1
|
Mathias RA, Velkoska E, Didichenko SA, Greene BH, Tan X, Navdaev AV, Collins HL, Adelman SJ, Young K, Gille A, Duffy D, Gibson CM, Pelzing M, Kingwell BA. Apolipoprotein A1 (CSL112) Increases Lecithin-Cholesterol Acyltransferase Levels in HDL Particles and Promotes Reverse Cholesterol Transport. JACC Basic Transl Sci 2025; 10:405-418. [PMID: 40306849 DOI: 10.1016/j.jacbts.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 05/02/2025]
Abstract
Although high-density lipoprotein (HDL) cholesterol is inversely correlated with cardiovascular risk, an emerging paradigm is focused on increasing reverse cholesterol transport (RCT) and HDL function via apolipoprotein A1 (ApoA1). The objective of this study was to investigate the effect of ApoA1 (CSL112) infusion on HDL protein composition, cholesterol esterification rate (CER), and cholesterol efflux capacity (CEC) in patients treated after acute myocardial infarction. CSL112 reduced levels of apolipoproteins A2, B, C, and E and serum amyloids A1 and A4, whereas ApoA1, ApoM, and lecithin-cholesterol acyltransferase were significantly elevated. Increased CEC, plasma HDL cholesterol levels, CER, and CEC also were observed in CSL112-treated patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - C Michael Gibson
- PERFUSE Study Group, Boston and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
2
|
Stadler JT, Borenich A, Stattau Bisgaard L, Bjergfelt SS, Vijayakumar S, Melholt L, Emrich IE, Hansen D, Bro S, Christoffersen C, Heine GH, Marsche G. ApoM and Major Adverse Cardiovascular Events in Chronic Kidney Disease: A Prospective Cohort Study. Arterioscler Thromb Vasc Biol 2025; 45:496-505. [PMID: 40047074 PMCID: PMC11936471 DOI: 10.1161/atvbaha.124.322367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Cardiovascular disease is the leading cause of mortality in patients with chronic kidney disease (CKD). APOM plays a critical role in reverse cholesterol transport by facilitating the formation of pre-β-HDL (high-density lipoprotein) and enabling the binding of S1P (sphingosine-1-phosphate) to HDL, a complex involved in several antiatherogenic processes. In this study, we sought to investigate the potential association between plasma APOM levels and the risk of adverse cardiovascular outcomes in individuals with CKD. METHODS Plasma APOM levels were quantified using a sandwich ELISA-based assay. Plasma S1P levels were measured by high-performance liquid chromatography. The primary end point was a composite of major adverse cardiovascular events (MACE) and all-cause mortality. RESULTS In this secondary analysis of the CARE FOR HOMe study (Cardiovascular and Renal Outcome in CKD 2-4 Patients-The Fourth Homburg Evaluation), 463 nondialysis patients with CKD stages G2 to G4 were included. Plasma APOM levels exhibited a significant inverse association with the risk of MACE (standardized hazard ratio, 0.60 [95% CI, 0.49-0.75]; P<0.001) and all-cause mortality (standardized hazard ratio, 0.63 [95% CI, 0.48-0.83]; P<0.001). This inverse association with MACE remained robust after adjusting for established cardiovascular and renal risk factors. These findings were further corroborated in an independent cohort of 822 patients with CKD from the Copenhagen CKD study. Plasma S1P levels showed an inverse association with MACE in univariable analyses; however, this relationship lost statistical significance after multivariable adjustments. CONCLUSIONS Our findings demonstrate a significant association between low plasma APOM levels and an increased risk of MACE in patients with CKD. These results suggest that APOM may play a role in cardiovascular protection in this vulnerable population.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation (J.T.S., G.M.), Medical University of Graz, Austria
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Austria (J.T.S.)
| | - Andrea Borenich
- Institute for Medical Informatics, Statistics and Documentation (A.B.), Medical University of Graz, Austria
| | - Line Stattau Bisgaard
- Departments of Clinical Biochemistry (L.S.B., S.V., L.M., C.C.), Copenhagen University Hospital, Rigshospitalet, Denmark
- Departments of Biomedical Sciences (L.S.B., S.S.B., C.C.), University of Copenhagen, Denmark
| | - Sasha S. Bjergfelt
- Nephrology (S.S.B., S.B.), Copenhagen University Hospital, Rigshospitalet, Denmark
- Departments of Biomedical Sciences (L.S.B., S.S.B., C.C.), University of Copenhagen, Denmark
| | - Sarunja Vijayakumar
- Departments of Clinical Biochemistry (L.S.B., S.V., L.M., C.C.), Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Line Melholt
- Departments of Clinical Biochemistry (L.S.B., S.V., L.M., C.C.), Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Insa E. Emrich
- Faculty of Medicine, Saarland University, Homburg/Saarbrücken, Germany (I.E.E., G.H.H.)
| | - Ditte Hansen
- Clinical Medicine (D.H.), University of Copenhagen, Denmark
- Department of Nephrology, Copenhagen University Hospital, Herlev-Gentofte, Denmark (D.H.)
| | - Susanne Bro
- Nephrology (S.S.B., S.B.), Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Christina Christoffersen
- Departments of Clinical Biochemistry (L.S.B., S.V., L.M., C.C.), Copenhagen University Hospital, Rigshospitalet, Denmark
- Departments of Biomedical Sciences (L.S.B., S.S.B., C.C.), University of Copenhagen, Denmark
| | - Gunnar H. Heine
- Faculty of Medicine, Saarland University, Homburg/Saarbrücken, Germany (I.E.E., G.H.H.)
- Department of Nephrology, Agaplesion Markus Krankenhaus, Frankfurt am Main, Germany (G.H.H.)
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation (J.T.S., G.M.), Medical University of Graz, Austria
| |
Collapse
|
3
|
Alidjan FM, Hoedt SD, Rashid M, Zee-van Vark LCVD, Voortman GJ, Dorst-Lagerwerf KY, Christoffersen C, Lafeber M, Roeters van Lennep JE, Verhoeven AJM, Friesema ECH, Mulder MT. Triglyceride-rich lipoprotein sphingolipids are altered in primary hypertension: A pilot case-control study. J Clin Lipidol 2025:S1933-2874(25)00067-4. [PMID: 40263067 DOI: 10.1016/j.jacl.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Sphingolipids modulate vascular function and alterations in plasma sphingolipid profiles have been associated with hypertension. Plasma sphingolipids, such as ceramides (Cer) and sphingosine-1-phosphate (S1P), are predominantly carried by lipoproteins. OBJECTIVE We compared sphingolipid profiles in plasma and isolated lipoproteins of patients with primary hypertension with those of normotensive controls. METHODS Blood was obtained from 19 patients with hypertension and 19 age- and sex-matched normotensive controls. S1P and the 7 most abundant Cer were quantified by liquid chromatography-tandem mass spectrometry in plasma and in lipoproteins. RESULTS Total plasma Cer were significantly higher in patients with hypertension compared to controls (14.3 ± 1.0 vs 11.9 ± 0.7 µM; P = .047), while there were no differences in plasma S1P levels (1.8 ± 0.1 vs 2.1 ± 0.1 µM; P = .128). Total Cer carried by patient triglyceride-rich lipoproteins (TRL; ie, predominantly very low-density lipoproteins) were also significantly higher (1.33 ± 0.15 vs 0.58 ± 0.10 µM; P = .001), which held for all Cer tested. Systolic blood pressure positively correlated with plasma levels of Cer(d18:1/20:0) and Cer(d18:1/24:1), and diastolic blood pressure positively correlated with total Cer, Cer(d18:1/18:0), Cer(d18:1/20:0) and Cer(d18:1/24:0). Relative to plasma Cer(d18:1/24:0), levels of Cer(d18:1/18:0), Cer(d18:1/20:0) and Cer(d18:1/24:1) were significantly higher in patients with hypertension than in controls. CONCLUSION Patients with hypertension display higher plasma Cer levels than normotensive controls, which is mainly explained by elevated concentrations in TRLs. Cer levels positively correlate with systolic and diastolic blood pressure, and ratios of Cer relative to Cer(d18:1/24:0) suggest an increased cardiovascular risk.
Collapse
Affiliation(s)
- Fazil M Alidjan
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder); Department of Internal Vascular Medicine, Haga Hospital, the Hague, the Netherlands (Dr Alidjan)
| | - Sandra den Hoedt
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Mardin Rashid
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Leonie C van der Zee-van Vark
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Gardi J Voortman
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Kristien Y Dorst-Lagerwerf
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (Dr Christoffersen)
| | - Melvin Lafeber
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder); Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands (Dr Lafeber)
| | - Jeanine E Roeters van Lennep
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Adrie J M Verhoeven
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Edith C H Friesema
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder).
| | - Monique T Mulder
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| |
Collapse
|
4
|
Nielsen VW, Bundgaard Vad O, Holgersen N, Paludan-Müller C, Meseguer Monfort L, Beyer AF, Jemec GBE, Kjærsgaard Andersen R, Egeberg A, Thyssen JP, Svendsen JH, Rosenø NAL, Hansen PR, Thomsen SF, Salling Olesen M. Genetic Susceptibility to Hidradenitis Suppurativa and Predisposition to Cardiometabolic Disease. JAMA Dermatol 2025; 161:22-30. [PMID: 39382891 PMCID: PMC11465120 DOI: 10.1001/jamadermatol.2024.3779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/06/2024] [Indexed: 10/10/2024]
Abstract
Importance Hidradenitis suppurativa (HS) is associated with an increased prevalence of cardiovascular diseases compared with the general population. Any association between polygenic risk for HS, risk of incident cardiometabolic outcomes, and the plasma proteome is unclear. Objective To investigate the genetic correlation between HS and cardiometabolic disease. Design, Setting, and Participants This cohort study used a polygenic risk score (PRS) for HS to examine the risks of coronary artery disease (CAD) and diabetes and identify changes in the plasma proteome in individuals of European ancestry from the UK Biobank. Participants were enrolled from January 1, 2006, to December 31, 2010. End of follow-up was January 1, 2023. Correlations were assessed between HS susceptibility and cardiometabolic traits using linkage disequilibrium score regression. Odds ratios were assessed in logistic regressions. The risk of incident CAD and diabetes was estimated in cause-specific survival models designed as time-to-event analyses. Exposure The PRS for HS. Main Outcomes and Measures Main outcomes were CAD and diabetes diagnosis measured by logistic regressions and incident disease measured by Cox proportional hazards regression models adjusted for sex, age, body mass index, and smoking status. Results The study included 391 481 individuals (median [IQR] age, 58 [51-64] years; 209 235 [53%] female). Genetic variants for HS correlated significantly with variants associated with CAD, diabetes, and plasma levels of high-density lipoprotein cholesterol, triglycerides, and C-reactive protein. Compared with the low-risk group, a high PRS for HS (≥75th percentile) conferred odds ratios of 1.09 (95% CI, 1.06-1.12; P < .001) for CAD and 1.13 (95% CI, 1.10-1.17; P < .001) for diabetes. Estimates remained consistent when examining only incident CAD and diabetes. The PRS for HS was significantly associated with altered expression of 58 plasma proteins. Integrating this proteomic profile and the PRS for HS in a machine learning model improved prediction of CAD and diabetes compared with a reference model based on sex, age, and body mass index. Conclusions and Relevance These findings suggest that a high genetic risk of HS is associated with increased risk of subsequent CAD and diabetes and altered composition of the plasma proteome. Additional investigation into the identified proteins and their potential roles as drug targets is warranted.
