1
|
Kitsou K, Kokkotis G, Rivera-Nieves J, Bamias G. Targeting the Sphingosine-1-Phosphate Pathway: New Opportunities in Inflammatory Bowel Disease Management. Drugs 2024; 84:1179-1197. [PMID: 39322927 DOI: 10.1007/s40265-024-02094-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
Inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC) are chronic immune-mediated diseases which primarily target the intestines. In recent years, the development and regulatory approval of various immunotherapies, both biological agents and small molecules, that target specific pathways of the IBD-associated inflammatory cascade have revolutionized the treatment of IBD. Small molecules offer the advantages of oral administration and short wash-out times. Sphingosine-1-phosphate (S1P) is a bioactive metabolite of ceramide, which exerts its functions after binding to five G-protein-coupled receptors (S1PR1-S1PR5). Concerning IBD, S1P participates in the egress of lymphocytes from the secondary lymphoid tissue and their re-circulation to sites of inflammation, mainly through S1PR1 binding. In addition, this system facilitates the differentiation of T-helper cells towards proinflammatory immunophenotypes. Recently, S1P modulators have offered a valuable addition to the IBD treatment armamentarium. They exert their anti-inflammatory function via sequestration of T cell subsets in the lymphoid tissues and prevention of gut homing. In this review, we revisit the role of the S1P/S1PR axis in the pathogenesis of IBD and discuss efficacy and safety data from clinical trials and real-world reports on the two S1PR modulators, ozanimod and etrasimod, that are currently approved for IBD treatment, and comment on their potential positioning in the IBD day-to-day management. We also present recent data on emerging S1P modulators. Finally, based on the successes and failures of S1PR modulators in IBD, we discuss future avenues of IBD treatments targeting the S1P/S1PR axis.
Collapse
Affiliation(s)
| | - Georgios Kokkotis
- GI-Unit, 3rd Department of Internal Medicine, Sotiria Hospital, 152 Mesogeion Av., 11528, Athens, Greece
| | - Jesús Rivera-Nieves
- San Diego VA Medical Center (SDVAMC), San Diego, CA, USA
- Division of Gastroenterology, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Giorgos Bamias
- GI-Unit, 3rd Department of Internal Medicine, Sotiria Hospital, 152 Mesogeion Av., 11528, Athens, Greece.
| |
Collapse
|
2
|
Bisgaard LS, Christensen PM, Oh J, Torta F, Füchtbauer EM, Nielsen LB, Christoffersen C. Kidney derived apolipoprotein M and its role in acute kidney injury. Front Pharmacol 2024; 15:1328259. [PMID: 38313311 PMCID: PMC10834784 DOI: 10.3389/fphar.2024.1328259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
Aim: Apolipoprotein M (apoM) is mainly expressed in liver and in proximal tubular epithelial cells in the kidney. In plasma, apoM associates with HDL particles via a retained signal peptide and carries sphingosine-1-phosphate (S1P), a small bioactive lipid. ApoM is undetectable in urine from healthy individuals but lack of megalin receptors in proximal tubuli cells induces loss of apoM into the urine. Besides this, very little is known about kidney-derived apoM. The aim of this study was to address the role of apoM in kidney biology and in acute kidney injury. Methods: A novel kidney-specific human apoM transgenic mouse model (RPTEC-hapoMTG) was generated and subjected to either cisplatin or ischemia/reperfusion injury. Further, a stable transfection of HK-2 cells overexpressing human apoM (HK-2-hapoMTG) was developed to study the pattern of apoM secretion in proximal tubuli cells. Results: Human apoM was present in plasma from RPTEC-hapoMTG mice (mean 0.18 μM), with a significant increase in plasma S1P levels. In vitro apoM was secreted to both the apical (urine) and basolateral (blood) compartment from proximal tubular epithelial cells. However, no differences in kidney injury score was seen between RPTEC-hapoMTG and wild type (WT) mice upon kidney injury. Further, gene expression of inflammatory markers (i.e., IL6, MCP-1) was similar upon ischemia/reperfusion injury. Conclusion: Our study suggests that kidney-derived apoM is secreted to plasma, supporting a role for apoM in sequestering molecules from excretion in urine. However, overexpression of human apoM in the kidney did not protect against acute kidney injury.
Collapse
Affiliation(s)
- Line S. Bisgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pernille M. Christensen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jeongah Oh
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Lars Bo Nielsen
- The Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
von Eckardstein A, Nordestgaard BG, Remaley AT, Catapano AL. High-density lipoprotein revisited: biological functions and clinical relevance. Eur Heart J 2022; 44:1394-1407. [PMID: 36337032 PMCID: PMC10119031 DOI: 10.1093/eurheartj/ehac605] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/16/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Previous interest in high-density lipoproteins (HDLs) focused on their possible protective role in atherosclerotic cardiovascular disease (ASCVD). Evidence from genetic studies and randomized trials, however, questioned that the inverse association of HDL-cholesterol (HDL-C) is causal. This review aims to provide an update on the role of HDL in health and disease, also beyond ASCVD. Through evolution from invertebrates, HDLs are the principal lipoproteins, while apolipoprotein B-containing lipoproteins first developed in vertebrates. HDLs transport cholesterol and other lipids between different cells like a reusable ferry, but serve many other functions including communication with cells and the inactivation of biohazards like bacterial lipopolysaccharides. These functions are exerted by entire HDL particles or distinct proteins or lipids carried by HDL rather than by its cholesterol cargo measured as HDL-C. Neither does HDL-C measurement reflect the efficiency of reverse cholesterol transport. Recent studies indicate that functional measures of HDL, notably cholesterol efflux capacity, numbers of HDL particles, or distinct HDL proteins are better predictors of ASCVD events than HDL-C. Low HDL-C levels are related observationally, but also genetically, to increased risks of infectious diseases, death during sepsis, diabetes mellitus, and chronic kidney disease. Additional, but only observational, data indicate associations of low HDL-C with various autoimmune diseases, and cancers, as well as all-cause mortality. Conversely, extremely high HDL-C levels are associated with an increased risk of age-related macular degeneration (also genetically), infectious disease, and all-cause mortality. HDL encompasses dynamic multimolecular and multifunctional lipoproteins that likely emerged during evolution to serve several physiological roles and prevent or heal pathologies beyond ASCVD. For any clinical exploitation of HDL, the indirect marker HDL-C must be replaced by direct biomarkers reflecting the causal role of HDL in the respective disease.
