinic acid was used to reduce the concentration of plasma free fatty acids (a substrate for the de novo ceramide synthesis). The sphingolipids were quantified by means of liquid chromatography/mass spectrometry. RESULTS Myriocin reduced the level of each compound in plasma. It reduced the level of sphinganine, sphingosine-1-phosphate and total ceramide and elevated the level of sphingosine in erythrocytes. In platelets, myriocin reduced the total level of ceramide. Nicotinic acid reduced the plasma level of sphinganine, sphingosine and total ceramide. It increased the level of sphingosine-1-phosphate in erythrocytes. In platelets, nicotinioc acid increased the level of sphinganine and sphingosine and reduced the level of sphingosine-1-phosphate and total ceramide. CONCLUSIONS Inhibition of serine palmitoyltransferase activity in solid tissues and reduction in plasma free fatty acids concentration affects sphingolipid level in plasma, erythrocytes and platelets. The changes in erythrocytes and platelets depend both on the cell type and the sphingolipid studied and only partially follow the changes in the plasma.
Collapse
|
44
|
Farrokhi M, Beni AA, Etemadifar M, Rezaei A, Rivard L, Zadeh AR, Sedaghat N, Ghadimi M. Effect of Fingolimod on Platelet Count Among Multiple Sclerosis Patients. Int J Prev Med 2016; 6:125. [PMID: 26900439 PMCID: PMC4736130 DOI: 10.4103/2008-7802.172539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 08/23/2015] [Indexed: 11/10/2022] Open
Abstract
Background: While many studies have previously focused on fingolimod's effect on immune cells, the effect it has on circulating and local central nervous system platelets (Plts) has not yet been investigated. This study will elucidate what effects fingolimod treatment has on multiple sclerosis (MS) patients’ plasma Plt levels. In addition, it will propose possible reasoning for these effects and suggest further investigation into this topic. Methods: This quasi-experimental study used patients from the Isfahan Multiple Sclerosis Society to produce a subject pool of 80 patients, including 14 patients who ceased fingolimod use due to complications. The patients had their blood analyzed to determine Plt levels both 1-month prior to fingolimod treatment and 1-month after fingolimod treatment had been started. Results: The mean level of Plts before initiation of fingolimod therapy (Plt1) among these MS patients was 256.53 ± 66.26. After 1-month of fingolimod treatment, the Plt level yielded an average of 229.96 ± 49.67 (Plt2). This number is significantly lower than the average Plt count before treatment (P < 0.01). Conclusions: MS patients taking oral fingolimod treatment may be at risk for side-effects caused by low Plt levels. This may not be a factor for patients with higher or normal Plt levels. However, a patient with naturally low Plt levels may experience a drop below the normal level and be at risk for excessive bleeding. In addition to these possible harmful side-effects, the decreased Plt population may pose positive effects for MS patients.
