1
|
Dunn A, Ditmarsch M, Kastelein JJP, Kling D, Neild A, Davidson MH, Gobburu J. Guiding Dyslipidemia Treatment: A Population Pharmacokinetic-Pharmacodynamic Framework for Obicetrapib. J Clin Pharmacol 2024; 64:1150-1164. [PMID: 38720593 DOI: 10.1002/jcph.2448] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 08/29/2024]
Abstract
Obicetrapib is a selective inhibitor of cholesteryl ester transfer protein that is currently in phase 3 of development for the treatment of dyslipidemia as adjunct therapy. The purpose of this study was to comprehensively characterize the pharmacokinetic (PK) and pharmacodynamic (PD) disposition of obicetrapib. Data from 7 clinical trials conducted in healthy adults and those with varying degrees of dyslipidemia were included for model development. The structural model that best described obicetrapib PK was a 3-compartment model with 4-compartment transit absorption and first-order elimination. Body weight was the only covariate found to significantly explain observed variability and was therefore included using allometric scaling on all disposition parameters. For a typical patient weighing 75 kg, the estimated apparent total body clearance and apparent volume of distribution of the central compartment was 0.81 L/h and 36.1 L, respectively. The final PK model parameters were estimated with good precision and were ultimately leveraged to sequentially inform 2 turnover models that describe obicetrapib's effect on low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) concentrations. The maximum stimulatory effect of obicetrapib on LDL-C loss was estimated to be 1.046, while the maximum inhibitory effect of obicetrapib on HDL-C loss was 0.691. This corresponds to a predicted typical maximum percent change from baseline LDL-C and HDL-C of 51.1% and 224%, respectively. The final sequential model described obicetrapib PKPD well and was ultimately able to both demonstrate evidence of internal consistency and support decision-making throughout the development lifecycle.
Collapse
Affiliation(s)
- Allison Dunn
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | | | - John J P Kastelein
- NewAmsterdam Pharma B.V., Naarden, The Netherlands
- Academic Medical Center, Amsterdam, The Netherlands
| | | | - Annie Neild
- NewAmsterdam Pharma B.V., Naarden, The Netherlands
| | | | - Joga Gobburu
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
2
|
Guo Y, Luo L, Zhu J, Li C. Advance in Multi-omics Research Strategies on Cholesterol Metabolism in Psoriasis. Inflammation 2024; 47:839-852. [PMID: 38244176 DOI: 10.1007/s10753-023-01961-9] [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: 09/24/2023] [Revised: 11/29/2023] [Accepted: 12/25/2023] [Indexed: 01/22/2024]
Abstract
The skin is a complex and dynamic organ where homeostasis is maintained through the intricate interplay between the immune system and metabolism, particularly cholesterol metabolism. Various factors such as cytokines, inflammatory mediators, cholesterol metabolites, and metabolic enzymes play crucial roles in facilitating these interactions. Dysregulation of this delicate balance contributes to the pathogenic pathways of inflammatory skin conditions, notably psoriasis. In this article, we provide an overview of omics biomarkers associated with psoriasis in relation to cholesterol metabolism. We explore multi-omics approaches that reveal the communication between immunometabolism and psoriatic inflammation. Additionally, we summarize the use of multi-omics strategies to uncover the complexities of multifactorial and heterogeneous inflammatory diseases. Finally, we highlight potential future perspectives related to targeted drug therapies and research areas that can advance precise medicine. This review aims to serve as a valuable resource for those investigating the role of cholesterol metabolism in psoriasis.
Collapse
Affiliation(s)
- Youming Guo
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Lingling Luo
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
| | - Jing Zhu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
| | - Chengrang Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China.
| |
Collapse
|
3
|
Nagesh PT, Nishi H, Rawal S, Zahr T, Miano JM, Sorci-Thomas M, Xu H, Akbar N, Choudhury RP, Misra A, Fisher EA. HDL regulates TGFß-receptor lipid raft partitioning, restoring contractile features of cholesterol-loaded vascular smooth muscle cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.562786. [PMID: 37905061 PMCID: PMC10614922 DOI: 10.1101/2023.10.19.562786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background Cholesterol-loading of mouse aortic vascular smooth muscle cells (mVSMCs) downregulates miR-143/145, a master regulator of the contractile state downstream of TGFβ signaling. In vitro, this results in transitioning from a contractile mVSMC to a macrophage-like state. This process likely occurs in vivo based on studies in mouse and human atherosclerotic plaques. Objectives To test whether cholesterol-loading reduces VSMC TGFβ signaling and if cholesterol efflux will restore signaling and the contractile state in vitro and in vivo. Methods Human coronary artery (h)VSMCs were cholesterol-loaded, then treated with HDL (to promote cholesterol efflux). For in vivo studies, partial conditional deletion of Tgfβr2 in lineage-traced VSMC mice was induced. Mice wild-type for VSMC Tgfβr2 or partially deficient (Tgfβr2+/-) were made hypercholesterolemic to establish atherosclerosis. Mice were then treated with apoA1 (which forms HDL). Results Cholesterol-loading of hVSMCs downregulated TGFβ signaling and contractile gene expression; macrophage markers were induced. TGFβ signaling positively regulated miR-143/145 expression, increasing Acta2 expression and suppressing KLF4. Cholesterol-loading localized TGFβ receptors into lipid rafts, with consequent TGFβ signaling downregulation. Notably, in cholesterol-loaded hVSMCs HDL particles displaced receptors from lipid rafts and increased TGFβ signaling, resulting in enhanced miR-145 expression and decreased KLF4-dependent macrophage features. ApoA1 infusion into Tgfβr2+/- mice restored Acta2 expression and decreased macrophage-marker expression in plaque VSMCs, with evidence of increased TGFβ signaling. Conclusions Cholesterol suppresses TGFβ signaling and the contractile state in hVSMC through partitioning of TGFβ receptors into lipid rafts. These changes can be reversed by promotion of cholesterol efflux, consistent with evidence in vivo.
Collapse
Affiliation(s)
- Prashanth Thevkar Nagesh
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Hitoo Nishi
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Shruti Rawal
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Tarik Zahr
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Joseph M Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Mary Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Hao Xu
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Oxford University Hospitals, NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Oxford University Hospitals, NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, NSW, Australia
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| |
Collapse
|
4
|
Yang Y, Zhang J, Jia L, Su J, Ma M, Lin X. The interaction between uric acid and high-density lipoprotein cholesterol on the prognosis of patients with acute myocardial infarction. Front Cardiovasc Med 2023; 10:1226108. [PMID: 37492158 PMCID: PMC10363914 DOI: 10.3389/fcvm.2023.1226108] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Background The significance of uric acid (UA) and high-density lipoprotein cholesterol (HDL-C) in the prognosis of acute myocardial infarction (AMI) remains controversial. This study investigated the effect of the interaction between UA and HDL-C on the prognosis of patients with AMI. Methods In total, 480 patients with AMI were included in this study. Baseline and follow-up data were collected, and the primary endpoint was major adverse cardiovascular events (MACE). The secondary endpoint was all-cause death. Both additive and multiplicative interactions were calculated to evaluate their interaction with prognosis. Then, the impact of UA and HDL-C ratio (UHR) on prognosis was assessed. Results Over a median follow-up period of 41 (30,46) months, 136 (28.3%) MACEs, and 44 (9.2%) deaths were recorded. There was a positive additive interaction between UA and HDL-C for MACEs. The attributable proportion (AP) showed that 46% of the estimated effect (MACE in patients) was attributable to this interaction. The synergy index (SI) was 2.04 (1.07,3.88) for MACE, indicating that the risk for patients presenting with both risk factors was greater than the sum of the risk factors alone. Multivariate Cox regression analysis revealed that UHR independently predicted MACEs and mortality. Kaplan-Meier survival curves according to tertiles of UHR showed statistically significant differences in MACE (log-rank test, P < 0.001). Receiver operating characteristic (ROC) analysis showed that the area under the curve (AUC) of UHR for predicting MACE was 0.716. Conclusion The coexistence of high UA and low HDL-C has a synergistic effect and provides further information for risk stratification of patients with AMI. UHR is a simple and easily available prognostic indicator independent of traditional risk factors.
Collapse
|
5
|
Abstract
Dyslipidemia is an important risk factor for coronary artery disease and stroke. All persons with dyslipidemia should be advised to focus on lifestyle interventions, including regular aerobic exercise, a healthy diet, maintenance of a healthy weight, and abstinence from smoking. In addition to lifestyle interventions, lipid-lowering therapy should be considered for persons at moderate to high risk for atherosclerotic cardiovascular disease based on validated risk equations. Statin therapy is the first-line medical treatment for dyslipidemia due to its effectiveness and favorable adverse effect profile, but newer treatments provide additional tools for clinicians to effectively treat dyslipidemia.
Collapse
Affiliation(s)
- Marios Arvanitis
- Johns Hopkins University School of Medicine, Baltimore, Maryland (M.A., C.J.L.)
| | | |
Collapse
|
6
|
Stadler JT, van Poppel MNM, Wadsack C, Holzer M, Pammer A, Simmons D, Hill D, Desoye G, Marsche G. Obesity Affects Maternal and Neonatal HDL Metabolism and Function. Antioxidants (Basel) 2023; 12:antiox12010199. [PMID: 36671061 PMCID: PMC9854613 DOI: 10.3390/antiox12010199] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Pregravid obesity is one of the major risk factors for pregnancy complications such as gestational diabetes mellitus (GDM) and an increased risk of cardiovascular events in children of affected mothers. However, the biological mechanisms that underpin these adverse outcomes are not well understood. High-density lipoproteins (HDLs) are antiatherogenic by promoting the efflux of cholesterol from macrophages and by suppression of inflammation. Functional impairment of HDLs in obese and GDM-complicated pregnancies may have long-term effects on maternal and offspring health. In the present study, we assessed metrics of HDL function in sera of pregnant women with overweight/obesity of the DALI lifestyle trial (prepregnancy BMI ≥ 29 kg/m2) and women with normal weight (prepregnancy BMI < 25 kg/m2), as well as HDL functionalities in cord blood at delivery. We observed that pregravid obesity was associated with impaired serum antioxidative capacity and lecithin−cholesterol acyltransferase activity in both mothers and offspring, whereas maternal HDL cholesterol efflux capacity was increased. Interestingly, functionalities of maternal and fetal HDL correlated robustly. GDM did not significantly further alter the parameters of HDL function and metabolism in women with obesity, so obesity itself appears to have a major impact on HDL functionality in mothers and their offspring.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, 8010 Graz, Austria
| | - Mireille N. M. van Poppel
- Institute of Human Movement Science, Sport and Health, University of Graz, 8010 Graz, Austria
- Correspondence: (M.N.M.v.P.); (G.M.); Tel.: +43-(0)-316-380-2335 (M.N.M.v.P.); +43-316-385-74128 (G.M.)