Collapse
Affiliation(s)
- Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich and University of Zurich , Zurich , Switzerland
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital , Herlev , Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital, Herlev and Gentofte Hospital , Herlev , Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, MD , USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan , Milan , Italy
- IRCCS MultiMedica, Sesto S. Giovanni , Milan , Italy
| |
Collapse
|
4
|
Cui GY, Rao BC, Zeng ZH, Wang XM, Ren T, Wang HY, Luo H, Ren HY, Liu C, Ding SY, Tan JJ, Liu ZG, Zou YW, Ren ZG, Yu ZJ. Characterization of oral and gut microbiome and plasma metabolomics in COVID-19 patients after 1-year follow-up. Mil Med Res 2022; 9:32. [PMID: 35715833 PMCID: PMC9204369 DOI: 10.1186/s40779-022-00387-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/26/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Due to the outbreak and rapid spread of coronavirus disease 2019 (COVID-19), more than 160 million patients have become convalescents worldwide to date. Significant alterations have occurred in the gut and oral microbiome and metabonomics of patients with COVID-19. However, it is unknown whether their characteristics return to normal after the 1-year recovery. METHODS We recruited 35 confirmed patients to provide specimens at discharge and one year later, as well as 160 healthy controls. A total of 497 samples were prospectively collected, including 219 tongue-coating, 129 stool and 149 plasma samples. Tongue-coating and stool samples were subjected to 16S rRNA sequencing, and plasma samples were subjected to untargeted metabolomics testing. RESULTS The oral and gut microbiome and metabolomics characteristics of the 1-year convalescents were restored to a large extent but did not completely return to normal. In the recovery process, the microbial diversity gradually increased. Butyric acid-producing microbes and Bifidobacterium gradually increased, whereas lipopolysaccharide-producing microbes gradually decreased. In addition, sphingosine-1-phosphate, which is closely related to the inflammatory factor storm of COVID-19, increased significantly during the recovery process. Moreover, the predictive models established based on the microbiome and metabolites of patients at the time of discharge reached high efficacy in predicting their neutralizing antibody levels one year later. CONCLUSIONS This study is the first to characterize the oral and gut microbiome and metabonomics in 1-year convalescents of COVID-19. The key microbiome and metabolites in the process of recovery were identified, and provided new treatment ideas for accelerating recovery. And the predictive models based on the microbiome and metabolomics afford new insights for predicting the recovery situation which benefited affected individuals and healthcare.
Collapse
Affiliation(s)
- Guang-Ying Cui
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Gene Hospital of Henan Province/Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ben-Chen Rao
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Gene Hospital of Henan Province/Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhao-Hai Zeng
- Department of Infectious Diseases, Guangshan County People's Hospital, Guangshan County, Xinyang, 465450, Henan, China
| | - Xue-Mei Wang
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Gene Hospital of Henan Province/Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tong Ren
- Department of Breast Surgery, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hai-Yu Wang
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Gene Hospital of Henan Province/Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hong Luo
- Department of Infectious Diseases, Guangshan County People's Hospital, Guangshan County, Xinyang, 465450, Henan, China
| | - Hong-Yan Ren
- Shanghai Mobio Biomedical Technology Co., Ltd, Shanghai, 201111, China
| | - Chao Liu
- Shanghai Mobio Biomedical Technology Co., Ltd, Shanghai, 201111, China
| | - Su-Ying Ding
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jun-Jie Tan
- Department of Infectious Diseases, Guangshan County People's Hospital, Guangshan County, Xinyang, 465450, Henan, China
| | - Zhen-Guo Liu
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Gene Hospital of Henan Province/Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ya-Wen Zou
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Gene Hospital of Henan Province/Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhi-Gang Ren
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Gene Hospital of Henan Province/Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Zu-Jiang Yu
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Gene Hospital of Henan Province/Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
5
|
Dong W, Wong KHY, Liu Y, Levy-Sakin M, Hung WC, Li M, Li B, Jin SC, Choi J, Lopez-Giraldez F, Vaka D, Poon A, Chu C, Lao R, Balamir M, Movsesyan I, Malloy MJ, Zhao H, Kwok PY, Kane JP, Lifton RP, Pullinger CR. Whole-exome sequencing reveals damaging gene variants associated with hypoalphalipoproteinemia. J Lipid Res 2022; 63:100209. [PMID: 35460704 PMCID: PMC9126845 DOI: 10.1016/j.jlr.2022.100209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 12/02/2022] Open
Abstract
Low levels of high density lipoprotein-cholesterol (HDL-C) are associated with an elevated risk of arteriosclerotic coronary heart disease. Heritability of HDL-C levels is high. In this research discovery study, we used whole-exome sequencing to identify damaging gene variants that may play significant roles in determining HDL-C levels. We studied 204 individuals with a mean HDL-C level of 27.8 ± 6.4 mg/dl (range: 4-36 mg/dl). Data were analyzed by statistical gene burden testing and by filtering against candidate gene lists. We found 120 occurrences of probably damaging variants (116 heterozygous; four homozygous) among 45 of 104 recognized HDL candidate genes. Those with the highest prevalence of damaging variants were ABCA1 (n = 20), STAB1 (n = 9), OSBPL1A (n = 8), CPS1 (n = 8), CD36 (n = 7), LRP1 (n = 6), ABCA8 (n = 6), GOT2 (n = 5), AMPD3 (n = 5), WWOX (n = 4), and IRS1 (n = 4). Binomial analysis for damaging missense or loss-of-function variants identified the ABCA1 and LDLR genes at genome-wide significance. In conclusion, whole-exome sequencing of individuals with low HDL-C showed the burden of damaging rare variants in the ABCA1 and LDLR genes is particularly high and revealed numerous occurrences in HDL candidate genes, including many genes identified in genome-wide association study reports. Many of these genes are involved in cancer biology, which accords with epidemiologic findings of the association of HDL deficiency with increased risk of cancer, thus presenting a new area of interest in HDL genomics.
Collapse
Affiliation(s)
- Weilai Dong
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Karen H Y Wong
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Youbin Liu
- Department of Cardiology, The Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Michal Levy-Sakin
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Wei-Chien Hung
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Mo Li
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Boyang Li
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Sheng Chih Jin
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jungmin Choi
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA; Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | | | - Dedeepya Vaka
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Annie Poon
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Catherine Chu
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Richard Lao
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Melek Balamir
- Department of Internal Medicine, Istanbul University, Istanbul, Turkey
| | - Irina Movsesyan
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Mary J Malloy
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Pui-Yan Kwok
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA; Department of Dermatology, University of California, San Francisco, CA, USA
| | - John P Kane
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Clive R Pullinger
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA; Physiological Nursing, University of California, San Francisco, CA, USA.