Collapse
Affiliation(s)
- Mehrdad Farrokhi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Neurosciences Research Centre, Affiliated to Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Amani Beni
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Neurosciences Research Centre, Affiliated to Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Etemadifar
- Isfahan Neurosciences Research Centre, Affiliated to Isfahan University of Medical Sciences, Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Multiple Sclerosis and Neuroimmunology Research Center, Isfahan, Iran
| | - Ali Rezaei
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912, USA; Department of Neuroscience, Brown University, Providence, Rhode Island 02912, USA
| | - Leah Rivard
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912, USA; Department of Neuroscience, Brown University, Providence, Rhode Island 02912, USA
| | - Aryan Rafiee Zadeh
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Sedaghat
- Isfahan Neurosciences Research Centre, Affiliated to Isfahan University of Medical Sciences, Isfahan, Iran
| | - Milad Ghadimi
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran; Department of Immunology, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
45
|
Inhibition of ceramide de novo synthesis as a postischemic strategy to reduce myocardial reperfusion injury. Basic Res Cardiol 2016; 111:12. [PMID: 26786259 DOI: 10.1007/s00395-016-0533-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/11/2016] [Indexed: 12/17/2022]
Abstract
The injury caused by myocardial reperfusion after ischemia can be contained by interventions aimed at reducing the inflammation and the oxidative stress that underlie exacerbation of tissue damage. Sphingolipids are a class of structural and signaling lipid molecules; among them, the inflammation mediator ceramide accumulates in the myocardium upon ischemia/reperfusion. Here, we show that, after transient coronary occlusion in mice, an increased de novo ceramide synthesis takes place at reperfusion in the ischemic area surrounding necrosis (area at risk). This correlates with the enhanced expression of the first and rate-limiting enzyme of the de novo pathway, serine palmitoyltransferase (SPT). The intraventricular administration at reperfusion of myriocin, an inhibitor of SPT, significantly protected the area at risk from damage, reducing the infarcted area by 40.9 % relative to controls not treated with the drug. In the area at risk, myriocin downregulated ceramide, reduced the content in other mediators of inflammation and reactive oxygen species, and activated the Nrf2-HO1 cytoprotective response. We conclude that an enhanced ceramide synthesis takes part in ischemia/reperfusion injury and that myriocin treatment can be proposed as a strategy for myocardial pharmacological postconditioning.
Collapse
|
46
|
Stöhr R, Kappel BA, Carnevale D, Cavalera M, Mavilio M, Arisi I, Fardella V, Cifelli G, Casagrande V, Rizza S, Cattaneo A, Mauriello A, Menghini R, Lembo G, Federici M. TIMP3 interplays with apelin to regulate cardiovascular metabolism in hypercholesterolemic mice. Mol Metab 2015; 4:741-52. [PMID: 26500845 PMCID: PMC4588459 DOI: 10.1016/j.molmet.2015.07.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 07/23/2015] [Accepted: 07/27/2015] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Tissue inhibitor of metalloproteinase 3 (TIMP3) is an extracellular matrix (ECM) bound protein, which has been shown to be downregulated in human subjects and experimental models with cardiometabolic disorders, including type 2 diabetes mellitus, hypertension and atherosclerosis. The aim of this study was to investigate the effects of TIMP3 on cardiac energy homeostasis during increased metabolic stress conditions. METHODS ApoE(-/-)TIMP3(-/-) and ApoE(-/-) mice on a C57BL/6 background were subjected to telemetric ECG analysis and experimental myocardial infarction as models of cardiac stress induction. We used Western blot, qRT-PCR, histology, metabolomics, RNA-sequencing and in vivo phenotypical analysis to investigate the molecular mechanisms of altered cardiac energy metabolism. RESULTS ApoE(-/-)TIMP3(-/-) revealed decreased lifespan. Telemetric ECG analysis showed increased arrhythmic episodes, and experimental myocardial infarction by left anterior descending artery (LAD) ligation resulted in increased peri-operative mortality together with increased scar formation, ventricular dilatation and a reduction of cardiac function after 4 weeks in the few survivors. Hearts of ApoE(-/-)TIMP3(-/-) exhibited accumulation of neutral lipids when fed a chow diet, which was exacerbated by a high fat, high cholesterol diet. Metabolomics analysis revealed an increase in circulating markers of oxidative stress with a reduction in long chain fatty acids. Using whole heart mRNA sequencing, we identified apelin as a putative modulator of these metabolic defects. Apelin is a regulator of fatty acid oxidation, and we found a reduction in the levels of enzymes involved in fatty acid oxidation in the left ventricle of ApoE(-/-)TIMP3(-/-) mice. Injection of apelin restored the hitherto identified metabolic defects of lipid oxidation. CONCLUSION TIMP3 regulates lipid metabolism as well as oxidative stress response via apelin. These findings therefore suggest that TIMP3 maintains metabolic flexibility in the heart, particularly during episodes of increased cardiac stress.