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Michael Holzer
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, 8010 Graz, Austria
| | - Anja Pammer
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, 8010 Graz, Austria
| | - David Simmons
- Macarthur Clinical School, Western Sydney University, Sydney, NSW 2560, Australia
| | - David Hill
- Lawson Health Research Institute, London, ON N6C 2R5, Canada
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, 8010 Graz, Austria
- Correspondence: (M.N.M.v.P.); (G.M.); Tel.: +43-(0)-316-380-2335 (M.N.M.v.P.); +43-316-385-74128 (G.M.)
| | | |
Collapse
|
7
|
Cui K, Gao X, Wang B, Wu H, Arulsamy K, Dong Y, Xiao Y, Jiang X, Malovichko MV, Li K, Peng Q, Lu YW, Zhu B, Zheng R, Wong S, Cowan DB, Linton M, Srivastava S, Shi J, Chen K, Chen H. Epsin Nanotherapy Regulates Cholesterol Transport to Fortify Atheroma Regression. Circ Res 2023; 132:e22-e42. [PMID: 36444722 PMCID: PMC9822875 DOI: 10.1161/circresaha.122.321723] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Excess cholesterol accumulation in lesional macrophages elicits complex responses in atherosclerosis. Epsins, a family of endocytic adaptors, fuel the progression of atherosclerosis; however, the underlying mechanism and therapeutic potential of targeting Epsins remains unknown. In this study, we determined the role of Epsins in macrophage-mediated metabolic regulation. We then developed an innovative method to therapeutically target macrophage Epsins with specially designed S2P-conjugated lipid nanoparticles, which encapsulate small-interfering RNAs to suppress Epsins. METHODS We used single-cell RNA sequencing with our newly developed algorithm MEBOCOST (Metabolite-mediated Cell Communication Modeling by Single Cell Transcriptome) to study cell-cell communications mediated by metabolites from sender cells and sensor proteins on receiver cells. Biomedical, cellular, and molecular approaches were utilized to investigate the role of macrophage Epsins in regulating lipid metabolism and transport. We performed this study using myeloid-specific Epsin double knockout (LysM-DKO) mice and mice with a genetic reduction of ABCG1 (ATP-binding cassette subfamily G member 1; LysM-DKO-ABCG1fl/+). The nanoparticles targeting lesional macrophages were developed to encapsulate interfering RNAs to treat atherosclerosis. RESULTS We revealed that Epsins regulate lipid metabolism and transport in atherosclerotic macrophages. Inhibiting Epsins by nanotherapy halts inflammation and accelerates atheroma resolution. Harnessing lesional macrophage-specific nanoparticle delivery of Epsin small-interfering RNAs, we showed that silencing of macrophage Epsins diminished atherosclerotic plaque size and promoted plaque regression. Mechanistically, we demonstrated that Epsins bound to CD36 to facilitate lipid uptake by enhancing CD36 endocytosis and recycling. Conversely, Epsins promoted ABCG1 degradation via lysosomes and hampered ABCG1-mediated cholesterol efflux and reverse cholesterol transport. In a LysM-DKO-ABCG1fl/+ mouse model, enhanced cholesterol efflux and reverse transport due to Epsin deficiency was suppressed by the reduction of ABCG1. CONCLUSIONS Our findings suggest that targeting Epsins in lesional macrophages may offer therapeutic benefits for advanced atherosclerosis by reducing CD36-mediated lipid uptake and increasing ABCG1-mediated cholesterol efflux.
Collapse
Affiliation(s)
- Kui Cui
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Xinlei Gao
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Beibei Wang
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Hao Wu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Kulandaisamy Arulsamy
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Yunzhou Dong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Yuling Xiao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Xingya Jiang
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Marina V. Malovichko
- Division of Environmental Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Kathryn Li
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Qianman Peng
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Yao Wei Lu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Bo Zhu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Rongbin Zheng
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Scott Wong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Douglas B. Cowan
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - MacRae Linton
- Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center; Nashville, TN, 37232, USA
| | - Sanjay Srivastava
- Division of Environmental Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Kaifu Chen
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| |
Collapse
|
8
|
Stadler JT, Mangge H, Rani A, Curcic P, Herrmann M, Prüller F, Marsche G. Low HDL Cholesterol Efflux Capacity Indicates a Fatal Course of COVID-19. Antioxidants (Basel) 2022; 11:1858. [PMID: 36290581 PMCID: PMC9598682 DOI: 10.3390/antiox11101858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/02/2022] Open
Abstract
Plasma membrane cholesterol is required for proper trafficking and localization of receptors that facilitate severe acute respiratory syndrome coronavirus 2 infection. High-density lipoproteins (HDL) mobilize plasma membrane cholesterol, and HDL-cholesterol levels are associated with the severity of COVID-19 disease and mortality. However, HDL-cholesterol levels poorly reflect the function of this complex family of particles, and a detailed assessment of COVID-19-associated changes in HDL functionality and its prognostic value is lacking. In the present study, we assessed HDL cholesterol efflux capacity, HDL anti-inflammatory and antioxidant properties, and changes in HDL composition and metabolism in COVID-19 (n = 48) and non-COVID pneumonia patients (n = 32). COVID-19 infection markedly reduced the activity of lecithin-cholesteryl-acyltransferase and functional parameters of HDL, such as the cholesterol efflux capacity, arylesterase activity of paraoxonase 1, and anti-oxidative capacity of apoB-depleted serum when compared to non-COVID pneumonia at baseline, paralleled by markedly reduced levels of HDL-cholesterol. Of particular interest, low HDL cholesterol efflux capacity was associated with increased mortality risk in COVID-19 patients, independent of HDL-C levels. Our results highlight profound effects of COVID-19 infection on HDL function, metabolism, and composition. Low HDL cholesterol efflux capacity indicates a fatal course of COVID-19, independent of HDL-cholesterol levels.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Pero Curcic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Florian Prüller
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| |
Collapse
|
9
|
Park S, Jang M, Park MY, Kim J, Shin S. Interactive effects of the low-carbohydrate diet score and genetic risk score on Hypo-HDL-cholesterolemia among Korean adults: A cross-sectional analysis from the Ansan and Ansung Study of the Korean Genome and Epidemiology Study. Food Sci Nutr 2022; 10:3106-3116. [PMID: 36171780 PMCID: PMC9469851 DOI: 10.1002/fsn3.2909] [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] [Indexed: 11/13/2022] Open
Abstract
This cross-sectional study investigated the interaction between the genetic risk score (GRS) and abnormal high-density lipoprotein (HDL) cholesterol lipid levels, which are modified by low-carbohydrate diets (LCDs) and their effects on the prevalence of hypo-HDL-cholesterolemia (hypo-HDL-C) in Korean adults. Baseline data were obtained from the Ansan and Ansung study of the Korean Genome and Epidemiology Study (KoGES), conducted from 2001 to 2002, that targeted 8,314 Korean adults aged 40-69 years, including old men (47.6%) and women (52.4%), and whole genomic single nucleotide polymorphism (SNP) genotyping was performed. We identified 18 SNPs significantly associated with hypo-HDL-C in the proximity of several genes, including LPL, APOA5, LIPC, and CETP, and calculated the GRS. The low-carbohydrate diet score (LCDS) was calculated on the basis of energy intake information from food frequency questionnaires. Furthermore, we performed multivariable-adjusted logistic modeling to examine the odds ratio (OR) for hypo-HDL-C across tertiles of LCDS and GRS, adjusted for several covariates. Among participants in the highest GRS tertile, those in the highest tertile of the LCDS had a significantly lower risk of hypo-HDL-C (OR: 0.759, 95% CI (confidence interval): 0.625-0.923) than those in the lowest tertile of the LCDS. In the joint effect model, the group with the lowest GRS and highest LCDS was found to have the lowest risk of hypo-HDL-C prevalence. This study suggests that individuals with a high genetic risk for low HDL concentrations may have a beneficial effect on a lower intake of carbohydrates.
Collapse
Affiliation(s)
- SoHyun Park
- Department of Food and NutritionChung‐Ang UniversityGyeonggi‐doKorea
| | - Min‐Jae Jang
- Department of Animal Science and TechnologyChung‐Ang UniversityGyeonggi‐doKorea
| | - Min Young Park
- Department of Molecular PathobiologyNYU College of DentistryNew YorkNew YorkUSA
| | - Jun‐Mo Kim
- Department of Animal Science and TechnologyChung‐Ang UniversityGyeonggi‐doKorea
| | - Sangah Shin
- Department of Food and NutritionChung‐Ang UniversityGyeonggi‐doKorea
| |
Collapse
|
10
|
Sirtori CR, Corsini A, Ruscica M. The Role of High-Density Lipoprotein Cholesterol in 2022. Curr Atheroscler Rep 2022; 24:365-377. [PMID: 35274229 PMCID: PMC8913032 DOI: 10.1007/s11883-022-01012-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE OF THE REVIEW High-density lipoproteins (HDL) are responsible for the transport in plasma of a large fraction of circulating lipids, in part from tissue mobilization. The evaluation of HDL-associated cholesterol (HDL-C) has provided a standard method for assessing cardiovascular (CV) risk, as supported by many contributions on the mechanism of this arterial benefit. The present review article will attempt to investigate novel findings on the role and mechanism of HDL in CV risk determination. RECENT FINDINGS The most recent research has been aimed to the understanding of how a raised functional capacity of HDL, rather than elevated levels per se, may be responsible for the postulated CV protection. Markedly elevated HDL-C levels appear instead to be associated to a raised coronary risk, indicative of a U-shaped relationship. While HDL-C reduction is definitely related to a raised CV risk, HDL-C elevations may be linked to non-vascular diseases, such as age-related macular disease. The description of anti-inflammatory, anti-oxidative and anti-infectious properties has indicated potential newer areas for diagnostic and therapeutic approaches. In the last two decades inconclusive data have arisen from clinical trials attempting to increase HDL-C pharmacologically or by way of recombinant protein infusions (most frequently with the mutant A-I Milano); prevention of stent occlusion or heart failure treatment have shown instead significant promise. Targeted clinical studies are still ongoing.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy.
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
11
|
Abstract
Stroke remains a leading cause of death and disability, with limited therapeutic options and suboptimal tools for diagnosis and prognosis. High throughput technologies such as proteomics generate large volumes of experimental data at once, thus providing an advanced opportunity to improve the status quo by facilitating identification of novel therapeutic targets and molecular biomarkers. Proteomics studies in animals are largely designed to decipher molecular pathways and targets altered in brain tissue after stroke, whereas studies in human patients primarily focus on biomarker discovery in biofluids and, more recently, in thrombi and extracellular vesicles. Here, we offer a comprehensive review of stroke proteomics studies conducted in both animal and human specimen and present our view on limitations, challenges, and future perspectives in the field. In addition, as a unique resource for the scientific community, we provide extensive lists of all proteins identified in proteomic studies as altered by stroke and perform postanalysis of animal data to reveal stroke-related cellular processes and pathways.