| |
Collapse
|
6
|
Duan M, Gao P, Chen SX, Novák P, Yin K, Zhu X. Sphingosine-1-phosphate in mitochondrial function and metabolic diseases. Obes Rev 2022; 23:e13426. [PMID: 35122459 DOI: 10.1111/obr.13426] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/02/2022] [Accepted: 01/02/2022] [Indexed: 01/23/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite. The past decade has witnessed exponential growth in the field of S1P research, partly attributed to drugs targeting its receptors or kinases. Accumulating evidence indicates that changes in the S1P axis (i.e., S1P production, transport, and receptors) may modify metabolism and eventually mediate metabolic diseases. Dysfunction of the mitochondria on a master monitor of cellular metabolism is considered the leading cause of metabolic diseases, with aberrations typically induced by abnormal biogenesis, respiratory chain complex disorders, reactive oxygen species overproduction, calcium deposition, and mitophagy impairment. Accordingly, we discuss decades of investigation into changes in the S1P axis and how it controls mitochondrial function. Furthermore, we summarize recent scientific advances in disorders associated with the S1P axis and their involvement in the pathogenesis of metabolic diseases in humans, including type 2 diabetes mellitus and cardiovascular disease, from the perspective of mitochondrial function. Finally, we review potential challenges and prospects for S1P axis application to the regulation of mitochondrial function and metabolic diseases; these data may provide theoretical guidance for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Meng Duan
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Pan Gao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Sheng-Xi Chen
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Kai Yin
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China.,Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
7
|
Xin JY, Huang X, Sun Y, Jiang HS, Fan J, Yu NW, Guo FQ, Ye F, Xiao J, Le WD, Yang SJ, Xiang Y. Association Between Plasma Apolipoprotein M With Alzheimer’s Disease: A Cross-Sectional Pilot Study From China. Front Aging Neurosci 2022; 14:838223. [PMID: 35370599 PMCID: PMC8973919 DOI: 10.3389/fnagi.2022.838223] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/17/2022] [Indexed: 12/19/2022] Open
Abstract
BackgroundRecent evidence of genetics and metabonomics indicated a potential role of apolipoprotein M (ApoM) in the pathogenesis of Alzheimer’s disease (AD). Here, we aimed to investigate the association between plasma ApoM with AD.MethodsA multicenter, cross-sectional study recruited patients with AD (n = 67), age- and sex-matched cognitively normal (CN) controls (n = 73). After the data collection of demographic characteristics, lifestyle risk factors, and medical history, we examined and compared the plasma levels of ApoM, tau phosphorylated at threonine 217 (p-tau217) and neurofilament light (NfL). Multivariate logistic regression analysis was applied to determine the association of plasma ApoM with the presence of AD. The correlation analysis was used to explore the correlations between plasma ApoM with cognitive function [Mini–Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA)], activities of daily living (ADL), and the representative blood-based biomarkers (plasma p-tau217 and NfL). Receiver operating characteristic (ROC) analysis and Delong’s test were used to determine the diagnostic power of plasma ApoM.ResultsPlasma ApoM and its derived indicators (ratios of ApoM/TC, ApoM/TG, ApoM/HDL-C, and ApoM/LDL-C) were significantly higher in AD group than those in CN group (each p < 0.0001). After adjusted for the risk factors of AD, the plasma ApoM and its derived indicators were significantly associated with the presence of AD, respectively. ApoM (OR = 1.058, 95% CI: 1.027–1.090, p < 0.0001), ApoM/TC ratio (OR = 1.239, 95% CI: 1.120–1.372, p < 0.0001), ApoM/TG ratio (OR = 1.064, 95% CI: 1.035–1.095, p < 0.0001), ApoM/HDL-C ratio (OR = 1.069, 95% CI: 1.037–1.102, p < 0.0001), and ApoM/LDL-C ratio (OR = 1.064, 95% CI:1.023–1.106, p = 0.002). In total participants, plasma ApoM was significantly positively correlated with plasma p-tau217, plasma NfL, and ADL (each p < 0.0001) and significantly negatively correlated with MMSE and MoCA (each p < 0.0001), respectively. In further subgroup analyses, these associations remained in different APOEϵ 4 status participants and sex subgroups. ApoM/TC ratio (ΔAUC = 0.056, p = 0.044) and ApoM/TG ratio (ΔAUC = 0.097, p = 0.011) had a statistically remarkably larger AUC than ApoM, respectively. The independent addition of ApoM and its derived indicators to the basic model [combining age, sex, APOEϵ 4, and body mass index (BMI)] led to the significant improvement in diagnostic power, respectively (each p < 0.05).ConclusionAll the findings preliminarily uncovered the association between plasma ApoM and AD and provided more evidence of the potential of ApoM as a candidate biomarker of AD.
Collapse
Affiliation(s)
- Jia-Yan Xin
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
- Department of Neurology, General Hospital of Western Theater Command, Chengdu, China
| | - Xiao Huang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
- Department of Neurology, General Hospital of Western Theater Command, Chengdu, China
| | - Ying Sun
- Department of Geriatrics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hai-Song Jiang
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Jin Fan
- Department of Neurology, General Hospital of Western Theater Command, Chengdu, China
| | - Neng-wei Yu
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fu-Qiang Guo
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fang Ye
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Xiao
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei-dong Le
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Shao-Jie Yang
- Department of Neurology, Chengdu Eighth People’s Hospital, Chengdu, China
- *Correspondence: Shao-Jie Yang,
| | - Yang Xiang
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Yang Xiang,
| |
Collapse
|
8
|
Diab A, Valenzuela Ripoll C, Guo Z, Javaheri A. HDL Composition, Heart Failure, and Its Comorbidities. Front Cardiovasc Med 2022; 9:846990. [PMID: 35350538 PMCID: PMC8958020 DOI: 10.3389/fcvm.2022.846990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Although research on high-density lipoprotein (HDL) has historically focused on atherosclerotic coronary disease, there exists untapped potential of HDL biology for the treatment of heart failure. Anti-oxidant, anti-inflammatory, and endothelial protective properties of HDL could impact heart failure pathogenesis. HDL-associated proteins such as apolipoprotein A-I and M may have significant therapeutic effects on the myocardium, in part by modulating signal transduction pathways and sphingosine-1-phosphate biology. Furthermore, because heart failure is a complex syndrome characterized by multiple comorbidities, there are complex interactions between heart failure, its comorbidities, and lipoprotein homeostatic mechanisms. In this review, we will discuss the effects of heart failure and associated comorbidities on HDL, explore potential cardioprotective properties of HDL, and review novel HDL therapeutic targets in heart failure.
Collapse
|
9
|
Bisgaard LS, Christoffersen C. The apoM/S1P Complex-A Mediator in Kidney Biology and Disease? Front Med (Lausanne) 2021; 8:754490. [PMID: 34722589 PMCID: PMC8553247 DOI: 10.3389/fmed.2021.754490] [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: 08/06/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022] Open
Abstract
Kidney disease affects more than 10% of the population, can be both acute and chronic, and is linked to other diseases such as cardiovascular disease, diabetes, and sepsis. Despite the detrimental consequences for patients, no good treatment options directly targeting the kidney are available. Thus, a better understanding of the pathology and new treatment modalities are required. Accumulating evidence suggests that the apolipoprotein M/sphingosine-1-phosphate (apoM/S1P) axis is a likely drug target, but significant gaps in our knowledge remain. In this review, we present what has so far been elucidated about the role of apoM in normal kidney biology and describe how changes in the apoM/S1P axis are thought to affect the development of kidney disease. ApoM is primarily produced in the liver and kidneys. From the liver, apoM is secreted into circulation, where it is attached to lipoproteins (primarily HDL). Importantly, apoM is a carrier of the bioactive lipid S1P. S1P acts by binding to five different receptors. Together, apoM/S1P plays a role in several biological mechanisms, such as inflammation, endothelial cell permeability, and lipid turnover. In the kidney, apoM is primarily expressed in the proximal tubular cells. S1P can be produced locally in the kidney, and several of the five S1P receptors are present in the kidney. The functional role of kidney-derived apoM as well as plasma-derived apoM is far from elucidated and will be discussed based on both experimental and clinical studies. In summary, the current studies provide evidence that support a role for the apoM/S1P axis in kidney disease; however, additional pre-clinical and clinical studies are needed to reveal the mechanisms and target potential in the treatment of patients.