Collapse
Affiliation(s)
- Valdemar Wendelboe Nielsen
- Department of Dermato-Venereology and Wound Healing Centre, Copenhagen University Hospital–Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Oliver Bundgaard Vad
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nikolaj Holgersen
- Department of Dermato-Venereology and Wound Healing Centre, Copenhagen University Hospital–Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Christian Paludan-Müller
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Laia Meseguer Monfort
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Astrid Filt Beyer
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gregor Borut Ernst Jemec
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rune Kjærsgaard Andersen
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark
- Department of Immunology and Microbiology, Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Egeberg
- Department of Dermato-Venereology and Wound Healing Centre, Copenhagen University Hospital–Bispebjerg and Frederiksberg, Copenhagen, Denmark
- LEO Pharma, Ballerup, Denmark
| | - Jacob P. Thyssen
- Department of Dermato-Venereology and Wound Healing Centre, Copenhagen University Hospital–Bispebjerg and Frederiksberg, Copenhagen, Denmark
- LEO Pharma, Ballerup, Denmark
| | - Jesper Hastrup Svendsen
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nana Aviaaja Lippert Rosenø
- Department of Dermato-Venereology and Wound Healing Centre, Copenhagen University Hospital–Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Peter Riis Hansen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital–Herlev and Gentofte, Hellerup, Denmark
| | - Simon Francis Thomsen
- Department of Dermato-Venereology and Wound Healing Centre, Copenhagen University Hospital–Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Salling Olesen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Jones Q, Zheng J, Li Z, He M, Li X, Dai K, Worgall TS, Yu Y, Jiang XC. Effect of phospholipid transfer protein on plasma sphingosine-1-phosphate. J Biol Chem 2024; 300:107837. [PMID: 39343001 PMCID: PMC11532956 DOI: 10.1016/j.jbc.2024.107837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Plasma phospholipid transfer protein (PLTP) is a risk factor for cardiovascular diseases. Sphingosine-1-phosphate (S1P), carried by high-density lipoprotein (HDL), is a potent lipid mediator and is also associated with cardiovascular diseases. We found that germline Pltp gene knockout (KO) mice have decreased circulating S1P without influencing apoM, a major S1P carrier on HDL. We then hypothesized that, like apoM, PLTP is another S1P carrier. We established inducible Pltp-KO, Apom-KO, and Pltp/Apom double KO mice and measured plasma lipoprotein and S1P levels under different diets. We found that PLTP deficiency, and the double deficiency have a similar effect on HDL reduction. Importantly, we found that all mice have about 50% reduction in plasma S1P levels, compared to WT mice, and PLTP deficiency significantly reduces apoM levels (about 40%), while apoM deficiency has no effect on PLTP activity, indicating that PLTP depletion reduces S1P through HDL reduction. To further evaluate this HDL reduction-mediated effect, we overexpressed PLTP which also caused a reduction of HDL. We found that the overexpression reduces S1P and apoM as well as apoA-I, a major apolipoprotein on HDL. Furthermore, we found that albumin (another reported S1P carrier) deficiency in mice has no effect on plasma S1P. We also found that the influence of PLTP on HDL may not require its direct binding to the particle. In conclusion, PLTP is not a direct S1P carrier. PLTP depletion or overexpression in adulthood dramatically reduces plasma S1P through HDL reduction. ApoM, but not albumin, deficiency reduces plasma S1P levels.
Collapse
Affiliation(s)
- Quiana Jones
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Jiao Zheng
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Zhiqiang Li
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Mulin He
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Xiang Li
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Kezhi Dai
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Tilla S Worgall
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Yang Yu
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA.
| |
Collapse
|
6
|
Bhale AS, Meilhac O, d'Hellencourt CL, Vijayalakshmi MA, Venkataraman K. Cholesterol transport and beyond: Illuminating the versatile functions of HDL apolipoproteins through structural insights and functional implications. Biofactors 2024; 50:922-956. [PMID: 38661230 DOI: 10.1002/biof.2057] [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: 01/18/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
High-density lipoproteins (HDLs) play a vital role in lipid metabolism and cardiovascular health, as they are intricately involved in cholesterol transport and inflammation modulation. The proteome of HDL particles is indeed complex and distinct from other components in the bloodstream. Proteomics studies have identified nearly 285 different proteins associated with HDL; however, this review focuses more on the 15 or so traditionally named "apo" lipoproteins. Important lipid metabolizing enzymes closely working with the apolipoproteins are also discussed. Apolipoproteins stand out for their integral role in HDL stability, structure, function, and metabolism. The unique structure and functions of each apolipoprotein influence important processes such as inflammation regulation and lipid metabolism. These interactions also shape the stability and performance of HDL particles. HDLs apolipoproteins have multifaceted roles beyond cardiovascular diseases (CVDs) and are involved in various physiological processes and disease states. Therefore, a detailed exploration of these apolipoproteins can offer valuable insights into potential diagnostic markers and therapeutic targets. This comprehensive review article aims to provide an in-depth understanding of HDL apolipoproteins, highlighting their distinct structures, functions, and contributions to various physiological processes. Exploiting this knowledge holds great potential for improving HDL function, enhancing cholesterol efflux, and modulating inflammatory processes, ultimately benefiting individuals by limiting the risks associated with CVDs and other inflammation-based pathologies. Understanding the nature of all 15 apolipoproteins expands our knowledge of HDL metabolism, sheds light on their pathological implications, and paves the way for advancements in the diagnosis, prevention, and treatment of lipid and inflammatory-related disorders.
Collapse
Affiliation(s)
- Aishwarya Sudam Bhale
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Olivier Meilhac
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | - Christian Lefebvre d'Hellencourt
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | | | - Krishnan Venkataraman
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
7
|
Ikeda T, Komiyama H, Miyakuni T, Takano M, Asai K. Exploring Possible Links: Thigh Muscle Mass, Apolipoproteins, and Glucose Metabolism in Peripheral Artery Disease-Insights from a Pilot Sub-Study following Endovascular Treatment. Metabolites 2024; 14:192. [PMID: 38668320 PMCID: PMC11052193 DOI: 10.3390/metabo14040192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Peripheral artery disease (PAD) compromises walking and physical activity, which results in further loss of skeletal muscle. The cross-sectional area of the thigh muscle has been shown to be correlated with systemic skeletal muscle volume. In our previous pilot study, we observed an increase in thigh muscle mass following endovascular treatment (EVT) in patients with proximal vascular lesions affecting the aortoiliac and femoropopliteal arteries. Considering the potential interactions between skeletal muscle, lipid profile, and glucose metabolism, we aimed to investigate the relationship between thigh muscle mass and apolipoproteins as well as glucose metabolism in PAD patients undergoing EVT. This study is a prespecified sub-study conducted as part of a pilot study. We prospectively enrolled 22 symptomatic patients with peripheral artery disease (PAD) and above-the-knee lesions, specifically involving the blood vessels supplying the thigh muscle. The mid-thigh muscle area was measured with computed tomography before and 6 months after undergoing EVT. Concurrently, we measured levels of apolipoproteins A1 (Apo A1) and B (Apo B), fasting blood glucose, 2 h post-load blood glucose (using a 75 g oral glucose tolerance test), and glycated hemoglobin A1c (HbA1c). Changes in thigh muscle area (delta muscle area: 2.5 ± 8.1 cm2) did not show significant correlations with changes in Apo A1, Apo B, fasting glucose, 2 h post-oral glucose tolerance test blood glucose, HbA1c, or Rutherford classification. However, among patients who experienced an increase in thigh muscle area following EVT (delta muscle area: 8.41 ± 5.93 cm2), there was a significant increase in Apo A1 (pre: 121.8 ± 15.1 mg/dL, 6 months: 136.5 ± 19.5 mg/dL, p < 0.001), while Apo B remained unchanged (pre: 76.4 ± 19.2 mg/dL, 6 months: 80.5 ± 4.9 mg/dL). Additionally, post-oral glucose tolerance test 2 h blood glucose levels showed a decrease (pre: 189.7 ± 67.5 mg/dL, 6 months: 170.6 ± 69.7 mg/dL, p = 0.075). Patients who exhibited an increase in thigh muscle area demonstrated more favorable metabolic changes compared to those with a decrease in thigh muscle area (delta muscle area: -4.67 ± 2.41 cm2). This pilot sub-study provides insights into the effects of EVT on thigh muscle, apolipoproteins, and glucose metabolism in patients with PAD and above-the-knee lesions. Further studies are warranted to validate these findings and establish their clinical significance. The trial was registered on the University Hospital Medical Information Network Clinical Trials Registry (UMIN000047534).
Collapse
Affiliation(s)
- Takeshi Ikeda
- Cardiovascular Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (T.I.); (K.A.)
| | - Hidenori Komiyama
- Cardiovascular Medicine, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan
| | - Tomoyo Miyakuni
- Cardiovascular Medicine, Nippon Medical School Chiba Hokusoh Hospital, Chiba 270-1613, Japan; (T.M.)
| | - Masamichi Takano
- Cardiovascular Medicine, Nippon Medical School Chiba Hokusoh Hospital, Chiba 270-1613, Japan; (T.M.)
| | - Kuniya Asai
- Cardiovascular Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (T.I.); (K.A.)
| |
Collapse
|
8
|
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
|
9
|
Abstract
Epidemiologic studies detected an inverse relationship between HDL (high-density lipoprotein) cholesterol (HDL-C) levels and atherosclerotic cardiovascular disease (ASCVD), identifying HDL-C as a major risk factor for ASCVD and suggesting atheroprotective functions of HDL. However, the role of HDL-C as a mediator of risk for ASCVD has been called into question by the failure of HDL-C-raising drugs to reduce cardiovascular events in clinical trials. Progress in understanding the heterogeneous nature of HDL particles in terms of their protein, lipid, and small RNA composition has contributed to the realization that HDL-C levels do not necessarily reflect HDL function. The most examined atheroprotective function of HDL is reverse cholesterol transport, whereby HDL removes cholesterol from plaque macrophage foam cells and delivers it to the liver for processing and excretion into bile. Indeed, in several studies, HDL has shown inverse associations between HDL cholesterol efflux capacity and ASCVD in humans. Inflammation plays a key role in the pathogenesis of atherosclerosis and vulnerable plaque formation, and a fundamental function of HDL is suppression of inflammatory signaling in macrophages and other cells. Oxidation is also a critical process to ASCVD in promoting atherogenic oxidative modifications of LDL (low-density lipoprotein) and cellular inflammation. HDL and its proteins including apoAI (apolipoprotein AI) and PON1 (paraoxonase 1) prevent cellular oxidative stress and LDL modifications. Importantly, HDL in humans with ASCVD is oxidatively modified rendering HDL dysfunctional and proinflammatory. Modification of HDL with reactive carbonyl species, such as malondialdehyde and isolevuglandins, dramatically impairs the antiatherogenic functions of HDL. Importantly, treatment of murine models of atherosclerosis with scavengers of reactive dicarbonyls improves HDL function and reduces systemic inflammation, atherosclerosis development, and features of plaque instability. Here, we discuss the HDL antiatherogenic functions in relation to oxidative modifications and the potential of reactive dicarbonyl scavengers as a therapeutic approach for ASCVD.
Collapse
Affiliation(s)
- MacRae F. Linton
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
- 2. Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Patricia G. Yancey
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Huan Tao
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Sean S. Davies
- 2. Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
10
|
Drexler Y, Molina J, Elfassy T, Ma R, Christoffersen C, Kurano M, Yatomi Y, Mariani LH, Contreras G, Merscher S, Fornoni A. Identification of Glomerular and Plasma Apolipoprotein M as Novel Biomarkers in Glomerular Disease. Kidney Int Rep 2023; 8:884-897. [PMID: 37069998 PMCID: PMC10105063 DOI: 10.1016/j.ekir.2023.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Dysregulation of sphingolipid and cholesterol metabolism contributes to the pathogenesis of glomerular diseases (GDs). Apolipoprotein M (ApoM) promotes cholesterol efflux and modulates the bioactive sphingolipid sphingosine-1-phosphate (S1P). Glomerular ApoM expression is decreased in patients with focal segmental glomerulosclerosis (FSGS). We hypothesized that glomerular ApoM deficiency occurs in GD and that ApoM expression and plasma ApoM correlate with outcomes. Methods Patients with GD from the Nephrotic Syndrome Study Network (NEPTUNE) were studied. We compared glomerular mRNA expression of ApoM (gApoM), sphingosine kinase 1 (SPHK1), and S1P receptors 1 to 5 (S1PR1-5) in patients (n = 84) and controls (n = 6). We used correlation analyses to determine associations between gApoM, baseline plasma ApoM (pApoM), and urine ApoM (uApoM/Cr). We used linear regression to determine whether gApoM, pApoM, and uApoM/Cr were associated with baseline estimated glomerular filtration rate (eGFR) and proteinuria. Using Cox models, we determined whether gApoM, pApoM, and uApoM/Cr were associated with complete remission (CR) and the composite of end-stage kidney disease (ESKD) or ≥40% eGFR decline. Results gApoM was reduced (P < 0.01) and SPHK1 and S1PR1 to 5 expression was increased (P < 0.05) in patients versus controls, consistent with ApoM/S1P pathway modulation. gApoM positively correlated with pApoM in the overall cohort (r = 0.34, P < 0.01) and in the FSGS (r = 0.48, P < 0.05) and minimal change disease (MCD) (r = 0.75, P < 0.05) subgroups. Every unit decrease in gApoM and pApoM (log2) was associated with a 9.77 ml/min per 1.73 m2 (95% confidence interval [CI]: 3.96-15.57) and 13.26 ml/min per 1.73 m2 (95% CI: 3.57-22.96) lower baseline eGFR, respectively (P < 0.01). From Cox models adjusted for age, sex, or race, pApoM was a significant predictor of CR (hazard ratio [HR]: 1.85; 95% CI: 1.06-3.23). Conclusions pApoM is a potential noninvasive biomarker of gApoM deficiency and strongly associates with clinical outcomes in GD.