Collapse
Affiliation(s)
- Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (K.H.)
| | - Wei Yang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University School of Medicine, Durham, NC (W.Y.)
| |
Collapse
|
12
|
The effect of a low carbohydrate ketogenic diet with or without exercise on postpartum weight retention, metabolic profile and physical activity performance in postpartum mice. J Nutr Biochem 2022; 102:108941. [PMID: 35017000 DOI: 10.1016/j.jnutbio.2022.108941] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/28/2021] [Accepted: 12/07/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE the present study examined the effect of the isocaloric low-carbohydrate ketogenic diet (LCKD) with or without exercise training for 6 weeks on postpartum weight retention (PPWR), body composition, metabolic profile and physical activity performance in postpartum mice. METHODS postpartum mice were assigned to 4 groups (n=8/group) as follows: (1) those on a control diet without aerobic exercise (CN); (2) those on a control diet with aerobic exercise (CN+EX), (3); those on a LCKD without aerobic exercise (LCKD); (4) those on a LCKD with aerobic exercise (LCKD+EX). CN+EX and LCKD+EX mice performed 6 weeks of exercise training on a treadmill. After the 6-week intervention, physical activity performance was determined. RESULTS postpartum mice in all groups experienced progressive reductions in body weight over the study period. The LCKD group had the smallest reduction in PPWR (p<0.05). The LCKD group had significantly higher total cholesterol, low-density lipoprotein cholesterol and lactate dehydrogenase levels, and liver lipid concentrations with a worsened glucose tolerance, compared to the CN group (p<0.05). The LCKD group showed significant reductions in physical activity performance, whilst the LCKD+EX group showed significantly improvement in endurance performance, and paralleled the concomitant elevation in blood ketone levels. CONCLUSIONS 6-week LCKD feeding on its own was less effective for reducing PPWR, and more detrimental to postpartum metabolic outcomes and physical activity performance of the postpartum mice. The feasibility of a LCKD with or without exercise during the postpartum period as a strategy for managing PPWR and improving postpartum metabolic profiles should be carefully considered.
Collapse
|
13
|
Ferrari F, Santos RD. Physical Activity and HDL-C: Are There Gender Differences in the Dose-response Effect? Arq Bras Cardiol 2021; 117:501-502. [PMID: 34550235 PMCID: PMC8462943 DOI: 10.36660/abc.20210551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Filipe Ferrari
- Programa de Pós-graduação em Cardiologia e Ciências Cardiovasculares - Universidade Federal do Rio Grande do Sul (UFRGS), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS - Brasil.,Grupo de Pesquisa em Cardiologia do Exercício (CardioEx) - UFRGS, HCPA, Porto Alegre, RS - Brasil
| | - Raul D Santos
- Unidade Clínica de Lípides do Instituto do Coração (InCor) - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), São Paulo, SP - Brasil.,Hospital Israelita Albert Einstein, São Paulo, SP - Brasil
| |
Collapse
|
14
|
Franczyk B, Rysz J, Ławiński J, Rysz-Górzyńska M, Gluba-Brzózka A. Is a High HDL-Cholesterol Level Always Beneficial? Biomedicines 2021; 9:1083. [PMID: 34572269 PMCID: PMC8466913 DOI: 10.3390/biomedicines9091083] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 01/02/2023] Open
Abstract
The specific interest concerning HDL cholesterol (HDL-C) is related to its ability to uptake and return surplus cholesterol from peripheral tissues back to the liver and, therefore, to its role in the prevention of cardiovascular diseases, such as atherosclerosis and myocardial infarction, but also transient ischemic attack and stroke. Previous epidemiological studies have indicated that HDL-C concentration is inversely associated with the risk of cardiovascular disease and that it can be used for risk prediction. Some genetic disorders are characterized by markedly elevated levels of HDL-C; however, they do not translate into diminished cardiovascular risk. The search of the potential causative relationship between HDL-C and adverse events has shifted the attention of researchers towards the composition and function of the HDL molecule/subfractions. HDL possesses various cardioprotective properties. However, currently, it appears that higher HDL-C is not necessarily protective against cardiovascular disease, but it can even be harmful in extremely high quantities.
Collapse
Affiliation(s)
- Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| | - Janusz Ławiński
- Department of Urology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-549 Rzeszow, Poland;
| | - Magdalena Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| |
Collapse
|
15
|
Effect of a High-Protein High-Fibre Nutritional Supplement on Lipid Profile in Overweight/Obese Adults with Type 2 Diabetes Mellitus: A 24-Week Randomized Controlled Trial. J Nutr Metab 2021; 2021:6634225. [PMID: 33953977 PMCID: PMC8064784 DOI: 10.1155/2021/6634225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/19/2021] [Accepted: 04/01/2021] [Indexed: 12/01/2022] Open
Abstract
Background Foods rich in protein and dietary fibre could potentially improve lipid profile in overweight or obese diabetic patients with dyslipidemia and, thereby, mitigate their risk of cardiovascular disease (CVD). In this study, the effect of providing high-protein high-fibre (HPHF) nutritional supplement in addition to standard care of type 2 diabetes mellitus (T2DM) on lipid profile was evaluated. Methods In this open-label, parallel-arm, prospective, randomized study, a total of 100 overweight/obese participants with T2DM were randomized to either an intervention group (25 g HPHF nutritional supplement given twice daily along with a standard care of T2DM) or a control group (standard care of T2DM) for 24 weeks. Change from baseline in lipid parameters such as total cholesterol (TChol), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG) was assessed between the intervention and control group at week 12 and week 24. Participant compliance was assessed using the dietary 24-hour recall. Statistical analysis was performed to assess the main effects on within- and between-group changes from baseline to end of 24 weeks. Results Participants in the HPHF nutritional supplement group showed a statistically significant improvement in HDL-C levels by the end of 24 weeks (p=0.04) and a significant increase in protein and total dietary fibre intake (p=0.002 and p=0.00, respectively) compared to the control group. The TChol/HDL-C ratio was significantly lower (p=0.03) in the HPHF group from baseline to 24 weeks. Conclusion Twice-daily consumption of a HPHF nutritional supplement significantly improved HDL-C levels. Inclusion of the HPHF supplement would be a useful effective aid for managing dyslipidemia in overweight/obese individuals with T2DM.
Collapse
|
16
|
Nogoy KMC, Kim HJ, Lee DH, Smith SB, Seong HA, Choi SH. Oleic acid in Angus and Hanwoo (Korean native cattle) fat reduced the fatty acid synthase activity in rat adipose tissues. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 63:380-393. [PMID: 33987612 PMCID: PMC8071735 DOI: 10.5187/jast.2021.e4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/03/2020] [Accepted: 10/20/2020] [Indexed: 12/04/2022]
Abstract
This study aimed to determine the blood lipid profiles, fatty acid composition,
and lipogenic enzyme activities in rat adipose tissues as affected by the Angus
beef fat (ABF) and Hanwoo beef fat (HBF) containing high oleic acid (OA)
content. We assigned 60 Sprague Dawley rats with a mean bodyweight of 249
± 3.04 g to three groups (n = 20 each) to receive diets containing 7%
coconut oil (CON), 7% ABF, or 7% HBF. The OA content was highest in the HBF
(45.23%) followed by ABF (39.51%) and CON (6.10%). The final body weight of the
HBF-fed group was significantly increased, probably due to increased feed
intake, indicating the palatability of the diet. The HBF and ABF significantly
increased high-density lipoprotein cholesterol (HDL-C), decreased triglyceride
(TG) and total cholesterol (TC) levels, and also tended to attenuate glutamic
oxaloacetic transaminase (GOT) and glutamic pyruvic transaminase (GPT) levels in
the bloodstream of the rats compared to CON. As compared to CON, lauric,
myristic, and palmitic acids were significantly lower, and those of OA and
α-linolenic acid (ALA) were significantly higher in the adipose tissues
of HBF and ABF-fed groups. The HBF and ABF also reduced lipogenesis as induced
by depleted fatty acid synthase (FAS) activity in rat adipose tissues.
Nevertheless, between the two fats, HBF showed high feed intake due to its high
palatability but reduced lipogenic enzyme activity, specifically that of FAS,
and increased HDL-C, decreased TC and TG levels in the bloodstream, reduced
saturated fatty acids (SFA), and increased oleic and ALA contents in rat adipose
tissues indicating that HBF consumption does not pose significant risks of
cardiovascular disease.
Collapse
Affiliation(s)
| | - Hyoun Ju Kim
- National Institute of Animal Science, Wanju 55365, Korea
| | - Dong Hoon Lee
- Department of Biosystems Engineering, Chungbuk National University, Cheongju 28644, Korea
| | - Stephen B Smith
- Department of Animal Science, Texas A&M University, College Station, Texas 77843, USA
| | - Hyun A Seong
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Korea
| | - Seong Ho Choi
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
17
|
Ibrahim RYM, Saber AA, Hammad HBI. The possible role of the seaweed Ulva fasciata on ameliorating hyperthyroidism-associated heart inflammations in a rat model. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:6830-6842. [PMID: 33011948 DOI: 10.1007/s11356-020-11036-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/28/2020] [Indexed: 06/11/2023]
Abstract
Cardiovascular diseases are key complications primarily associated with hyperthyroidism disorders. The present study sought to ameliorate hyperthyroidism-mediated cardiovascular inflammations and related oxidative stress paradigms in experimental rats using the broadly distributed green seaweed Ulva fasciata. Forty-eight adult male albino rats were recruited and randomly classified into six groups. Hyperthyroidism was stimulated using L-thyroxine sodium at a dose of 100 μg/kg i.p. for 3 weeks daily. Further, 200 mg/kg b.wt. concentration of the U. fasciata methanolic (U. fasciata-MeOH) extract was the recommended dose and administrated orally to the hyperthyroid rats. The standard commercial drug "propranolol hydrochloride" was also tested at a dose of 10 mg/kg i.p. to compare the findings obtained from the seaweed extract. A combined treatment with the U. fasciata-MeOH extract and propranolol hydrochloride was also assessed. Our results implied that the treatment of hyperthyroid rats with the U. fasciata-MeOH extract significantly reduced serum levels of the thyroid hormones T3 and T4, proinflammatory cytokines (TNF-α, MPO, and CRP), triglycerides and total cholesterol, as well as the cardiac biomarkers CK-MB, LDH, and troponin to thresholds close to those of the standard drug. In addition, levels of high-density lipoprotein cholesterol (HDL-C) and interleukin 10 (IL-10) were significantly upregulated. Hyperthyroid rats only treated with propranolol hydrochloride, or with a combination of the drug and the seaweed extract, conferred the same observations. Histopathological architecture boosted our interesting findings where the myocardium tissues in hyperthyroid rats, administrated the U. fasciata-MeOH extract or/and propranolol hydrochloride, exhibited more or less a normal structure as the control, reflecting the potential cardiovascular recovery exerted by this seaweed extract. In vitro DPPH, ABTS, and FRAP antioxidant assays of the U. fasciata-MeOH extract showed an outstanding ROS-scavenging potential. HPLC analysis of the U. fasciata-MeOH extract unraveled an inestimable valuable array of phenolics (mainly p-coumaric, gallic, ferulic, chlorogenic, and syringic acids) and flavonoids (hesperidin, kaempferol, catechin, quercetin, and rutin). Conclusively, the seaweed U. fasciata is a profitable source of antioxidant polyphenolics characterized by having a pharmaceutical potential against hyperthyroidism-linked cardiovascular inflammations and oxidative stress patterns due to their substantial free radical quenching properties, and also via regulating the signalling pathways of the proinflammatory, lipid profile, and cardiac biomarkers.