Collapse
Affiliation(s)
- Line S Bisgaard
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Velagapudi S, Rohrer L, Poti F, Feuerborn R, Perisa D, Wang D, Panteloglou G, Potapenko A, Yalcinkaya M, Hülsmeier AJ, Hesse B, Lukasz A, Liu M, Parks JS, Christoffersen C, Stoffel M, Simoni M, Nofer JR, von Eckardstein A. Apolipoprotein M and Sphingosine-1-Phosphate Receptor 1 Promote the Transendothelial Transport of High-Density Lipoprotein. Arterioscler Thromb Vasc Biol 2021; 41:e468-e479. [PMID: 34407633 PMCID: PMC8458249 DOI: 10.1161/atvbaha.121.316725] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective: ApoM enriches S1P (sphingosine-1-phosphate) within HDL (high-density lipoproteins) and facilitates the activation of the S1P1 (S1P receptor type 1) by S1P, thereby preserving endothelial barrier function. Many protective functions exerted by HDL in extravascular tissues raise the question of how S1P regulates transendothelial HDL transport. Approach and Results: HDL were isolated from plasma of wild-type mice, Apom knockout mice, human apoM transgenic mice or humans and radioiodinated to trace its binding, association, and transport by bovine or human aortic endothelial cells. We also compared the transport of fluorescently-labeled HDL or Evans Blue, which labels albumin, from the tail vein into the peritoneal cavity of apoE-haploinsufficient mice with (apoE-haploinsufficient mice with endothelium-specific knockin of S1P1) or without (control mice, ie, apoE-haploinsufficient mice without endothelium-specific knockin of S1P1) endothelium-specific knockin of S1P1. The binding, association, and transport of HDL from Apom knockout mice and human apoM-depleted HDL by bovine aortic endothelial cells was significantly lower than that of HDL from wild-type mice and human apoM-containing HDL, respectively. The binding, uptake, and transport of 125I-HDL by human aortic endothelial cells was increased by an S1P1 agonist but decreased by an S1P1 inhibitor. Silencing of SR-BI (scavenger receptor BI) abrogated the stimulation of 125I-HDL transport by the S1P1 agonist. Compared with control mice, that is, apoE-haploinsufficient mice without endothelium-specific knockin of S1P1, apoE-haploinsufficient mice with endothelium-specific knockin of S1P1 showed decreased transport of Evans Blue but increased transport of HDL from blood into the peritoneal cavity and SR-BI expression in the aortal endothelium. Conclusions: ApoM and S1P1 promote transendothelial HDL transport. Their opposite effect on transendothelial transport of albumin and HDL indicates that HDL passes endothelial barriers by specific mechanisms rather than passive filtration.
Collapse
Affiliation(s)
- Srividya Velagapudi
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Francesco Poti
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, Italy (F.P.)
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy (F.P., M. Simoni, J.-R.N.)
| | - Renate Feuerborn
- Central Laboratory Facility, University Hospital of Münster, Germany (R.F., J.-R.N.)
| | - Damir Perisa
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Dongdong Wang
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Grigorios Panteloglou
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Anton Potapenko
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Andreas J Hülsmeier
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Bettina Hesse
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, Germany (B.H., A.L.)
| | - Alexander Lukasz
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, Germany (B.H., A.L.)
| | - Mingxia Liu
- Department of Internal Medicine/Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (M.L., J.S.P.)
| | - John S Parks
- Department of Internal Medicine/Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (M.L., J.S.P.)
| | - Christina Christoffersen
- Department of Biomedical Science, University of Copenhagen, Denmark (C.C.)
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (C.C.)
| | - Markus Stoffel
- Institute of Molecular Health Sciences, ETH Zurich, Switzerland (M. Stoffel)
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy (F.P., M. Simoni, J.-R.N.)
| | - Jerzy-Roch Nofer
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy (F.P., M. Simoni, J.-R.N.)
- Central Laboratory Facility, University Hospital of Münster, Germany (R.F., J.-R.N.)
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (J.-R.N.)
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| |
Collapse
|
11
|
Patanapirunhakit P, Karlsson H, Mulder M, Ljunggren S, Graham D, Freeman D. Sphingolipids in HDL - Potential markers for adaptation to pregnancy? Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158955. [PMID: 33933650 DOI: 10.1016/j.bbalip.2021.158955] [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: 01/20/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 11/15/2022]
Abstract
Plasma high density lipoprotein (HDL) exhibits many functions that render it an effective endothelial protective agent and may underlie its potential role in protecting the maternal vascular endothelium during pregnancy. In non-pregnant individuals, the HDL lipidome is altered in metabolic disease compared to healthy individuals and is linked to reduced cholesterol efflux, an effect that can be reversed by lifestyle management. Specific sphingolipids such as sphingosine-1-phosphate (S1P) have been shown to mediate the vaso-dilatory effects of plasma HDL via interaction with the endothelial nitric oxide synthase pathway. This review describes the relationship between plasma HDL and vascular function during healthy pregnancy and details how this is lost in pre-eclampsia, a disorder of pregnancy associated with widespread endothelial dysfunction. Evidence of a role for HDL sphingolipids, in particular S1P and ceramide, in cardiovascular disease and in healthy pregnancy and pre-eclampsia is discussed. Available data suggest that HDL-S1P and HDL-ceramide can mediate vascular protection in healthy pregnancy but not in preeclampsia. HDL sphingolipids thus are of potential importance in the healthy maternal adaptation to pregnancy.