Collapse
Affiliation(s)
- Yelena Drexler
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Judith Molina
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Tali Elfassy
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ruixuan Ma
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Christina Christoffersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Laura H. Mariani
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Gabriel Contreras
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
11
|
Yang HH, Wang X, Li S, Liu Y, Akbar R, Fan GC. Lipocalin family proteins and their diverse roles in cardiovascular disease. Pharmacol Ther 2023; 244:108385. [PMID: 36966973 PMCID: PMC10079643 DOI: 10.1016/j.pharmthera.2023.108385] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023]
Abstract
The lipocalin (LCN) family members, a group of small extracellular proteins with 160-180 amino acids in length, can be detected in all kingdoms of life from bacteria to human beings. They are characterized by low similarity of amino acid sequence but highly conserved tertiary structures with an eight-stranded antiparallel β-barrel which forms a cup-shaped ligand binding pocket. In addition to bind small hydrophobic ligands (i.e., fatty acids, odorants, retinoids, and steroids) and transport them to specific cells, lipocalins (LCNs) can interact with specific cell membrane receptors to activate their downstream signaling pathways, and with soluble macromolecules to form the complex. Consequently, LCNs exhibit great functional diversity. Accumulating evidence has demonstrated that LCN family proteins exert multiple layers of function in the regulation of many physiological processes and human diseases (i.e., cancers, immune disorders, metabolic disease, neurological/psychiatric disorders, and cardiovascular disease). In this review, we firstly introduce the structural and sequence properties of LCNs. Next, six LCNs including apolipoprotein D (ApoD), ApoM, lipocalin 2 (LCN2), LCN10, retinol-binding protein 4 (RBP4), and Lipocalin-type prostaglandin D synthase (L-PGDS) which have been characterized so far are highlighted for their diagnostic/prognostic values and their potential effects on coronary artery disease and myocardial infarction injury. The roles of these 6 LCNs in cardiac hypertrophy, heart failure, diabetes-induced cardiac disorder, and septic cardiomyopathy are also summarized. Finally, their therapeutic potential for cardiovascular disease is discussed in each section.
Collapse
Affiliation(s)
- Hui-Hui Yang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xiaohong Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Siru Li
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yueying Liu
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rubab Akbar
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
12
|
Guo Z, Valenzuela Ripoll C, Picataggi A, Rawnsley DR, Ozcan M, Chirinos JA, Chendamarai E, Girardi A, Riehl T, Evie H, Diab A, Kovacs A, Hyrc K, Ma X, Asnani A, Shewale SV, Scherrer-Crosbie M, Cowart LA, Parks JS, Zhao L, Gordon D, Ramirez-Valle F, Margulies KB, Cappola TP, Desai AA, Pedersen LN, Bergom C, Stitziel NO, Rettig MP, DiPersio JF, Hajny S, Christoffersen C, Diwan A, Javaheri A. Apolipoprotein M Attenuates Anthracycline Cardiotoxicity and Lysosomal Injury. JACC Basic Transl Sci 2023; 8:340-355. [PMID: 37034289 PMCID: PMC10077122 DOI: 10.1016/j.jacbts.2022.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 01/06/2023]
Abstract
Apolipoprotein M (ApoM) binds sphingosine-1-phosphate (S1P) and is inversely associated with mortality in human heart failure (HF). Here, we show that anthracyclines such as doxorubicin (Dox) reduce circulating ApoM in mice and humans, that ApoM is inversely associated with mortality in patients with anthracycline-induced heart failure, and ApoM heterozygosity in mice increases Dox-induced mortality. In the setting of Dox stress, our studies suggest ApoM can help sustain myocardial autophagic flux in a post-transcriptional manner, attenuate Dox cardiotoxicity, and prevent lysosomal injury.
Collapse
Affiliation(s)
- Zhen Guo
- Washington University School of Medicine, St Louis, Missouri, USA
| | | | | | | | - Mualla Ozcan
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Julio A. Chirinos
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Amanda Girardi
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Terrence Riehl
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Hosannah Evie
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Ahmed Diab
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Attila Kovacs
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Krzysztof Hyrc
- Hope Center, Washington University School of Medicine, St Louis, Missouri, USA
| | - Xiucui Ma
- Washington University School of Medicine, St Louis, Missouri, USA
- John Cochran Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Aarti Asnani
- Beth Israel Deaconess, Harvard Medical School, Boston, Massachusetts, USA
| | - Swapnil V. Shewale
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marielle Scherrer-Crosbie
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lauren Ashley Cowart
- Virginia Commonwealth University, Richmond, Virginia, USA
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, Virginia, USA
| | - John S. Parks
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Lei Zhao
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - David Gordon
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Kenneth B. Margulies
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas P. Cappola
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Carmen Bergom
- Washington University School of Medicine, St Louis, Missouri, USA
| | | | | | - John F. DiPersio
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Stefan Hajny
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Abhinav Diwan
- Washington University School of Medicine, St Louis, Missouri, USA
- John Cochran Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Ali Javaheri
- Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
13
|
Levesque MV, Hla T. Signal Transduction and Gene Regulation in the Endothelium. Cold Spring Harb Perspect Med 2023; 13:a041153. [PMID: 35667710 PMCID: PMC9722983 DOI: 10.1101/cshperspect.a041153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Extracellular signals act on G-protein-coupled receptors (GPCRs) to regulate homeostasis and adapt to stress. This involves rapid intracellular post-translational responses and long-lasting gene-expression changes that ultimately determine cellular phenotype and fate changes. The lipid mediator sphingosine 1-phosphate (S1P) and its receptors (S1PRs) are examples of well-studied GPCR signaling axis essential for vascular development, homeostasis, and diseases. The biochemical cascades involved in rapid S1P signaling are well understood. However, gene-expression regulation by S1PRs are less understood. In this review, we focus our attention to how S1PRs regulate nuclear chromatin changes and gene transcription to modulate vascular and lymphatic endothelial phenotypic changes during embryonic development and adult homeostasis. Because S1PR-targeted drugs approved for use in the treatment of autoimmune diseases cause substantial vascular-related adverse events, these findings are critical not only for general understanding of stimulus-evoked gene regulation in the vascular endothelium, but also for therapeutic development of drugs for autoimmune and perhaps vascular diseases.
Collapse
Affiliation(s)
- Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
14
|
Association of apolipoprotein M and sphingosine-1-phosphate with brown adipose tissue after cold exposure in humans. Sci Rep 2022; 12:18753. [PMID: 36335116 PMCID: PMC9637161 DOI: 10.1038/s41598-022-21938-2] [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: 11/27/2021] [Accepted: 10/06/2022] [Indexed: 11/07/2022] Open
Abstract
The HDL-associated apolipoprotein M (apoM) and its ligand sphingosine-1-phosphate (S1P) may control energy metabolism. ApoM deficiency in mice is associated with increased vascular permeability, brown adipose tissue (BAT) mass and activity, and protection against obesity. In the current study, we explored the connection between plasma apoM/S1P levels and parameters of BAT as measured via 18F-FDG PET/CT after cold exposure in humans. Fixed (n = 15) vs personalized (n = 20) short-term cooling protocols decreased and increased apoM (- 8.4%, P = 0.032 vs 15.7%, P < 0.0005) and S1P (- 41.0%, P < 0.0005 vs 19.1%, P < 0.005) plasma levels, respectively. Long-term cooling (n = 44) did not affect plasma apoM or S1P levels. Plasma apoM and S1P did not correlate significantly to BAT volume and activity in the individual studies. However, short-term studies combined, showed that increased changes in plasma apoM correlated with BAT metabolic activity (β: 0.44, 95% CI [0.06-0.81], P = 0.024) after adjusting for study design but not BAT volume (β: 0.39, 95% CI [- 0.01-0.78], P = 0.054). In conclusion, plasma apoM and S1P levels are altered in response to cold exposure and may be linked to changes in BAT metabolic activity but not BAT volume in humans. This contrasts partly with observations in animals and highlights the need for further studies to understand the biological role of apoM/S1P complex in human adipose tissue and lipid metabolism.
Collapse
|
15
|
Meszaros M, Bikov A. Obstructive Sleep Apnoea and Lipid Metabolism: The Summary of Evidence and Future Perspectives in the Pathophysiology of OSA-Associated Dyslipidaemia. Biomedicines 2022; 10:2754. [PMID: 36359273 PMCID: PMC9687681 DOI: 10.3390/biomedicines10112754] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
Obstructive sleep apnoea (OSA) is associated with cardiovascular and metabolic comorbidities, including hypertension, dyslipidaemia, insulin resistance and atherosclerosis. Strong evidence suggests that OSA is associated with an altered lipid profile including elevated levels of triglyceride-rich lipoproteins and decreased levels of high-density lipoprotein (HDL). Intermittent hypoxia; sleep fragmentation; and consequential surges in the sympathetic activity, enhanced oxidative stress and systemic inflammation are the postulated mechanisms leading to metabolic alterations in OSA. Although the exact mechanisms of OSA-associated dyslipidaemia have not been fully elucidated, three main points have been found to be impaired: activated lipolysis in the adipose tissue, decreased lipid clearance from the circulation and accelerated de novo lipid synthesis. This is further complicated by the oxidisation of atherogenic lipoproteins, adipose tissue dysfunction, hormonal changes, and the reduced function of HDL particles in OSA. In this comprehensive review, we summarise and critically evaluate the current evidence about the possible mechanisms involved in OSA-associated dyslipidaemia.
Collapse
Affiliation(s)
- Martina Meszaros
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary
| | - Andras Bikov
- North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester M13 9MT, UK
| |
Collapse
|
16
|
Vyletelová V, Nováková M, Pašková Ľ. Alterations of HDL's to piHDL's Proteome in Patients with Chronic Inflammatory Diseases, and HDL-Targeted Therapies. Pharmaceuticals (Basel) 2022; 15:1278. [PMID: 36297390 PMCID: PMC9611871 DOI: 10.3390/ph15101278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/03/2022] [Accepted: 10/14/2022] [Indexed: 09/10/2023] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, steatohepatitis, periodontitis, chronic kidney disease, and others are associated with an increased risk of atherosclerotic cardiovascular disease, which persists even after accounting for traditional cardiac risk factors. The common factor linking these diseases to accelerated atherosclerosis is chronic systemic low-grade inflammation triggering changes in lipoprotein structure and metabolism. HDL, an independent marker of cardiovascular risk, is a lipoprotein particle with numerous important anti-atherogenic properties. Besides the essential role in reverse cholesterol transport, HDL possesses antioxidative, anti-inflammatory, antiapoptotic, and antithrombotic properties. Inflammation and inflammation-associated pathologies can cause modifications in HDL's proteome and lipidome, transforming HDL from atheroprotective into a pro-atherosclerotic lipoprotein. Therefore, a simple increase in HDL concentration in patients with inflammatory diseases has not led to the desired anti-atherogenic outcome. In this review, the functions of individual protein components of HDL, rendering them either anti-inflammatory or pro-inflammatory are described in detail. Alterations of HDL proteome (such as replacing atheroprotective proteins by pro-inflammatory proteins, or posttranslational modifications) in patients with chronic inflammatory diseases and their impact on cardiovascular health are discussed. Finally, molecular, and clinical aspects of HDL-targeted therapies, including those used in therapeutical practice, drugs in clinical trials, and experimental drugs are comprehensively summarised.
