1
|
Packard CJ, Taskinen MR, Björnson E, Matikainen N, Söderlund S, Andersson L, Adiels M, Borén J. Genetically determined increase in apolipoprotein C-III (APOC3 gain-of-function) delays very low-density lipoprotein clearance in humans. Atherosclerosis 2025; 404:119166. [PMID: 40203662 DOI: 10.1016/j.atherosclerosis.2025.119166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/02/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
AIM Apolipoprotein C-III (apoC-III) is an important regulator of triglyceride (TG) metabolism and a target for intervention. The present study examined the effects of gain-of-function (GOF) variants in APOC3 on apolipoprotein B kinetics to understand further how changes in the synthesis of this apolipoprotein impact triglyceride-rich lipoprotein (TRL) metabolism. METHODS Two groups of subjects were recruited by population screening, 9 carriers of known APOC3 GOF variants and 9 age-, sex- and BMI-matched non-carriers. The kinetics of TRL were determined using stable isotope tracers of apoprotein and triglyceride metabolism in a non-steady-state protocol involving administration of a fat-rich meal. RESULTS APOC3 GOF carriers had 47 % higher plasma apoC-III levels compared to non-carriers (P = 0.022) and higher production rates for the apolipoprotein. Post-prandial response (total area-under-curve) for plasma TG was 108 % greater in GOF carriers compared to non-carriers (P = 0.002) due specifically to higher levels of VLDL1. In contrast, no difference was seen in the chylomicron apoB48 response. Comparison of TRL kinetics between groups showed that APOC3 GOF carriers had lower fractional clearance rates for VLDL1-apoB100 and VLDL1-apoB48-containing particles (P < 0.02), but no difference in VLDL1-apoB100 or chylomicron apoB48 production rates. Both the rate of VLDL lipolysis and the rate of clearance of VLDL particles from the circulation were lower in APOC3 GOF carriers than in non-carriers. In contrast, chylomicron apoB clearance rates did not differ between APOC3 GOF carriers and non-carriers. CONCLUSION APOC3 GOF carriers showed specific alterations in TRL metabolism (compared to matched non-carriers), namely slower lipolysis and delayed clearance of VLDL1-sized particles, but no difference in chylomicron metabolism. Our findings suggest that intervention to reduce apoC-III production can be modelled as a reduction in TRL, particularly VLDL particle levels, without deleterious effects on fat absorption or hepatic VLDL production.
Collapse
Affiliation(s)
- C J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - M R Taskinen
- Research Programs Unit, Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - E Björnson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden
| | - N Matikainen
- Research Programs Unit, Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland; Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - S Söderlund
- Research Programs Unit, Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland; Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - L Andersson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden
| | - M Adiels
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden
| | - J Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden; Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
2
|
Hirano T. Clinical significance of small dense low-density lipoprotein cholesterol measurement in type 2 diabetes. J Diabetes Investig 2025; 16:370-383. [PMID: 39778086 PMCID: PMC11871407 DOI: 10.1111/jdi.14398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/18/2024] [Accepted: 12/22/2024] [Indexed: 01/11/2025] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) is known to be a causal substance of atherosclerosis, but its usefulness as a predictive biomarker for atherosclerotic cardiovascular disease (ASCVD) is limited. In patients with type 2 diabetes (T2D), LDL-C concentrations do not markedly increase, while triglycerides (TG) concentrations are usually elevated. Although TG is associated with ASCVD risk, they do not play a direct role in the formation of atheromatous plaques. TG changes the risk of ASCVD in a way that is dependent on LDL-C, and TG is the primary factor in reducing LDL particle size. Small dense (sd)LDL, a potent atherogenic LDL subfraction, best explains the "Atherogenic Duo" of TG and LDL-C. Although hypertriglyceridemia is associated with small-sized LDL, patients with severe hypertriglyceridemia and low LDL-C rarely develop ASCVD. This suggests that quantifying sdLDL is more clinically relevant than measuring LDL size. We developed a full-automated direct sdLDL-C assay, and it was proven that sdLDL-C is a better predictor of ASCVD than LDL-C. The sdLDL-C level is specifically elevated in patients with metabolic syndrome and T2D who have insulin resistance. Due to its clear link to metabolic dysfunction, sdLDL-C could be named "metabolic LDL-C." Insulin resistance/hyperinsulinemia promotes TG production in the liver, causing steatosis and overproduction of VLDL1, a precursor of sdLDL. sdLDL-C is closely associated with steatotic liver disease and chronic kidney disease, which are common complications in T2D. This review focuses on T2D and discusses the clinical significance of sdLDL-C including its composition, pathophysiology, measurements, association with ASCVD, and treatments.
Collapse
Affiliation(s)
- Tsutomu Hirano
- Diabetes CenterEbina General HospitalEbina CityKanagawaJapan
| |
Collapse
|
3
|
Dai LR, Lyu L, Zhan WY, Jiang S, Zhou PZ. Genetic Evidence for Causal Effects of Circulating Remnant Lipid Profile on Cerebral Hemorrhage and Ischemic Stroke: A Mendelian Randomization Study. World Neurosurg 2025; 195:123649. [PMID: 39889961 DOI: 10.1016/j.wneu.2024.123649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/27/2024] [Indexed: 02/03/2025]
Abstract
BACKGROUND Mendelian randomization was employed to investigate the impact of circulating lipids, specifically residual lipids, on the risk of susceptibility to cerebral hemorrhage and ischemic stroke. METHODS According to the previous studies, we chose 19 circulating lipids, comprising 6 regular lipids and 13 residual lipids, to investigate their potential causal relationship with intracranial hemorrhage and ischemic stroke. The effect estimates were computed utilizing the random-effects inverse-variance-weighted methodology. RESULTS The findings revealed negative correlations between high-density lipoprotein cholesterol (HDL-C) and cerebral hemorrhage and large artery stroke. HDL-C, apolipoprotein A1 (Apo A1), TG in very small VLDL, and TG in IDL were found to be negatively correlated with any ischemic stroke. apolipoprotein B (Apo B), triglycerides (TG), low-density lipoprotein cholestrol (LDL-C), L.VLDL-TG, TG in medium VLDL, and TG in small VLDL exhibited positive correlations with large artery stroke. TG in very large HDL and TG in IDL were positively correlated with cardioembolic stroke. No significant causal relationship was observed between circulating lipids, with the exception of HDL-C and cerebral hemorrhage. No causal relationship was identified between any circulating lipids and small vessel stroke. Furthermore, the causal relationships were only found between residual lipids and ischemic stroke. CONCLUSIONS This study provides evidence for the beneficial impact of Apo A1 and HDL-C in reducing the risk of ischemic stroke, as well as the protective effect of HDL-C against cerebral hemorrhage. It highlights the detrimental effects of Apo B, TG, and LDL-C in increasing the risk of ischemic stroke, particularly in cases of large artery stroke. Furthermore, the study underscores the heterogeneity and 2-sided effects of the causal relationship between triglyceride-rich lipoproteins and ischemic stroke, offering a promising avenue for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Li-Rui Dai
- Department of Neurosurgery, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan, China
| | - Liang Lyu
- Department of Neurosurgery, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan, China
| | - Wen-Yi Zhan
- Department of Neurosurgery, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan, China
| | - Shu Jiang
- Department of Neurosurgery, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan, China
| | - Pei-Zhi Zhou
- Department of Neurosurgery, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Fu L, Liu Q, Cheng H, Zhao X, Xiong J, Mi J. Insights Into Causal Effects of Genetically Proxied Lipids and Lipid-Modifying Drug Targets on Cardiometabolic Diseases. J Am Heart Assoc 2025; 14:e038857. [PMID: 39868518 DOI: 10.1161/jaha.124.038857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The differential impact of serum lipids and their targets for lipid modification on cardiometabolic disease risk is debated. This study used Mendelian randomization to investigate the causal relationships and underlying mechanisms. METHODS Genetic variants related to lipid profiles and targets for lipid modification were sourced from the Global Lipids Genetics Consortium. Summary data for 10 cardiometabolic diseases were compiled from both discovery and replication data sets. Expression quantitative trait loci data from relevant tissues were employed to evaluate significant lipid-modifying drug targets. Comprehensive analyses including colocalization, mediation, and bioinformatics were conducted to validate the results and investigate potential mediators and mechanisms. RESULTS Significant causal associations were identified between lipids, lipid-modifying drug targets, and various cardiometabolic diseases. Notably, genetic enhancement of LPL (lipoprotein lipase) was linked to reduced risks of myocardial infarction (odds ratio [OR]1, 0.65 [95% CI, 0.57-0.75], P1=2.60×10-9; OR2, 0.59 [95% CI, 0.49-0.72], P2=1.52×10-7), ischemic heart disease (OR1, 0.968 [95% CI, 0.962-0.975], P1=5.50×10-23; OR2, 0.64 [95% CI, 0.55-0.73], P2=1.72×10-10), and coronary heart disease (OR1, 0.980 [95% CI, 0.975-0.985], P1=3.63×10-14; OR2, 0.64 [95% CI, 0.54-0.75], P2=6.62×10-8) across 2 data sets. Moreover, significant Mendelian randomization and strong colocalization associations for the expression of LPL in blood and subcutaneous adipose tissue were linked with myocardial infarction (OR, 0.918 [95% CI, 0.872-0.967], P=1.24×10-3; PP.H4, 0.99) and coronary heart disease (OR, 0.991 [95% CI, 0.983-0.999], P=0.041; PP.H4=0.92). Glucose levels and blood pressure were identified as mediators in the total effect of LPL on cardiometabolic outcomes. CONCLUSIONS The study substantiates the causal role of lipids in specific cardiometabolic diseases, highlighting LPL as a potent drug target. The effects of LPL are suggested to be influenced by changes in glucose and blood pressure, providing insights into its mechanism of action.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non-Communicable Disease Management Beijing Children's Hospital, Capital Medical University, National Center for Children's Health Beijing China
| | - Qin Liu
- Department of Ultrasound Children's Hospital of the Capital Institute of Pediatrics Beijing China
| | - Hong Cheng
- Department of Epidemiology Capital Institute of Pediatrics Beijing China
| | - Xiaoyuan Zhao
- Department of Epidemiology Capital Institute of Pediatrics Beijing China
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control Department Shenzhen Center for Chronic Disease Control Shenzhen China
| | - Jie Mi
- Center for Non-Communicable Disease Management Beijing Children's Hospital, Capital Medical University, National Center for Children's Health Beijing China
- Key Laboratory of Major Diseases in Children, Ministry of Education China
| |
Collapse
|
5
|
Puig N, Camps-Renom P, Hermansson M, Aguilera-Simón A, Marín R, Bautista O, Rotllan N, Blanco-Sanroman N, Domine MC, Öörni K, Sánchez-Quesada JL, Benitez S. Alterations in LDL and HDL after an ischemic stroke associated with carotid atherosclerosis are reversed after 1 year. J Lipid Res 2025; 66:100739. [PMID: 39746448 PMCID: PMC11815653 DOI: 10.1016/j.jlr.2024.100739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025] Open
Abstract
Approximately, 20% of ischemic strokes are attributed to the presence of atherosclerosis. Lipoproteins play a crucial role in the development of atherosclerosis, with LDL promoting atherogenesis and HDL inhibiting it. Therefore, both their concentrations and their biological properties are decisive factors in atherosclerotic processes. In this study, we examined the qualitative properties of lipoproteins in ischemic stroke patients with carotid atherosclerosis. Lipoproteins were isolated from the blood of healthy controls (n = 27) and patients with carotid atherosclerosis (n = 64) at 7 days and 1 year postischemic stroke. Compared to controls, patients' LDL 7 days poststroke showed increased levels of apoC-III, triacylglycerol, and ceramide, along with decreased cholesterol and phospholipid content. LDL from patients induced more inflammation in macrophages than did LDL from controls. HDL isolated from patients 7 days after stroke showed alterations in the apolipoprotein cargo, with reduced levels of apoA-I and increased levels of apoA-II, and apoC-III compared to controls. Patients' HDL also showed a higher electronegative charge than that of controls and partially lost its ability to counteract the modification of LDL and the inflammatory effects of modified LDL. One year after stroke onset, the composition of patients' LDL and HDL resembled those of the controls. In parallel, LDL and HDL gained positive charge, LDL became less prone to oxidation and aggregation, and HDL regained protective properties. In conclusion, LDL and HDL in ischemic stroke patients with carotid atherosclerosis exhibited alterations in composition and function, which were partially reversed 1 year after stroke.