Collapse
Affiliation(s)
- Patamat Patanapirunhakit
- Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand; Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| | - Helen Karlsson
- Occupational and Environmental Medicine Center, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Monique Mulder
- Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Stefan Ljunggren
- Occupational and Environmental Medicine Center, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| | - Dilys Freeman
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
12
|
Cheng G, Zheng L. Regulation of the apolipoprotein M signaling pathway: a review. J Recept Signal Transduct Res 2021; 42:285-292. [PMID: 34006168 DOI: 10.1080/10799893.2021.1924203] [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] [Indexed: 12/17/2022]
Abstract
Apolipoprotein M (apoM), an apolipoprotein predominantly associated with high-density lipoprotein (HDL), is considered a mediator of the numerous roles of HDL, including reverse cholesterol transport, anti-atherosclerotic, anti-inflammatory and anti-oxidant, and mediates pre-β-HDL formation. ApoM expression is known to be regulated by a variety of in vivo and in vitro factors. The transcription factors farnesoid X receptor, small heterodimer partner, liver receptor homolog-1, and liver X receptor comprise the signaling cascade network that regulates the expression and secretion of apoM. Moreover, hepatocyte nuclear factor-1α and c-Jun/JunB have been demonstrated to exert opposing regulatory effects on apoM through competitive binding to the same sites in the proximal region of the apoM gene. Furthermore, as a carrier and modulator of sphingosine 1-phosphate (S1P), apoM binds to S1P within its hydrophobic-binding pocket. The apoM/S1P axis has been discovered to play a crucial role in the apoM signaling pathway through its ability to regulate glucose and lipid metabolism, vascular barrier homeostasis, inflammatory response and other pathological and physiological processes. Using the findings of previous studies, the present review aimed to summarize the regulation of apoM expression by various factors and its role in different physiological and pathological conditions, and provide a new perspective for the further treatment of these diseases.
Collapse
Affiliation(s)
- Gangli Cheng
- Clinical Medical Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lu Zheng
- Clinical Medical Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
13
|
Wigger D, Schumacher F, Schneider-Schaulies S, Kleuser B. Sphingosine 1-phosphate metabolism and insulin signaling. Cell Signal 2021; 82:109959. [PMID: 33631318 DOI: 10.1016/j.cellsig.2021.109959] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022]
Abstract
Insulin is the main anabolic hormone secreted by β-cells of the pancreas stimulating the assimilation and storage of glucose in muscle and fat cells. It modulates the postprandial balance of carbohydrates, lipids and proteins via enhancing lipogenesis, glycogen and protein synthesis and suppressing glucose generation and its release from the liver. Resistance to insulin is a severe metabolic disorder related to a diminished response of peripheral tissues to the insulin action and signaling. This leads to a disturbed glucose homeostasis that precedes the onset of type 2 diabetes (T2D), a disease reaching epidemic proportions. A large number of studies reported an association between elevated circulating fatty acids and the development of insulin resistance. The increased fatty acid lipid flux results in the accumulation of lipid droplets in a variety of tissues. However, lipid intermediates such as diacylglycerols and ceramides are also formed in response to elevated fatty acid levels. These bioactive lipids have been associated with the pathogenesis of insulin resistance. More recently, sphingosine 1-phosphate (S1P), another bioactive sphingolipid derivative, has also been shown to increase in T2D and obesity. Although many studies propose a protective role of S1P metabolism on insulin signaling in peripheral tissues, other studies suggest a causal role of S1P on insulin resistance. In this review, we critically summarize the current state of knowledge of S1P metabolism and its modulating role on insulin resistance. A particular emphasis is placed on S1P and insulin signaling in hepatocytes, skeletal muscle cells, adipocytes and pancreatic β-cells. In particular, modulation of receptors and enzymes that regulate S1P metabolism can be considered as a new therapeutic option for the treatment of insulin resistance and T2D.
Collapse
Affiliation(s)
- Dominik Wigger
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany
| | | | - Burkhard Kleuser
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany.
| |
Collapse
|
14
|
Chun J, Giovannoni G, Hunter SF. Sphingosine 1-phosphate Receptor Modulator Therapy for Multiple Sclerosis: Differential Downstream Receptor Signalling and Clinical Profile Effects. Drugs 2021; 81:207-231. [PMID: 33289881 PMCID: PMC7932974 DOI: 10.1007/s40265-020-01431-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Lysophospholipids are a class of bioactive lipid molecules that produce their effects through various G protein-coupled receptors (GPCRs). Sphingosine 1-phosphate (S1P) is perhaps the most studied lysophospholipid and has a role in a wide range of physiological and pathophysiological events, via signalling through five distinct GPCR subtypes, S1PR1 to S1PR5. Previous and continuing investigation of the S1P pathway has led to the approval of three S1PR modulators, fingolimod, siponimod and ozanimod, as medicines for patients with multiple sclerosis (MS), as well as the identification of new S1PR modulators currently in clinical development, including ponesimod and etrasimod. S1PR modulators have complex effects on S1PRs, in some cases acting both as traditional agonists as well as agonists that produce functional antagonism. S1PR subtype specificity influences their downstream effects, including aspects of their benefit:risk profile. Some S1PR modulators are prodrugs, which require metabolic modification such as phosphorylation via sphingosine kinases, resulting in different pharmacokinetics and bioavailability, contrasting with others that are direct modulators of the receptors. The complex interplay of these characteristics dictates the clinical profile of S1PR modulators. This review focuses on the S1P pathway, the characteristics and S1PR binding profiles of S1PR modulators, the mechanisms of action of S1PR modulators with regard to immune cell trafficking and neuroprotection in MS, together with a summary of the clinical effectiveness of the S1PR modulators that are approved or in late-stage development for patients with MS. Sphingosine 1-phosphate receptor modulator therapy for multiple sclerosis: differential downstream receptor signalling and clinical profile effects (MP4 65540 kb).
Collapse
Affiliation(s)
- Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark St, London, E1 2AT UK
| | - Samuel F. Hunter
- Advanced Neurosciences Institute, 101 Forrest Crossing Blvd STE 103, Franklin, TN 37064 USA
| |
Collapse
|
15
|
Marfia G, Navone S, Guarnaccia L, Campanella R, Mondoni M, Locatelli M, Barassi A, Fontana L, Palumbo F, Garzia E, Ciniglio Appiani G, Chiumello D, Miozzo M, Centanni S, Riboni L. Decreased serum level of sphingosine-1-phosphate: a novel predictor of clinical severity in COVID-19. EMBO Mol Med 2021; 13:e13424. [PMID: 33190411 PMCID: PMC7744841 DOI: 10.15252/emmm.202013424] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
The severity of coronavirus disease 2019 (COVID-19) is a crucial problem in patient treatment and outcome. The aim of this study is to evaluate circulating level of sphingosine-1-phosphate (S1P) along with severity markers, in COVID-19 patients. One hundred eleven COVID-19 patients and forty-seven healthy subjects were included. The severity of COVID-19 was found significantly associated with anemia, lymphocytopenia, and significant increase of neutrophil-to-lymphocyte ratio, ferritin, fibrinogen, aminotransferases, lactate dehydrogenase (LDH), C-reactive protein (CRP), and D-dimer. Serum S1P level was inversely associated with COVID-19 severity, being significantly correlated with CRP, LDH, ferritin, and D-dimer. The decrease in S1P was strongly associated with the number of erythrocytes, the major source of plasma S1P, and both apolipoprotein M and albumin, the major transporters of blood S1P. Not last, S1P was found to be a relevant predictor of admission to an intensive care unit, and patient's outcome. Circulating S1P emerged as negative biomarker of severity/mortality of COVID-19 patients. Restoring abnormal S1P levels to a normal range may have the potential to be a therapeutic target in patients with COVID-19.