Collapse
Affiliation(s)
| | | | - Ľudmila Pašková
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University, 83232 Bratislava, Slovakia
| |
Collapse
|
17
|
Mietus-Snyder M, Suslovic W, Delaney M, Playford MP, Ballout RA, Barber JR, Otvos JD, DeBiasi RL, Mehta NN, Remaley AT. Changes in HDL cholesterol, particles, and function associate with pediatric COVID-19 severity. Front Cardiovasc Med 2022; 9:1033660. [PMID: 36312284 PMCID: PMC9597312 DOI: 10.3389/fcvm.2022.1033660] [Citation(s) in RCA: 10] [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: 08/31/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Background Myriad roles for high-density lipoprotein (HDL) beyond atheroprotection include immunologic functions implicated in the severity of coronavirus disease-2019 (COVID-19) in adults. We explored whether there is an association between HDL and COVID-19 severity in youth. Methods A pediatric cohort (N = 102), who tested positive for COVID-19 across a range of disease manifestations from mild or no symptoms, to acute severe symptoms, to the multisystem inflammatory syndrome of children (MIS-C) was identified. Clinical data were collected from the medical record and reserve plasma aliquots were assessed for lipoproteins by NMR spectroscopy and assayed for HDL functional cholesterol efflux capacity (CEC). Findings were compared by COVID-19 status and symptom severity. Lipoprotein, NMR spectroscopy and CEC data were compared with 30 outpatient COVID negative children. Results Decreasing HDL cholesterol (HDL-c), apolipoprotein AI (ApoA-I), total, large and small HDL particles and HDL CEC showed a strong and direct linear dose-response relationship with increasing severity of COVID-19 symptoms. Youth with mild or no symptoms closely resembled the uninfected. An atypical lipoprotein that arises in the presence of severe hepatic inflammation, lipoprotein Z (LP-Z), was absent in COVID-19 negative controls but identified more often in youth with the most severe infections and the lowest HDL parameters. The relationship between HDL CEC and symptom severity and ApoA-I remained significant in a multiply adjusted model that also incorporated age, race/ethnicity, the presence of LP-Z and of GlycA, a composite biomarker reflecting multiple acute phase proteins. Conclusion HDL parameters, especially HDL function, may help identify youth at risk of more severe consequences of COVID-19 and other novel infectious pathogens.
Collapse
Affiliation(s)
- Michele Mietus-Snyder
- Children's National Hospital, Washington, DC, United States
- The Children's National Clinical and Translational Science Institute, Washington, DC, United States
- Division of Cardiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | | | - Meghan Delaney
- Children's National Hospital, Washington, DC, United States
- Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Division of Clinical and Laboratory Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Martin P. Playford
- Cardiovascular and Pulmonary Branch, National Institutes of Health, Bethesda, MD, United States
| | - Rami A. Ballout
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - John R. Barber
- The Children's National Clinical and Translational Science Institute, Washington, DC, United States
| | - James D. Otvos
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Roberta L. DeBiasi
- Children's National Hospital, Washington, DC, United States
- The Children's National Clinical and Translational Science Institute, Washington, DC, United States
- Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Division of Infectious Diseases, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Nehal N. Mehta
- Cardiovascular and Pulmonary Branch, National Institutes of Health, Bethesda, MD, United States
| | - Alan T. Remaley
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- Clinical Center, Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
18
|
Baker NL, Hammad SM, Hunt KJ, Semler A, Klein RL, Lopes-Virella MF. Plasma apoM Levels and Progression to Kidney Dysfunction in Patients With Type 1 Diabetes. Diabetes 2022; 71:1795-1799. [PMID: 35554520 PMCID: PMC9490352 DOI: 10.2337/db21-0920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/30/2022] [Indexed: 11/13/2022]
Abstract
Apolipoprotein M (apoM), primarily carried by HDL, has been associated with several conditions, including cardiovascular disease and diabetic nephropathy. This study proposes to examine whether plasma apoM levels are associated with the development of diabetic kidney disease, assessed as progression to macroalbuminuria (MA) and chronic kidney disease (CKD). Plasma apoM was measured using an enzyme immunoassay in 386 subjects from the Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications (EDIC) cohort at DCCT entry and closeout and the concentrations used to determine the association with risk of progression to kidney dysfunction from the time of measurement through 18 years of EDIC follow-up. apoM levels, at DCCT baseline, were higher in patients who developed CKD than in those who retained normal renal function. At DCCT closeout, participants who progressed to MA, CKD, or both MA and CKD also had significantly higher apoM levels than those who remained normal, and increased levels of apoM were associated with increased risk of progression to both MA (risk ratio [RR] 1.30 [95% CI 1.01, 1.66]) and CKD (RR 1.69 [95% CI 1.18, 2.44]). Our results strongly suggest that alterations in apoM and therefore in the composition and function of HDL in type 1 diabetes are present early in the disease process and are associated with the development of nephropathy.
Collapse
Affiliation(s)
- Nathaniel L. Baker
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC
| | - Samar M. Hammad
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC
| | - Kelly J. Hunt
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC
| | - Andrea Semler
- Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Richard L. Klein
- Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC
- Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC
| | - Maria F. Lopes-Virella
- Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC
- Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC
- Corresponding author: Maria F. Lopes-Virella,
| |
Collapse
|
19
|
Linton MF, Yancey PG, Leuthner ZM, Brown JD. The FoxOs are in the ApoM house. J Clin Invest 2022; 132:158471. [PMID: 35362476 PMCID: PMC8970665 DOI: 10.1172/jci158471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The prevalence of metabolic syndrome continues to increase globally and heightens the risk for cardiovascular disease (CVD). Insulin resistance is a core pathophysiologic mechanism that causes abnormal carbohydrate metabolism and atherogenic changes in circulating lipoprotein quantity and function. In particular, dysfunctional HDL is postulated to contribute to CVD risk in part via loss of HDL-associated sphingosine-1-phosphate (S1P). In this issue of the JCI, Izquierdo et al. demonstrate that HDL from humans with insulin resistance contained lower levels of S1P. Apolipoprotein M (ApoM), a protein constituent of HDL that binds S1P and controls bioavailability was decreased in insulin-resistant db/db mice. Gain- and loss-of-function mouse models implicated the forkhead box O transcription factors (FoxO1,3,4) in the regulation of both ApoM and HDL-associated S1P. These data have important implications for potential FoxO-based therapies designed to treat lipid and carbohydrate abnormalities associated with human metabolic disease and CVD.
Collapse
Affiliation(s)
- MacRae F Linton
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine and.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Patricia G Yancey
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine and
| | - Zoe M Leuthner
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine and
| | - Jonathan D Brown
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine and
| |
Collapse
|
20
|
Izquierdo MC, Shanmugarajah N, Lee SX, Kraakman MJ, Westerterp M, Kitamoto T, Harris M, Cook JR, Gusarova GA, Zhong K, Marbuary E, O-Sullivan I, Rasmus NF, Camastra S, Unterman TG, Ferrannini E, Hurwitz BE, Haeusler RA. Hepatic FoxOs link insulin signaling with plasma lipoprotein metabolism through an apolipoprotein M/sphingosine-1-phosphate pathway. J Clin Invest 2022; 132:146219. [PMID: 35104242 PMCID: PMC8970673 DOI: 10.1172/jci146219] [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: 12/02/2020] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple beneficial cardiovascular effects of HDL depend on sphingosine-1-phosphate (S1P). S1P associates with HDL by binding to apolipoprotein M (ApoM). Insulin resistance is a major driver of dyslipidemia and cardiovascular risk. However, the mechanisms linking alterations in insulin signaling with plasma lipoprotein metabolism are incompletely understood. The insulin-repressible FoxO transcription factors mediate key effects of hepatic insulin action on glucose and lipoprotein metabolism. This work tested whether hepatic insulin signaling regulates HDL-S1P and aimed to identify the underlying molecular mechanisms. We report that insulin-resistant, nondiabetic individuals had decreased HDL-S1P levels, but no change in total plasma S1P. This also occurred in insulin-resistant db/db mice, which had low ApoM and a specific reduction of S1P in the HDL fraction, with no change in total plasma S1P levels. Using mice lacking hepatic FoxOs (L-FoxO1,3,4), we found that hepatic FoxOs were required for ApoM expression. Total plasma S1P levels were similar to those in controls, but S1P was nearly absent from HDL and was instead increased in the lipoprotein-depleted plasma fraction. This phenotype was restored to normal by rescuing ApoM in L-FoxO1,3,4 mice. Our findings show that insulin resistance in humans and mice is associated with decreased HDL-associated S1P. Our study shows that hepatic FoxO transcription factors are regulators of the ApoM/S1P pathway.
Collapse
Affiliation(s)
- María Concepción Izquierdo
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Niroshan Shanmugarajah
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Samuel X Lee
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Michael J Kraakman
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Marit Westerterp
- Department of Pediatrics, University of Groningen, Groningen, Netherlands
| | - Takumi Kitamoto
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Michael Harris
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Joshua R Cook
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Galina A Gusarova
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Kendra Zhong
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Elijah Marbuary
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - InSug O-Sullivan
- Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, United States of America
| | - Nikolaus F Rasmus
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, United States of America
| | - Stefania Camastra
- Department of Clinical and Experimental Medicine, University of Pisa School of Medicine, Pisa, Italy
| | - Terry G Unterman
- Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, United States of America
| | - Ele Ferrannini
- Department of Internal Medicine, CNR Institute of Clinical Physiology, Pisa, Italy
| | - Barry E Hurwitz
- Department of Psychology, University of Miami, Miami, United States of America
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, United States of America
| |
Collapse
|
21
|
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: 7] [Impact Index Per Article: 1.8] [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
|
22
|
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: 11] [Impact Index Per Article: 2.8] [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
|
23
|
Yao Mattisson I, Christoffersen C. Apolipoprotein M and its impact on endothelial dysfunction and inflammation in the cardiovascular system. Atherosclerosis 2021; 334:76-84. [PMID: 34482091 DOI: 10.1016/j.atherosclerosis.2021.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/09/2023]
Abstract
Apolipoprotein M (apoM) is a member of the lipocalin superfamily and is predominantly associated with high-density lipoprotein (HDL). It was found that apoM is the chaperon to the bioactive sphingolipid, sphingosine-1-phosphate (S1P). Several studies have since contributed to expand the knowledge on apoM, S1P, and the apoM/S1P-complex in cardiovascular diseases. For instance, the HDL-bound apoM/S1P complex serves as a bridge between HDL and endothelial cells, maintaining a healthy endothelial barrier. Evidence indicates, however, that the apoM/S1P complex may has both protective and harmful effects on the cardiovascular system, which suggests the need for more research to understand the interplay between these molecules. This review aims to shed light on the most recent findings on apoM/S1P-signaling and its impact on endothelial dysfunction, inflammation, and cardiovascular diseases. Finally, it will be discussed whether drugs that target apoM and/or S1P-signaling may be beneficial to patients with cardiovascular and inflammatory diseases.
Collapse
Affiliation(s)
- Ingrid Yao Mattisson
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark; Department of Clinical Sciences Malmö, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3A, 2200, Copenhagen, Denmark.
| |
Collapse
|
24
|
Hajny S, Borup A, Elsøe S, Christoffersen C. Increased plasma apoM levels impair triglyceride turnover in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158969. [PMID: 34051379 DOI: 10.1016/j.bbalip.2021.158969] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Apolipoprotein M (apoM) is an essential transporter of plasma Sphingosine-1-Phosphate (S1P), typically attached to all lipoprotein classes, but with a majority bound to high density lipoproteins (HDL). ApoM-deficient mice display an increased activity in brown adipose tissue and a concomitant fast turnover of triglycerides. In what manner apoM/S1P affect the triglyceride metabolism is however still unknown and explored in the present study. METHODS Triglyceride turnover and potentially associated metabolic pathways were studied in the female human apoM transgenic mouse model (apoM-Tg) with increased plasma apoM and S1P levels. The model was compared with wild type (WT) mice. RESULTS ApoM-Tg mice had a reduced plasma triglyceride turnover rate and a lower free fatty acid uptake in subcutaneous adipocytes compared to WT mice. Screening for potential molecular mechanisms furthermore revealed a reduction in plasma lipase activity in apoM-Tg animals. Overexpression of apoM also reduced the plasma levels of fibroblast growth factor 21 (FGF21). CONCLUSIONS The study features the significant role of the apoM/S1P axis in maintaining a balanced triglyceride metabolism. Further, it also highlights the risk of inducing dyslipidaemia in patients receiving S1P-analouges and additionlly emphasizes the apoM/S1P axis as a potential therapeutic target in treatment of hypertriglyceridemia.