Collapse
Affiliation(s)
| | - Abdullah Antar Saber
- Botany Department, Faculty of Science, Ain Shams University, Abbassia Square, Cairo, 11566, Egypt.
| | | |
Collapse
|
18
|
Effects of dietary saturated fatty acids on serum high density lipoprotein, non-high-density lipoprotein and remnant-cholesterol in the Reading Imperial Surrey Saturated fat Cholesterol Intervention (RISSCI-1) study. Proc Nutr Soc 2021. [DOI: 10.1017/s0029665121003396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
19
|
Azeez T. Diabesity lipid index: A potential novel marker of 10-year cardiovascular risk. VASCULAR INVESTIGATION AND THERAPY 2021. [DOI: 10.4103/2589-9686.321923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
20
|
Athinarayanan SJ, Hallberg SJ, McKenzie AL, Lechner K, King S, McCarter JP, Volek JS, Phinney SD, Krauss RM. Impact of a 2-year trial of nutritional ketosis on indices of cardiovascular disease risk in patients with type 2 diabetes. Cardiovasc Diabetol 2020; 19:208. [PMID: 33292205 PMCID: PMC7724865 DOI: 10.1186/s12933-020-01178-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/15/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND We have previously reported that in patients with type 2 diabetes (T2D) consumption of a very low carbohydrate diet capable of inducing nutritional ketosis over 2 years (continuous care intervention, CCI) resulted in improved body weight, glycemic control, and multiple risk factors for cardiovascular disease (CVD) with the exception of an increase in low density lipoprotein cholesterol (LDL-C). In the present study, we report the impact of this intervention on markers of risk for atherosclerotic cardiovascular disease (CVD), with a focus on lipoprotein subfraction particle concentrations as well as carotid-artery intima-media thickness (CIMT). METHODS Analyses were performed in patients with T2D who completed 2 years of this study (CCI; n = 194; usual care (UC): n = 68). Lipoprotein subfraction particle concentrations were measured by ion mobility at baseline, 1, and 2 years and CIMT was measured at baseline and 2 years. Principal component analysis (PCA) was used to assess changes in independent clusters of lipoprotein particles. RESULTS At 2 years, CCI resulted in a 23% decrease of small LDL IIIb and a 29% increase of large LDL I with no change in total LDL particle concentration or ApoB. The change in proportion of smaller and larger LDL was reflected by reversal of the small LDL subclass phenotype B in a high proportion of CCI participants (48.1%) and a shift in the principal component (PC) representing the atherogenic lipoprotein phenotype characteristic of T2D from a major to a secondary component of the total variance. The increase in LDL-C in the CCI group was mainly attributed to larger cholesterol-enriched LDL particles. CIMT showed no change in either the CCI or UC group. CONCLUSION Consumption of a very low carbohydrate diet with nutritional ketosis for 2 years in patients with type 2 diabetes lowered levels of small LDL particles that are commonly increased in diabetic dyslipidemia and are a marker for heightened CVD risk. A corresponding increase in concentrations of larger LDL particles was responsible for higher levels of plasma LDL-C. The lack of increase in total LDL particles, ApoB, and in progression of CIMT, provide supporting evidence that this dietary intervention did not adversely affect risk of CVD.
Collapse
Affiliation(s)
| | - Sarah J Hallberg
- Virta Health, 501 Folsom Street, San Francisco, CA, 94105, USA
- Indiana University Health Arnett, Lafayette, IN, USA
- Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Amy L McKenzie
- Virta Health, 501 Folsom Street, San Francisco, CA, 94105, USA
| | - Katharina Lechner
- Department of Cardiology, German Heart Centre Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Sarah King
- School of Medicine, University of California, San Francisco, CA, 94143, USA
| | - James P McCarter
- Abbott Diabetes Care, Alameda, CA, 94502, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeff S Volek
- Virta Health, 501 Folsom Street, San Francisco, CA, 94105, USA
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | | | - Ronald M Krauss
- School of Medicine, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
21
|
Ahn SS, Lee LE, Pyo JY, Song JJ, Park YB, Lee SW. Atherogenic index of plasma predicts cerebrovascular accident occurrence in antineutrophil cytoplasmic antibody-associated vasculitis. Lipids Health Dis 2020; 19:184. [PMID: 32799861 PMCID: PMC7429760 DOI: 10.1186/s12944-020-01360-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
Background To investigate whether atherogenic index of plasma (AIP) at diagnosis is associated with the occurrence of cerebrovascular accident (CVA) or coronary artery disease (CAD) in antineutrophil cytoplasmic antibody-associated vasculitis (AAV). Methods The medical records of 167 AAV patients on initial diagnosis was reviewed, and 300 healthy controls were included. AIP was calculated using the following equation: AIP = Log (triglyceride [mg/dL] / high-density lipoprotein cholesterol [mg/dL]). AAV patients were divided into two groups according to the AIP cut-off of 0.11. The event of stroke, transient ischemic attack, and cerebral hemorrhage was recorded as CVA, and CAD events consisted of either myocardial infarction and angina pectoris. CVA- and CAD- free survival rate between those with AIP ≥ 0.11 and < 0.11 were compared by the Kaplan-Meier analysis, and Cox hazard analysis was conducted to identify predictors of CVA. Results The median age of AAV patients were 59.0 years, and 54 (32.3%) patients were male. One-hundred and fifteen (68.9%) patients had AIP < 0.11 and 52 (31.1%) had AIP ≥ 0.11. The mean Birmingham vasculitis activity score in AAV patients with AIP < 0.11 was lower than that seen in patients with AIP ≥ 0.11 (12.0 vs. 14.0, P = 0.041). AAV patients had a significantly higher AIP compared to controls (mean − 0.01 vs. -0.10, P < 0.001). During follow-up, the occurrence of CVA and CAD was observed in 16 (9.6%) and 14 (8.4%) patients, respectively. In Kaplan-Meier analysis, AAV patients with AIP ≥ 0.11 had significantly lower CVA-free survival rates than in those with AIP < 0.11 (P = 0.027), whereas there was no difference in CAD according to AIP (P = 0.390). Multivariable Cox analysis indicated that AIP ≥ 0.11 at diagnosis was the sole predictor of CVA (Hazard ratio 3.392, 95% confidence interval 1.076, 10.696, P = 0.037). Conclusions AIP is significantly higher in AAV patients than in healthy controls, and AIP ≥ 0.11 at diagnosis is a significant predictor of CVA during follow-up. Stringent surveillance should be provided in AAV patients with AIP ≥ 0.11 regarding the occurrence of CVA. Trial registration Retrospectively registered (4–2017-0673).
Collapse
Affiliation(s)
- Sung Soo Ahn
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea, 03722
| | - Lucy Eunju Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea, 03722
| | - Jung Yoon Pyo
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea, 03722
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea, 03722.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea, 03722.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea, 03722. .,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Abstract
Cardiovascular disease, with atherosclerosis as the major underlying factor, remains the leading cause of death worldwide. It is well established that cholesterol ester-enriched foam cells are the hallmark of atherosclerotic plaques. Multiple lines of evidence support that enhancing foam cell cholesterol efflux by HDL (high-density lipoprotein) particles, the first step of reverse cholesterol transport (RCT), is a promising antiatherogenic strategy. Yet, excitement towards the therapeutic potential of manipulating RCT for the treatment of cardiovascular disease has faded because of the lack of the association between cardiovascular disease risk and what was typically measured in intervention trials, namely HDL cholesterol, which has an inconsistent relationship to HDL function and RCT. In this review, we will summarize some of the potential reasons for this inconsistency, update the mechanisms of RCT, and highlight conditions in which impaired HDL function or RCT contributes to vascular disease. On balance, the evidence still argues for further research to better understand how HDL functionality contributes to RCT to develop prevention and treatment strategies to reduce the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Mireille Ouimet
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa Heart Institute, University of Ottawa, Canada (M.O.)
| | - Tessa J Barrett
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.)
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.)
| |
Collapse
|
23
|
Descamps OS, Verhaegen A, Demeure F, Langlois M, Rietzschel E, Mertens A, De Sutter J, Wallemacq C, Lancellotti P, De Backer G. Evolving concepts on the management of dyslipidaemia. Acta Clin Belg 2020; 75:80-90. [PMID: 31846601 DOI: 10.1080/17843286.2019.1702823] [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] [Indexed: 12/25/2022]
Abstract
It has been well established that low-density lipoproteins (LDL) and other apolipoprotein B-containing lipoproteins are causally related to atherosclerotic cardiovascular disease (ASCVD) and that lowering these lipoproteins reduces the risk of ASCVD. By lowering LDL particles as much as possible, ASCVD can be prevented. There seems to be no LDL-cholesterol (LDL-C) threshold below which no further ASCVD prevention can be achieved. Furthermore, a low (an even very low) LDL-C appears to be safe. The new ESC/EAS guidelines based on these concepts are a step towards a benefit-based strategy by focusing on the clinical benefit that can be achieved by treating the cause of ASCVD. It is recommended to lower LDL-C as much as possible to prevent ASCVD, especially in high and very high-risk patients. With these new recommendations come recognition of the importance of combination therapies in high and very high-risk patients, first with statins and ezetimibe, and if needed with a PCSK9 inhibitor. The present paper is a review of some new concepts arising during the past 10 years in the field of lipidology and the description of what is new in the 2019 EAS/ESC guidelines.
Collapse
Affiliation(s)
- Olivier S. Descamps
- Department of internal medicine, Centres Hospitaliers Jolimont, Haine Saint-Paul and department of cardiology, UCL Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
| | - Ann Verhaegen
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Antwerpen, Belgium
| | - Fabien Demeure
- Department of Cardiology, CHU UCL Namur site de Godinne, Yvoir, Belgium
| | - Michel Langlois
- department of Laboratory Medicine, Algemeen Ziekenhuis Sint-Jan, Brugge, Belgium
| | - Ernst Rietzschel
- department of Cardiology, University Hospital Ghent and Ghent University, Ghent, Belgium
| | - Ann Mertens
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - Johan De Sutter
- Department of Cardiology, AZ Maria Middelars Ghent and University Ghent, Belgium
| | - Caroline Wallemacq
- department of Diabetes, Nutrition and Metabolic diseases, Centre Hospitalier Universitaire Sart Tilman, Liège, Belgium
| | - Patrizio Lancellotti
- GIGA Cardiovascular Sciences, department of Cardiology, Centre Hospitalier Universitaire Sart Tilman, Liège, Belgium
| | - Guy De Backer
- Gent Department of public health and primary care, Ghent University, Gent, Belgium
| |
Collapse
|
24
|
The Effect of High Carbohydrate-to-fat Intake Ratios on Hypo-HDL-cholesterolemia Risk and HDL-cholesterol Levels over a 12-year Follow-up. Sci Rep 2020; 10:913. [PMID: 31969639 PMCID: PMC6976611 DOI: 10.1038/s41598-020-57931-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/08/2020] [Indexed: 01/26/2023] Open
Abstract
Considering the strong correlation between carbohydrate and fat intake, we defined and assessed the association of the carbohydrate-to-fat ratio with the high-density lipoprotein cholesterol (HDL-c) level using 12-year follow-up data from the community-based cohort of the Korean Genome Epidemiology Study. We evaluated the long-term changes in HDL-c levels according to quartiles of carbohydrate-to-fat ratio using a mixed model. We also assessed the effect of the carbohydrate-to-fat ratio on the prevalence and incidence of hypo-HDL-cholesterolemia. Of 6,627 subjects, the prevalence of undiagnosed hypo-HDL-cholesterolemia at baseline was 35.3% (n = 2,339). Among the disease-free subjects, 56.8% developed hypo-HDL-cholesterolemia (incidence = 92/1,000 person-years). The prevalence and incidence of hypo-HDL-cholesterolemia were higher in females than in males. The highest carbohydrate-to-fat ratio quartile, which was characterized by high and low intake of carbohydrate and fat, was consistently associated with a lower HDL-c level during the 12-year follow up. Moreover, those in the highest quartile had a 1.14-fold greater risk of incident hypo-HDL-cholesterolemia than those in the lowest quartile, with a significant dose-response relationship. We found that high and low intake of carbohydrate and fat, respectively, was consistently associated with a low HDL-c level over a prolonged period. More research is needed to promote appropriate intake of macronutrients.