Collapse
Affiliation(s)
- Núria Puig
- Cardiovascular Biochemistry Group, Institut de Recerca Sant Pau, (IR Sant Pau), Barcelona, Spain
| | - Pol Camps-Renom
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IR Sant Pau, Barcelona, Spain
| | - Martin Hermansson
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Ana Aguilera-Simón
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IR Sant Pau, Barcelona, Spain
| | - Rebeca Marín
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IR Sant Pau, Barcelona, Spain
| | - Olga Bautista
- Cardiovascular Biochemistry Group, Institut de Recerca Sant Pau, (IR Sant Pau), Barcelona, Spain
| | - Noemi Rotllan
- Pathophysiology of Lipid-Related Diseases, Research Institute Sant Pau (Institut de Recerca Sant Pau, IR Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | | | | | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - José Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Institut de Recerca Sant Pau, (IR Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain.
| | - Sonia Benitez
- Cardiovascular Biochemistry Group, Institut de Recerca Sant Pau, (IR Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain.
| |
Collapse
|
6
|
Mocciaro G, George AL, Allison M, Frontini M, Huang‐Doran I, Reiman F, Gribble F, Griffin JL, Vidal‐Puig A, Azzu V, Kay R, Vacca M. Oxidised Apolipoprotein Peptidome Characterises Metabolic Dysfunction-Associated Steatotic Liver Disease. Liver Int 2025; 45:e16200. [PMID: 39822152 PMCID: PMC11740006 DOI: 10.1111/liv.16200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) encompasses a spectrum of histological conditions ranging from simple steatosis to fibrosing steatohepatitis, and is a risk factor for cardiovascular diseases (CVD). While oxidised apolipoproteins A and B have been linked to obesity and CVD, the association between other oxidised apolipoproteins and MASLD is yet to be established. To fill this gap, we characterised the circulating serum peptidome of patients with MASLD. METHODS We studied the serum of 87 biopsy-confirmed MASLD patients and 20 age- and sex-matched control (CTRL) subjects. We first employed an untargeted LC-MS/MS peptidomics approach (9 CTRL, 32 MASLD) to identify key hits differentially modulated, and subsequently validated the most relevant findings through targeted peptidomics in an enlarged study population (87 MASLD and 20 CTRL). RESULTS Untargeted serum peptidomics identified several oxidised apolipoprotein peptide fragments, including ApoE and ApoC-III, significantly upregulated in MASLD compared to CTRL. Specifically focusing on the oxidative status of intact ApoC-III, studied through its major glycoforms (ApoC-III0, ApoC-IIIi and ApoC-IIIii), we observed a marked reduction in non-oxidised forms of these circulating peptides alongside substantially increased levels of their oxidised proteoforms in MASLD versus controls (but not within the disease stages). Oxidised ApoE and ApoC-III peptide fragments were also significantly correlated with obesity, insulin resistance, dyslipidaemia and transaminases, suggesting a potential link between circulating apolipoprotein oxidation and systemic/hepatic metabolic dysfunction. CONCLUSION Our data reveals a previously unreported oxidised apolipoprotein profile associated with MASLD. The functional and clinical implications of these findings warrant further mechanistic investigation.
Collapse
Grants
- Wellcome Trust
- MR/S010483/1 Medical Research Council
- This research was supported by NIHR Cambridge Biomedical Research Centre (BRC-1215-20014) funding to V.A., M.A., and M.V.The initial establishment of the cohorts was supported by the Evelyn Trust (funding to M.A. and A.V.P.), Mason Medical Research Trust (funding to V.A.), and the Academy of Medical Sciences (funding to V.A.). V.A. was supported by the University of Cambridge. M.A. is supported by Cambridge University Hospitals NHS Foundation Trust. M.V. is supported by the University of Bari (Horizon Europe Seed cod. id. S06-miRNASH), the Foundation for Liver Research (Intramural Funding), Associazione Italiana Ricerca sul Cancro (IG2022 Grant n. 27521) and Ministry of University and Research on Next Generation EU Funds [COD: P202222FCC, CUP: H53D23009960001, D.D. MUR 1366 (01-09-2023), Title: "System Biology" approaches in HCV Patients with Residual Hepatic Steatosis after Viral Eradication; Cod PE00000003, CUP: H93C22000630001, DD MUR 1550, Title: "ON Foods - Research and innovation network on food and nutrition Sustainability, Safety and Security - Working ON Foods"; Cod: CN00000041, CUP: H93C22000430007, Title PNRR "National Center for Gene Therapy and Drugs based on RNA Technology", M4C2-Investment 1.4; Code: CN00000013, CUP: H93C22000450007, Title PNNR: "National Centre for HPC, Big Data and Quantum Computing").A.V.P. is funded by MRC MDU, MRC Metabolic Diseases Unit (MC_UU_00014/5): Disease Model Core, Biochemistry Assay Lab, Histology Core and British Heart Foundation. M.F. is supported by the British Heart Foundation (FS/18/53/33863) and the British Heart Foundation Cambridge Centre for Research Excellence (RE/18/1/34212). This study was supported by the National Institute for Health and Care Research Exeter Biomedical Research Centre. We would like to thank all participants in this study and the NIHR National BioResource. The views expressed are those of the author(s) and not necessarily those of the NIHR or the Department of Health and Social Care. JLG is funded by the Medical Research Council (MR/S010483/1; MR/P011705/1; MR/P01836X/1; MR/X012700/1). F.G., F.R., R.K. and A.L.G. were funded by the Wellcome Trust (grants 106262/Z/14/Z, 106263/Z/14/Z), the MRC Metabolic Diseases Unit (grants MRC MC UU 12012/3, MRC MC UU12012/5) and by the NIHR Cambridge Biomedical Research Centre. The mass spectrometers were obtained using the Medical Research Council "Enhancing UK Clinical Research" grant (MR/M009041/1). The views expressed are those of the author(s) and not necessarily those of the relevant funding or supporting institutions. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript
Collapse
Affiliation(s)
- Gabriele Mocciaro
- Roger Williams Institute of Liver StudiesFoundation for Liver ResearchLondonUK
| | - Amy L. George
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Michael Allison
- Liver Unit, Cambridge NIHR Biomedical Research CentreCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Mattia Frontini
- Faculty of Health and Life Sciences, Clinical and Biomedical SciencesUniversity of Exeter Medical SchoolExeterUK
| | - Isabel Huang‐Doran
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Frank Reiman
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Fiona Gribble
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Julian L. Griffin
- The Rowett Institute, Foresterhill CampusUniversity of AberdeenAberdeenUK
| | - Antonio Vidal‐Puig
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Vian Azzu
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
- Liver Unit, Cambridge NIHR Biomedical Research CentreCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Richard Kay
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Michele Vacca
- Roger Williams Institute of Liver StudiesFoundation for Liver ResearchLondonUK
- University of Bari "Aldo Moro"Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni"BariItaly
| |
Collapse
|
7
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025:10.1038/s41569-024-01111-0. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Zhuang Y, Zhang Y, Liu C, Zhong Y. Interplay Between the Circadian Clock and Sirtuins. Int J Mol Sci 2024; 25:11469. [PMID: 39519022 PMCID: PMC11545976 DOI: 10.3390/ijms252111469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
The circadian clock is an autonomous timekeeping system evolved by organisms to adapt to external changes, regulating a variety of important physiological and behavioral processes. Recent studies have shown that the sirtuin family of histone deacetylases is involved in regulating the expression of clock genes and plays an important role in maintaining the normal rhythm of clock gene expression and behavior. Moreover, sirtuins are regulated directly or indirectly by the circadian clock system. The mutual regulation between the circadian clock and sirtuins is likely involved in a variety of signal transduction and metabolism processes. In this review, we discuss the molecular mechanisms and research progress on the intertwined relationship between the circadian clock and sirtuins, mainly in mammals, highlighting sirtuins as molecular links between metabolic control and circadian rhythms and offering our perspectives on future developments in the field.
Collapse
Affiliation(s)
- Yan Zhuang
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yantong Zhang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Chao Liu
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yingbin Zhong
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
9
|
Liu D, Mai D, Jahn AN, Murray TA, Aitchison JD, Gern BH, Urdahl KB, Aderem A, Diercks AH, Gold ES. APOE Protects Against Severe Infection with Mycobacterium tuberculosis by Restraining Production of Neutrophil Extracellular Traps. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616580. [PMID: 39605723 PMCID: PMC11601580 DOI: 10.1101/2024.10.04.616580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
While neutrophils are the predominant cell type in the lungs of humans with active tuberculosis (TB), they are relatively scarce in the lungs of most strains of mice that are used to study the disease. However, similar to humans, neutrophils account for approximately 45% of CD45+ cells in the lungs of Apoe -/- mice on a high-cholesterol (HC) diet following infection with Mycobacterium tuberculosis (Mtb). We hypothesized that the susceptibility of Apoe -/- HC mice might arise from an unrestrained feed-forward loop in which production of neutrophil extracellular traps (NETs) stimulates production of type I interferons by pDCs which in turn leads to the recruitment and activation of more neutrophils, and demonstrated that depleting neutrophils, depleting plasmacytoid dendritic cells (pDCs), or blocking type I interferon signaling, improved the outcome of infection. In concordance with these results, we found that Mtb-infected in Apoe -/- HC mice produce high levels of LTB4 and 12-HETE, two eicosanoids known to act as neutrophil chemoattractants and showed that blocking leukotriene B4 (LTB4) receptor signaling also improved the outcome of tuberculosis. While production of NETs has been associated with severe tuberculosis in other mouse models and in humans, a causative role for NETs in the pathology has not been directly established. We demonstrate that blocking the activation of peptidylarginine deiminase 4 (PAD4), an enzyme critical to NET formation, leads to fewer NETs in the lungs and, strikingly, completely reverses the hypersusceptibility of Apoe -/- HC mice to tuberculosis.
Collapse
Affiliation(s)
- Dong Liu
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Dat Mai
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Ana N. Jahn
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Tara A. Murray
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - John D. Aitchison
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Benjamin H. Gern
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
- University of Washington, Dept. of Pediatrics; Seattle, Washington, USA
| | - Kevin B. Urdahl
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
- University of Washington, Dept. of Immunology; Seattle, Washington, USA
- University of Washington, Dept. of Pediatrics; Seattle, Washington, USA
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Alan H. Diercks
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Elizabeth S. Gold
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
- Virginia Mason Franciscan Health; Seattle, WA, 98101, USA
| |
Collapse
|
10
|
Besin V, Humardani FM, Yulianti T, Putra SED, Triana R, Justyn M. The Apo gene's genetic variants: hidden role in Asian vascular risk. Neurogenetics 2024; 25:157-164. [PMID: 38625441 DOI: 10.1007/s10048-024-00757-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
Vascular risk factors, including diabetes, hypertension, hyperlipidemia, and obesity, pose significant health threats with implications extending to neuropsychiatric disorders such as stroke and Alzheimer's disease. The Asian population, in particular, appears to be disproportionately affected due to unique genetic predispositions, as well as epigenetic factors such as dietary patterns and lifestyle habits. Existing management strategies often fall short of addressing these specific needs, leading to greater challenges in prevention and treatment. This review highlights a significant gap in our understanding of the impact of genetic screening in the early detection and tailored treatment of vascular risk factors among the Asian population. Apolipoprotein, a key player in cholesterol metabolism, is primarily associated with dyslipidemia, yet emerging evidence suggests its involvement in conditions such as diabetes, hypertension, and obesity. While genetic variants of vascular risk are ethnic-dependent, current evidence indicates that epigenetics also exhibits ethnic specificity. Understanding the interplay between Apolipoprotein and genetics, particularly within diverse ethnic backgrounds, has the potential to refine risk stratification and enhance precision in management. For Caucasian carrying the APOA5 rs662799 C variant, pharmacological interventions are recommended, as dietary interventions may not be sufficient. In contrast, for Asian populations with the same genetic variant, dietary modifications are initially advised. Should dyslipidemia persist, the consideration of pharmaceutical agents such as statins is recommended.
Collapse
Affiliation(s)
| | - Farizky Martriano Humardani
- Faculty of Medicine, Universitas Surabaya, Surabaya, Indonesia.
- Magister in Biomedical Science Program, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia.
- Bioinformatics Research Center, Indonesian Bioinformatics and Biomolecular, Malang, Indonesia.
| | - Trilis Yulianti
- Prodia Education and Research Institute, Jakarta, Indonesia
- Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | | | - Rina Triana
- Prodia Clinical Laboratories, Jakarta, Indonesia
| | - Matthew Justyn
- Faculty of Pharmacy, Universitas Padjajaran, Jatinangor, Indonesia
| |
Collapse
|
11
|
Lusiki Z, Blom D, Soko ND, Malema S, Jones E, Rayner B, Blackburn J, Sinxadi P, Dandara MT, Dandara C. Major Genetic Drivers of Statin Treatment Response in African Populations and Pharmacogenetics of Dyslipidemia Through a One Health Lens. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:261-279. [PMID: 37956269 DOI: 10.1089/omi.2023.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
A One Health lens is increasingly significant to address the intertwined challenges in planetary health concerned with the health of humans, nonhuman animals, plants, and ecosystems. A One Health approach can benefit the public health systems in Africa that are overburdened by noncommunicable, infectious, and environmental diseases. Notably, the COVID-19 pandemic revealed the previously overlooked two-fold importance of pharmacogenetics (PGx), for individually tailored treatment of noncommunicable diseases and environmental pathogens. For example, dyslipidemia, a common cardiometabolic risk factor, has been identified as an independent COVID-19 severity risk factor. Observational data suggest that patients with COVID-19 infection receiving lipid-lowering therapy may have better outcomes. However, among African patients, the response to these drugs varies from patient to patient, pointing to the possible contribution of genetic variation in important pharmacogenes. The PGx of lipid-lowering therapies may underlie differences in treatment responses observed among dyslipidemia patients as well as patients comorbid with COVID-19 and dyslipidemia. Genetic variations in APOE, ABCB1, CETP, CYP2C9, CYP3A4, CYP3A5, HMGCR, LDLR, NPC1L1, and SLCO1B1 genes affect the pharmacogenomics of statins, and they have individually been linked to differential responses to dyslipidemia and COVID-19 treatment. African populations are underrepresented in PGx research. This leads to poor accounting of additional diverse genetic variants that could be important in understanding interindividual and between-population variations in therapeutic responses to dyslipidemia and COVID-19. This expert review examines and synthesizes the salient and priority PGx variations, as seen through a One Health lens in Africa, to improve and inform personalized medicine in both dyslipidemia and COVID-19.