Collapse
Affiliation(s)
- Robert Stöhr
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Internal Medicine I, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Ben Arpad Kappel
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Internal Medicine I, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Michele Cavalera
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Maria Mavilio
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ivan Arisi
- Genomics Facility, European Brain Research Institute, Rome, Italy
| | - Valentina Fardella
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | - Giuseppe Cifelli
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | - Viviana Casagrande
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Stefano Rizza
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute, Rome, Italy
- Scuola Normale Superiore, Pisa, Italy
| | - Alessandro Mauriello
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Center for Atherosclerosis, Department of Medicine, Policlinico Tor Vergata, 00133 Rome, Italy
- Corresponding author. Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy. Tel.: +39 06 72596889; fax: +39 06 72596890.
| |
Collapse
|
47
|
Rose JJ, Voora D, Cyr DD, Lucas JE, Zaas AK, Woods CW, Newby LK, Kraus WE, Ginsburg GS. Gene Expression Profiles Link Respiratory Viral Infection, Platelet Response to Aspirin, and Acute Myocardial Infarction. PLoS One 2015; 10:e0132259. [PMID: 26193668 PMCID: PMC4507878 DOI: 10.1371/journal.pone.0132259] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 06/12/2015] [Indexed: 01/09/2023] Open
Abstract
Background Influenza infection is associated with myocardial infarction (MI), suggesting that respiratory viral infection may induce biologic pathways that contribute to MI. We tested the hypotheses that 1) a validated blood gene expression signature of respiratory viral infection (viral GES) was associated with MI and 2) respiratory viral exposure changes levels of a validated platelet gene expression signature (platelet GES) of platelet function in response to aspirin that is associated with MI. Methods A previously defined viral GES was projected into blood RNA data from 594 patients undergoing elective cardiac catheterization and used to classify patients as having evidence of viral infection or not and tested for association with acute MI using logistic regression. A previously defined platelet GES was projected into blood RNA data from 81 healthy subjects before and after exposure to four respiratory viruses: Respiratory Syncytial Virus (RSV) (n=20), Human Rhinovirus (HRV) (n=20), Influenza A virus subtype H1N1 (H1N1) (n=24), Influenza A Virus subtype H3N2 (H3N2) (n=17). We tested for the change in platelet GES with viral exposure using linear mixed-effects regression and by symptom status. Results In the catheterization cohort, 32 patients had evidence of viral infection based upon the viral GES, of which 25% (8/32) had MI versus 12.2% (69/567) among those without evidence of viral infection (OR 2.3; CI [1.03-5.5], p=0.04). In the infection cohorts, only H1N1 exposure increased platelet GES over time (time course p-value = 1e-04). Conclusions A viral GES of non-specific, respiratory viral infection was associated with acute MI; 18% of the top 49 genes in the viral GES are involved with hemostasis and/or platelet aggregation. Separately, H1N1 exposure, but not exposure to other respiratory viruses, increased a platelet GES previously shown to be associated with MI. Together, these results highlight specific genes and pathways that link viral infection, platelet activation, and MI especially in the case of H1N1 influenza infection.
Collapse
Affiliation(s)
- Jason J. Rose
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Deepak Voora
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Derek D. Cyr
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Joseph E. Lucas
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Aimee K. Zaas
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Christopher W. Woods
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - L. Kristin Newby
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - William E. Kraus
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Geoffrey S. Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
48
|
Borodzicz S, Czarzasta K, Kuch M, Cudnoch-Jedrzejewska A. Sphingolipids in cardiovascular diseases and metabolic disorders. Lipids Health Dis 2015; 14:55. [PMID: 26076974 PMCID: PMC4470334 DOI: 10.1186/s12944-015-0053-y] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/01/2015] [Indexed: 12/11/2022] Open
Abstract
Many investigations suggest the pivotal role of sphingolipids in the pathogenesis of lifestyle diseases such as myocardial infarction, hypertension, stroke, diabetes mellitus type 2 and obesity. Some studies suggest that sphingolipids are important factors in cellular signal transduction. They serve as biologically active components of cell membrane and are involved in many processes such as proliferation, maturation and apoptosis. Recently, ceramide and sphingosine-1-phosphate have become the target of many investigations. Ceramide is generated in three metabolic pathways and many factors induce its production as a cellular stress response. Ceramide has proapoptotic properties and acts as a precursor for many other sphingolipids. Sphingosine-1-phosphate is a ceramide derivative, acting antiapoptotically and mitogenically and it is importantly involved in cardioprotection. Further research on the involvement of sphingolipids in cellular pathophysiology may improve the prevention and therapy of lifestyle diseases.