Collapse
|
25
|
Mu Q, Zhu J, Si Y, Chen X, Duan G, Sun S, Fang G, Zeng Y, Yang N. Overexpression of PID1 reduces high density lipoprotein level and functionality in swine. IUBMB Life 2019; 71:1946-1951. [PMID: 31339628 DOI: 10.1002/iub.2135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/10/2019] [Indexed: 01/10/2023]
Abstract
Phosphotyrosine interaction domain 1 (PID1), a protein with a phosphotyrosine-binding (PTB) domain, interacts with the lipoprotein receptor-related protein 1 (LRP1) to reduce the insulin sensitivity of adipocyte. Considering the role of LRP1 in lipid metabolism, we investigated the effect of PID1 on the content and biological activities of serum lipoproteins in pigs. PID1-transgenic pigs were genetated by sperm and magnetic nanoparticles-mediated method. The levels of PID1 in PID1-transgenic pig's liver were higher than that in the wild-type pig's liver. We found that serum levels of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I (apoA-I) were significantly reduced in PID1-transgenic pigs. On the other hand, PID1-transgenic pigs displayed increased non-HDL-C levels. Serum levels of total cholesterol and triglycerides were comparable between the PID1-transgenic and the wild-type pigs. Further, the HDL isolated from PID1-transgenic pigs showed a significant reduction in cholesterol efflux ability. In addition, serum superoxide dismutase activity of PID1-transgenic pigs was also obviously lowered compared with that of wild type pigs. In conclusion, these results suggest that PID1 might be able to adjust HDL-C levels in serum and HDL cholesterol efflux ability.
Collapse
Affiliation(s)
- Qingjie Mu
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Jing Zhu
- Shandong Laboratory Animal Center of Shandong Academy of Medical Science, Jinan, Shandong, People's Republic of China
| | - Yanhong Si
- Basic Medical College, Shandong First Medical University, Taian, Shandong, People's Republic of China
| | - Xiaofeng Chen
- Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Guangqing Duan
- Experimental Center for Medical Research, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Shiyu Sun
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Guofeng Fang
- Academy of Swine Research, New Hope Liuhe Limited Liability Company, Tsingtao, Shandong, People's Republic of China
| | - Yongqing Zeng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, People's Republic of China
| | - Nana Yang
- Experimental Center for Medical Research, Weifang Medical University, Weifang, Shandong, People's Republic of China.,School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| |
Collapse
|
26
|
The association between the chromosome 9p21 CDKN2B-AS1 gene variants and the lipid metabolism: A pre-diagnostic biomarker for coronary artery disease. Anatol J Cardiol 2019; 21:31-38. [PMID: 30587704 PMCID: PMC6382903 DOI: 10.14744/anatoljcardiol.2018.90907] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Objective: Recent genome-wide association studies have established that polymorphisms within CDKN2B-AS1 of chr9p21.3 locus increased susceptibility to coronary artery disease (CAD) or myocardial infarction. Common variants of CDKN2B-AS1 (including rs4977574 A>G and rs1333040 C>T) are determined to be directly associated with CADs in many populations worldwide and suggested biomarkers for the early detection of CAD. There is a lack of investigation for the association between CDKN2B-AS1 rs4977574 A>G and rs1333040 C>T genetic modifiers and CAD in a Turkish Cypriot population. The aim of the present study was to investigate the potential effects of these variants on susceptibility to developing CAD in a Turkish Cypriot population and their contribution to lipid metabolism. Methods: Seventy-one patients with angiography-confirmed CAD were recruited to the CAD group, whereas 153 voluntary subjects without CAD symptoms were enrolled to the control group. Genotyping for the CDKN2B-AS1 gene polymorphisms was performed by polymerase chain reaction, followed by restriction fragment length polymorphism analysis. Results: There is no statistical significant association observed between rs4977574 and rs1333040 single-nucleotide polymorphisms and two studied groups [odds ratio (OR): 0.763, p=0.185, 95% confidence interval (CI): 0.511–1.139 and OR: 1.060, p=0.802, 95% CI 0.672–1.671, respectively]. However, rs2977574 G and rs1333040 T alleles–the risk alleles–were found to be associated with higher level of serum total cholesterol and lower level of high-density lipoprotein-cholesterol in the CAD group (p=0.019, p=0.006 and p=0.022, p=0.031, respectively). To our knowledge, this is the first study that establishes the effect of rs1333040 on lipid metabolism. Conclusion: The presence of rs4977574 G and rs1333040 T alleles and interaction may exist as environmental factors associated with lipid metabolism and might be responsible for the development of CAD in a Turkish Cypriot population.
Collapse
|
27
|
Du D, Bruno R, Blizzard L, Venn A, Dwyer T, Smith KJ, Magnussen CG, Gall S. The metabolomic signatures of alcohol consumption in young adults. Eur J Prev Cardiol 2019; 27:840-849. [PMID: 30857428 DOI: 10.1177/2047487319834767] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Metabolomic analysis may help us to understand the association between alcohol consumption and cardio-metabolic health. We aimed to: (i) replicate a previous study of alcohol consumption and metabolic profiles, (ii) examine associations between types of alcoholic beverages and metabolites and (iii) include potential confounders not examined in previous studies. METHODS Cross-sectional data of 1785 participants (age 26-36 years, 52% women) from the 2004-2006 Childhood Determinants of Adult Health study were used. Consumption of beer, wine and spirits was assessed by questionnaires. Metabolites were measured by a high-throughput nuclear magnetic resonance platform and multivariable linear regression examined their association with alcohol consumption (combined total and types) adjusted for covariates including socio-demographics, health behaviours and mental health. RESULTS Alcohol consumption was associated with 23 out of 37 lipids, 12 out of 16 fatty acids and six out of 20 low-molecular-weight metabolites independent of confounders with similar associations for combined total alcohol consumption and different types of alcohol. Many metabolites (lipoprotein lipids in high-density lipoprotein (HDL) subclasses, HDL cholesterol, apolipoprotein A-1, phosphotriglycerides, total fatty acids, monounsaturated fatty acids, omega-3 fatty acids) had positive linear associations with alcohol consumption but some showed negative linear (low-density lipoprotein particle size, omega-6 fatty acids ratio to total fatty acids, citrate) or U-shaped (lipoprotein lipids in very-low-density lipoprotein (VLDL) subclasses, VLDL triglycerides) associations. CONCLUSIONS Our results were similar to those of the only previous study. Associations with metabolites were similar for total and types of alcohol. Alcohol consumption in young adults is related to a diverse range of metabolomic signatures associated with benefits and harms to health.
Collapse
Affiliation(s)
- Duc Du
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Raimondo Bruno
- School of Medicine, University of Tasmania, Hobart, Australia
| | - Leigh Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Alison Venn
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Terence Dwyer
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Kylie J Smith
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Costan G Magnussen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Finland
| | - Seana Gall
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| |
Collapse
|
28
|
Gupta MD, Girish MP, Sarkar PG, Gupta A, Kategari A, Bansal A, Saijpaul R, Batra V, Rain M, Tyagi S, Pasha Q. Role of ApoE gene polymorphism and nonconventional biochemical risk factors among very young individuals (aged less than 35 years) presenting with acute myocardial infarction. Indian Heart J 2018; 70 Suppl 3:S146-S156. [PMID: 30595248 PMCID: PMC6310748 DOI: 10.1016/j.ihj.2018.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/09/2018] [Accepted: 08/12/2018] [Indexed: 12/11/2022] Open
Abstract
Background Incidence rate of acute myocardial infarction (MI) has increased in younger population over the years. The young patients have a different risk profile, presentation, and prognosis than the elderly. Hence, it is essential to understand the risk factors in young patients for proper treatment. Methods Apolipoprotein E (ApoE) polymorphism and biochemicals such as total cholesterol, serum triglycerides, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), lipoprotein(a), insulin, interleukin-6, homocysteine, fibrinogen, and highly sensitive C-reactive protein were investigated in very young MI (yMI patients; age ≤ 35 years; n = 125), in old MI (oMI patients; age >35 and < 80 years; n = 111), and healthy controls (age ≤35 years; n = 103). Results HDL-C was significantly lower in yMI patients than in controls (p = 2.63E-04) and oMI patients (p = 1.29E-05). ApoA1 was also lowest in yMI patients, but significant only in comparison to controls (p = 2.62E.04). The yMI group had the highest ratios of total cholesterol:HDL-C (p = 0.027 in yMI patients versus controls and p = 0.018 in yMI patients versus oMI patients), LDL-C:HDL-C (p = 0.002 in yMI patients versus controls and p = 0.005 in yMI patients versus oMI patients), and ApoB:ApoA1 (p = 8.75E-05 in yMI patients versus controls and p > 0.05 in yMI patients versus oMI patients). No significant pattern of ApoE polymorphisms was observed. Conclusion The lower level of HDL-C and ApoA1 and higher ratios of total cholesterol:HDL-C, LDL-C:HDL-C, and ApoB:ApoA1 are risk factors for MI in young patients.