Collapse
Affiliation(s)
- Zizo Lusiki
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Dirk Blom
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Lipidology and Cape Heart Institute, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Nyarai D Soko
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Smangele Malema
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Erika Jones
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Nephrology and Hypertension, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Brian Rayner
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Nephrology and Hypertension, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Jonathan Blackburn
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Phumla Sinxadi
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Michelle T Dandara
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| |
Collapse
|
12
|
Heianza Y, Xue Q, Rood J, Clish CB, Bray GA, Sacks FM, Qi L. Changes in bile acid subtypes and improvements in lipid metabolism and atherosclerotic cardiovascular disease risk: the Preventing Overweight Using Novel Dietary Strategies (POUNDS Lost) trial. Am J Clin Nutr 2024; 119:1293-1300. [PMID: 38428740 PMCID: PMC11130658 DOI: 10.1016/j.ajcnut.2024.02.019] [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: 08/30/2023] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Distinct circulating bile acid (BA) subtypes may play roles in regulating lipid homeostasis and atherosclerosis. OBJECTIVES We investigated whether changes in circulating BA subtypes induced by weight-loss dietary interventions were associated with improved lipid profiles and atherosclerotic cardiovascular disease (ASCVD) risk estimates. METHODS This study included adults with overweight or obesity (n = 536) who participated in a randomized weight-loss dietary intervention trial. Circulating primary and secondary unconjugated BAs and their taurine-/glycine-conjugates were measured at baseline and 6 mo after the weight-loss diet intervention. The ASCVD risk estimates were calculated using the validated equations. RESULTS At baseline, higher concentrations of specific BA subtypes were related to higher concentrations of atherogenic very low-density lipoprotein lipid subtypes and ASCVD risk estimates. Weight-loss diet-induced decreases in primary BAs were related to larger reductions in triglycerides and total cholesterol [every 1 standard deviation (SD) decrease of glycocholate, glycochenodeoxycholate, or taurochenodeoxycholate was related to β (standard error) -3.3 (1.3), -3.4 (1.3), or -3.8 (1.3) mg/dL, respectively; PFDR < 0.05 for all]. Greater decreases in specific secondary BA subtypes were also associated with improved lipid metabolism at 6 mo; there was β -4.0 (1.1) mg/dL per 1-SD decrease of glycoursodeoxycholate (PFDR =0.003) for changes in low-density lipoprotein cholesterol. We found significant interactions (P-interaction < 0.05) between dietary fat intake and changes in BA subtypes on changes in ASCVD risk estimates; decreases in primary and secondary BAs (such as conjugated cholate or deoxycholate) were significantly associated with improved ASCVD risk after consuming a high-fat diet, but not after consuming a low-fat diet. CONCLUSIONS Decreases in distinct BA subtypes were associated with improved lipid profiles and ASCVD risk estimates, highlighting the importance of changes in circulating BA subtypes as significant factors linked to improved lipid metabolism and ASCVD risk estimates in response to weight-loss dietary interventions. Habitual dietary fat intake may modify the associations of changes in BAs with ASCVD risk. This trial was registered at clinicaltrials.gov as NCT00072995.
Collapse
Affiliation(s)
- Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States.
| | - Qiaochu Xue
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Jennifer Rood
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Clary B Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - George A Bray
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Frank M Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| |
Collapse
|
13
|
Yang CH, Ho YH, Tang HY, Lo CJ. NMR-Based Analysis of Plasma Lipoprotein Subclass and Lipid Composition Demonstrate the Different Dietary Effects in ApoE-Deficient Mice. Molecules 2024; 29:988. [PMID: 38474500 DOI: 10.3390/molecules29050988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Plasma lipid levels are commonly measured using traditional methods such as triglycerides (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), and cholesterol (CH). However, the use of newer technologies, such as nuclear magnetic resonance (NMR) with post-analysis platforms, has made it easier to assess lipoprotein profiles in research. In this study involving ApoE-deficient mice that were fed high-fat diets, significant changes were observed in TG, CH, free cholesterol (FC), and phospholipid (PL) levels within the LDL fraction. The varied proportions of TG in wild-type mice and CH, FC, and PL in ApoE-/- mice were strikingly different in very low-density lipoproteins (VLDL), LDL, intermediate-density lipoprotein (IDL), and HDL. This comprehensive analysis expands our understanding of lipoprotein subfractions and the impacts of the APOE protein and high-fat diet in mouse models. The new testing method allows for a complete assessment of plasma lipids and their correlation with genetic background and diet in mice.
Collapse
Affiliation(s)
- Cheng-Hung Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Yu-Hsuan Ho
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Hsiang-Yu Tang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Chi-Jen Lo
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan City 33302, Taiwan
| |
Collapse
|
14
|
Packard CJ, Pirillo A, Tsimikas S, Ference BA, Catapano AL. Exploring apolipoprotein C-III: pathophysiological and pharmacological relevance. Cardiovasc Res 2024; 119:2843-2857. [PMID: 38039351 PMCID: PMC11484501 DOI: 10.1093/cvr/cvad177] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/21/2022] [Accepted: 02/07/2023] [Indexed: 12/03/2023] Open
Abstract
The availability of pharmacological approaches able to effectively reduce circulating LDL cholesterol (LDL-C) has led to a substantial reduction in the risk of atherosclerosis-related cardiovascular disease (CVD). However, a residual cardiovascular (CV) risk persists in treated individuals with optimal levels of LDL-C. Additional risk factors beyond LDL-C are involved, and among these, elevated levels of triglycerides (TGs) and TG-rich lipoproteins are causally associated with an increased CV risk. Apolipoprotein C-III (apoC-III) is a key regulator of TG metabolism and hence circulating levels through several mechanisms including the inhibition of lipoprotein lipase activity and alterations in the affinity of apoC-III-containing lipoproteins for both the hepatic receptors involved in their removal and extracellular matrix in the arterial wall. Genetic studies have clarified the role of apoC-III in humans, establishing a causal link with CVD and showing that loss-of-function mutations in the APOC3 gene are associated with reduced TG levels and reduced risk of coronary heart disease. Currently available hypolipidaemic drugs can reduce TG levels, although to a limited extent. Substantial reductions in TG levels can be obtained with new drugs that target specifically apoC-III; these include two antisense oligonucleotides, one small interfering RNA and an antibody.
Collapse
Affiliation(s)
- Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Milan, Italy
- Center for the Study of Dyslipidaemias, IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK
| | - Alberico L Catapano
- Center for the Study of Dyslipidaemias, IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
15
|
Haring B, Schumacher H, Mancia G, Teo KK, Lonn EM, Mahfoud F, Schmieder R, Mann JFE, Sliwa K, Yusuf S, Böhm M. Triglyceride-glucose index, low-density lipoprotein levels, and cardiovascular outcomes in chronic stable cardiovascular disease: results from the ONTARGET and TRANSCEND trials. Eur J Prev Cardiol 2024; 31:311-319. [PMID: 37890035 DOI: 10.1093/eurjpc/zwad340] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 09/22/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023]
Abstract
AIMS The triglyceride-glucose index (TyG) has been proposed as an alternative to insulin resistance and as a predictor of cardiovascular outcomes. Little is known on its role in chronic stable cardiovascular disease and its predictive power at controlled low density lipoprotein (LDL) levels. METHODS AND RESULTS Our study population consisted of 29 960 participants in the ONTARGET and TRANSCEND trials that enrolled patients with known atherosclerotic disease. Triglycerides and glucose were measured at baseline. TyG was calculated as the logarithmized product of fasting triglycerides and glucose divided by 2. The primary endpoint of both trials was a composite of cardiovascular death, myocardial infarction, stroke, or hospitalization for heart failure. The secondary endpoint was all-cause death and the components of the primary endpoint. Cox proportional hazards models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CI) with extensive covariate adjustment for demographic, medical history, and lifestyle factors. During a mean follow-up of 4.3 years, 4895 primary endpoints and 3571 all-cause deaths occurred. In fully adjusted models, individuals in the highest compared to the lowest quartile of the TyG index were at higher risk for the primary endpoint (HR 1.14; 95% CI 1.05-1.25) and for myocardial infarction (HR 1.30; 95% CI 1.11-1.53). A higher TyG index did not associate with the primary endpoint in individuals with LDL levels < 100 mg/dL. CONCLUSION A higher TyG index is associated with a modestly increased cardiovascular risk in chronic stable cardiovascular disease. This association is largely attenuated when LDL levels are controlled. REGISTRATION www.clinicaltrials.gov: NCT00153101.
Collapse
Affiliation(s)
- Bernhard Haring
- Department of Medicine III, Saarland University, Kirrberger Strasse 100, 66421 Homburg, Germany
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Giuseppe Mancia
- Instituto Clinico Universitario Policlinico di Monza, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo, 1, Milano, Italy
| | - Koon K Teo
- Population Health Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Canada
| | - Eva M Lonn
- Population Health Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Canada
| | - Felix Mahfoud
- Department of Medicine III, Saarland University, Kirrberger Strasse 100, 66421 Homburg, Germany
| | - Roland Schmieder
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen, Germany
| | - Johannes F E Mann
- KfH Kidney Centre, München, Germany
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen, Germany
| | - Karen Sliwa
- Faculty of Health Sciences, Hatter Institute for Cardiovascular Research in Africa & IIDMM, University of Cape Town, Cape Town, South Africa
| | - Salim Yusuf
- Population Health Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Canada
| | - Michael Böhm
- Department of Medicine III, Saarland University, Kirrberger Strasse 100, 66421 Homburg, Germany
| |
Collapse
|
16
|
Zheng C, Andraski AB, Khoo C, Furtado JD, Sacks FM. Food Intake Suppresses ApoB Secretion and Fractional Catabolic Rates in Humans. Arterioscler Thromb Vasc Biol 2024; 44:435-451. [PMID: 38126174 DOI: 10.1161/atvbaha.123.319769] [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: 06/25/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Humans spend much of the day in the postprandial state. However, most research and clinical guidelines on plasma lipids pertain to blood drawn after a 12-hour fast. We aimed to study the metabolic differences of apoB lipoproteins between the fasting and postprandial states. METHODS We investigated plasma apoB metabolism using stable isotope tracers in 12 adult volunteers under fasting and continuous postprandial conditions in a randomized crossover study. We determined the metabolism of apoB in multiple lipoprotein subfractions, including light and dense VLDLs (very-low-density lipoproteins), IDLs (intermediate-density lipoproteins), and light and dense LDLs (low-density lipoproteins) that do or do not contain apoE or apoC3. RESULTS A major feature of the postprandial state is 50% lower secretion rate of triglyceride-rich lipoproteins and concurrent slowdown of their catabolism in circulation, as shown by 34% to 55% lower rate constants for the metabolic pathways of conversion by lipolysis from larger to smaller lipoproteins and direct clearance of lipoproteins from the circulation. In addition, the secretion pattern of apoB lipoprotein phenotypes was shifted from particles containing apoE and apoC3 in the fasting state to those without either protein in the postprandial state. CONCLUSIONS Overall, during the fasting state, hepatic apoB lipoprotein metabolism is activated, characterized by increased production, transport, and clearance. After food intake, endogenous apoB lipoprotein metabolism is globally reduced as appropriate to balance dietary input to maintain the supply of energy to peripheral tissues.
Collapse
Affiliation(s)
- Chunyu Zheng
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.Z., A.B.A., C.K., J.D.F., F.M.S.)
- National Resilience, Inc, La Jolla, CA (C.Z.)
| | - Allison B Andraski
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.Z., A.B.A., C.K., J.D.F., F.M.S.)
| | - Christina Khoo
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.Z., A.B.A., C.K., J.D.F., F.M.S.)
- Ocean Spray Cranberries, Inc, Middleboro-Lakeville, MA (C.K.)
| | - Jeremy D Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.Z., A.B.A., C.K., J.D.F., F.M.S.)
- Biogen, Cambridge, MA (J.D.F.)
| | - Frank M Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.Z., A.B.A., C.K., J.D.F., F.M.S.)
| |
Collapse
|
17
|
Bhatt V, Tiwari AK. Sirtuins, a key regulator of ageing and age-related neurodegenerative diseases. Int J Neurosci 2023; 133:1167-1192. [PMID: 35549800 DOI: 10.1080/00207454.2022.2057849] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
Abstract
Sirtuins are Nicotinamide Adenine Dinucleotide (NAD+) dependent class ІΙΙ histone deacetylases enzymes (HDACs) present from lower to higher organisms such as bacteria (Sulfolobus solfataricus L. major), yeasts (Saccharomyces cerevisiae), nematodes (Caenorhabditis elegans), fruit flies (Drosophila melanogaster), humans (Homo sapiens sapiens), even in plants such as rice (Oryza sativa), thale cress (Arabidopsis thaliana), vine (Vitis vinifera L.) tomato (Solanum lycopersicum). Sirtuins play an important role in the regulation of various vital cellular functions during metabolism and ageing. It also plays a neuroprotective role by modulating several biological pathways such as apoptosis, DNA repair, protein aggregation, and inflammatory processes associated with ageing and neurodegenerative diseases. In this review, we have presented an updated Sirtuins and its role in ageing and age-related neurodegenerative diseases (NDDs). Further, this review also describes the therapeutic potential of Sirtuins and the use of Sirtuins inhibitor/activator for altering the NDDs disease pathology.