Collapse
Affiliation(s)
- Stefan Hajny
- Department of Clinical Biochemistry, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Science, University of Copenhagen, Blegdamsvej 3A, 2200 Copenhagen, Denmark.
| | - Anna Borup
- Department of Clinical Biochemistry, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Science, University of Copenhagen, Blegdamsvej 3A, 2200 Copenhagen, Denmark
| | - Sara Elsøe
- Department of Clinical Biochemistry, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Science, University of Copenhagen, Blegdamsvej 3A, 2200 Copenhagen, Denmark.
| |
Collapse
|
25
|
Eskandari M, Mellati AA. Liver X Receptor as a Possible Drug Target for Blood-Brain Barrier Integrity. Adv Pharm Bull 2021; 12:466-475. [PMID: 35935038 PMCID: PMC9348539 DOI: 10.34172/apb.2022.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/13/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose: blood-brain barrier (BBB) is made of specialized cells that are responsible for the selective passage of substances directed to the brain. The integrated BBB is essential for precise controlling of the different substances passage as well as protecting the brain from various damages. In this article, we attempted to explain the role of liver X receptor (LXR) in maintaining BBB integrity as a possible drug target.
Methods: In this study, various databases, including PubMed, Google Scholar, and Scopus were searched using the following keywords: blood-brain barrier, BBB, liver X receptor, and LXR until July, 2020. Additionally, contents close to the subject of our study were surveyed.
Results: LXR is a receptor the roles of which in various diseases have been investigated. LXR can affect maintaining BBB by affecting various ways such as ATP-binding cassette transporter A1 (ABCA1), matrix metalloproteinase-9 (MMP9), insulin-like growth factor 1 (IGF1), nuclear factor-kappa B (NF-κB) signaling, mitogen-activated protein kinase (MAPK), tight junction molecules, both signal transducer and activator of transcription 1 (STAT1), Wnt/β-catenin Signaling, transforming growth factor beta (TGF-β) signaling, and expressions of Smad 2/3 and Snail.
Conclusion: LXR could possibly be used either as a target for drug delivery to brain tissue or as a target for maintaining the BBB integrity in different diseases; thereby the drug will be conducted to tissues, other than the brain. If it is verified that only LXRα is necessary for protecting BBB, some specific LXRα ligands must be found and then used in medication.
Collapse
Affiliation(s)
- Mahsa Eskandari
- Medical school, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Awsat Mellati
- Zanjan Metabolic Disease Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
26
|
Yao S, Zheng F, Yu Y, Zhan Y, Xu N, Luo G, Zheng L. Apolipoprotein M promotes cholesterol uptake and efflux from mouse macrophages. FEBS Open Bio 2021; 11:1607-1620. [PMID: 33830664 PMCID: PMC8167864 DOI: 10.1002/2211-5463.13157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/14/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Apolipoprotein M (ApoM) exhibits various anti-atherosclerotic functions as a component of high-density lipoprotein (HDL) particles. Scavenger receptor class B type I (SR-BI) is a classic HDL receptor that mediates selective cholesterol uptake and enhances the efflux of cellular cholesterol to HDL. However, the effect of ApoM on cholesterol transport in macrophages remains unclear. In this study, we identified for the first time that ApoM is expressed in mouse macrophages and is involved in cholesterol uptake, similar to SR-BI. NBD-cholesterol uptake and efflux in cells were characterized using fluorescence spectrophotometry. The uptake ratios of cholesterol by macrophages from ApoM-/- SR-BI-/- mice were significantly lower than those from ApoM+/+ SR-BI-/- and ApoM-/- SR-BI+/+ mice. Real-time fluorescence quantitative PCR was used to analyze the expression of cholesterol transport-related genes involved in cholesterol uptake. ApoM-enriched HDL (ApoM+ HDL) facilitated more cholesterol efflux from murine macrophage Ana-1 cells than ApoM-free HDL (ApoM- HDL). However, recombinant human ApoM protein inhibited the ability of ApoM- HDL to induce cholesterol efflux. In conclusion, ApoM promotes cholesterol uptake and efflux in mouse macrophages. A better understanding of ApoM function may lead to the development of novel therapeutic strategies for treating atherosclerotic diseases.
Collapse
Affiliation(s)
- Shuang Yao
- Clinical Medical Research Centerthe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| | - Fan Zheng
- Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Yang Yu
- Clinical Medical Research Centerthe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| | - Yuxia Zhan
- Clinical Medical Research Centerthe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| | - Ning Xu
- Section of Clinical Chemistry and PharmacologyInstitute of Laboratory MedicineLund UniversitySweden
| | - Guanghua Luo
- Clinical Medical Research Centerthe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| | - Lu Zheng
- Clinical Medical Research Centerthe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| |
Collapse
|
27
|
Trakaki A, Marsche G. Current Understanding of the Immunomodulatory Activities of High-Density Lipoproteins. Biomedicines 2021; 9:biomedicines9060587. [PMID: 34064071 PMCID: PMC8224331 DOI: 10.3390/biomedicines9060587] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
Lipoproteins interact with immune cells, macrophages and endothelial cells - key players of the innate and adaptive immune system. High-density lipoprotein (HDL) particles seem to have evolved as part of the innate immune system since certain HDL subspecies contain combinations of apolipoproteins with immune regulatory functions. HDL is enriched in anti-inflammatory lipids, such as sphingosine-1-phosphate and certain saturated lysophospholipids. HDL reduces inflammation and protects against infection by modulating immune cell function, vasodilation and endothelial barrier function. HDL suppresses immune cell activation at least in part by modulating the cholesterol content in cholesterol/sphingolipid-rich membrane domains (lipid rafts), which play a critical role in the compartmentalization of signaling pathways. Acute infections, inflammation or autoimmune diseases lower HDL cholesterol levels and significantly alter HDL metabolism, composition and function. Such alterations could have a major impact on disease progression and may affect the risk for infections and cardiovascular disease. This review article aims to provide a comprehensive overview of the immune cell modulatory activities of HDL. We focus on newly discovered activities of HDL-associated apolipoproteins, enzymes, lipids, and HDL mimetic peptides.
Collapse
|
28
|
Effects of lipoproteins on endothelial cells and macrophages function and its possible implications on fetal adverse outcomes associated to maternal hypercholesterolemia during pregnancy. Placenta 2021; 106:79-87. [PMID: 33706211 DOI: 10.1016/j.placenta.2021.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/03/2021] [Accepted: 02/25/2021] [Indexed: 11/23/2022]
Abstract
Hypercholesterolemia is one of the main risk factors associated with atherosclerosis and cardiovascular disease, the leading cause of death worldwide. During pregnancy, maternal hypercholesterolemia develops, and it can occur in a physiological (MPH) or supraphysiological (MSPH) manner, where MSPH is associated with endothelial dysfunction and early atherosclerotic lesions in the fetoplacental vasculature. In the pathogenesis of atherosclerosis, endothelial activation and endothelial dysfunction, characterized by an imbalance in the bioavailability of nitric oxide, contribute to the early stages of this disease. Macrophages conversion to foam cells, cholesterol efflux from these cells and its differentiation into a pro- or anti-inflammatory phenotype are also important processes that contribute to atherosclerosis. In adults it has been reported that native and modified HDL and LDL play an important role in endothelial and macrophage function. In this review it is proposed that fetal lipoproteins could be also relevant factors involved in the detrimental vascular effects described in MSPH. Changes in the composition and function of neonatal lipoproteins compared to adults has been reported and, although in MSPH pregnancies the fetal lipid profile does not differ from MPH, differences in the lipidomic profiles of umbilical venous blood have been reported, which could have implications in the vascular function. In this review we summarize the available information regarding the effects of lipoproteins on endothelial and macrophage function, emphasizing its possible implications on fetal adverse outcomes associated to maternal hypercholesterolemia during pregnancy.
Collapse
|
29
|
Morris G, Puri BK, Bortolasci CC, Carvalho A, Berk M, Walder K, Moreira EG, Maes M. The role of high-density lipoprotein cholesterol, apolipoprotein A and paraoxonase-1 in the pathophysiology of neuroprogressive disorders. Neurosci Biobehav Rev 2021; 125:244-263. [PMID: 33657433 DOI: 10.1016/j.neubiorev.2021.02.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/29/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Lowered high-density lipoprotein (HDL) cholesterol has been reported in major depressive disorder, bipolar disorder, first episode of psychosis, and schizophrenia. HDL, its major apolipoprotein component, ApoA1, and the antioxidant enzyme paraoxonase (PON)1 (which is normally bound to ApoA1) all have anti-atherogenic, antioxidant, anti-inflammatory, and immunomodulatory roles, which are discussed in this paper. The paper details the pathways mediating the anti-inflammatory effects of HDL, ApoA1 and PON1 and describes the mechanisms leading to compromised HDL and PON1 levels and function in an environment of chronic inflammation. The molecular mechanisms by which changes in HDL, ApoA1 and PON1 might contribute to the pathophysiology of the neuroprogressive disorders are explained. Moreover, the anti-inflammatory actions of ApoM-mediated sphingosine 1-phosphate (S1P) signalling are reviewed as well as the deleterious effects of chronic inflammation and oxidative stress on ApoM/S1P signalling. Finally, therapeutic interventions specifically aimed at improving the levels and function of HDL and PON1 while reducing levels of inflammation and oxidative stress are considered. These include the so-called Mediterranean diet, extra virgin olive oil, polyphenols, flavonoids, isoflavones, pomegranate juice, melatonin and the Mediterranean diet combined with the ketogenic diet.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | | | - Chiara C Bortolasci
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.
| | - Andre Carvalho
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Estefania G Moreira
- Post-Graduation Program in Health Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Michael Maes
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
30
|
Tageldeen MM, Badrawy H, Abdelmeguid M, Zaghlol M, Gaber N, Kenawy EM. Apolipoprotein M Gene Polymorphism Rs805297 (C-1065A): Association With Type 2 Diabetes Mellitus and Related Microvascular Complications in South Egypt. Am J Med Sci 2021; 362:48-55. [PMID: 33621527 DOI: 10.1016/j.amjms.2021.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 11/06/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Apolipoprotein M (ApoM) may have a role in the susceptibility of type 2 diabetes mellitus (T2DM). Polymorphisms in the promoter region of the ApoM gene were found to be significantly associated with diabetes. The aim of this study was to investigate the association of ApoM SNP rs805297 (C-1065A) with the susceptibility of T2DM and related microvascular complications in South Egypt. METHODS We conducted a case-control study of 60 T2DM patients and 60 healthy control subjects. Lipid profile, fasting, and 2 hours postprandial glucose and creatinine levels were estimated. Patients were subjected to general and Fundus examinations, and screening for nephropathy by urinary albumin levels. ApoM level was assayed by ELISA. Genotyping of the human ApoM gene polymorphism SNP rs805297 (C-1065A) was done by PCR-restriction fragment length polymorphism followed by sequencing to confirm the polymorphism results. RESULTS ApoM was not different between T2DM and the control group (p=0.075) and was negatively correlated with LDL-c (p=0.029). There were significant differences in ApoM genotypes (p=0.001) and allele frequencies (p=0.019) between T2DM and the control group. A significant reduction in FBG, 2hPPG, and HbA1c levels in the heterozygous than the wild genotype in the group with diabetes with no difference in other lab parameters and microvascular complications. The C-allele is associated with lower blood glucose levels and retinopathy. The wild (CC) genotype is considered as a risk factor for developing T2DM in South Egyptians but not CA+AA genotypes. CONCLUSIONS In South Egyptians the ApoM polymorphism rs805297 (C-1065A) wild type (CC) was associated with T2DM susceptibility and may have a role in controlling hyperglycemia in these patients. The A-allele is associated with hyperglycemia and diabetic retinopathy.