Collapse
Affiliation(s)
- Mohit D Gupta
- GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, India; Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India.
| | - M P Girish
- GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, India; Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India
| | - Prattay G Sarkar
- GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, India; Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India
| | - Amit Gupta
- GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, India; Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India
| | - Anand Kategari
- GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, India; Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India
| | - Ankit Bansal
- GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, India; Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India
| | - Rajni Saijpaul
- Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India
| | - Vishal Batra
- GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, India; Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India
| | - Manjari Rain
- Institute of Genomics and Integrative Biology, New Delhi, India
| | - Sanjay Tyagi
- GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, India; Safdarjung Hospital and Vardhman Institute of Medical Sciences, New Delhi, India
| | - Qadar Pasha
- Institute of Genomics and Integrative Biology, New Delhi, India
| |
Collapse
|
29
|
Lee Y, Pędziwiatr M, Major P, Brar K, Doumouras AG, Hong D. The effect of omentectomy added to bariatric surgery on metabolic outcomes: a systematic review and meta-analysis of randomized controlled trials. Surg Obes Relat Dis 2018; 14:1766-1782. [PMID: 30228082 DOI: 10.1016/j.soard.2018.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 08/02/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Excess visceral adipose tissue has been identified as an important risk factor for obesity-related co-morbidities. Conflicting information exists on whether omentectomy added to bariatric surgery is beneficial to metabolic variables. OBJECTIVE To evaluate the impact of omentectomy added to bariatric surgery on metabolic outcomes SETTING: University Hospital, Canada. METHODS MEDLINE, EMBASE, and PubMed were searched up to May 2018. Studies were eligible for inclusion if they were randomized controlled trials comparing omentectomy added to bariatric surgery with bariatric surgery alone. Primary outcome measures were absolute change in metabolic variables (body mass index, insulin, glucose, cholesterol, lipoproteins, and triglycerides); secondary outcomes were changes in adipocytokines. Pooled mean differences (mean deviation; MD) were calculated using random effects meta-analyses, and heterogeneity was quantified using the I2 statistic. RESULTS Ten trials involving a total of 366 patients met the inclusion criteria with a median follow-up time of 1 year after surgery. Adding omentectomy to bariatric surgery demonstrated a minimal but statistically significant decrease in body mass index compared with bariatric surgery alone (MD 1.29, 95% confidence interval .35-2.23, P = .007, I2 = 0%, 10 trials). Conversely, patients who underwent bariatric surgery alone had significant increases in high-density lipoprotein (MD -2.12, 95% confidence interval -4.13 to -.11, P = .04, I2 = 0%, 6 trials). Other metabolic outcomes and adipocytokines showed no significant difference between procedures. CONCLUSION The addition of omentectomy to bariatric surgery results in minimal reduction of body mass index. Considering no overall improvement in metabolic outcomes and the time and effort required, the therapeutic use of omentectomy added to bariatric surgery is not warranted.
Collapse
Affiliation(s)
- Yung Lee
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Michał Pędziwiatr
- 2nd Department of General Surgery, Jagiellonian University, Krakow, Poland; Centre for Research, Training and Innovation in Surgery (CERTAIN Surgery), Krakow, Poland
| | - Piotr Major
- 2nd Department of General Surgery, Jagiellonian University, Krakow, Poland; Centre for Research, Training and Innovation in Surgery (CERTAIN Surgery), Krakow, Poland
| | - Karanbir Brar
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Aristithes G Doumouras
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Dennis Hong
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
30
|
Borén J, Williams KJ. The central role of arterial retention of cholesterol-rich apolipoprotein-B-containing lipoproteins in the pathogenesis of atherosclerosis: a triumph of simplicity. Curr Opin Lipidol 2016; 27:473-83. [PMID: 27472409 DOI: 10.1097/mol.0000000000000330] [Citation(s) in RCA: 317] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Today, it is no longer a hypothesis, but an established fact, that increased plasma concentrations of cholesterol-rich apolipoprotein-B (apoB)-containing lipoproteins are causatively linked to atherosclerotic cardiovascular disease (ASCVD) and that lowering plasma LDL concentrations reduces cardiovascular events in humans. Here, we review evidence behind this assertion, with an emphasis on recent studies supporting the 'response-to-retention' model - namely, that the key initiating event in atherogenesis is the retention, or trapping, of cholesterol-rich apoB-containing lipoproteins within the arterial wall. RECENT FINDINGS New clinical trials have shown that ezetimibe and anti-PCSK9 antibodies - both nonstatins - lower ASCVD events, and they do so to the same extent as would be expected from comparable plasma LDL lowering by a statin. These studies demonstrate beyond any doubt the causal role of apoB-containing lipoproteins in atherogenesis. In addition, recent laboratory experimentation and human Mendelian randomization studies have revealed novel information about the critical role of apoB-containing lipoproteins in atherogenesis. New information has also emerged on mechanisms for the accumulation in plasma of harmful cholesterol-rich and triglyceride-rich apoB-containing remnant lipoproteins in states of overnutrition. Like LDL, these harmful cholesterol-rich and triglyceride-rich apoB-containing remnant lipoprotein remnants become retained and modified within the arterial wall, causing atherosclerosis. SUMMARY LDL and other cholesterol-rich, apoB-containing lipoproteins, once they become retained and modified within the arterial wall, cause atherosclerosis. This simple, robust pathophysiologic understanding may finally allow us to eradicate ASCVD, the leading killer in the world.
Collapse
Affiliation(s)
- Jan Borén
- aDepartment of Molecular and Clinical Medicine, University of Gothenburg bSahlgrenska University Hospital, Gothenburg, Sweden cSection of Endocrinology, Diabetes, & Metabolism, Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
31
|
Einfluss der Lipidstoffwechselparameter auf die Entstehung und Progression der koronaren Herzerkrankung. Herz 2016; 41:273-80. [DOI: 10.1007/s00059-016-4430-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
32
|
Corsetti JP, Salzman P, Ryan D, Moss AJ, Zareba W, Sparks CE. Influences on plasminogen activator inhibitor-2 polymorphism-associated recurrent cardiovascular disease risk in patients with high HDL cholesterol and inflammation. Atherosclerosis 2016; 250:1-8. [PMID: 27174532 DOI: 10.1016/j.atherosclerosis.2016.04.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 03/29/2016] [Accepted: 04/19/2016] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS Evidence continues to accumulate that athero-protective effects of high-density lipoprotein (HDL) depend to some degree on effective HDL functionality and that such functionality can become degraded in the setting of chronic inflammation. To investigate this issue, we have studied a group of post-myocardial infarction patients with high levels of C-reactive protein as an indicator of chronic inflammation and with concurrently high levels of HDL cholesterol. For these patients we have demonstrated high-risk for recurrent cardiac events as well as a strong association of risk with a polymorphism of the gene (SERPINB2) for plasminogen activator inhibitor-2 (PAI-2) presumptively reflective of an important role for fibrinolysis in risk. However, additional processes might be involved. The current work sought to characterize processes underlying how PAI-2 might be involved in the generation of risk. METHODS Multivariate population data were leveraged using Bayesian network modeling, a graphical probabilistic approach for knowledge discovery, to generate networks reflective of influences on PAI-2 polymorphism-associated risk. RESULTS Modeling results revealed three individual networks centering on the PAI-2 polymorphism with specific features providing information relating to how the polymorphism might associate with risk. These included racial dependency, platelet clot initiation and propagation, oxidative stress, inflammation effects on HDL metabolism and coagulation, and induction and termination of fibrinolysis. CONCLUSIONS Beyond direct association of a PAI-2 polymorphism with recurrent risk in post-myocardial infarction patients, results suggest that PAI-2 likely plays a key role leading to risk through multiple pathophysiologic processes. Such knowledge could potentially be valuable with individualization of patient care.
Collapse
Affiliation(s)
- James P Corsetti
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Peter Salzman
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Dan Ryan
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Arthur J Moss
- Department of Medicine - Cardiology Unit, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Wojciech Zareba
- Department of Medicine - Cardiology Unit, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Charles E Sparks
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
33
|
Morin EE, Guo L, Schwendeman A, Li XA. HDL in sepsis - risk factor and therapeutic approach. Front Pharmacol 2015; 6:244. [PMID: 26557091 PMCID: PMC4616240 DOI: 10.3389/fphar.2015.00244] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022] Open
Abstract
High-density lipoprotein (HDL) is a key component of circulating blood and plays essential roles in regulation of vascular endothelial function and immunity. Clinical data demonstrate that HDL levels drop by 40-70% in septic patients, which is associated with a poor prognosis. Experimental studies using Apolipoprotein A-I (ApoAI) null mice showed that HDL deficient mice are susceptible to septic death, and overexpressing ApoAI in mice to increase HDL levels protects against septic death. These clinical and animal studies support our hypothesis that a decrease in HDL level is a risk factor for sepsis, and raising circulating HDL levels may provide an efficient therapy for sepsis. In this review, we discuss the roles of HDL in sepsis and summarize the efforts of using synthetic HDL as a potential therapy for sepsis.
Collapse
Affiliation(s)
- Emily E. Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann ArborMI, USA
- Biointerfaces Institute, University of Michigan, Ann ArborMI, USA
| | - Ling Guo
- Department of Pediatrics, Saha Cardiovascular Research Center, University of Kentucky College of Medicine, LexingtonKY, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann ArborMI, USA
- Biointerfaces Institute, University of Michigan, Ann ArborMI, USA
| | - Xiang-An Li
- Department of Pediatrics, Saha Cardiovascular Research Center, University of Kentucky College of Medicine, LexingtonKY, USA
| |
Collapse
|
34
|
Thacker SG, Rousset X, Esmail S, Zarzour A, Jin X, Collins HL, Sampson M, Stonik J, Demosky S, Malide DA, Freeman L, Vaisman BL, Kruth HS, Adelman SJ, Remaley AT. Increased plasma cholesterol esterification by LCAT reduces diet-induced atherosclerosis in SR-BI knockout mice. J Lipid Res 2015; 56:1282-95. [PMID: 25964513 PMCID: PMC4479333 DOI: 10.1194/jlr.m048629] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/27/2015] [Indexed: 12/15/2022] Open
Abstract
LCAT, a plasma enzyme that esterifies cholesterol, has been proposed to play an antiatherogenic role, but animal and epidemiologic studies have yielded conflicting results. To gain insight into LCAT and the role of free cholesterol (FC) in atherosclerosis, we examined the effect of LCAT over- and underexpression in diet-induced atherosclerosis in scavenger receptor class B member I-deficient [Scarab(-/-)] mice, which have a secondary defect in cholesterol esterification. Scarab(-/-)×LCAT-null [Lcat(-/-)] mice had a decrease in HDL-cholesterol and a high plasma ratio of FC/total cholesterol (TC) (0.88 ± 0.033) and a marked increase in VLDL-cholesterol (VLDL-C) on a high-fat diet. Scarab(-/-)×LCAT-transgenic (Tg) mice had lower levels of VLDL-C and a normal plasma FC/TC ratio (0.28 ± 0.005). Plasma from Scarab(-/-)×LCAT-Tg mice also showed an increase in cholesterol esterification during in vitro cholesterol efflux, but increased esterification did not appear to affect the overall rate of cholesterol efflux or hepatic uptake of cholesterol. Scarab(-/-)×LCAT-Tg mice also displayed a 51% decrease in aortic sinus atherosclerosis compared with Scarab(-/-) mice (P < 0.05). In summary, we demonstrate that increased cholesterol esterification by LCAT is atheroprotective, most likely through its ability to increase HDL levels and decrease pro-atherogenic apoB-containing lipoprotein particles.