Collapse
Affiliation(s)
- Vidhi Bhatt
- Department of Biological Sciences & Biotechnology, Institute of Advanced Research, Koba, Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Department of Biological Sciences & Biotechnology, Institute of Advanced Research, Koba, Gandhinagar, Gujarat, India
| |
Collapse
|
18
|
Syed-Abdul MM, Tian L, Lewis GF. Unanticipated Enhancement of Intestinal TG Output by Apoc3 ASO Inhibition. Arterioscler Thromb Vasc Biol 2023; 43:2133-2142. [PMID: 37675633 DOI: 10.1161/atvbaha.123.319765] [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: 06/20/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The objective of this study was to investigate whether apoC3 (apolipoprotein C3) inhibition with an antisense oligonucleotide (ASO) modulates intestinal triglyceride secretion. METHODS Sprague-Dawley rats were treated with subcutaneous injections of apoC3 ASO 25 mg/kg twice weekly or inactive ASO for 4 weeks before the assessment of lymph flow, triglyceride and apoB48 (apolipoprotein B48) appearance in the lymph. Rats were surgically implanted with catheters in the mesenteric lymph duct and duodenum. Following an overnight fast, an intraduodenal lipid bolus (1.5-mL intralipid) was administered. Lymph fluid was collected for the following 4 hours to compare effects on lymph flow, lymph triglyceride and apoB48 concentration, and secretion. To assess suppression of apoC3 expression and protein abundance by apoC3 ASO compared with inactive ASO (placebo), intestinal and hepatic tissues were collected from a subset of animals before (fasting) and after an enteral lipid bolus (post-lipid). RESULTS ApoC3 ASO significantly reduced apoC3 mRNA expression in the liver compared with inactive ASO (fasting: 42%, P=0.0048; post-lipid: 66%, P<0.001) and in the duodenum (fasting: 29%, P=0.0424; post-lipid: 53%, P=0.0120). As expected, plasma triglyceride also decreased significantly (fasting: 74%, P<0.001; post-lipid: 33%, P=0.0276). Lymph flow and cumulative lymph volume remained unchanged following apoC3 ASO therapy; however, lymph triglyceride, but not apoB48 output, increased by 38% (ANOVA, P<0.001). Last, no changes were observed in stool triglyceride, intestinal fat (quantified via oil red O staining), and expression of mRNAs involved in triglyceride synthesis, lipid droplet formation, and chylomicron transport and secretion. CONCLUSIONS Despite the marked reduction in plasma triglyceride concentration that occurs with apoC3 ASO inhibition, intestinal triglyceride output surprisingly increased rather than decreased. These data demonstrate that the reduction of intestinal triglyceride output does not contribute to the potent plasma triglyceride-lowering observed with this novel therapy for hypertriglyceridemia. Further studies are required to explore the mechanism of this intestinal effect.
Collapse
Affiliation(s)
- Majid Mufaqam Syed-Abdul
- Division of Endocrinology, Department of Medicine and Banting & Best Diabetes Centre, University of Toronto, ON, Canada
| | - Lili Tian
- Division of Endocrinology, Department of Medicine and Banting & Best Diabetes Centre, University of Toronto, ON, Canada
| | - Gary F Lewis
- Division of Endocrinology, Department of Medicine and Banting & Best Diabetes Centre, University of Toronto, ON, Canada
| |
Collapse
|
19
|
Michaeli DT, Michaeli JC, Albers S, Boch T, Michaeli T. Established and Emerging Lipid-Lowering Drugs for Primary and Secondary Cardiovascular Prevention. Am J Cardiovasc Drugs 2023; 23:477-495. [PMID: 37486464 PMCID: PMC10462544 DOI: 10.1007/s40256-023-00594-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2023] [Indexed: 07/25/2023]
Abstract
Despite treatment with statins, patients with elevated low-density lipoprotein cholesterol (LDL-C) and triglycerides remain at increased risk for adverse cardiovascular events. Consequently, novel pharmaceutical drugs have been developed to control and modify the composition of blood lipids to ultimately prevent fatal cardiovascular events in patients with dyslipidaemia. This article reviews established and emerging lipid-lowering drugs regarding their mechanism of action, development stage, ongoing clinical trials, side effects, effect on blood lipids and reduction in cardiovascular morbidity and mortality. We conducted a keyword search to identify studies on established and emerging lipid modifying drugs. Results were summarized in a narrative overview. Established pharmaceutical treatment options include the Niemann-Pick-C1 like-1 protein (NPC1L1) inhibitor ezetimibe, the protein convertase subtilisin-kexin type 9 (PCSK9) inhibitors alirocumab and evolocumab, fibrates as peroxisome proliferator receptor alpha (PPAR-α) activators, and the omega-3 fatty acid icosapent ethyl. Statins are recommended as the first-line therapy for primary and secondary cardiovascular prevention in patients with hypercholesterinaemia and hypertriglyceridemia. For secondary prevention in hypercholesterinaemia, second-line options such as statin add-on or statin-intolerant treatments are ezetimibe, alirocumab and evolocumab. For secondary prevention in hypertriglyceridemia, second-line options such as statin add-on or statin-intolerant treatments are icosapent ethyl and fenofibrate. Robust data for these add-on therapeutics in primary cardiovascular prevention remains scarce. Recent biotechnological advances have led to the development of innovative small molecules (bempedoic acid, lomitapide, pemafibrate, docosapentaenoic and eicosapentaenoic acid), antibodies (evinacumab), antisense oligonucleotides (mipomersen, volanesorsen, pelcarsen, olezarsen), small interfering RNA (inclisiran, olpasiran), and gene therapies for patients with dyslipidemia. These molecules specifically target new cellular pathways, such as the adenosine triphosphate-citrate lyase (bempedoic acid), PCSK9 (inclisiran), angiopoietin-like 3 (ANGPTL3: evinacumab), microsomal triglyceride transfer protein (MTP: lomitapide), apolipoprotein B-100 (ApoB-100: mipomersen), apolipoprotein C-III (ApoC-III: volanesorsen, olezarsen), and lipoprotein (a) (Lp(a): pelcarsen, olpasiran). The authors are hopeful that the development of new treatment modalities alongside new therapeutic targets will further reduce patients' risk of adverse cardiovascular events. Apart from statins, data on new drugs' use in primary cardiovascular prevention remain scarce. For their swift adoption into clinical routine, these treatments must demonstrate safety and efficacy as well as cost-effectiveness in randomized cardiovascular outcome trials.
Collapse
Affiliation(s)
- Daniel Tobias Michaeli
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany.
| | - Julia Caroline Michaeli
- Department of Obstetrics and Gynaecology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Albers
- Department of Orthopaedics and Sport Orthopaedics, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boch
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Heidelberg University, Heidelberg, Germany
| | - Thomas Michaeli
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
20
|
Li X, Shao X, Xue Q, Kou M, Champagne CM, Koseva BS, Heianza Y, Grundberg E, Bazzano LA, Bray GA, Sacks FM, Qi L. DNA Methylation Near CPT1A and Changes in Triglyceride-rich Lipoproteins in Response to Weight-loss Diet Interventions. J Clin Endocrinol Metab 2023; 108:e542-e549. [PMID: 36800272 PMCID: PMC10348458 DOI: 10.1210/clinem/dgad086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023]
Abstract
CONTEXT Carnitine palmitoyltransferase-1A, encoded by the CPT1A gene, plays a key role in the oxidation of long-chain fatty acids in the mitochondria and may be important in triglyceride metabolism. Previous work has shown that high fat intake was negatively associated with CPT1A methylation and positively associated with CPT1A expression. OBJECTIVE We aim to investigate the association of DNA methylation (DNAm) at the CPT1A gene with reductions in triglycerides and triglyceride-rich lipoproteins (TRLs) in response to weight-loss diet interventions. METHODS The current study included 538 White participants, who were randomly assigned to 1 of 4 diets varying in macronutrient components. We defined the regional DNAm at CPT1A as the average methylation level over CpGs within 500 bp of the 3 triglyceride-related DNAm sites. RESULTS Dietary fat intake significantly modified the association between baseline DNAm at CPT1A and 2-year changes in total plasma triglycerides, independent of concurrent weight loss. Among participants assigned to a low-fat diet, a higher regional DNAm level at CPT1A was associated with a greater reduction in total plasma triglycerides at 2 years (P = .01), compared with those assigned to a high-fat diet (P = .64) (P interaction = .018). Further investigation on lipids and apolipoproteins in very low-density lipoprotein (VLDL) revealed similar interaction patterns for 2-year changes in VLDL-triglycerides, VLDL-cholesterol, and VLDL-apolipoprotein B (P interaction = .009, .002, and .016, respectively), but not for VLDL-apoC-III (P interaction = .36). CONCLUSION Participants with a higher regional DNAm level at CPT1A benefit more in long-term improvement in triglycerides, particularly in the TRLs and related apolipoproteins when consuming a low-fat weight-loss diet.
Collapse
Affiliation(s)
- Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Xiaojian Shao
- Digital Technologies Research Centre, National Research Council Canada, Ottawa, Ontario K1C 0R6, Canada
| | - Qiaochu Xue
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Minghao Kou
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Catherine M Champagne
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Boryana S Koseva
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, Kansas City, MO 64108, USA
| | - Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Elin Grundberg
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, Kansas City, MO 64108, USA
| | - Lydia A Bazzano
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - George A Bray
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Frank M Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
21
|
Li J, Sun H, Wang Y, Liu J, Wang G. Apolipoprotein C3 is negatively associated with estrogen and mediates the protective effect of estrogen on hypertriglyceridemia in obese adults. Lipids Health Dis 2023; 22:29. [PMID: 36855114 PMCID: PMC9972754 DOI: 10.1186/s12944-023-01797-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Both estrogen and apolipoprotein C3 (ApoC3) play crucial roles in lipid metabolism. But the link between them remains unclear, and it is unknown whether estrogen regulates triglyceride (TG) levels via ApoC3. Researchers hypothesized that estrogen exerts a regulatory effect on ApoC3 metabolism, and that this regulation could play a significant role in lipid metabolism. To explore this potential link, the present investigation aimed to examine the associations between estradiol (E2), ApoC3, and TG levels in both males and females. METHODS A total of 519 obese people (133 males and 386 premenopausal females) were recruited. Based on their TG levels, the participants were split into two groups [hypertriglyceridemia (HTG) group: TG ≥ 1.7 mmol/L; control group: TG < 1.7 mmol/L]. Serum ApoC3, E2, and TG levels were measured and compared in those two groups for both sexes separately. To ascertain the connection among E2, ApoC3, and TG, linear regression and mediation analysis were used. RESULTS Participants in the HTG group presented higher levels of ApoC3 (P < 0.001). In contrast, they tend to have lower E2 levels than the control. Linear regression analysis proposed that in both sexes, E2 was negatively associated with ApoC3 levels. The relationship remained significant after adjustment for confounding factors (male: standardized β = -0.144, t = -2.392, P < 0.05; female: standardized β = -0.077, t = -2.360, P < 0.001). Furthermore, mediation analysis revealed the relationship between reduced E2 levels and elevated TG levels is directly mediated by ApoC3. CONCLUSIONS In obese men and premenopausal women, ApoC3 was negatively and linearly correlated with serum E2 levels. The findings showed that estrogen may suppress ApoC3 expression and thus lower TG levels.
Collapse
Affiliation(s)
- Jinman Li
- grid.411607.5Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020 China
| | - Honglin Sun
- grid.411607.5Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020 China
| | - Ying Wang
- grid.411607.5Department of Medical Examination, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China.
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
22
|
Genetic Heterogeneity of Familial Hypercholesterolemia: Repercussions for Molecular Diagnosis. Int J Mol Sci 2023; 24:ijms24043224. [PMID: 36834635 PMCID: PMC9961636 DOI: 10.3390/ijms24043224] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Genetics of Familial Hypercholesterolemia (FH) is ascribable to pathogenic variants in genes encoding proteins leading to an impaired LDL uptake by the LDL receptor (LDLR). Two forms of the disease are possible, heterozygous (HeFH) and homozygous (HoFH), caused by one or two pathogenic variants, respectively, in the three main genes that are responsible for the autosomal dominant disease: LDLR, APOB and PCSK9 genes. The HeFH is the most common genetic disease in humans, being the prevalence about 1:300. Variants in the LDLRAP1 gene causes FH with a recessive inheritance and a specific APOE variant was described as causative of FH, contributing to increase FH genetic heterogeneity. In addition, variants in genes causing other dyslipidemias showing phenotypes overlapping with FH may mimic FH in patients without causative variants (FH-phenocopies; ABCG5, ABCG8, CYP27A1 and LIPA genes) or act as phenotype modifiers in patients with a pathogenic variant in a causative gene. The presence of several common variants was also considered a genetic basis of FH and several polygenic risk scores (PRS) have been described. The presence of a variant in modifier genes or high PRS in HeFH further exacerbates the phenotype, partially justifying its variability among patients. This review aims to report the updates on the genetic and molecular bases of FH with their implication for molecular diagnosis.