Collapse
Affiliation(s)
| | - Hosny Badrawy
- Clinical Pathology, South Egypt Cancer Institute, Assiut, Egypt
| | - Mona Abdelmeguid
- Clinical Pathology, Faculty of Medicine (Boys), Al-Azhar University, Assiut, Egypt.
| | - Mohammed Zaghlol
- Clinical Pathology, Faculty of Medicine (Boys), Al-Azhar University, Assiut, Egypt
| | - Noha Gaber
- Clinical Pathology, South Egypt Cancer Institute, Assiut, Egypt
| | - Eglal Mohamed Kenawy
- Internal Medicine, Faculty of Medicine (Boys), Al-Azhar University, Assiut, Egypt
| |
Collapse
|
31
|
Christoffersen C. Apolipoprotein M-A Marker or an Active Player in Type II Diabetes? Front Endocrinol (Lausanne) 2021; 12:665393. [PMID: 34093440 PMCID: PMC8176018 DOI: 10.3389/fendo.2021.665393] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/03/2021] [Indexed: 11/15/2022] Open
Abstract
Apolipoprotein M (apoM) is a member of the lipocalin superfamily and an important carrier of the small bioactive lipid sphingosine-1-phosphate (S1P). The apoM/S1P complex is attached to all lipoproteins, but exhibits a significant preference for high-density lipoproteins. Although apoM, S1P, and the apoM/S1P complex have been discovered more than a decade earlier, the overall function of the apoM/S1P complex remains controversial. Evidence suggests that the complex plays a role in inflammation and cholesterol metabolism and is important for maintaining a healthy endothelial barrier, regulating the turnover of triglycerides from lipoproteins, and reducing cholesterol accumulation in vessel walls. Recent studies have also addressed the role of apoM and S1P in the development of diabetes and obesity. However, limited evidence is available, and the data published so far deviates. This review discusses the specific elements indicative of the protective or harmful effects of apoM, S1P, and the apoM/S1P complex on type 2 diabetes development. Since drugs targeting the S1P system and its receptors are available and could be potentially used for treating diabetes, this research topic is a pertinent one.
Collapse
Affiliation(s)
- Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Christina Christoffersen,
| |
Collapse
|
32
|
Márquez AB, Nazir S, van der Vorst EP. High-Density Lipoprotein Modifications: A Pathological Consequence or Cause of Disease Progression? Biomedicines 2020; 8:biomedicines8120549. [PMID: 33260660 PMCID: PMC7759904 DOI: 10.3390/biomedicines8120549] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
High-density lipoprotein (HDL) is well-known for its cardioprotective effects, as it possesses anti-inflammatory, anti-oxidative, anti-thrombotic, and cytoprotective properties. Traditionally, studies and therapeutic approaches have focused on raising HDL cholesterol levels. Recently, it became evident that, not HDL cholesterol, but HDL composition and functionality, is probably a more fruitful target. In disorders, such as chronic kidney disease or cardiovascular diseases, it has been observed that HDL is modified and becomes dysfunctional. There are different modification that can occur, such as serum amyloid, an enrichment and oxidation, carbamylation, and glycation of key proteins. Additionally, the composition of HDL can be affected by changes to enzymes such as cholesterol ester transfer protein (CETP), lecithin-cholesterol acyltransferase (LCAT), and phospholipid transfer protein (PLTP) or by modification to other important components. This review will highlight some main modifications to HDL and discuss whether these modifications are purely a consequential result of pathology or are actually involved in the pathology itself and have a causal role. Therefore, HDL composition may present a molecular target for the amelioration of certain diseases, but more information is needed to determine to what extent HDL modifications play a causal role in disease development.
Collapse
Affiliation(s)
- Andrea Bonnin Márquez
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| | - Sumra Nazir
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P.C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, 80336 Munich, Germany
- Correspondence: ; Tel.: +49-241-80-36914
| |
Collapse
|
33
|
Pedrini S, Chatterjee P, Hone E, Martins RN. High‐density lipoprotein‐related cholesterol metabolism in Alzheimer’s disease. J Neurochem 2020; 159:343-377. [DOI: 10.1111/jnc.15170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Steve Pedrini
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Pratishtha Chatterjee
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
| | - Eugene Hone
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Ralph N. Martins
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Nedlands WA Australia
| |
Collapse
|
34
|
Chen Z, Hu M. The apoM-S1P axis in hepatic diseases. Clin Chim Acta 2020; 511:235-242. [PMID: 33096030 DOI: 10.1016/j.cca.2020.10.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
Liver dysfunction is always accompanied by lipid metabolism dysfunction. Apolipoprotein M (apoM), a member of the apolipoprotein family, is primarily expressed and secreted from the liver. apoM is the main chaperone of sphingosine-1-phosphate (S1P), a small signalling molecule associated with numerous physiologic and pathophysiologic processes. In addition to transport, apoM also influences the biologic effects of S1P. Most recently, numerous studies have investigated the potential role of the apoM-S1P axis in a variety of hepatic diseases. These include liver fibrosis, viral hepatitis B and C infection, hepatobiliary disease, non-alcoholic and alcoholic steatohepatitis, acute liver injury and hepatocellular carcinoma. In this review, the roles of apoM and S1P in the development of hepatic diseases are summarized, and novel insights into the diagnosis and treatment of hepatic diseases are discussed.
Collapse
Affiliation(s)
- Zhiyang Chen
- Department of Laboratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, PR China
| | - Min Hu
- Department of Laboratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, PR China.
| |
Collapse
|
35
|
Chirinos JA, Zhao L, Jia Y, Frej C, Adamo L, Mann D, Shewale SV, Millar JS, Rader DJ, French B, Brandimarto J, Margulies KB, Parks JS, Wang Z, Seiffert DA, Fang J, Sweitzer N, Chistoffersen C, Dahlbäck B, Car BD, Gordon DA, Cappola TP, Javaheri A. Reduced Apolipoprotein M and Adverse Outcomes Across the Spectrum of Human Heart Failure. Circulation 2020; 141:1463-1476. [PMID: 32237898 PMCID: PMC7200273 DOI: 10.1161/circulationaha.119.045323] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Apo (apolipoprotein) M mediates the physical interaction between high-density lipoprotein (HDL) particles and sphingosine-1-phosphate (S1P). Apo M exerts anti-inflammatory and cardioprotective effects in animal models. METHODS In a subset of PHFS (Penn Heart Failure Study) participants (n=297), we measured apo M by Enzyme-Linked ImmunoSorbent Assay (ELISA). We also measured total S1P by liquid chromatography-mass spectrometry and isolated HDL particles to test the association between apo M and HDL-associated S1P. We confirmed the relationship between apo M and outcomes using modified aptamer-based apo M measurements among 2170 adults in the PHFS and 2 independent cohorts: the Washington University Heart Failure Registry (n=173) and a subset of TOPCAT (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist Trial; n=218). Last, we examined the relationship between apo M and ≈5000 other proteins (SomaScan assay) to identify biological pathways associated with apo M in heart failure. RESULTS In the PHFS, apo M was inversely associated with the risk of death (standardized hazard ratio, 0.56 [95% CI, 0.51-0.61]; P<0.0001) and the composite of death/ventricular assist device implantation/heart transplantation (standardized hazard ratio, 0.62 [95% CI, 0.58-0.67]; P<0.0001). This relationship was independent of HDL cholesterol or apo AI levels. Apo M remained associated with death (hazard ratio, 0.78 [95% CI, 0.69-0.88]; P<0.0001) and the composite of death/ventricular assist device/heart transplantation (hazard ratio, 0.85 [95% CI, 0.76-0.94]; P=0.001) in models that adjusted for multiple confounders. This association was present in both heart failure with reduced and preserved ejection fraction and was replicated in the Washington University cohort and a cohort with heart failure with preserved ejection fraction only (TOPCAT). The S1P and apo M content of isolated HDL particles strongly correlated (R=0.81, P<0.0001). The top canonical pathways associated with apo M were inflammation (negative association), the coagulation system (negative association), and liver X receptor/retinoid X receptor activation (positive association). The relationship with inflammation was validated with multiple inflammatory markers measured with independent assays. CONCLUSIONS Reduced circulating apo M is independently associated with adverse outcomes across the spectrum of human heart failure. Further research is needed to assess whether the apo M/S1P axis is a suitable therapeutic target in heart failure.
Collapse
Affiliation(s)
- Julio A. Chirinos
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Lei Zhao
- Bristol-Myers Squibb Company, Lawrenceville, NJ
| | - Yi Jia
- SomaLogic Inc., Boulder, CO
| | | | - Luigi Adamo
- Washington University School of Medicine, St. Louis, MO
| | - Douglas Mann
- Washington University School of Medicine, St. Louis, MO
| | - Swapnil V. Shewale
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - John S. Millar
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Daniel J. Rader
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Benjamin French
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Jeff Brandimarto
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Kenneth B. Margulies
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - John S. Parks
- Dept. of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC
| | | | | | - James Fang
- University of Utah. Salt Lake City, Utah
| | - Nancy Sweitzer
- Sarver Heart Institute, University of Arizona, Tuscon, AZ
| | - Christina Chistoffersen
- Dept. of Clinical Biochemistry, Rigshospitalet and Dept. of Biomedical Sciences, Copenhagen, Denmark
| | | | | | | | - Thomas P. Cappola
- Perelman School of Medicine. University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania. Philadelphia, PA
| | - Ali Javaheri
- Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
36
|
Potential sphingosine-1-phosphate-related therapeutic targets in the treatment of cerebral ischemia reperfusion injury. Life Sci 2020; 249:117542. [PMID: 32169519 DOI: 10.1016/j.lfs.2020.117542] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/29/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that regulates lymphocyte trafficking, glial cell activation, vasoconstriction, endothelial barrier function, and neuronal death pathways in the brain. Research has increasingly implicated S1P in the pathology of cerebral ischemia reperfusion (IR) injury. As a high-affinity agonist of S1P receptor, fingolimod exhibits excellent neuroprotective effects against ischemic challenge both in vivo and in vitro. By summarizing recent progress on how S1P participates in the development of brain IR injury, this review identifies potential therapeutic targets for the treatment of brain IR injury.
Collapse
|
37
|
Luo G, Xu N. Apolipoprotein M: Research Progress and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:85-103. [PMID: 32705596 DOI: 10.1007/978-981-15-6082-8_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Apolipoprotein M (apoM) was first identified and characterized to the apolipoprotein family in 1999. Human apoM gene is located in a highly conserved segment in the major histocompatibility complex (MHC) class III locus on chromosome 6 and codes for an about 23 kDa protein that structurally belongs to the lipocalin superfamily. ApoM is selectively expressed in hepatocytes and in the tubular epithelium of kidney. In human plasma, apoM is mainly confined to the high-density lipoprotein (HDL) particles, but it may also occur in other lipoprotein classes, such as in the triglyceride-rich particles after fat intake. It has been demonstrated that apoM is critical for the formation of HDL, notably pre-beta HDL1. The antiatherogenic function of HDL is well established, and its ability to promote cholesterol efflux from foam cells in the atherosclerotic lesions is generally regarded as one of the key mechanisms behind this protective function. However, HDL could also display a variety of properties that may affect the complex atherosclerotic processes by other mechanisms, thus being involved in processes related to antioxidant defense, immune system, and systemic effects in septicemia, which may be partly contributed via its apolipoproteins and/or phospholipids. Moreover, it has been demonstrated that apoM functions as a natural carrier of sphingosin-1-phosphate (S1P) in vivo which may be related to its antiatherosclerotic and protective effects on endothelial cell barrier and anti-inflammatory properties. These may also provide a link between the diverse effects of HDL.