Collapse
Affiliation(s)
- Seth G. Thacker
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xavier Rousset
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Safiya Esmail
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Abdalrahman Zarzour
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xueting Jin
- Experimental Atherosclerosis Section, Center for Molecular, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | - Maureen Sampson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - John Stonik
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Stephen Demosky
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Daniela A. Malide
- Light Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Lita Freeman
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Boris L. Vaisman
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Howard S. Kruth
- Experimental Atherosclerosis Section, Center for Molecular, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | - Alan T. Remaley
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
35
|
|
36
|
El Khoury P, Ghislain M, Villard EF, Le Goff W, Lascoux-Combe C, Yeni P, Meyer L, Vigouroux C, Goujard C, Guerin M. Plasma cholesterol efflux capacity from human THP-1 macrophages is reduced in HIV-infected patients: impact of HAART. J Lipid Res 2015; 56:692-702. [PMID: 25573889 DOI: 10.1194/jlr.m054510] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The capacity of HDL to remove cholesterol from macrophages is inversely associated with the severity of angiographic coronary artery disease. The effect of human immunodeficiency virus (HIV) infection or its treatment on the ability of HDL particles to stimulate cholesterol efflux from human macrophages has never been studied. We evaluated the capacity of whole plasma and isolated HDL particles from HIV-infected subjects (n = 231) and uninfected controls (n = 200), as well as in a subset of 41 HIV subjects receiving highly active antiretroviral therapy (HAART) to mediate cholesterol efflux from human macrophages. Plasma cholesterol efflux capacity was reduced (-12%; P = 0.001) in HIV patients as compared with controls. HIV infection reduced by 27% (P < 0.05) the capacity of HDL subfractions to promote cholesterol efflux from macrophages. We observed a reduced ABCA1-dependent efflux capacity of plasma (-27%; P < 0.0001) from HIV-infected subjects as a result of a reduction in the efflux capacity of HDL3 particles. HAART administration restored the capacity of plasma from HIV patients to stimulate cholesterol efflux from human macrophages (9.4%; P = 0.04). During HIV infection, the capacity of whole plasma to remove cholesterol from macrophages is reduced, thus potentially contributing to the increased coronary heart disease in the HIV population. HAART administration restored the removal of cholesterol from macrophages by increasing HDL functionality.
Collapse
Affiliation(s)
- Petra El Khoury
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS1166, Hôpital de la Pitié, Paris, France; Sorbonne Universités, UMPC Univ Paris 6, Paris, France; Université Saint Joseph, Faculté de pharmacie, Beyrouth, Liban
| | - Mathilde Ghislain
- INSERM, UMRS 1018, CESP, Le Kremlin-Bicêtre, France; Service d'Epidémiologie et de Santé Publique, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin-Bicêtre, France
| | - Elise F Villard
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS1166, Hôpital de la Pitié, Paris, France; Sorbonne Universités, UMPC Univ Paris 6, Paris, France
| | - Wilfried Le Goff
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS1166, Hôpital de la Pitié, Paris, France; Sorbonne Universités, UMPC Univ Paris 6, Paris, France; ICAN Institute of Cardiometabolism and Nutrition, Hôpital de la Pitié, Paris, France
| | | | - Patrick Yeni
- Service de Maladies Infectieuses, Hôpital Bichat, AP-HP, Paris, France
| | - Laurence Meyer
- INSERM, UMRS 1018, CESP, Le Kremlin-Bicêtre, France; Service d'Epidémiologie et de Santé Publique, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin-Bicêtre, France; Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Corinne Vigouroux
- Sorbonne Universités, UMPC Univ Paris 6, Paris, France; ICAN Institute of Cardiometabolism and Nutrition, Hôpital de la Pitié, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Antoine, Laboratoire de Biologie et Génétique Moléculaires, Paris, France; INSERM UMRS938, Centre de recherche Saint-Antoine, Paris, France
| | - Cécile Goujard
- INSERM, UMRS 1018, CESP, Le Kremlin-Bicêtre, France; Université Paris-Sud, Le Kremlin-Bicêtre, France; Service de Médecine Interne, Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - Maryse Guerin
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS1166, Hôpital de la Pitié, Paris, France; Sorbonne Universités, UMPC Univ Paris 6, Paris, France; ICAN Institute of Cardiometabolism and Nutrition, Hôpital de la Pitié, Paris, France.
| |
Collapse
|
37
|
Birner-Gruenberger R, Schittmayer M, Holzer M, Marsche G. Understanding high-density lipoprotein function in disease: recent advances in proteomics unravel the complexity of its composition and biology. Prog Lipid Res 2014; 56:36-46. [PMID: 25107698 DOI: 10.1016/j.plipres.2014.07.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/21/2014] [Accepted: 07/24/2014] [Indexed: 10/24/2022]
Abstract
Although the epidemiology of high-density lipoprotein (HDL) cholesterol and cardiovascular risk has been consistent, pharmacologic interventions to increase HDL-cholesterol by delaying HDL catabolism did not translate into reduction in cardiovascular risk. HDL particles are small, protein-rich when compared to other plasma lipoprotein classes. Latest progresses in proteomics technology have dramatically increased our understanding of proteins carried by HDL. In addition to proteins with well-established functions in lipid transport, iron transport proteins, members of the complement pathway, and proteins involved in immune function and acute phase response were repeatedly identified on HDL particles. With the unraveling of the complexity of the HDL proteome, different laboratories have started to monitor its changes in various disease states. In addition, dynamic aspects of HDL subgroups are being discovered. These recent studies clearly illustrate the promise of HDL proteomics for deriving new biomarkers for disease diagnosis and to measure the effectiveness of current and future treatment regimens. This review summarizes recent advances in proteomics and lipidomics helping to understand HDL function in health and disease.
Collapse
Affiliation(s)
- Ruth Birner-Gruenberger
- Institute of Pathology, Medical University of Graz, Graz, Austria; Omics Center Graz, BioTechMed, Graz, Austria.
| | - Matthias Schittmayer
- Institute of Pathology, Medical University of Graz, Graz, Austria; Omics Center Graz, BioTechMed, Graz, Austria
| | - Michael Holzer
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Gunther Marsche
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
38
|
Wool GD, Reardon CA, Getz GS. Mimetic peptides of human apoA-I helix 10 get together to lower lipids and ameliorate atherosclerosis: is the action in the gut? J Lipid Res 2014; 55:1983-5. [PMID: 25085258 DOI: 10.1194/jlr.e053538] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | | | - Godfrey S Getz
- Department of Pathology, University of Chicago, Chicago, IL
| |
Collapse
|
39
|
Tomey MI, Narula J, Kovacic JC. Advances in the Understanding of Plaque Composition and Treatment Options. J Am Coll Cardiol 2014; 63:1604-16. [DOI: 10.1016/j.jacc.2014.01.042] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 01/02/2014] [Accepted: 01/28/2014] [Indexed: 12/11/2022]
|
40
|
Guo L, Zheng Z, Ai J, Howatt DA, Mittelstadt PR, Thacker S, Daugherty A, Ashwell JD, Remaley AT, Li XA. Scavenger receptor BI and high-density lipoprotein regulate thymocyte apoptosis in sepsis. Arterioscler Thromb Vasc Biol 2014; 34:966-75. [PMID: 24603680 DOI: 10.1161/atvbaha.113.302484] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Thymocyte apoptosis is a major event in sepsis; however, how this process is regulated remains poorly understood. APPROACH AND RESULTS Septic stress induces glucocorticoids production which triggers thymocyte apoptosis. Here, we used scavenger receptor BI (SR-BI)-null mice, which are completely deficient in inducible glucocorticoids in sepsis, to investigate the regulation of thymocyte apoptosis in sepsis. Cecal ligation and puncture induced profound thymocyte apoptosis in SR-BI(+/+) mice, but no thymocyte apoptosis in SR-BI(-/-) mice because of lack of inducible glucocorticoids. Unexpectedly, supplementation of glucocorticoids only partly restored thymocyte apoptosis in SR-BI(-/-) mice. We demonstrated that high-density lipoprotein (HDL) is a critical modulator for thymocyte apoptosis. SR-BI(+/+) HDL significantly enhanced glucocorticoid-induced thymocyte apoptosis, but SR-BI(-/-) HDL had no such activity. Further study revealed that SR-BI(+/+) HDL modulates glucocorticoid-induced thymocyte apoptosis via promoting glucocorticoid receptor translocation, but SR-BI(-/-) HDL loses such regulatory activity. To understand why SR-BI(-/-) HDL loses its regulatory activity, we analyzed HDL cholesterol contents. There was 3-fold enrichment of unesterified cholesterol in SR-BI(-/-) HDL compared with SR-BI(+/+) HDL. Normalization of unesterified cholesterol in SR-BI(-/-) HDL by probucol administration or lecithin cholesteryl acyltransferase expression restored glucocorticoid-induced thymocyte apoptosis, and incorporating unesterified cholesterol into SR-BI(+/+) HDL rendered SR-BI(+/+) HDL dysfunctional. Using lckCre-GR(fl/fl) mice in which thymocytes lack cecal ligation and puncture-induced thymocyte apoptosis, we showed that lckCre-GR(fl/fl) mice were significantly more susceptible to cecal ligation and puncture-induced septic death than GR(fl/fl) control mice, suggesting that glucocorticoid-induced thymocyte apoptosis is required for protection against sepsis. CONCLUSIONS The findings in this study reveal a novel regulatory mechanism of thymocyte apoptosis in sepsis by SR-BI and HDL.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cecum/microbiology
- Cecum/surgery
- Cells, Cultured
- Cholesterol, HDL/blood
- Corticosterone/metabolism
- Disease Models, Animal
- Female
- Humans
- Ligation
- Male
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Phosphatidylcholine-Sterol O-Acyltransferase/genetics
- Phosphatidylcholine-Sterol O-Acyltransferase/metabolism
- Probucol/pharmacology
- Protein Transport
- Punctures
- Receptors, Glucocorticoid/metabolism
- Scavenger Receptors, Class B/deficiency
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Sepsis/blood
- Sepsis/metabolism
- Sepsis/microbiology
- Sepsis/pathology
- Signal Transduction
- Thymocytes/drug effects
- Thymocytes/metabolism
- Thymocytes/pathology
Collapse
Affiliation(s)
- Ling Guo
- From the Department of Pediatrics (L.G., Z.Z., J.A., X.-A.L.), Graduate Center for Nutritional Sciences (Z.Z., J.A., X.-A.L.), and Saha Cardiovascular Research Center (D.A.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington; and Laboratory of Immune Cell Biology, National Cancer Institute (P.R.M., J.D.A.) and Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute (S.T., A.T.R.), National Institutes of Health, Bethesda, MD
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Feig JE, Hewing B, Smith JD, Hazen SL, Fisher EA. High-density lipoprotein and atherosclerosis regression: evidence from preclinical and clinical studies. Circ Res 2014; 114:205-13. [PMID: 24385513 DOI: 10.1161/circresaha.114.300760] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
High-density lipoprotein (HDL) particles transport (among other molecules) cholesterol (HDL-C). In epidemiological studies, plasma HDL-C levels have an inverse relationship to the risk of atherosclerotic cardiovascular disease. It has been assumed that this reflects the protective functions of HDL, which include their ability to promote cholesterol efflux. Yet, several recent pharmacological and genetic studies have failed to demonstrate that increased plasma levels of HDL-C resulted in decreased cardiovascular disease risk, giving rise to a controversy regarding whether plasma levels of HDL-C reflect HDL function, or that HDL is even as protective as assumed. The evidence from preclinical and (limited) clinical studies shows that HDL can promote the regression of atherosclerosis when the levels of functional particles are increased from endogenous or exogenous sources. The data show that regression results from a combination of reduced plaque lipid and macrophage contents, as well as from a reduction in its inflammatory state. Although more research will be needed regarding basic mechanisms and to establish that these changes translate clinically to reduced cardiovascular disease events, that HDL can regress plaques suggests that the recent trial failures do not eliminate HDL from consideration as an atheroprotective agent but rather emphasizes the important distinction between HDL function and plasma levels of HDL-C.