Collapse
|
23
|
Proteomic analysis of postprandial high-density lipoproteins in healthy subjects. Int J Biol Macromol 2023; 225:1280-1290. [PMID: 36427620 DOI: 10.1016/j.ijbiomac.2022.11.187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
The relationship between the functionality and composition of high-density lipoproteins (HDL) is yet not fully studied, and little is known about the influence of the diet in HDL proteome. Therefore, the aim of this research was to elucidate the HDL proteome associated to postprandial hyperlipidemia. Male volunteers were recruited for an interventional study with high fatty acid-based meals. Blood samples were collected before the intake (baseline), and 2-3 (postprandial peak) and 5-6 (postprandial post peak) hours later. HDL were purified and the protein composition was quantified by LC-MS/MS. Statistical analysis was performed by lineal models (amica) and by ANOVA and multi-t-test of the different conditions (MetaboAnalyst). Additionally, a clustering of the expression profiles of each protein was done with coseq R package (RStudio). Initially, 320 proteins were identified but only 119 remained after the filtering. APOM, APOE, APOB, and APOA2, proteins previously identified in the HDL proteome, were the only proteins with a statistically significant altered expression in postprandial hyperlipidemia when compared to baseline (p values <0.05 and logFC >1). In conclusion, we have been able to describe several behaviors of the whole HDL proteome during the postprandial hyperlipidemic metabolism.
Collapse
|
24
|
Recio-López P, Valladolid-Acebes I, Hadwiger P, Hossbach M, Krampert M, Prata C, Berggren PO, Juntti-Berggren L. Treatment of the metabolic syndrome by siRNA targeting apolipoprotein CIII. Biofactors 2023; 49:153-172. [PMID: 36039858 DOI: 10.1002/biof.1885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/05/2022] [Indexed: 11/06/2022]
Abstract
Apolipoprotein CIII (apoCIII) is increased in obesity-induced insulin resistance and type-2 diabetes. Emerging evidences support the advantages of small interfering RNAs (siRNAs) to target disease-causing genes. The aim of this study was to develop siRNAs for in vivo silencing of apoCIII and investigate if this results in metabolic improvements comparable to what we have seen using antisense oligonucelotides against apoCIII. Twenty-four siRNAs were synthesized and tested in a dual luciferase reporter assay. The eight best were selected, based on knockdown at 20 nM, and of these, two were selected based on IC50 values. In vivo experiments were performed in ob/ob mice, an obese animal model for diabetes. To determine the dose-dependency, efficacy, duration of effect and therapeutic dose we used a short protocol giving the apoCIII-siRNA mix for three days. To evaluate long-term metabolic effects mice were treated for three days, every second week for eight weeks. The siRNA mix effectively and selectively reduced expression of apoCIII in liver in vivo. Treatment had to be repeated every two weeks to maintain a suppression of apoCIII. The reduction of apoCIII resulted in increased LPL activity, lower triglycerides, reduced liver fat, ceased weight gain, enhanced insulin sensitivity, and improved glucose homeostasis. No off-target or side effects were observed during the eight-week treatment period. These results suggest that in vivo silencing of apoCIII with siRNA, is a promising approach with the potential to be used in the battle against obesity-induced metabolic disorders.
Collapse
Affiliation(s)
- Patricia Recio-López
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | | | | | | | | | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Lisa Juntti-Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| |
Collapse
|
25
|
Giammanco A, Spina R, Cefalù AB, Averna M. APOC-III: a Gatekeeper in Controlling Triglyceride Metabolism. Curr Atheroscler Rep 2023; 25:67-76. [PMID: 36689070 PMCID: PMC9947064 DOI: 10.1007/s11883-023-01080-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW Apolipoprotein C-III (ApoC-III) is a widely known player in triglyceride metabolism, and it has been recently recognized as a polyhedric factor which may regulate several pathways beyond lipid metabolism by influencing cardiovascular, metabolic, and neurological disease risk. This review summarizes the different functions of ApoC-III and underlines the recent findings related to its multifaceted pathophysiological role. RECENT FINDINGS The role of ApoC-III has been implicated in HDL metabolism and in the development of atherosclerosis, inflammation, and ER stress in endothelial cells. ApoC-III has been recently considered an important player in insulin resistance mechanisms, lipodystrophy, diabetic dyslipidemia, and postprandial hypertriglyceridemia (PPT). The emerging evidence of the involvement of ApoC-III in the in the pathogenesis of Alzheimer's disease open the way to further study if modification of ApoC-III level slows disease progression. Furthermore, ApoC-III is clearly linked to cardiovascular disease (CVD) risk, and progression of coronary artery disease (CAD) as well as the calcification of aortic valve and recent clinical trials has pointed out the inhibition of ApoC-III as a promising approach to manage hypertriglyceridemia and prevent CVD. Several evidences highlight the role of ApoC-III not only in triglyceride metabolism but also in several cardio-metabolic pathways. Results from recent clinical trials underline that the inhibition of ApoC-III is a promising therapeutical strategy for the management of severe hypertriglyceridemia and in CVD prevention.
Collapse
Affiliation(s)
- Antonina Giammanco
- grid.10776.370000 0004 1762 5517Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), University of Palermo, Palermo, Italy
| | - Rossella Spina
- grid.10776.370000 0004 1762 5517Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), University of Palermo, Palermo, Italy
| | - Angelo B. Cefalù
- grid.10776.370000 0004 1762 5517Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), University of Palermo, Palermo, Italy
| | - Maurizio Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro" (PROMISE), University of Palermo, Palermo, Italy. .,Institute of Biophysics (IBF), National Research Council (CNR), Palermo, Italy.
| |
Collapse
|
26
|
Sex-related differences in single nucleotide polymorphisms associated with dyslipidemia in a Korean population. Lipids Health Dis 2022; 21:124. [PMID: 36419087 PMCID: PMC9685854 DOI: 10.1186/s12944-022-01736-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/14/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The prevalence of dyslipidemia has increased steadily in Korea, and the incidence of dyslipidemia differs by sex. In this study, we identified single nucleotide polymorphisms (SNPs) related to dyslipidemia in Korean cohorts through genome-wide association study (GWAS) analysis. METHODS Genotyping was conducted to determine the genotypes of 72,298 participants and investigate genotypes for 7,079,946 SNPs. Sex, age, and BMI were set as covariates for GWAS, and significant SNPs were identified in the discovery and replication stages using logistic regression. RESULTS GWAS of the entire cohort revealed a total of five significant SNPs: rs117026536 (LPL), rs651821 (APOA5), rs9804646 (APOA5), rs9926440 (CETP), and rs429358 (APOE). GWAS of the male subjects revealed a total of four significant SNPs. While rs9804646 (APOA5) and rs429358 (APOE) were significant for all the subjects, rs662799 (APOA5) and rs56156922 (CETP) were significant only for the male subjects. GWAS of the female subjects revealed two significant SNPs, rs651821 (APOA5) and rs9804646 (APOA5), both of which were significant in all the subjects. CONCLUSION This is the first study to identify sex-related differences in genetic polymorphisms in Korean populations with dyslipidemia. Further studies considering environmental variables will be needed to elucidate these sex-related genetic differences in dyslipidemia.
Collapse
|
27
|
Vyletelová V, Nováková M, Pašková Ľ. Alterations of HDL's to piHDL's Proteome in Patients with Chronic Inflammatory Diseases, and HDL-Targeted Therapies. Pharmaceuticals (Basel) 2022; 15:1278. [PMID: 36297390 PMCID: PMC9611871 DOI: 10.3390/ph15101278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/03/2022] [Accepted: 10/14/2022] [Indexed: 09/10/2023] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, steatohepatitis, periodontitis, chronic kidney disease, and others are associated with an increased risk of atherosclerotic cardiovascular disease, which persists even after accounting for traditional cardiac risk factors. The common factor linking these diseases to accelerated atherosclerosis is chronic systemic low-grade inflammation triggering changes in lipoprotein structure and metabolism. HDL, an independent marker of cardiovascular risk, is a lipoprotein particle with numerous important anti-atherogenic properties. Besides the essential role in reverse cholesterol transport, HDL possesses antioxidative, anti-inflammatory, antiapoptotic, and antithrombotic properties. Inflammation and inflammation-associated pathologies can cause modifications in HDL's proteome and lipidome, transforming HDL from atheroprotective into a pro-atherosclerotic lipoprotein. Therefore, a simple increase in HDL concentration in patients with inflammatory diseases has not led to the desired anti-atherogenic outcome. In this review, the functions of individual protein components of HDL, rendering them either anti-inflammatory or pro-inflammatory are described in detail. Alterations of HDL proteome (such as replacing atheroprotective proteins by pro-inflammatory proteins, or posttranslational modifications) in patients with chronic inflammatory diseases and their impact on cardiovascular health are discussed. Finally, molecular, and clinical aspects of HDL-targeted therapies, including those used in therapeutical practice, drugs in clinical trials, and experimental drugs are comprehensively summarised.
Collapse
Affiliation(s)
| | | | - Ľudmila Pašková
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University, 83232 Bratislava, Slovakia
| |
Collapse
|
28
|
Cinnamaldehyde Mitigates Atherosclerosis Induced by High-Fat Diet via Modulation of Hyperlipidemia, Oxidative Stress, and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4464180. [PMID: 35774377 PMCID: PMC9239836 DOI: 10.1155/2022/4464180] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/16/2022] [Accepted: 05/19/2022] [Indexed: 12/26/2022]
Abstract
Atherosclerosis is a disease in which plaque builds up inside arteries. Cinnamaldehyde (Ci) has many biological properties that include anti-inflammatory and antioxidant activities. Thus, this study was designed to explore the protective effect of Ci against atherosclerosis induced by a high-fat diet (HFD) in Wistar rats. Atherosclerosis was induced by an oral administration of an HFD for 10 weeks. Atherosclerosis-induced rats were supplemented with Ci at a dose of 20 mg/kg bw dissolved in 0.5% dimethyl sulfoxide (DMSO), daily by oral gavage for the same period. Rats were divided into three groups of 10 rats each fed with (a) ND, (b) HFD, and (c) HFD+Ci, daily for 10 weeks. Treatment of rats with Ci significantly reduced the elevated levels of serum total cholesterol (T.Ch), triglycerides (TG), low-density lipoprotein-cholesterol (LDL-Ch), very low-density lipoprotein-cholesterol (VLDL-Ch), and free fatty acids (FFAs) and significantly increased the lowered levels of high-density lipoprotein-cholesterol (HDL-Ch) level. Ci ameliorated the increased cardiovascular risk indices 1 and 2 and the decreased antiatherogenic index. Moreover, Ci reduced the elevated serum creatine kinase (CK), creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), and aspartate aminotransferase (AST) activities. Ci also improved the heart antioxidant activities by decreasing malondialdehyde (MDA) and increasing glutathione S-transferase (GST), superoxide dismutase (SOD), catalase (CAT), reduced glutathione (GSH), and glutathione peroxidase (Gpx) activities. Furthermore, the supplementation with Ci downregulated the mRNA expression levels of interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-17 (IL-17), and tumor necrosis factor-α (TNF-α). Thus, Ci successfully elicited a therapeutic impact against atherosclerosis induced by HFD via its hypolipidemic, antioxidant, and anti-inflammatory actions.
Collapse
|
29
|
Identification of different lipoprotein response types in people following a Mediterranean diet pattern with and without whole eggs. Nutr Res 2022; 105:82-96. [DOI: 10.1016/j.nutres.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 11/19/2022]
|
30
|
Abstract
Apolipoproteins are important structural components of plasma lipoproteins that influence vascular biology and atherosclerotic disease pathophysiology by regulating lipoprotein metabolism. Clinically important apolipoproteins related to lipid metabolism and atherogenesis include apolipoprotein B-100, apolipoprotein B-48, apolipoprotein A-I, apolipoprotein C-II, apolipoprotein C-III, apolipoprotein E and apolipoprotein(a). Apolipoprotein B-100 is the major structural component of VLDL, IDL, LDL and lipoprotein(a). Apolipoprotein B-48 is a truncated isoform of apolipoprotein B-100 that forms the backbone of chylomicrons. Apolipoprotein A-I provides the scaffolding for lipidation of HDL and has an important role in reverse cholesterol transport. Apolipoproteins C-II, apolipoprotein C-III and apolipoprotein E are involved in triglyceride-rich lipoprotein metabolism. Apolipoprotein(a) covalently binds to apolipoprotein B-100 to form lipoprotein(a). In this Review, we discuss the mechanisms by which these apolipoproteins regulate lipoprotein metabolism and thereby influence vascular biology and atherosclerotic disease. Advances in the understanding of apolipoprotein biology and their translation into therapeutic agents to reduce the risk of cardiovascular disease are also highlighted.
Collapse
|
31
|
de la Parra Soto LG, Gutiérrez-Uribe JA, Sharma A, Ramírez-Jiménez AK. Is Apo-CIII the new cardiovascular target? An analysis of its current clinical and dietetic therapies. Nutr Metab Cardiovasc Dis 2022; 32:295-308. [PMID: 34895805 DOI: 10.1016/j.numecd.2021.09.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/21/2021] [Accepted: 09/30/2021] [Indexed: 11/23/2022]
Abstract
AIMS Recently, Apolipoprotein CIII (Apo-CIII) has gained remarkable attention since its overexpression has been strongly correlated to cardiovascular disease (CVD) occurrence. The aim of this review was to summarize the latest findings of Apo-CIII as a CVDs and diabetes risk factor, as well as the plausible mechanisms involved in the development of these pathologies, with particular emphasis on current clinical and dietetic therapies. DATA SYNTHESIS Apo-CIII is a small protein (∼8.8 kDa) that, among other functions, inhibits lipoprotein lipase, a key enzyme in lipid metabolism. Apo-CIII plays a fundamental role in the physiopathology of atherosclerosis, type-1, and type-2 diabetes. Apo-CIII has become a potential clinical target to tackle these multifactorial diseases. Dietetic (omega-3 fatty acids, stanols, polyphenols, lycopene) and non-dietetic (fibrates, statins, and antisense oligonucleotides) therapies have shown promising results to regulate Apo-CIII and triglyceride levels. However, more information from clinical trials is required to validate it as a new target for atherosclerosis and diabetes types 1 and 2. CONCLUSIONS There are still several pathways involving Apo-CIII regulation that might be affected by bioactive compounds that need further research. The mechanisms that trigger metabolic responses following bioactive compounds consumption are mainly related to higher LPL expression and PPARα activation, although the complete pathways are yet to be elucidated.