Collapse
Affiliation(s)
- Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Istituto di Medicina Aerospaziale "A. Mosso"Aeronautica MilitareMilanItaly
- Aldo Ravelli” Research CenterMilanItaly
| | - Stefania Navone
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Aldo Ravelli” Research CenterMilanItaly
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Michele Mondoni
- Respiratory UnitASST Santi Paolo e CarloDepartment of Health SciencesUniversità degli Studi di MilanoMilanItaly
| | - Marco Locatelli
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Aldo Ravelli” Research CenterMilanItaly
- Department of Medical‐Surgical Physiopathology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Alessandra Barassi
- Laboratory of Clinical BiochemistryASST Santi Paolo e CarloDepartment of Health SciencesUniversità degli Studi di MilanoMilanItaly
| | - Laura Fontana
- Department of Medical‐Surgical Physiopathology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Fabrizio Palumbo
- Istituto di Medicina Aerospaziale "A. Mosso"Aeronautica MilitareMilanItaly
| | - Emanuele Garzia
- Istituto di Medicina Aerospaziale "A. Mosso"Aeronautica MilitareMilanItaly
- Reproductive Medicine UnitASST Santi Paolo e CarloUniversità degli Studi di MilanoMilanItaly
| | | | | | - Monica Miozzo
- Department of Medical‐Surgical Physiopathology and TransplantationUniversità degli Studi di MilanoMilanItaly
- Unit of Research Laboratories CoordinationFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Stefano Centanni
- Respiratory UnitASST Santi Paolo e CarloDepartment of Health SciencesUniversità degli Studi di MilanoMilanItaly
| | - Laura Riboni
- Department of Medical Biotechnology and Translational MedicineLITA‐Segrate, Università degli Studi di MilanoMilanItaly
| |
Collapse
|
16
|
Zhong F, Guan L, Lin H, Zhao M, Qin Y, Li Q, Yuan Z, Yang G, Gao L, Zhao J. Red Blood Cell Count: An Unrecognized Risk Factor for Nonalcoholic Fatty Liver Disease. Front Endocrinol (Lausanne) 2021; 12:760981. [PMID: 34950103 PMCID: PMC8688742 DOI: 10.3389/fendo.2021.760981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD) is becoming a global public health challenge. A convenient NAFLD indicator will greatly facilitate risk appraisal and prevention. As a readily available and inexpensive hematological index in routine clinical examinations, red blood cells (RBCs) are gaining increasing attention in many diseases, such as metabolic syndrome, but their association with NAFLD is unknown. METHODS This health management cohort study included 27,112 subjects (17,383 non-NAFLD and 9,729 NAFLD) with up to 5 years of follow-up (median 2.8 years). NAFLD was diagnosed by ultrasonography. NAFLD severity was categorized as mild, moderate, or severe. The generalized estimation equation (GEE), an extension of generalized linear models that allows for analysis of repeated measurements, was used to analyze the association between RBC count and NAFLD. RESULTS Overall, 4,332 of 17,383 (24.9%) subjects without NAFLD at baseline developed NAFLD. Incident NAFLD risk was positively associated with RBC count. After adjustment for hemoglobin and other confounders, the risk of incident NAFLD was 21%, 32%, and 51% higher in the second, third, and fourth RBC count quartiles, respectively, than in the lowest quartile. In 1,798 of 9,476 (19.0%) subjects with NAFLD at baseline, the severity of NAFLD increased. NAFLD progression risk increased progressively as RBC count increased (P for trend < 0.001). Every one-unit (1012 cells/L) increase in RBC count was associated with a 53% [OR 1.53 (95% CI 1.32-1.77)] increased risk for NAFLD progression. CONCLUSIONS Elevated RBC count was independently associated with a high risk of NAFLD incidence and progression. This finding revealed a convenient NAFLD risk indicator.
Collapse
Affiliation(s)
- Fang Zhong
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
| | - Liying Guan
- Health Management Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haiyan Lin
- Health Management Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Meng Zhao
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yiming Qin
- College of Chemical Engineering and Materials Science, Shandong Normal University, Jinan, China
| | - Qihang Li
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, China
| | - Guang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Jiajun Zhao,
| |
Collapse
|
17
|
Papadopoulos C, Panopoulou M, Anagnostopoulos K, Tentes I. Immune and Metabolic Interactions of Human Erythrocytes: A Molecular Perspective. Endocr Metab Immune Disord Drug Targets 2020; 21:843-853. [PMID: 33148159 DOI: 10.2174/1871530320666201104115016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 11/22/2022]
Abstract
Apart from their main function as oxygen carriers in vertebrates, erythrocytes are also involved in immune regulation. By circulating throughout the body, the erythrocytes are exposed and interact with tissues that are damaged as a result of a disease. In this study, we summarize the literature regarding the contribution of erythrocytes to immune regulation and metabolism. Under the circumstances of a disease state, the erythrocytes may lose their antioxidant capacity and release Damage Associated Molecular Patterns, resulting in the regulation of innate and adaptive immunity. In addition, the erythrocytes scavenge and affect the levels of chemokines, circulating cell-free mtDNA, and C3b attached immune complexes. Furthermore, through surface molecules, erythrocytes control the function of T lymphocytes, macrophages, and dendritic cells. Through an array of enzymes, red blood cells contribute to the pool of blood's bioactive lipids. Finally, the erythrocytes contribute to reverse cholesterol transport through various mechanisms. Our study is highlighting overlooked molecular interactions between erythrocytes and immunity and metabolism, which could lead to the discovery of potent therapeutic targets for immunometabolic diseases.
Collapse
Affiliation(s)
| | - Maria Panopoulou
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Ioannis Tentes
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
18
|
Emerging roles of lysophospholipids in health and disease. Prog Lipid Res 2020; 80:101068. [PMID: 33068601 DOI: 10.1016/j.plipres.2020.101068] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 12/22/2022]
Abstract
Lipids are abundant and play essential roles in human health and disease. The main functions of lipids are building blocks for membrane biogenesis. However, lipids are also metabolized to produce signaling molecules. Here, we discuss the emerging roles of circulating lysophospholipids. These lysophospholipids consist of lysoglycerophospholipids and lysosphingolipids. They are both present in cells at low concentration, but their concentrations in extracellular fluids are significantly higher. The biological functions of some of these lysophospholipids have been recently revealed. Remarkably, some of the lysophospholipids play pivotal signaling roles as well as being precursors for membrane biogenesis. Revealing how circulating lysophospholipids are produced, released, transported, and utilized in multi-organ systems is critical to understand their functions. The discovery of enzymes, carriers, transporters, and membrane receptors for these lysophospholipids has shed light on their physiological significance. In this review, we summarize the biological roles of these lysophospholipids via discussing about the proteins regulating their functions. We also discuss about their potential impacts to human health and diseases.