Collapse
Affiliation(s)
- Sonia Borodzicz
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, First Faculty of Medicine, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland. .,1st Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland.
| | - Katarzyna Czarzasta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, First Faculty of Medicine, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| | - Marek Kuch
- Department of Heart Failure and Cardiac Rehabilitation of the Chair and Department of Cardiology, Hypertension and Internal Diseases, Second Faculty of Medicine, Medical University of Warsaw, Kondratowicza 8, 03-242, Warsaw, Poland.
| | - Agnieszka Cudnoch-Jedrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, First Faculty of Medicine, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| |
Collapse
|
49
|
Charkiewicz K, Blachnio-Zabielska A, Zbucka-Kretowska M, Wolczynski S, Laudanski P. Maternal plasma and amniotic fluid sphingolipids profiling in fetal Down syndrome. PLoS One 2015; 10:e0127732. [PMID: 26000716 PMCID: PMC4441425 DOI: 10.1371/journal.pone.0127732] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 04/20/2015] [Indexed: 01/03/2023] Open
Abstract
Introduction Sphingolipids can be potentially involved in the formation of the central and peripheral nervous systems, which are particularly connected with the pathogenesis of Down syndrome. The aim of the study was to determine the concentration of selected sphingolipids in the plasma and amniotic fluid of pregnant patients with fetal Down syndrome. Material and Methods Out of 190 amniocentesis we had 10 patients with confirmed Down syndrome. For the purpose of our control we chose 14 women without confirmed chromosomal aberration. To assess the concentration of 11 sphingolipids in the blood plasma and amniotic fluid we used an ultra-high performance liquid chromatography coupled with triple quadrupole mass spectrometry (UHPLC/MS/MS). Results We showed a significant increase in the concentration of 2 ceramides, C22-Cer and C24:1-Cer, in the plasma of women with fetal Down syndrome. Furthermore we showed a decrease in the concentration of 7 ceramides—C16-Cer, C18-Cer, C18:1-Cer, C20-Cer, C22-Cer, C24:1-Cer, and C24-Cer—in the amniotic fluid of women with fetal Down syndrome. We created ROC curves for all significant sphingolipids in maternal plasma, which set the threshold values and allowed for predicting the likelihood of Down syndrome in the fetus with specific sensitivity and specificity. We demonstrated a significantly higher risk of Down syndrome when the plasma concentration of C22-Cer > 12.66 ng/100ul (sens. 0.9, sp. 0.79, P value = 0.0007) and C24:1-Cer > 33,19 ng/100ul (sens. 0.6, sp. 0.86, P value = 0.0194). Conclusion On the basis of our findings, it seems that the sphingolipids may play a role in the pathogenesis of Down syndrome. Defining their potential as biochemical markers of Down syndrome requires further investigation on a larger group of patients.
Collapse
Affiliation(s)
- Karol Charkiewicz
- Department of Perinatology and Obstetrics, Medical University of Bialystok, Marii Sklodowskiej-Curie 24a, Bialystok, 15-276, Poland
| | | | - Monika Zbucka-Kretowska
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Marii Sklodowskiej Curie 24a, Bialystok, 15-273, Poland
| | - Slawomir Wolczynski
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Marii Sklodowskiej Curie 24a, Bialystok, 15-273, Poland
| | - Piotr Laudanski
- Department of Perinatology and Obstetrics, Medical University of Bialystok, Marii Sklodowskiej-Curie 24a, Bialystok, 15-276, Poland
| |
Collapse
|
50
|
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: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|