Collapse
Affiliation(s)
- Lorenzo G de la Parra Soto
- Tecnologico de Monterrey, School of Engineering and Sciences, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico
| | - Janet A Gutiérrez-Uribe
- Tecnologico de Monterrey, School of Engineering and Sciences, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico
| | - Ashutosh Sharma
- Tecnologico de Monterrey, School of Engineering and Sciences, Centre of Bioengineering, Campus Queretaro, Av. Epigmenio González, No. 500, Fracc. San Pablo, 76130, Querétaro, Mexico
| | - Aurea K Ramírez-Jiménez
- Tecnologico de Monterrey, School of Engineering and Sciences, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico.
| |
Collapse
|
32
|
Lewkowicz E, Gursky O. Dynamic protein structures in normal function and pathologic misfolding in systemic amyloidosis. Biophys Chem 2022; 280:106699. [PMID: 34773861 PMCID: PMC9416430 DOI: 10.1016/j.bpc.2021.106699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 02/08/2023]
Abstract
Dynamic and disordered regions in native proteins are often critical for their function, particularly in ligand binding and signaling. In certain proteins, however, such regions can contribute to misfolding and pathologic deposition as amyloid fibrils in vivo. For example, dynamic and disordered regions can promote amyloid formation by destabilizing the native structure, by directly triggering the aggregation, by promoting protein condensation, or by acting as sites of early proteolytic cleavage that favor a release of aggregation-prone fragments or facilitate fibril maturation. At the same time, enhanced dynamics in the native protein state accelerates proteolytic degradation that counteracts amyloid accumulation in vivo. Therefore, the functional need for dynamic protein regions must be balanced against their inherently labile nature. How exactly this balance is achieved and how is it shifted upon amyloidogenic mutations or post-translational modifications? To illustrate possible scenarios, here we review the beneficial and pathologic roles of dynamic and disordered regions in the native states of three families of human plasma proteins that form amyloid precursors in systemic amyloidoses: immunoglobulin light chain, apolipoproteins, and serum amyloid A. Analysis of structure, stability and local dynamics of these diverse proteins and their amyloidogenic variants exemplifies how disordered/dynamic regions can provide a functional advantage as well as an Achilles heel in pathologic amyloid formation.
Collapse
|
33
|
Jayaraman S, Pérez A, Miñambres I, Sánchez-Quesada JL, Gursky O. Heparin binding triggers human VLDL remodeling by circulating lipoprotein lipase: Relevance to VLDL functionality in health and disease. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159064. [PMID: 34610468 PMCID: PMC8595799 DOI: 10.1016/j.bbalip.2021.159064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/13/2021] [Accepted: 09/29/2021] [Indexed: 02/06/2023]
Abstract
Hydrolysis of VLDL triacylglycerol (TG) by lipoprotein lipase (LpL) is a major step in energy metabolism and VLDL-to-LDL maturation. Most functional LpL is anchored to the vascular endothelium, yet a small amount circulates on TG-rich lipoproteins. As circulating LpL has low catalytic activity, its role in VLDL remodeling is unclear. We use pre-heparin plasma and heparin-sepharose affinity chromatography to isolate VLDL fractions from normolipidemic, hypertriglyceridemic, or type-2 diabetic subjects. LpL is detected only in the heparin-bound fraction. Transient binding to heparin activates this VLDL-associated LpL, which hydrolyses TG, leading to gradual VLDL remodeling into IDL/LDL and HDL-size particles. The products and the timeframe of this remodeling closely resemble VLDL-to-LDL maturation in vivo. Importantly, the VLDL fraction that does not bind heparin is not remodeled. This relatively inert LpL-free VLDL is rich in TG and apoC-III, poor in apoE and apoC-II, shows impaired functionality as a substrate for the exogenous LpL or CETP, and likely has prolonged residence time in blood, which is expected to promote atherogenesis. This non-bound VLDL fraction increases in hypertriglyceridemia and in type-2 diabetes but decreases upon diabetes treatment that restores the glycemic control. In stark contrast, heparin binding by LDL increases in type-2 diabetes triggering pro-atherogenic LDL modifications. Therefore, the effects of heparin binding are associated negatively with atherogenesis for VLDL but positively for LDL. Collectively, the results reveal that binding to glycosaminoglycans initiates VLDL remodeling by circulating LpL, and suggest heparin binding as a marker of VLDL functionality and a readout for treatment of metabolic disorders.
Collapse
Affiliation(s)
- Shobini Jayaraman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118, USA.,Corresponding author.
| | - Antonio Pérez
- Endocrinology Department of the Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Spain
| | - Inka Miñambres
- Endocrinology Department of the Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jose Luis Sánchez-Quesada
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Spain.,Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau, CIBERDEM, Barcelona, Spain
| | - Olga Gursky
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
34
|
Hamid SB, Hamid AFA. Roles of Nutraceuticals and Functional Food in Prevention of Cardiovascular Disease. RESEARCH ANTHOLOGY ON RECENT ADVANCEMENTS IN ETHNOPHARMACOLOGY AND NUTRACEUTICALS 2022:810-839. [DOI: 10.4018/978-1-6684-3546-5.ch041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The chapter provides an overview of cardiovascular disease, a major cause of mortality worldwide. It relates economic and social impacts to the disease, especially in developing countries. One of the approaches to addressing this challenge is increasing awareness within society, through implementation of education programs. It is important for society to understand the types and roles of the risk factors leading to cardiovascular disease. Emphasis is on the role of functional food and nutraceuticals as dietary sources that could prevent development of cardiovascular disease. The chapter highlights roles of nutraceuticals and functional food sources from medical plants, seeds, berries, and tropical fruits in lowering risk factors. Key findings from trials conducted in Asia, China, Europe, and America provide supporting evidence for the importance of functional food to health, and its potential for modifying the level of risk factors related to cardiovascular diseases.
Collapse
|
35
|
Bermudez-Lopez M, Perpiñan H, Amigo N, Castro E, Alonso N, Mauricio D, Fernandez E, Valdivielso JM. Advanced lipoprotein parameters could better explain atheromatosis in non-diabetic chronic kidney disease patients. Clin Kidney J 2021; 14:2591-2599. [PMID: 34950470 PMCID: PMC8690051 DOI: 10.1093/ckj/sfab113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Background Chronic kidney disease (CKD) patients have a high burden of atheromatous cardiovascular disease (ASCVD) not fully explained by traditional lipid parameters. Lipoprotein composition and subclass particle number information could improve ASCVD risk assessment. The objective of this study is to investigate the association of advanced lipoprotein parameters with the risk of atheromatosis in a subpopulation of the NEFRONA study. Methods This was a cross-sectional study in 395 non-diabetic individuals (209 CKD and 186 non-diabetic and non-CKD) without statin therapy. Vascular ultrasound examination assessing 10 territories was combined with advanced lipoprotein testing performed by nuclear magnetic resonance spectroscopy. Logistic regression was used to estimate adjusted odds ratios (ORs) per 1 standard deviation increment. Results Atheromatosis was more prevalent in CKD patients (33.9% versus 64.6%). After adjusting for age, gender, smoking habit and CKD stage, the amount of triglycerides (TGs) within low-density lipoprotein (LDL) lipoproteins was independently and positively associated with atheromatosis [OR 1.33; 95% confidence interval (CI) 1.03–1.74; P = 0.03]. Similarly, total and medium LDL particles (LDL-Ps) showed a positive association (OR 1.29; 95% CI 1.00–1.68; P = 0.05 and OR 1.34; 95% CI 1.04–1.75; P = 0.03, respectively). TG-loaded medium LDL-Ps were higher in CKD patients compared with controls and showed an adjusted OR of 1.40 (95% CI 1.09–1.82; P = 0.01) in non-diabetic patients (CKD and non-CKD individuals). In contrast, non-diabetic CKD patients showed a similar coefficient but the significance was lost (OR 1.2; 95% CI 0.8–1.7; P = 0.359). Conclusions Non-diabetic CKD patients showed a higher amount of TG-loaded medium LDL-Ps compared with controls. These particles were independently associated with atheromatosis in non-diabetic patients.
Collapse
Affiliation(s)
- Marcelino Bermudez-Lopez
- Vascular and Renal Translational Research Group, Spanish Research Network for Renal Diseases (REDINREN del ISCIII), IRBLleida, Lleida, Spain
| | - Hector Perpiñan
- Conselleria de Sanitat Universal i Salut Pública, Generalitat Valenciana, Valencia, Spain
| | | | - Eva Castro
- Vascular and Renal Translational Research Group, Spanish Research Network for Renal Diseases (REDINREN del ISCIII), IRBLleida, Lleida, Spain
| | - Nuria Alonso
- Endocrinology and Nutrition Department, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Didac Mauricio
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Elvira Fernandez
- Vascular and Renal Translational Research Group, Spanish Research Network for Renal Diseases (REDINREN del ISCIII), IRBLleida, Lleida, Spain
| | - Jose M Valdivielso
- Vascular and Renal Translational Research Group, Spanish Research Network for Renal Diseases (REDINREN del ISCIII), IRBLleida, Lleida, Spain
| |
Collapse
|
36
|
Changes in gut-microbiota-related metabolites and long-term improvements in lipoprotein subspecies in overweight and obese adults: the POUNDS lost trial. Int J Obes (Lond) 2021; 45:2600-2607. [PMID: 34426648 PMCID: PMC8608703 DOI: 10.1038/s41366-021-00939-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/23/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND/OBJECTIVES Alterations in gut microbiota have been linked to obesity and impaired lipid metabolism. Lipoproteins are heterogeneous, and lipoprotein subspecies containing apolipoprotein C-III (apoCIII) have adverse associations with obesity and related cardiometabolic abnormalities. We investigated associations of weight-loss diet-induced decreases in atherogenic gut-microbial metabolites, trimethylamine N-oxide (TMAO) and L-carnitine, with improvements in atherogenic lipoproteins containing apoCIII among patients with obesity. SUBJECTS/METHODS This study included overweight and obese adults who participated in a 2-year weight-loss dietary intervention, the POUNDS Lost trial. Blood levels of TMAO and L-carnitine were measured at baseline and 6 months after the intervention; 6-month changes in the metabolites were calculated. We evaluated 2-year changes in lipid profiles (n = 395) and cholesterol [Chol] in lipoprotein (very-low-density lipoprotein (VLDL), low-density lipoprotein (LDL), and high-density lipoprotein (HDL)) subfractions defined by the presence or absence of apoCIII (n = 277). RESULTS The initial (6-month) decrease in L-carnitine was significantly associated with long-term (2-year) reductions in non-HDL-Chol and LDL-Chol (p < 0.05). Also, the decrease in L-carnitine was significantly related to decreases in Chol in LDL with apoCIII (p = 0.034) and Chol in [LDL + VLDL] with apoCIII (p = 0.018). We found significant interactions between dietary fat and TMAO on changes in LDL-Chol (Pinteraction = 0.013) and Chol in [LDL + VLDL] with apoCIII (Pinteraction = 0.0048); a greater increase in TMAO was related to lesser improvements in the lipoprotein outcomes if participants consumed a high-fat compared to a low-fat diet. CONCLUSIONS Changes in TMAO and L-carnitine induced by weight-loss diets were associated with long-term improvements in atherogenic lipoproteins containing apoCIII, implicating that these metabolic changes might be predictive of an individual's response to the dietary treatment to modify the unfavorable lipid profiles in obese patients. Dietary fat intake might modify associations of TMAO changes with long-term improvements of atherogenic cholesterol metabolism in overweight and obese adults. CLINICALTRIALS. GOV IDENTIFIER NCT00072995.
Collapse
|
37
|
Soohoo M, Hashemi L, Hsiung JT, Moradi H, Budoff MJ, Kovesdy CP, Kalantar-Zadeh K, Streja E. Risk of Atherosclerotic Cardiovascular Disease and Nonatherosclerotic Cardiovascular Disease Hospitalizations for Triglycerides Across Chronic Kidney Disease Stages Among 2.9 Million US Veterans. J Am Heart Assoc 2021; 10:e022988. [PMID: 34729994 PMCID: PMC9075381 DOI: 10.1161/jaha.121.022988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background High triglycerides are associated with atherosclerotic cardiovascular disease (ASCVD) risks. Among patients with advanced chronic kidney disease (CKD), the association of elevated triglycerides with mortality is diminished and, thus, we investigated the relationship of triglycerides with ASCVD and non‐ASCVD hospitalizations across CKD stages. Methods and Results The cohort comprised 2 963 176 veterans who received care in 2004 to 2006 (baseline) and were followed up to 2014. Using Cox models, we evaluated baseline and time‐varying triglycerides with time to ASCVD or non‐ASCVD hospitalizations, stratified by baseline CKD stage, and adjusted for demographics and baseline or time‐updated clinical characteristics. The cohort mean±SD age was 63±14 years, with a baseline median (interquartile range) triglycerides level of 127 (87–189) mg/dL, and a quarter had prevalent CKD. There was a linear association between baseline triglycerides and ASCVD risk; however, the risk with high triglycerides ≥240 mg/dL attenuated with worsening CKD stages (reference: triglycerides 120 to <160 mg/dL). Baseline triglycerides were associated with a U‐shaped relationship for non‐ASCVD events in patients with CKD 3A to 3B. Patients with late‐stage CKD had lower to null relationships between baseline triglycerides and non‐ASCVD events. Time‐varying triglycerides associations with ASCVD were similar to baseline analyses. Yet, the time‐varying triglycerides relationship with non‐ASCVD events was inverse and linear, where elevated triglycerides were associated with lower risks. Conclusions Associations of higher triglycerides with ASCVD and non‐ASCVD events declined across advancing CKD stages, where a lower to null risk was observed in patients with advanced CKD. Studies are needed to examine the impact of advanced CKD on triglycerides metabolism and its association with outcomes in this high‐risk population.