Collapse
Affiliation(s)
- Guanghua Luo
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, People's Republic of China
| | - Ning Xu
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lunds University, Lund, Sweden.
| |
Collapse
|
38
|
Ozaki T, Muramatsu R, Nakamura H, Kinoshita M, Kishima H, Yamashita T. Proteomic analysis of protein changes in plasma by balloon test occlusion. J Clin Neurosci 2019; 72:397-401. [PMID: 31875829 DOI: 10.1016/j.jocn.2019.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/01/2019] [Indexed: 10/25/2022]
Abstract
Transient ischemia provides the tolerance against prolonged ischemia in the brain. In mouse experimental model, transient ischemia changes the composition ratio of circulating proteins, which associate with neuroprotection; however, the human evidence is lacking. Here we mimicked balloon test occlusion (BTO) of carotid artery as a transient ischemia and investigated the change of composition ratio of the circulating protein in the human plasma. We collected blood samples from nine patients (5 men and 4 women; mean age 64.2 years; range 45 to 77 years) before and 48 h after BTO and investigated the changes of circulating molecules level in the proteome using LC-MS/MS analysis. Leucine-rich alpha-2-glycoprotein and serum amyloid A-1 increased and protein AMBP decreased in the blood samples after BTO. Transient change of blood flow in the brain alters molecular expression in the plasma. Because the alteration of plasma protein composition is involved in ischemic tolerance in animal models, the proteins whose level was changed by BTO may be also involved in neuroprotection against ischemia in human.
Collapse
Affiliation(s)
- Tomohiko Ozaki
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Neurosurgery, Graduate School of Medicine, Osaka University, Japan; Department of Neurosurgery, Osaka International Cancer Institute, Osaka, Japan
| | - Rieko Muramatsu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Japan; Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Neurology and Psychiatry, Kodaira, Japan.
| | - Hajime Nakamura
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, Japan
| | - Manabu Kinoshita
- Department of Neurosurgery, Osaka International Cancer Institute, Osaka, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Japan; Department of Molecular Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Japan
| |
Collapse
|
39
|
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: 22] [Impact Index Per Article: 3.7] [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
|
40
|
Apolipoprotein M Serum Levels Correlate with IgA Vasculitis and IgA Vasculitis Nephritis. DISEASE MARKERS 2019; 2019:1825849. [PMID: 31885732 PMCID: PMC6927057 DOI: 10.1155/2019/1825849] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022]
Abstract
Objective IgA vasculitis (lgAV) is the most frequent vessel vasculitis in children, and the prognosis is related to the children's age and degree of nephritis. This study is aimed at investigating serum apolipoprotein M (apoM) levels in patients with lgAV patients and at evaluating the association between apoM and disease severity. Methods A total of 109 lgAV patients and 76 age- and sex-matched healthy controls were included. The age and gender of the study participants were matched. ApoM levels were measured by an enzyme-linked immunosorbent assay. Additionally, the serum levels of lipids, apolipoproteins, kidney biochemical profiles, immunoglobulins (IgA, IgG, IgM, and IgE), and the complements (C3 and C4) were assessed using an automatic biochemical analyzer. Results ApoM was increased significantly in lgAV patients compared to healthy controls. ApoM, meanwhile, was lower in patients with nephritis than in those without nephritis. The apoM levels were higher in classes I and II IgA vasculitis nephritis (lgAVN) patients than in classes III and IV. Besides, the apoM serum level < 24.81 mg/L was an independent predictive factor for lgAVN and can be independently associated with the presence of nephritis in lgAV patients. Meanwhile, the serum apoM concentration negatively correlated with the ISKDC grading score in lgAVN patients. Conclusions Serum apoM was elevated in lgAV patients and decreased gradually with the ISKDC grading score. ApoM (OR = 0.32, 95%CI = 0.12-0.85, p = 0.023) was identified as a protective factor for nephritis in all lgAV patients.
Collapse
|
41
|
Mathiesen Janiurek M, Soylu-Kucharz R, Christoffersen C, Kucharz K, Lauritzen M. Apolipoprotein M-bound sphingosine-1-phosphate regulates blood-brain barrier paracellular permeability and transcytosis. eLife 2019; 8:e49405. [PMID: 31763978 PMCID: PMC6877292 DOI: 10.7554/elife.49405] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/01/2019] [Indexed: 12/18/2022] Open
Abstract
The blood-brain barrier (BBB) is formed by the endothelial cells lining cerebral microvessels, but how blood-borne signaling molecules influence permeability is incompletely understood. We here examined how the apolipoprotein M (apoM)-bound sphingosine 1-phosphate (S1P) signaling pathway affects the BBB in different categories of cerebral microvessels using ApoM deficient mice (Apom-/-). We used two-photon microscopy to monitor BBB permeability of sodium fluorescein (376 Da), Alexa Fluor (643 Da), and fluorescent albumin (45 kDA). We show that BBB permeability to small molecules increases in Apom-/- mice. Vesicle-mediated transfer of albumin in arterioles increased 3 to 10-fold in Apom-/- mice, whereas transcytosis in capillaries and venules remained unchanged. The S1P receptor 1 agonist SEW2871 rapidly normalized paracellular BBB permeability in Apom-/- mice, and inhibited transcytosis in penetrating arterioles, but not in pial arterioles. Thus, apoM-bound S1P maintains low paracellular BBB permeability in all cerebral microvessels and low levels of vesicle-mediated transport in penetrating arterioles.
Collapse
Affiliation(s)
| | | | - Christina Christoffersen
- Department of Clinical BiochemistryRigshospitaletCopenhagenDenmark
- Department of Biomedical SciencesCopenhagen UniversityCopenhagenDenmark
| | | | - Martin Lauritzen
- Department of NeuroscienceUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical NeurophysiologyRigshospitalet-GlostrupCopenhagenDenmark
| |
Collapse
|
42
|
Andraski AB, Singh SA, Lee LH, Higashi H, Smith N, Zhang B, Aikawa M, Sacks FM. Effects of Replacing Dietary Monounsaturated Fat With Carbohydrate on HDL (High-Density Lipoprotein) Protein Metabolism and Proteome Composition in Humans. Arterioscler Thromb Vasc Biol 2019; 39:2411-2430. [PMID: 31554421 DOI: 10.1161/atvbaha.119.312889] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Clinical evidence has linked low HDL (high-density lipoprotein) cholesterol levels with high cardiovascular disease risk; however, its significance as a therapeutic target remains unestablished. We hypothesize that HDLs functional heterogeneity is comprised of metabolically distinct proteins, each on distinct HDL sizes and that are affected by diet. Approach and Results: Twelve participants were placed on 2 healthful diets high in monounsaturated fat or carbohydrate. After 4 weeks on each diet, participants completed a metabolic tracer study. HDL was isolated by Apo (apolipoprotein) A1 immunopurification and separated into 5 sizes. Tracer enrichment and metabolic rates for 8 HDL proteins-ApoA1, ApoA2, ApoC3, ApoE, ApoJ, ApoL1, ApoM, and LCAT (lecithin-cholesterol acyltransferase)-were determined by parallel reaction monitoring and compartmental modeling, respectively. Each protein had a unique, size-specific distribution that was not altered by diet. However, carbohydrate, when replacing fat, increased the fractional catabolic rate of ApoA1 and ApoA2 on alpha3 HDL; ApoE on alpha3 and alpha1 HDL; and ApoM on alpha2 HDL. Additionally, carbohydrate increased the production of ApoC3 on alpha3 HDL and ApoJ and ApoL1 on the largest alpha0 HDL. LCAT was the only protein studied that diet did not affect. Finally, global proteomics showed that diet did not alter the distribution of the HDL proteome across HDL sizes. CONCLUSIONS This study demonstrates that HDL in humans is composed of a complex system of proteins, each with its own unique size distribution, metabolism, and diet regulation. The carbohydrate-induced hypercatabolic state of HDL proteins may represent mechanisms by which carbohydrate alters the cardioprotective properties of HDL.
Collapse
Affiliation(s)
- Allison B Andraski
- From the Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (A.B.A., N.S., B.Z., F.M.S.)
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences (S.A.S., L.H.L., H.H., M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lang Ho Lee
- Center for Interdisciplinary Cardiovascular Sciences (S.A.S., L.H.L., H.H., M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences (S.A.S., L.H.L., H.H., M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Nathaniel Smith
- From the Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (A.B.A., N.S., B.Z., F.M.S.)
| | - Bo Zhang
- From the Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (A.B.A., N.S., B.Z., F.M.S.).,Department of Biochemistry, Fukuoka University School of Medicine, Fukuoka, Japan (B.Z.)
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences (S.A.S., L.H.L., H.H., M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Channing Division of Network Medicine (M.A., F.M.S.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Frank M Sacks
- From the Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (A.B.A., N.S., B.Z., F.M.S.).,Channing Division of Network Medicine (M.A., F.M.S.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
43
|
Arnao V, Tuttolomondo A, Daidone M, Pinto A. Lipoproteins in Atherosclerosis Process. Curr Med Chem 2019; 26:1525-1543. [PMID: 31096892 DOI: 10.2174/0929867326666190516103953] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/17/2017] [Accepted: 12/10/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Dyslipidaemias is a recognized risk factor for atherosclerosis, however, new evidence brought to light by trials investigating therapies to enhance HDLcholesterol have suggested an increased atherosclerotic risk when HDL-C is high. RESULTS Several studies highlight the central role in atherosclerotic disease of dysfunctional lipoproteins; oxidised LDL-cholesterol is an important feature, according to "oxidation hypothesis", of atherosclerotic lesion, however, there is today a growing interest for dysfunctional HDL-cholesterol. The target of our paper is to review the functions of modified and dysfunctional lipoproteins in atherogenesis. CONCLUSION Taking into account the central role recognized to dysfunctional lipoproteins, measurements of functional features of lipoproteins, instead of conventional routine serum evaluation of lipoproteins, could offer a valid contribution in experimental studies as in clinical practice to stratify atherosclerotic risk.
Collapse
Affiliation(s)
- Valentina Arnao
- BioNeC Dipartimento di BioMedicina Sperimentale e Neuroscienze Cliniche, Universita degli Studi di Palermo, Palermo, Italy.,PhD School of: Medicina Clinica e Scienze del Comportamento-Biomedical Department of Internal and Specialistic Medicine. (Di.Bi.M.I.S), University of Palermo, Palermo, Italy
| | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, (PROMISE), University of Palermo, Palermo, Italy
| | - Mario Daidone
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, (PROMISE), University of Palermo, Palermo, Italy
| | - Antonio Pinto
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, (PROMISE), University of Palermo, Palermo, Italy
| |
Collapse
|
44
|
Liu M, Frej C, Langefeld CD, Divers J, Bowden DW, Carr JJ, Gebre AK, Xu J, Larsson B, Dahlbäck B, Freedman BI, Parks JS. Plasma apoM and S1P levels are inversely associated with mortality in African Americans with type 2 diabetes mellitus. J Lipid Res 2019; 60:1425-1431. [PMID: 31133557 DOI: 10.1194/jlr.p089409] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 04/27/2019] [Indexed: 12/21/2022] Open
Abstract
apoM is a minor HDL apolipoprotein and carrier for sphingosine-1-phosphate (S1P). HDL apoM and S1P concentrations are inversely associated with atherosclerosis progression in rodents. We evaluated associations between plasma concentrations of S1P, plasma concentrations of apoM, and HDL apoM levels with prevalent subclinical atherosclerosis and mortality in the African American-Diabetes Heart Study participants (N = 545). Associations between plasma S1P, plasma apoM, and HDL apoM with subclinical atherosclerosis and mortality were assessed using multivariate parametric, nonparametric, and Cox proportional hazards models. At baseline, participants' median (25th percentile, 75th percentile) age was 55 (49, 62) years old and their coronary artery calcium (CAC) mass score was 26.5 (0.0, 346.5). Plasma S1P, plasma apoM, and HDL apoM were not associated with CAC. After 64 (57.6, 70.3) months of follow-up, 81 deaths were recorded. Higher concentrations of plasma S1P [odds ratio (OR) = 0.14, P = 0.01] and plasma apoM (OR = 0.10, P = 0.02), but not HDL apoM (P = 0.89), were associated with lower mortality after adjusting for age, sex, statin use, CAC, kidney function, and albuminuria. We conclude that plasma S1P and apoM concentrations are inversely and independently associated with mortality, but not CAC, in African Americans with type 2 diabetes after accounting for conventional risk factors.