Collapse
Affiliation(s)
- Jonathan E Feig
- From the Departments of Medicine (Cardiology) and Cell Biology, Marc and Ruti Bell Vascular Biology Program, New York University School of Medicine, New York, NY (J.E.F., B.H., E.A.F.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH (J.D.S., S.L.H.). J.E.F. is currently affiliated with Department of Medicine (Cardiology), Mount Sinai School of Medicine, New York, NY. B.H. is currently affiliated with Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
42
|
Hewing B, Parathath S, Barrett T, Chung WKK, Astudillo YM, Hamada T, Ramkhelawon B, Tallant TC, Yusufishaq MSS, Didonato JA, Huang Y, Buffa J, Berisha SZ, Smith JD, Hazen SL, Fisher EA. Effects of native and myeloperoxidase-modified apolipoprotein a-I on reverse cholesterol transport and atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2014; 34:779-89. [PMID: 24407029 DOI: 10.1161/atvbaha.113.303044] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Preclinical and clinical studies have shown beneficial effects of infusions of apolipoprotein A-I (ApoA-I) on atherosclerosis. ApoA-I is also a target for myeloperoxidase-mediated oxidation, leading in vitro to a loss of its ability to promote ATP-binding cassette transporter A1-dependent macrophage cholesterol efflux. Therefore, we hypothesized that myeloperoxidase-mediated ApoA-I oxidation would impair its promotion of reverse cholesterol transport in vivo and the beneficial effects on atherosclerotic plaques. APPROACH AND RESULTS ApoA-I(-/-) or apolipoprotein E-deficient mice were subcutaneously injected with native human ApoA-I, oxidized human ApoA-I (myeloperoxidase/hydrogen peroxide/chloride treated), or carrier. Although early postinjection (8 hours) levels of total ApoA-I in plasma were similar for native versus oxidized human ApoA-I, native ApoA-I primarily resided within the high-density lipoprotein fraction, whereas the majority of oxidized human ApoA-I was highly cross-linked and not high-density lipoprotein particle associated, consistent with impaired ATP-binding cassette transporter A1 interaction. In ApoA-I(-/-) mice, ApoA-I oxidation significantly impaired reverse cholesterol transport in vivo. In advanced aortic root atherosclerotic plaques of apolipoprotein E-deficient mice, native ApoA-I injections led to significant decreases in lipid content, macrophage number, and an increase in collagen content; in contrast, oxidized human ApoA-I failed to mediate these changes. The decrease in plaque macrophages with native ApoA-I was accompanied by significant induction of their chemokine receptor CCR7. Furthermore, only native ApoA-I injections led to a significant reduction of inflammatory M1 and increase in anti-inflammatory M2 macrophage markers in the plaques. CONCLUSIONS Myeloperoxidase-mediated oxidation renders ApoA-I dysfunctional and unable to (1) promote reverse cholesterol transport, (2) mediate beneficial changes in the composition of atherosclerotic plaques, and (3) pacify the inflammatory status of plaque macrophages.
Collapse
Affiliation(s)
- Bernd Hewing
- From the Department of Medicine, Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine (B.H., S.P., T.B., W.K.K.C., Y.M.A., T.H., B.R., E.A.F.); Medizinische Klinik für Kardiologie und Angiologie, Charité-Universitaetsmedizin Berlin, Berlin, Germany (B.H.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (T.C.T., M.S.S.Y., J.A.D., Y.H., J.B., S.Z.B., J.D.S., S.L.H.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sontag TJ, Krishack PA, Lukens JR, Bhanvadia CV, Getz GS, Reardon CA. Apolipoprotein A-I protection against atherosclerosis is dependent on genetic background. Arterioscler Thromb Vasc Biol 2013; 34:262-9. [PMID: 24334873 DOI: 10.1161/atvbaha.113.302831] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Inbred mouse strains have different susceptibilities to experimental atherosclerosis. The C57BL/6 strain is among the most sensitive and has, therefore, been the most widely used in atherosclerosis studies, whereas many strains are resistant. The FVB/N strain is highly resistant to atherosclerosis on the apolipoprotein E (apoE)- and low-density lipoprotein (LDL) receptor-deficient backgrounds. High-density lipoprotein and its major apoprotein, apoA-I, have been shown to be protective against atherogenesis on the C57BL/6 background. We here examine the influence of genetic background on the atheroprotective nature of apoA-I. APPROACH AND RESULTS ApoE-deficient/apoA-I-deficient mice were generated in the C57BL/6 and FVB/N strains from apoE-deficient mice. After 6 to 10 weeks on a Western-type diet, plasma lipids and atherosclerotic lesion size were assessed. Macrophage recruitment, cholesterol regulation, and blood monocyte levels were examined as potential mechanisms driving lesion size differences. FVB/N knockout mice had higher plasma very-LDL/LDL cholesterol than their C57BL/6 counterparts. ApoA-I deficiency decreased very-LDL/LDL cholesterol in C57BL/6 mice but not in FVB/N mice. FVB/N single and double knockout mice had less lesion than C57BL/6 6 to 10 weeks on diet. ApoA-I deficiency augmented lesion development only in C57BL/6 mice. Macrophage recruitment to thioglycollate-treated peritoneum and diet-induced blood monocyte levels reflected the pattern of lesion development among the 4 genotypes. ApoA-I deficiency increased macrophage cholesterol content only in C57BL/6. FVB/N plasma was a better acceptor for macrophage cholesterol efflux than C57BL/6. CONCLUSIONS ApoA-I is atheroprotective only in certain genetic contexts. In the C57BL/6 context, but not FVB/N, apoA-I decreases inflammatory macrophage recruitment and monocytosis, contributors to lesion formation.
Collapse
|
44
|
Circulation Research
Thematic Synopsis Diabetes and Obesity. Circ Res 2013; 113:e62-75. [DOI: 10.1161/circresaha.113.302431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Sanson M, Distel E, Fisher EA. HDL induces the expression of the M2 macrophage markers arginase 1 and Fizz-1 in a STAT6-dependent process. PLoS One 2013; 8:e74676. [PMID: 23991225 PMCID: PMC3749183 DOI: 10.1371/journal.pone.0074676] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 08/03/2013] [Indexed: 12/22/2022] Open
Abstract
Our lab has previously shown in a mouse model that normalization of a low HDL level achieves atherosclerotic plaque regression. This included the shift from a pro ("M1") to an anti-inflammatory ("M2") phenotypic state of plaque macrophages. Whether HDL can directly cause this phenotypic change and, if so, what the signaling mechanism is, were explored in the present studies. Murine primary macrophages treated with HDL showed increased gene expression for the M2 markers Arginase-1 (Arg-1) and Fizz-1, which are classically induced by IL-4. HDL was able to potentiate the IL-4-induced changes in Arg-1, and tended to do the same for Fizz-1, while suppressing the expression of inflammatory genes in response to IFNγ. The effects of either IL-4 or HDL were suppressed when macrophages were from STAT6(-/-) mice, but inhibitor studies suggested differential utilization of JAK isoforms by IL-4 and HDL to activate STAT6 by phosphorylation. Overall, our results describe a new function of HDL, namely its ability to directly enrich macrophages in markers of the M2, anti-inflammatory, state in a process requiring STAT6.
Collapse
Affiliation(s)
- Marie Sanson
- The Leon H. Charney Division of Cardiology and Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Emilie Distel
- The Leon H. Charney Division of Cardiology and Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Edward A. Fisher
- The Leon H. Charney Division of Cardiology and Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
46
|
Abstract
Why patients with chronic kidney disease have elevated cardiovascular risk remains elusive. In this issue of Immunity, Speer et al. (2013) show that natural modification of high density lipoprotein promotes hypertension through a Toll-like receptor-dependent mechanism.
Collapse
Affiliation(s)
- Kathryn J Moore
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10029, USA.
| | | |
Collapse
|
47
|
Murphy AJ, Funt S, Gorman D, Tall AR, Wang N. Pegylation of high-density lipoprotein decreases plasma clearance and enhances antiatherogenic activity. Circ Res 2013; 113:e1-e9. [PMID: 23613182 DOI: 10.1161/circresaha.113.301112] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE Infusions of apolipoprotein AI (apoAI), mimetic peptides, or high-density lipoprotein (HDL) remain a promising approach for the treatment of atherosclerotic coronary disease. However, rapid clearance leads to a requirement for repeated administration of large amounts of material and limits effective plasma concentrations. OBJECTIVE Because pegylation of purified proteins is commonly used as a method to increase their half-life in the circulation, we determined whether pegylation of apoAI or HDL would increase its plasma half-life and in turn its antiatherogenic potential. METHODS AND RESULTS Initial pegylation attempts using lipid-poor apoAI showed a marked tendency to form multi-pegylated (PEG) species with reduced ability to promote cholesterol efflux from macrophage foam cells. However, pegylation of human holo-HDL or reconstituted phospholipid/apoAI particles (rHDL) led to selective N-terminal monopegylation of apoAI with full preservation of cholesterol efflux activity. The plasma clearance of PEG-rHDL was estimated after injection into hypercholesterolemic Apoe-/- mice; the half-life of pegylated PEG-apoAI after injection of PEG-rHDL was increased ≈7-fold compared with apoAI in nonpegylated rHDL. In comparison with nonpegylated rHDL, infusion of PEG-rHDL (40 mg/kg) into hypercholesterolemic Apoe-/- mice led to more pronounced suppression of bone marrow myeloid progenitor cell proliferation and monocytosis, as well as reduced atherosclerosis and a stable plaque phenotype. CONCLUSIONS We describe a novel method for effective monopegylation of apoAI in HDL particles, in which lipid binding seems to protect against pegylation of key functional residues. Pegylation of apoAI in rHDL markedly increases its plasma half-life and enhances antiatherogenic properties in vivo.
Collapse
Affiliation(s)
- Andrew J Murphy
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, New York 10032, USA
| | - Samuel Funt
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, New York 10032, USA
| | - Darren Gorman
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, New York 10032, USA
| | - Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, New York 10032, USA
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, New York 10032, USA
| |
Collapse
|
48
|
Fisher EA, Feig JE, Hewing B, Hazen SL, Smith JD. High-density lipoprotein function, dysfunction, and reverse cholesterol transport. Arterioscler Thromb Vasc Biol 2013; 32:2813-20. [PMID: 23152494 DOI: 10.1161/atvbaha.112.300133] [Citation(s) in RCA: 266] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although high high-density lipoprotein (HDL)-cholesterol levels are associated with decreased cardiovascular risk in epidemiological studies, recent genetic and pharmacological findings have raised doubts about the beneficial effects of HDL. Raising HDL levels in animal models by infusion or overexpression of apolipoprotein A-I has shown clear vascular improvements, such as delayed atherosclerotic lesion progression and accelerated lesion regression, along with increased reverse cholesterol transport. Inflammation and other factors, such as myeloperoxidase-mediated oxidation, can impair HDL production and HDL function, with regard to its reverse cholesterol transport, antioxidant, and anti-inflammatory activities. Thus, tests of HDL function, which have not yet been developed as routine diagnostic assays, may prove useful and be a better predictor of cardiovascular risk than HDL-cholesterol levels.
Collapse
Affiliation(s)
- Edward A Fisher
- Department of Cardiovascular Medicine, New York University School of Medicine, New York, NY 444195, USA
| | | | | | | | | |
Collapse
|