Collapse
Affiliation(s)
- Melissa Soohoo
- Division of Nephrology and Hypertension Harold Simmons Center for Kidney Disease Research and Epidemiology University of California Irvine Medical Center Orange CA.,Nephrology Section Tibor Rubin Veterans Affairs Medical Center Long Beach CA
| | - Leila Hashemi
- Department of General Internal Medicine Greater Los Angeles Healthcare System Los Angeles CA.,David Geffen School of Medicine University of California Los Angeles Los Angeles CA
| | - Jui-Ting Hsiung
- Division of Nephrology and Hypertension Harold Simmons Center for Kidney Disease Research and Epidemiology University of California Irvine Medical Center Orange CA.,Nephrology Section Tibor Rubin Veterans Affairs Medical Center Long Beach CA
| | - Hamid Moradi
- Division of Nephrology and Hypertension Harold Simmons Center for Kidney Disease Research and Epidemiology University of California Irvine Medical Center Orange CA.,Nephrology Section Tibor Rubin Veterans Affairs Medical Center Long Beach CA
| | - Matthew J Budoff
- Division of Cardiology The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center Torrance CA
| | - Csaba P Kovesdy
- Nephrology Section Memphis Veterans Affairs Medical Center Memphis TN.,Division of Nephrology University of Tennessee Health Science Center Memphis TN
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology and Hypertension Harold Simmons Center for Kidney Disease Research and Epidemiology University of California Irvine Medical Center Orange CA.,Nephrology Section Tibor Rubin Veterans Affairs Medical Center Long Beach CA
| | - Elani Streja
- Division of Nephrology and Hypertension Harold Simmons Center for Kidney Disease Research and Epidemiology University of California Irvine Medical Center Orange CA.,Nephrology Section Tibor Rubin Veterans Affairs Medical Center Long Beach CA
| |
Collapse
|
38
|
Abstract
Triglycerides are critical lipids as they provide an energy source that is both compact and efficient. Due to its hydrophobic nature triglyceride molecules can pack together densely and so be stored in adipose tissue. To be transported in the aqueous medium of plasma, triglycerides have to be incorporated into lipoprotein particles along with other components such as cholesterol, phospholipid and associated structural and regulatory apolipoproteins. Here we discuss the physiology of normal triglyceride metabolism, and how impaired metabolism induces hypertriglyceridemia and its pathogenic consequences including atherosclerosis. We also discuss established and novel therapies to reduce triglyceride-rich lipoproteins.
Collapse
|
39
|
Nour OA, Ghoniem HA, Nader MA, Suddek GM. Impact of protocatechuic acid on high fat diet-induced metabolic syndrome sequelae in rats. Eur J Pharmacol 2021; 907:174257. [PMID: 34129881 DOI: 10.1016/j.ejphar.2021.174257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 01/03/2023]
Abstract
The study aimed to assess the possible protective impact of protocatechuic acid (PCA) on high fat diet (HFD)-induced metabolic syndrome (Mets) sequelae in rats. Forty-two male Sprague-Dawley (SD) rats were randomly grouped as follows: CTR group; PCA group; HFD group; HFD-PCA group and HFD-MET group. Rats were fed on standard diet or HFD for 14 weeks. HFD-fed rats exhibited significant decreases in food intake and adiponectin (ADP) level; yet, body weight and anthropometrical parameters were significantly increased. Moreover, insulin sensitivity was impaired as indicated by significant elevation in glucose AUC during oral glucose tolerance test (OGTT), fasting serum glucose, fasting serum insulin and homeostasis model assessment of insulin resistance (HOMA-IR) index. Furthermore, chronic HFD feeding elicited significant increases in serum lipid profile and free fatty acids (FFAs) with concomitant hepatic steatosis. Additionally, serum C-reactive protein (CRP), interleukin 1b (Il-1b) and monocyte chemoattractant protein 1(MCP-1) levels were increased. Also, HFD-fed rats exhibited an increase in MDA level, while superoxide dismutase (SOD) and glutathione (GSH) activities were decreased. Moreover, the insulin-signaling pathway was markedly impaired in soleus muscles as indicated by a decrease in insulin-induced AKT phosphorylation. Histopathologically, adipose tissues showed significant increase in adipocyte size. Also, flow cytometry analysis of adipose tissue confirmed a significant increase in the percentage of number of CD68+ cells. PCA administration succeeded to attenuate HFD-induced obesity, insulin resistance, oxidative stress and inflammation. In conclusion, PCA administration could protect against HFD-induced Mets, possibly via its hypoglycemic, insulin-sensitizing, anti-oxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Omnia A Nour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Hamdy A Ghoniem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
40
|
Keirns BH, Sciarrillo CM, Koemel NA, Emerson SR. Fasting, non-fasting and postprandial triglycerides for screening cardiometabolic risk. J Nutr Sci 2021; 10:e75. [PMID: 34589207 PMCID: PMC8453457 DOI: 10.1017/jns.2021.73] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/23/2022] Open
Abstract
Fasting triacylglycerols have long been associated with cardiovascular disease (CVD) and other cardiometabolic conditions. Evidence suggests that non-fasting triglycerides (i.e. measured within 8 h of eating) better predict CVD than fasting triglycerides, which has led several organisations to recommend non-fasting lipid panels as the new clinical standard. However, unstandardised assessment protocols associated with non-fasting triglyceride measurement may lead to misclassification, with at-risk individuals being overlooked. A third type of triglyceride assessment, postprandial testing, is more controlled, yet historically has been difficult to implement due to the time and effort required to execute it. Here, we review differences in assessment, the underlying physiology and the pathophysiological relevance of elevated fasting, non-fasting and postprandial triglycerides. We also present data suggesting that there may be a distinct advantage of postprandial triglycerides, even over non-fasting triglycerides, for early detection of CVD risk and offer suggestions to make postprandial protocols more clinically feasible.
Collapse
Affiliation(s)
- Bryant H. Keirns
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK74075, USA
| | | | - Nicholas A. Koemel
- Boden Collaboration for Obesity, Nutrition, Exercise and Eating Disorders, The University of Sydney, Sydney, NSW2006, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW2006, Australia
| | - Sam R. Emerson
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK74075, USA
| |
Collapse
|
41
|
Shimizu-Albergine M, Basu D, Kanter JE, Kramer F, Kothari V, Barnhart S, Thornock C, Mullick AE, Clouet-Foraison N, Vaisar T, Heinecke JW, Hegele RA, Goldberg IJ, Bornfeldt KE. CREBH normalizes dyslipidemia and halts atherosclerosis in diabetes by decreasing circulating remnant lipoproteins. J Clin Invest 2021; 131:e153285. [PMID: 34491909 DOI: 10.1172/jci153285] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Loss-of-function mutations in the transcription factor CREB3L3 (CREBH) associate with severe hypertriglyceridemia in humans. CREBH is believed to lower plasma triglycerides by augmenting the action of lipoprotein lipase (LPL). However, by using a mouse model of type 1 diabetes mellitus (T1DM), we found that greater liver expression of active CREBH normalized both elevated plasma triglycerides and cholesterol. Residual triglyceride-rich lipoprotein (TRL) remnants were enriched in apolipoprotein E (APOE) and impoverished in APOC3, an apolipoprotein composition indicative of increased hepatic clearance. The underlying mechanism was independent of LPL as CREBH reduced both triglycerides and cholesterol in LPL-deficient mice. Instead, APOE was critical for CREBH's ability to lower circulating remnant lipoproteins because it failed to reduce TRL cholesterol in Apoe-/- mice. Importantly, humans with CREB3L3 loss-of-function mutations exhibited increased levels of remnant lipoproteins that were deprived of APOE. Recent evidence suggests that impaired clearance of TRL remnants promotes cardiovascular disease in patients with T1DM. Consistently, we found that hepatic expression of CREBH prevented the progression of diabetes-accelerated atherosclerosis. Our results support the proposal that CREBH acts through an APOE-dependent pathway to increase hepatic clearance of remnant lipoproteins. They also implicate elevated levels of remnants in the pathogenesis of atherosclerosis in T1DM.
Collapse
Affiliation(s)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, NYU Langone Medical Center, New York, United States of America
| | - Jenny E Kanter
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Farah Kramer
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Vishal Kothari
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Shelley Barnhart
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Carissa Thornock
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Adam E Mullick
- Cardiovascular Disease Research, Ionis Pharmaceuticals, Inc., Carlsbad, United States of America
| | | | - Tomas Vaisar
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Jay W Heinecke
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Robert A Hegele
- Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, London, Canada
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, NYU Langone Medical Center, New York, United States of America
| | - Karin E Bornfeldt
- Department of Medicine, University of Washington, Seattle, United States of America
| |
Collapse
|
42
|
Dong Y, Wang H, Shan D, Zhang L, Yu Z. [Correlation between Pretreatment Serum Apolipoprotein Level and Prognosis of Small Cell Lung Cancer Patients]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 23:845-851. [PMID: 33070513 PMCID: PMC7583878 DOI: 10.3779/j.issn.1009-3419.2020.104.21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related death, and small cell lung cancer (SCLC) has a poor prognosis in all types of lung cancer. This study evaluated the relationship between pretreatment serum apolipoprotein levels and prognosis in patients with SCLC, seeks a new index can guide diagnosis and treatment of SCLC. METHODS This study retrospectively analyzed the clinical data of 122 patients with SCLC. The clinical results of patients with serum apolipoprotein levels within 2 weeks before treatment were collected, including apolipoprotein AI (ApoA-I), apolipoprotein B (ApoB), and the ratio of apolipoprotein B to apolipoprotein AI (ApoB/ApoA-I). Patients' progression-free survival (PFS) and overall survival (OS) are the main outcome indicators. The best critical to determine the index's value by X-tile tool. For survival analysis, Kaplan-Meier method was used for analysis, and Cox regression analysis method was used for single factor analysis and multifactor analysis. RESULTS Compared with patients with low ApoA-I levels, patients with high ApoA-I levels (ApoA-I>1.12 g/L) had better OS (21.5 mon vs 12.3 mon, P=0.007) and PFS (7.3 mon vs 5.5 mon, P=0.017). In contrast, patients with higher ApoB/ApoA-I levels had worse median OS than patients with lower ApoB/ApoA-I levels (13.4 mon vs 20.7 mon, P=0.012). Multivariate Cox regression analysis showed that ApoA-I was an independent prognostic factor affecting PFS in SCLC patients (HR=0.67, 95%CI: 0.45-0.99, P=0.043). ApoB/ApoA-I is an independent risk factor for OS in patients with SCLC (HR=1.98, 95%CI: 1.21-3.23, P=0.007). CONCLUSIONS Serum ApoA-I level and ApoB/ApoA-I level before treatment can be important prognostic factors for SCLC, which is helpful to judge the prognosis of patients.
Collapse
Affiliation(s)
- Ya Dong
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Haocheng Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Dongfeng Shan
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Linwei Zhang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhuang Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
43
|
C Thambiah S, Lai LC. Diabetic dyslipidaemia. Pract Lab Med 2021; 26:e00248. [PMID: 34368411 PMCID: PMC8326412 DOI: 10.1016/j.plabm.2021.e00248] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 05/16/2021] [Accepted: 07/14/2021] [Indexed: 12/22/2022] Open
Abstract
Diabetes mellitus (DM) is an escalating pandemic and an established cardiovascular risk factor. An important aspect of the interaction between DM and atherosclerotic cardiovascular disease (ASCVD) is diabetic dyslipidaemia, an atherogenic dyslipidaemia encompassing quantitative [hypertriglyceridaemia (hyperTG) and decreased high density lipoprotein cholesterol (HDL)] and qualitative [increased small dense low density lipoprotein cholesterol (sdLDL) particles, large very low density lipoprotein cholesterol (VLDL) subfraction (VLDL1) and dysfunctional HDL] modifications in lipoproteins. Much of the pathophysiology linking DM and dyslipidaemia has been elucidated. This paper aims to review the pathophysiology and management of diabetic dyslipidaemia with respect to ASCVD. Briefly, the influence of diabetic kidney disease on lipid profile and lipid changes causing type 2 diabetes mellitus are highlighted. Biomarkers of diabetic dyslipidaemia, including novel markers and clinical trials that have demonstrated that non-lipid and lipid lowering therapies can lower cardiovascular risk in diabetics are discussed. The stands of various international guidelines on lipid management in DM are emphasised. It is important to understand the underlying mechanisms of diabetic dyslipidaemia in order to develop new therapeutic strategies against dyslipidaemia and diabetes. The various international guidelines on lipid management can be used to tailor a holistic approach specific to each patient with diabetic dyslipidaemia.