Collapse
|
19
|
Crivelli SM, Giovagnoni C, Visseren L, Scheithauer AL, de Wit N, den Hoedt S, Losen M, Mulder MT, Walter J, de Vries HE, Bieberich E, Martinez-Martinez P. Sphingolipids in Alzheimer's disease, how can we target them? Adv Drug Deliv Rev 2020; 159:214-231. [PMID: 31911096 DOI: 10.1016/j.addr.2019.12.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/09/2019] [Accepted: 12/31/2019] [Indexed: 01/06/2023]
Abstract
Altered levels of sphingolipids and their metabolites in the brain, and the related downstream effects on neuronal homeostasis and the immune system, provide a framework for understanding mechanisms in neurodegenerative disorders and for developing new intervention strategies. In this review we will discuss: the metabolites of sphingolipids that function as second messengers; and functional aberrations of the pathway resulting in Alzheimer's disease (AD) pathophysiology. Focusing on the central product of the sphingolipid pathway ceramide, we describ approaches to pharmacologically decrease ceramide levels in the brain and we argue on how the sphingolipid pathway may represent a new framework for developing novel intervention strategies in AD. We also highlight the possible use of clinical and non-clinical drugs to modulate the sphingolipid pathway and sphingolipid-related biological cascades.
Collapse
|
20
|
Druggable Sphingolipid Pathways: Experimental Models and Clinical Opportunities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:101-135. [PMID: 32894509 DOI: 10.1007/978-3-030-50621-6_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Intensive research in the field of sphingolipids has revealed diverse roles in cell biological responses and human health and disease. This immense molecular family is primarily represented by the bioactive molecules ceramide, sphingosine, and sphingosine 1-phosphate (S1P). The flux of sphingolipid metabolism at both the subcellular and extracellular levels provides multiple opportunities for pharmacological intervention. The caveat is that perturbation of any single node of this highly regulated flux may have effects that propagate throughout the metabolic network in a dramatic and sometimes unexpected manner. Beginning with S1P, the receptors for which have thus far been the most clinically tractable pharmacological targets, this review will describe recent advances in therapeutic modulators targeting sphingolipids, their chaperones, transporters, and metabolic enzymes.
Collapse
|
21
|
Diarte-Añazco EMG, Méndez-Lara KA, Pérez A, Alonso N, Blanco-Vaca F, Julve J. Novel Insights into the Role of HDL-Associated Sphingosine-1-Phosphate in Cardiometabolic Diseases. Int J Mol Sci 2019; 20:ijms20246273. [PMID: 31842389 PMCID: PMC6940915 DOI: 10.3390/ijms20246273] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023] Open
Abstract
Sphingolipids are key signaling molecules involved in the regulation of cell physiology. These species are found in tissues and in circulation. Although they only constitute a small fraction in lipid composition of circulating lipoproteins, their concentration in plasma and distribution among plasma lipoproteins appears distorted under adverse cardiometabolic conditions such as diabetes mellitus. Sphingosine-1-phosphate (S1P), one of their main representatives, is involved in regulating cardiomyocyte homeostasis in different models of experimental cardiomyopathy. Cardiomyopathy is a common complication of diabetes mellitus and represents a main risk factor for heart failure. Notably, plasma concentration of S1P, particularly high-density lipoprotein (HDL)-bound S1P, may be decreased in patients with diabetes mellitus, and hence, inversely related to cardiac alterations. Despite this, little attention has been given to the circulating levels of either total S1P or HDL-bound S1P as potential biomarkers of diabetic cardiomyopathy. Thus, this review will focus on the potential role of HDL-bound S1P as a circulating biomarker in the diagnosis of main cardiometabolic complications frequently associated with systemic metabolic syndromes with impaired insulin signaling. Given the bioactive nature of these molecules, we also evaluated its potential of HDL-bound S1P-raising strategies for the treatment of cardiometabolic disease.
Collapse
Affiliation(s)
- Elena M. G. Diarte-Añazco
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
| | - Karen Alejandra Méndez-Lara
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| | - Antonio Pérez
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei d’Endocrinologia, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Núria Alonso
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei d’Endocrinologia, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Francisco Blanco-Vaca
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| | - Josep Julve
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| |
Collapse
|
22
|
Sposito AC, de Lima-Junior JC, Moura FA, Barreto J, Bonilha I, Santana M, Virginio VW, Sun L, Carvalho LSF, Soares AA, Nadruz W, Feinstein SB, Nofer JR, Zanotti I, Kontush A, Remaley AT. Reciprocal Multifaceted Interaction Between HDL (High-Density Lipoprotein) and Myocardial Infarction. Arterioscler Thromb Vasc Biol 2019; 39:1550-1564. [DOI: 10.1161/atvbaha.119.312880] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite decades of therapeutic advances, myocardial infarction remains a leading cause of death worldwide. Recent studies have identified HDLs (high-density lipoproteins) as a potential candidate for mitigating coronary ischemia/reperfusion injury via a broad spectrum of signaling pathways. HDL ligands, such as S1P (sphingosine-1-phosphate), Apo (apolipoprotein) A-I, clusterin, and miRNA, may influence the opening of the mitochondrial channel, insulin sensitivity, and production of vascular autacoids, such as NO, prostacyclin, and endothelin-1. In parallel, antioxidant activity and sequestration of oxidized molecules provided by HDL can attenuate the oxidative stress that triggers ischemia/reperfusion. Nevertheless, during myocardial infarction, oxidation and the capture of oxidized and proinflammatory molecules generate large phenotypic and functional changes in HDL, potentially limiting its beneficial properties. In this review, new findings from cellular and animal models, as well as from clinical studies, will be discussed to describe the cardioprotective benefits of HDL on myocardial infarction. Furthermore, mechanisms by which HDL modulates cardiac function and potential strategies to mitigate postmyocardial infarction risk damage by HDL will be detailed throughout the review.
Collapse
Affiliation(s)
- Andrei C. Sposito
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - José Carlos de Lima-Junior
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Filipe A. Moura
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
- Department of Medicine, Weill-Cornell Medical College, New York, NY (F.A.M.)
| | - Joaquim Barreto
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Isabella Bonilha
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Michele Santana
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Vitor W. Virginio
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Lufan Sun
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (L.S., A.T.R.)