Collapse
Affiliation(s)
- Mingxia Liu
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Cecilia Frej
- Department of Translational Medicine Skåne University Hospital, Lund University, Malmö, Sweden
| | - Carl D Langefeld
- Division of Public Health Sciences, Department of Biostatistics and Data Science Wake Forest School of Medicine, Winston-Salem, NC
| | - Jasmin Divers
- Division of Public Health Sciences, Department of Biostatistics and Data Science Wake Forest School of Medicine, Winston-Salem, NC
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - J Jeffrey Carr
- Department of Radiology Vanderbilt University Medical Center, Nashville, TN
| | - Abraham K Gebre
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Jianzhao Xu
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - Benny Larsson
- Department of Clinical Chemistry Skåne University Hospital, Lund, Sweden
| | - Björn Dahlbäck
- Department of Translational Medicine Skåne University Hospital, Lund University, Malmö, Sweden
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC .,Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
45
|
Yafasova A, Mandrup CM, Egelund J, Nyberg M, Stallknecht B, Hellsten Y, Nielsen LB, Christoffersen C. Effect of menopause and exercise training on plasma apolipoprotein M and sphingosine-1-phosphate. J Appl Physiol (1985) 2019; 126:214-220. [DOI: 10.1152/japplphysiol.00527.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The axis of apolipoprotein M (apoM) and sphingosine-1-phosphate (S1P) is of importance to plasma lipid levels, endothelial function, and development of atherosclerosis. Menopause is accompanied by dyslipidemia and an increased risk of atherosclerosis, which can be lowered by exercise training. The aim of this study was to explore if effects of menopause and training are paralleled by changes in the apoM/S1P axis. Healthy, late premenopausal [ n = 38, age 49.2 (SD 2)] and recent postmenopausal [ n = 37, age 53.3 (SD 3)] women from the Copenhagen Women Study participated in a 3-mo, aerobic high-intensity exercise intervention. Before training, plasma apoM was higher in postmenopausal [1.08 µmol/l (SD 0.2)] compared with premenopausal [0.82 µmol/l (SD 0.2)] women ( P < 0.0001). Plasma S1P was similar in the two groups [0.44 µmol/l (SD 0.1) and 0.46 µmol/l (SD 0.1), respectively]. Thus, the pretraining S1P/apoM ratio was 26% lower in postmenopausal than premenopausal women ( P < 0.0001). After the training program, plasma apoM increased from 0.82 µmol/l (SD 0.2) to 0.90 µmol/l (SD 0.3) in premenopausal women and from 1.08 µmol/l (SD 0.2) to 1.16 µmol/l (SD 0.3) in postmenopausal women ( P < 0.05). Plasma S1P increased from 0.44 µmol/l (SD 0.1) to 0.47 µmol/l (SD 0.1) in premenopausal women and from 0.46 µmol/l (SD 0.1) to 0.48 µmol/l (SD 0.1) in postmenopausal women ( P < 0.05). The results suggest that menopause is accompanied by higher plasma apoM but not S1P concentrations and that exercise training increases plasma apoM and S1P in healthy middle-aged women irrespective of menopausal status. NEW & NOTEWORTHY The apolipoprotein M/sphingosine-1-phosphate (apoM/S1P) complex is involved in maintaining a healthy endothelial barrier function. Our study is the first, to our knowledge, to show how menopause affects the apoM/S1P axis. The results suggest that menopause is accompanied by higher plasma apoM but not S1P concentrations. Second, to our knowledge the study is also the first to show that exercise training increases both apoM/S1P in women irrespective of menopausal status.
Collapse
Affiliation(s)
- Adelina Yafasova
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Camilla M. Mandrup
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jon Egelund
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Michael Nyberg
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Bente Stallknecht
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Lars B. Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Bispebjerg Hospital, Copenhagen, Denmark
| |
Collapse
|
46
|
Liu J, Huang H, Shi S, Wang X, Yu Y, Hu Y, Sun J, Ren C, Yang J, Shen Z. Atorvastatin upregulates apolipoprotein M expression via attenuating LXRα expression in hyperlipidemic apoE-deficient mice. Exp Ther Med 2018; 16:3785-3792. [PMID: 30344653 PMCID: PMC6176103 DOI: 10.3892/etm.2018.6694] [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: 02/25/2018] [Accepted: 06/08/2018] [Indexed: 11/16/2022] Open
Abstract
Apolipoprotein M (apoM) is a recently identified human apolipoprotein that is associated with the formation of high-density lipoprotein (HDL). Studies have demonstrated that statins may affect the expression of apoM; however, the regulatory effects of statins on apoM are controversial. Furthermore, the underlying mechanisms by which statins regulate apoM remain unclear. In the present study, in vivo and in vitro models were used to investigate whether the anti-atherosclerotic effects of statins are associated with its apoM-regulating effects and the underlying mechanism. Hyperlipidemia was induced by in apolipoprotein E-deficient mice by providing a high-fat diet. Atorvastatin was administered to hyperlipidemic mice and HepG2 cells to investigate its effect on apoM expression. The liver X receptor α (LXRα) agonist T0901317 was also administered together with atorvastatin to hyperlipidemic mice and HepG2 cells. The results revealed that atorvastatin increased apoM expression, which was accompanied with decreased expression of LXRα in the liver of hyperlipidemic apolipoprotein E-deficient mice and HepG2 cells. Additionally, apoM upregulation was inhibited following treatment with T0901317. In summary, atorvastatin exhibited anti-atherosclerotic effects by upregulating apoM expression in hyperlipidemic mice, which may be mediated by the inhibition of LXRα.
Collapse
Affiliation(s)
- Jian Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Department of Cardiovascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Haoyue Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Sheng Shi
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Department of Cardiovascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Xu Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yunsheng Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yanqiu Hu
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiacheng Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Chuanlu Ren
- Department of Clinical Laboratory, The 100th Hospital of The People's Liberation Army, Suzhou, Jiangsu 215000, P.R. China
| | - Junjie Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Zhenya Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
47
|
Feng YH, Zheng L, Wei J, Yu MM, Zhang J, Luo GH, Xu N. Increased apolipoprotein M induced by lack of scavenger receptor BI is not activated via HDL-mediated cholesterol uptake in hepatocytes. Lipids Health Dis 2018; 17:200. [PMID: 30144814 PMCID: PMC6109342 DOI: 10.1186/s12944-018-0849-7] [Citation(s) in RCA: 7] [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: 04/23/2018] [Accepted: 08/13/2018] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Scavenger receptor BI (SR-BI) is a classic high-density lipoprotein (HDL) receptor, which mediates selective lipid uptake from HDL cholesterol esters (HDL-C). Apolipoprotein M (ApoM), as a component of HDL particles, could influence preβ-HDL formation and cholesterol efflux. The aim of this study was to determine whether SR-BI deficiency influenced the expression of ApoM. METHODS Blood samples and liver tissues were collected from SR-BI gene knockout mice, and serum lipid parameters, including total cholesterol (TC), triglyceride (TG), high and low-density lipoprotein cholesterol (HDL-C and LDL-C) and ApoM were measured. Hepatic ApoM and ApoAI mRNA levels were also determined. In addition, BLT-1, an inhibitor of SR-BI, was added to HepG2 cells cultured with cholesterol and HDL, under serum or serum-free conditions. The mRNA and protein expression levels of ApoM were detected by RT-PCR and western blot. RESULTS We found that increased serum ApoM protein levels corresponded with high hepatic ApoM mRNA levels in both male and female SR-BI-/- mice. Besides, serum TC and HDL-C were also significantly increased. Treatment of HepG2 hepatoma cells with SR-BI specific inhibitor, BLT-1, could up-regulate ApoM expression in serum-containing medium but not in serum-free medium, even in the presence of HDL-C and cholesterol. CONCLUSIONS Results suggested that SR-BI deficiency promoted ApoM expression, but the increased ApoM might be independent from HDL-mediated cholesterol uptake in hepatocytes.
Collapse
Affiliation(s)
- Yue-Hua Feng
- Comprehensive Laboratory, the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Lu Zheng
- Comprehensive Laboratory, the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Jiang Wei
- Comprehensive Laboratory, the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Miao-Mei Yu
- Comprehensive Laboratory, the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Jun Zhang
- Comprehensive Laboratory, the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Guang-Hua Luo
- Comprehensive Laboratory, the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China.
| | - Ning Xu
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lunds University, S-221 85, Lund, Sweden.
| |
Collapse
|
48
|
Simmons S, Erfinanda L, Bartz C, Kuebler WM. Novel mechanisms regulating endothelial barrier function in the pulmonary microcirculation. J Physiol 2018; 597:997-1021. [PMID: 30015354 DOI: 10.1113/jp276245] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary epithelial and vascular endothelial cell layers provide two sequential physical and immunological barriers that together form a semi-permeable interface and prevent alveolar and interstitial oedema formation. In this review, we focus specifically on the continuous endothelium of the pulmonary microvascular bed that warrants strict control of the exchange of gases, fluid, solutes and circulating cells between the plasma and the interstitial space. The present review provides an overview of emerging molecular mechanisms that permit constant transcellular exchange between the vascular and interstitial compartment, and cause, prevent or reverse lung endothelial barrier failure under experimental conditions, yet with a clinical perspective. Based on recent findings and at times seemingly conflicting results we discuss emerging paradigms of permeability regulation by altered ion transport as well as shifts in the homeostasis of sphingolipids, angiopoietins and prostaglandins.
Collapse
Affiliation(s)
- Szandor Simmons
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lasti Erfinanda
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bartz
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Sørensen IM, Bertelsen M, Freese E, Lindhard K, Ullum H, Feldt-Rasmussen B, Nielsen LB, Christoffersen C, Bro S. Apolipoprotein M in patients with chronic kidney disease. Atherosclerosis 2018; 275:304-311. [PMID: 29980057 DOI: 10.1016/j.atherosclerosis.2018.06.815] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 05/21/2018] [Accepted: 06/13/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND AIMS Plasma apolipoprotein M (APOM) is bound to HDL-particles and has anti-atherogenic effects. The present study explored whether plasma APOM is reduced in patients with chronic kidney disease (CKD), and associated with cardiovascular disease (CVD). In addition, we tested the hypothesis that the excretion of APOM into the urine is increased in patients with kidney disease. METHODS Plasma samples were collected from a cohort of patients with CKD stages 1 to 5D (N = 409) and controls (N = 35). Urine was collected from 47 subjects. Plasma APOM was measured with sandwich ELISA and urine APOM with competitive ELISA. RESULTS Plasma APOM levels were reduced in patients with CKD stages 3-5D as compared to patients with CKD stages 1 + 2 and controls (p < 0.01). CKD patients with known CVD displayed even further reduction in plasma APOM levels than CKD patients without known CVD (p < 0.001). Fast-phase liquid chromatography showed that plasma APOM was primarily associated with HDL-cholesterol (HDL-C) across CKD stages. Accordingly, when plasma APOM values were corrected for HDL-C, a significant difference only persisted between patients with CKD stage 3 and stages 1 + 2 (p < 0.05), and the difference between CKD patients with and without known CVD disappeared. Urine APOM/creatinine ratio was not significantly increased in patients with kidney disease. CONCLUSIONS The results show that the difference in plasma APOM levels observed between patients with mild and advanced CKD may mainly be due to differences in plasma HDL-C. Whether APOM plays a role in human uremic atherogenesis warrants further experimental studies.
Collapse
Affiliation(s)
- Ida Mh Sørensen
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
| | | | - Ellen Freese
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
| | | | - Henrik Ullum
- Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark
| | | | - Lars Bo Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry, Bispebjerg Hospital, Copenhagen, Denmark
| | - Susanne Bro
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
50
|
Christoffersen C, Federspiel CK, Borup A, Christensen PM, Madsen AN, Heine M, Nielsen CH, Kjaer A, Holst B, Heeren J, Nielsen LB. The Apolipoprotein M/S1P Axis Controls Triglyceride Metabolism and Brown Fat Activity. Cell Rep 2018; 22:175-188. [DOI: 10.1016/j.celrep.2017.12.029] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/25/2017] [Accepted: 12/07/2017] [Indexed: 01/22/2023] Open
|