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China (L.S.)
| | - Luiz Sergio F. Carvalho
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Alexandre A.S. Soares
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Wilson Nadruz
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Steve B. Feinstein
- Division of Cardiology, Rush University Medical Center, Chicago, IL (S.B.F.)
| | - Jerzy-Roch Nofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (J.-R.N.)
| | - Ilaria Zanotti
- Department of Food and Drugs, University of Parma, Italy (I.Z.)
| | - Anatol Kontush
- UMR-ICAN 1166, National Institute for Health and Medical Research (INSERM), Sorbonne University, Paris, France (A.K.)
| | - Alan T. Remaley
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (L.S., A.T.R.)
| |
Collapse
|
23
|
Bisgaard LS, Christoffersen C. Apolipoprotein M/sphingosine-1-phosphate: novel effects on lipids, inflammation and kidney biology. Curr Opin Lipidol 2019; 30:212-217. [PMID: 31008738 DOI: 10.1097/mol.0000000000000606] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In 2011, the crystal structure of apolipoprotein M (apoM) and its capacity to bind sphingosine-1-phosphate (S1P) was characterized. Since then, a variety of studies has increased our knowledge on apoM biology and functionality. From being an unknown and hardly significant player in overall metabolism, apoM has gained significant interest. RECENT FINDINGS Key discoveries in the last 2 years have indicated that the apoM/S1P complex has important roles in lipid metabolism (affecting triglyceride turnover), inflammation (a marker of severe sepsis and potentially providing anti-inflammatory signaling) and kidney biology (potential to protect against immunoglobulin A nephropathy). SUMMARY Several studies suggest a potential for apoM/S1P as biomarkers for inflammation, sepsis and nephropathy. Also, a novel chaperone is characterized and could have potential as a drug for treatment in inflammation and nephropathy.
Collapse
Affiliation(s)
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet
- Department of Clinical Biochemistry, Bispebjerg Hospital
- Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
24
|
Brunkhorst R, Pfeilschifter W, Rajkovic N, Pfeffer M, Fischer C, Korf HW, Christoffersen C, Trautmann S, Thomas D, Pfeilschifter J, Koch A. Diurnal regulation of sphingolipids in blood. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:304-311. [DOI: 10.1016/j.bbalip.2018.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 11/12/2018] [Accepted: 12/09/2018] [Indexed: 01/30/2023]
|
25
|
Biological function of SPNS2: From zebrafish to human. Mol Immunol 2018; 103:55-62. [DOI: 10.1016/j.molimm.2018.08.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 01/01/2023]
|
26
|
Dihydro-sphingosine 1-phosphate interacts with carrier proteins in a manner distinct from that of sphingosine 1-phosphate. Biosci Rep 2018; 38:BSR20181288. [PMID: 30279204 PMCID: PMC6209608 DOI: 10.1042/bsr20181288] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/17/2018] [Accepted: 09/26/2018] [Indexed: 01/02/2023] Open
Abstract
Dihydro-sphingosine 1-phosphate (DH-S1P) is an analog of sphingosine 1-phosphate (S1P), which is a potent lysophospholipid mediator. DH-S1P has been proposed to exert physiological properties similar to S1P. Although S1P is known to be carried on HDL via apolipoprotein M (apoM), the association between DH-S1P and HDL/apoM has not been fully elucidated. Therefore, in the present study, we aimed to elucidate this association and to compare it with that of S1P and HDL/apoM. First, we investigated the distributions of S1P and DH-S1P among lipoproteins and lipoprotein-depleted fractions in human serum and plasma samples and observed that both S1P and DH-S1P were detected on HDL; furthermore, elevated amounts of DH-S1P in serum samples were distributed to the lipoprotein-depleted fraction to a greater degree than to the HDL fraction. Concordantly, a preference for HDL over albumin was only observed for S1P, and not for DH-S1P, when the molecules were secreted from platelets. Regarding the association with HDL, although both S1P and DH-S1P prefer to bind to HDL, HDL preferentially accepts S1P over DH-S1P. For the association with apoM, S1P was not detected on HDL obtained from apoM knockout mice, while DH-S1P was detected. Moreover, apoM retarded the degradation of S1P, but not of DH-S1P. These results suggest that S1P binds to HDL via apoM, while DH-S1P binds to HDL in a non-specific manner. Thus, DH-S1P is not a mere analog of S1P and might possess unique clinical significance.
Collapse
|
27
|
MFSD2B is a sphingosine 1-phosphate transporter in erythroid cells. Sci Rep 2018; 8:4969. [PMID: 29563527 PMCID: PMC5862976 DOI: 10.1038/s41598-018-23300-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/02/2018] [Indexed: 12/22/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is an intercellular signaling molecule present in blood. Erythrocytes have a central role in maintaining the S1P concentration in the blood stream. We previously demonstrated that S1P is exported from erythrocytes by a glyburide-sensitive S1P transporter. However, the gene encoding the S1P transporter in erythrocytes is unknown. In this study, we found that the mouse erythroid cell line, MEDEP-E14, has S1P export activity and exhibits properties that are consistent with those of erythrocytes. Using microarray analysis of MEDEP-E14 cells and its parental cell line, E14TG2a, we identified several candidate genes for S1P export activity. Of those genes, only one gene, Mfsd2b, showed S1P transport activity. The properties of S1P release by MFSD2B were similar to those in erythrocytes. Moreover, knockout of MFSD2B in MEDEP-E14 cells decreased S1P export from the cells. These results strongly suggest that MFSD2B is a novel S1P transporter in erythroid cells.
Collapse
|
28
|
Kleuser B. Divergent Role of Sphingosine 1-Phosphate in Liver Health and Disease. Int J Mol Sci 2018; 19:ijms19030722. [PMID: 29510489 PMCID: PMC5877583 DOI: 10.3390/ijms19030722] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/08/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022] Open
Abstract
Two decades ago, sphingosine 1-phosphate (S1P) was discovered as a novel bioactive molecule that regulates a variety of cellular functions. The plethora of S1P-mediated effects is due to the fact that the sphingolipid not only modulates intracellular functions but also acts as a ligand of G protein-coupled receptors after secretion into the extracellular environment. In the plasma, S1P is found in high concentrations, modulating immune cell trafficking and vascular endothelial integrity. The liver is engaged in modulating the plasma S1P content, as it produces apolipoprotein M, which is a chaperone for the S1P transport. Moreover, the liver plays a substantial role in glucose and lipid homeostasis. A dysfunction of glucose and lipid metabolism is connected with the development of liver diseases such as hepatic insulin resistance, non-alcoholic fatty liver disease, or liver fibrosis. Recent studies indicate that S1P is involved in liver pathophysiology and contributes to the development of liver diseases. In this review, the current state of knowledge about S1P and its signaling in the liver is summarized with a specific focus on the dysregulation of S1P signaling in obesity-mediated liver diseases. Thus, the modulation of S1P signaling can be considered as a potential therapeutic target for the treatment of hepatic diseases.
Collapse
Affiliation(s)
- Burkhard Kleuser
- Department of Toxicology, Institute of Nutritional Science, Faculty of Mathematics and Natural Science, University of Potsdam, Arthur-Scheunert Allee 114-116, 14558 Nuthetal, Germany.
| |
Collapse
|