Collapse
Affiliation(s)
- Subashini C Thambiah
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | | |
Collapse
|
44
|
Riccardi G, Giosuè A, Calabrese I, Vaccaro O. Dietary recommendations for prevention of atherosclerosis. Cardiovasc Res 2021; 118:1188-1204. [PMID: 34229346 DOI: 10.1093/cvr/cvab173] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/07/2021] [Accepted: 05/18/2021] [Indexed: 12/17/2022] Open
Abstract
This review aims at summarizing updated evidence on cardiovascular disease (CVD) risk associated with consumption of specific food items to substantiate dietary strategies for atherosclerosis prevention. A systematic search on PubMed was performed to identify meta-analyses of cohort studies and RCTs with CVD outcomes. The evidence is highly concordant in showing that, for the healthy adult population, low consumption of salt and foods of animal origin, and increased intake of plant-based foods-whole grains, fruits, vegetables, legumes, and nuts-are linked with reduced atherosclerosis risk. The same applies for the replacement of butter and other animal/tropical fats with olive oil and other unsaturated-fat-rich oil. Although the literature reviewed overall endorses scientific society dietary recommendations, some relevant novelties emerge. With regard to meat, new evidence differentiates processed and red meat-both associated with increased CVD risk-from poultry, showing a neutral relationship with CVD for moderate intakes. Moreover, the preferential use of low-fat dairies in the healthy population is not supported by recent data, since both full-fat and low-fat dairies, in moderate amounts and in the context of a balanced diet, are not associated with increased CVD risk; furthermore, small quantities of cheese and regular yogurt consumption are even linked with a protective effect. Among other animal protein sources, moderate fish consumption is also supported by the latest evidence, although there might be sustainability concerns. New data endorse the replacement of most high glycemic index (GI) foods with both whole grain and low GI cereal foods. As for beverages, low consumption not only of alcohol, but also of coffee and tea is associated with a reduced atherosclerosis risk while soft drinks show a direct relationship with CVD risk. This review provides evidence-based support for promoting appropriate food choices for atherosclerosis prevention in the general population.
Collapse
Affiliation(s)
- Gabriele Riccardi
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples, Via Sergio Pansini, 5 - 80131, Naples, Italy
| | - Annalisa Giosuè
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples, Via Sergio Pansini, 5 - 80131, Naples, Italy
| | - Ilaria Calabrese
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples, Via Sergio Pansini, 5 - 80131, Naples, Italy
| | - Olga Vaccaro
- Department of Pharmacy, "Federico II" University of Naples, Via Domenico Montesano, 49 - 80131, Naples, Italy
| |
Collapse
|
45
|
Khalil YA, Rabès JP, Boileau C, Varret M. APOE gene variants in primary dyslipidemia. Atherosclerosis 2021; 328:11-22. [PMID: 34058468 DOI: 10.1016/j.atherosclerosis.2021.05.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/30/2021] [Accepted: 05/12/2021] [Indexed: 01/10/2023]
Abstract
Apolipoprotein E (apoE) is a major apolipoprotein involved in lipoprotein metabolism. It is a polymorphic protein and different isoforms are associated with variations in lipid and lipoprotein levels and thus cardiovascular risk. The isoform apoE4 is associated with an increase in LDL-cholesterol levels and thus a higher cardiovascular risk compared to apoE3. Whereas, apoE2 is associated with a mild decrease in LDL-cholesterol levels. In the presence of other risk factors, apoE2 homozygotes could develop type III hyperlipoproteinemia (familial dysbetalipoproteinemia or FD), an atherogenic disorder characterized by an accumulation of remnants of triglyceride-rich lipoproteins. Several rare APOE gene variants were reported in different types of dyslipidemias including FD, familial combined hyperlipidemia (FCH), lipoprotein glomerulopathy and bona fide autosomal dominant hypercholesterolemia (ADH). ADH is characterized by elevated LDL-cholesterol levels leading to coronary heart disease, and due to molecular alterations in three main genes: LDLR, APOB and PCSK9. The identification of the APOE-p.Leu167del variant as the causative molecular element in two different ADH families, paved the way to considering APOE as a candidate gene for ADH. Due to non mendelian interacting factors, common genetic and environmental factors and perhaps epigenetics, clinical presentation of lipid disorders associated with APOE variants often strongly overlap. More studies are needed to determine the spectrum of APOE implication in each of the diseases, notably ADH, in order to improve clinical and genetic diagnosis, prognosis and patient management. The purpose of this review is to comment on these APOE variants and on the molecular and clinical overlaps between dyslipidemias.
Collapse
Affiliation(s)
- Yara Abou Khalil
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalo-Universitaire Xavier Bichat, Paris, France; Université de Paris, Paris, France; Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé (PTS), Saint-Joseph University, Beirut, Lebanon
| | - Jean-Pierre Rabès
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalo-Universitaire Xavier Bichat, Paris, France; Laboratory of Biochemistry and Molecular Genetics, Centre Hospitalo-Universitaire Ambroise Paré, HUPIFO, AP-HP. Paris-Saclay, Boulogne-Billancourt, France; UFR Simone Veil-Santé, UVSQ, Montigny-Le-Bretonneux, France
| | - Catherine Boileau
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalo-Universitaire Xavier Bichat, Paris, France; Université de Paris, Paris, France; Genetics Department, AP-HP, CHU Xavier Bichat, Paris, France
| | - Mathilde Varret
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalo-Universitaire Xavier Bichat, Paris, France; Université de Paris, Paris, France.
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Triglycerides (TGs) are measured as part of routine lipid profiles but their relationship to cardiovascular disease (CVD) risk has been controversial and overshadowed by high-density lipoprotein cholesterol (HDL-C). RECENT FINDINGS Epidemiological studies show a clear relationship of TG-containing lipoproteins including remnant particles with CVD risk with the effect being most clearly demonstrated through the excess risk captured by non-HDL-C compared with low-density lipoprotein-cholesterol (LDL-C). Mendelian randomisation studies show a consistent relationship of gene variants linked to TG metabolism with rates of CVD. Furthermore, meta-analyses of intervention trials with statins and other nonstatin drugs also suggest that reducing TGs is associated with benefits on rates of CVD events. Historical subgroup data from fibrate trials suggest benefits in patients with high TG:HDL ratios but seem to add little to optimized statin therapy. Recent trials with omega-3 fatty acids (specifically eicosapentaenoic acid) have suggested that high-dose formulations in contrast to low dose formulations have benefits on CVD outcomes. SUMMARY Further studies with newer agents are required to determine the place of TG-lowering drugs in therapeutic pathways. Trials with agents such as pemafibrate and vupanorsen may finally answer these questions.
Collapse
Affiliation(s)
| | - Anthony S Wierzbicki
- Metabolic Medicine/Chemical Pathology, Guy's & St Thomas Hospitals, London SE1 7EH, UK
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW The functions, genetic variations and impact of apolipoprotein E on lipoprotein metabolism in general are placed in the context of clinical practice dealing with moderate dyslipidaemia as well as dysbetalipoproteinemia, a highly atherogenic disorder and lipoprotein glomerulopathy. RECENT FINDINGS Additional variants of apolipoprotein E and participation of apolipoprotein E in inflammation are of interest. The mostly favourable effects of apolipoprotein E2 as well as the atherogenic nature of apolipoproteinE4, which has an association with cognitive impairment, are confirmed. The contribution of remnant lipoproteins of triglyceride-rich lipoproteins, of which dysbetalipoproteinemia represents an extreme, is explored in atherosclerosis. Mimetic peptides may present new therapeutic approaches. Apolipoprotein E is an important determinant of the lipid profile and cardiovascular health in the population at large and can precipitate dysbetalipoproteinemia and glomerulopathy. Awareness of apolipoprotein E polymorphisms should improve medical care.
Collapse
|
48
|
Valladolid-Acebes I, Åvall K, Recio-López P, Moruzzi N, Bryzgalova G, Björnholm M, Krook A, Alonso EF, Ericsson M, Landfors F, Nilsson SK, Berggren PO, Juntti-Berggren L. Lowering apolipoprotein CIII protects against high-fat diet-induced metabolic derangements. SCIENCE ADVANCES 2021; 7:7/11/eabc2931. [PMID: 33712458 PMCID: PMC7954448 DOI: 10.1126/sciadv.abc2931] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 01/27/2021] [Indexed: 02/05/2023]
Abstract
Increased levels of apolipoprotein CIII (apoCIII), a key regulator of lipid metabolism, result in obesity-related metabolic derangements. We investigated mechanistically whether lowering or preventing high-fat diet (HFD)–induced increase in apoCIII protects against the detrimental metabolic consequences. Mice, first fed HFD for 10 weeks and thereafter also given an antisense (ASO) to lower apoCIII, already showed reduced levels of apoCIII and metabolic improvements after 4 weeks, despite maintained obesity. Prolonged ASO treatment reversed the metabolic phenotype due to increased lipase activity and receptor-mediated hepatic uptake of lipids. Fatty acids were transferred to the ketogenic pathway, and ketones were used in brown adipose tissue (BAT). This resulted in no fat accumulation and preserved morphology and function of liver and BAT. If ASO treatment started simultaneously with the HFD, mice remained lean and metabolically healthy. Thus, lowering apoCIII protects against and reverses the HFD-induced metabolic phenotype by promoting physiological insulin sensitivity.
Collapse
Affiliation(s)
- Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Karin Åvall
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Patricia Recio-López
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Noah Moruzzi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Galyna Bryzgalova
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Marie Björnholm
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, C3, Integrative Physiology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Elena Fauste Alonso
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden.,Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Madelene Ericsson
- Department of Medical Biosciences, Unit of Physiological Chemistry 6M, Umeå University, SE-901 85 Umeå, Sweden
| | - Fredrik Landfors
- Department of Medical Biosciences, Unit of Physiological Chemistry 6M, Umeå University, SE-901 85 Umeå, Sweden
| | - Stefan K Nilsson
- Department of Medical Biosciences, Unit of Physiological Chemistry 6M, Umeå University, SE-901 85 Umeå, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden.,Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea.,Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553, Singapore.,Center for Diabetes and Metabolism Research, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China
| | - Lisa Juntti-Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden.
| |
Collapse
|
49
|
Jimenes DR, Teixeira Junior NR, Pereira AV, Berti JA, Barbosa CP, Sant'Ana DDMG. Human apoCIII transgenic mice with epicardial adipose tissue inflammation and PRESERVATION of the cardiac plexus. Exp Gerontol 2021; 148:111261. [PMID: 33647361 DOI: 10.1016/j.exger.2021.111261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 12/16/2020] [Accepted: 01/24/2021] [Indexed: 10/22/2022]
Abstract
Hypertriglyceridemia is a result of the increase in the serum levels of lipoproteins, which are responsible for the transport of triglycerides and can be caused by genetic and/or metabolic factors. Animal models which either express or lack genes related to changes in the lipoproteins profile are useful to understand lipid metabolism. Apolipoprotein CIII (apoCIII) is an important modulator of hepatic production and peripheral removal of triglycerides. Mice that overexpress the apoCIII gene become hypertriglyceridemic, showing high concentrations of free fatty acids in the blood. Since hypertriglyceridemia is related to atherosclerosis, and the latter refers to cardiac alterations, this study aimed at evaluating the morphological, morphometric and quantitative profiles of the cardiac plexus, as well as the morphometric and histopathological aspects of the epicardial adipose tissue in human apoCIII transgenic mice. Therefore, 8-12-month-old male C57BL/6 mice that overexpressed human apoCIII (CIII) and their respective controls were used. Our results showed that overexpression of human apoCIII did not modify morphological or quantitative parameters of cardiac plexus neurons; however, age increased both, the area and the number of such cells. Furthermore, there was a direct correlation of this dyslipidemia to the thickening of periganglionar type 1 collagens. On the other hand, this overexpression caused epicardial adipose tissue inflammation and an increase in the area of the adipocytes, thus, favoring the recruitment of inflammatory cells in this tissue. In conclusion, this overexpression is harmful since it is related to an increase in cardiac adiposity, as well as to a predisposition to an inflammatory environment in the epicardial fat and to the incidence of cardiovascular diseases.
Collapse
Affiliation(s)
- Diogo Rodrigues Jimenes
- Program of Graduate Studies in Bioscience and Physiopathology - State University of Maringá (PBF-UEM), Brazil.
| | | | | | | | | | - Débora de Mello Gonçales Sant'Ana
- Program of Graduate Studies in Bioscience and Physiopathology - State University of Maringá (PBF-UEM), Brazil; Department of Physiological Sciences (DFS-UEM), Brazil
| |
Collapse
|
50
|
Valladolid-Acebes I, Berggren PO, Juntti-Berggren L. Apolipoprotein CIII Is an Important Piece in the Type-1 Diabetes Jigsaw Puzzle. Int J Mol Sci 2021; 22:ijms22020932. [PMID: 33477763 PMCID: PMC7832341 DOI: 10.3390/ijms22020932] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 12/05/2022] Open
Abstract
It is well known that type-2 diabetes mellitus (T2D) is increasing worldwide, but also the autoimmune form, type-1 diabetes (T1D), is affecting more people. The latest estimation from the International Diabetes Federation (IDF) is that 1.1 million children and adolescents below 20 years of age have T1D. At present, we have no primary, secondary or tertiary prevention or treatment available, although many efforts testing different strategies have been made. This review is based on the findings that apolipoprotein CIII (apoCIII) is increased in T1D and that in vitro studies revealed that healthy β-cells exposed to apoCIII became apoptotic, together with the observation that humans with higher levels of the apolipoprotein, due to mutations in the gene, are more susceptible to developing T1D. We have summarized what is known about apoCIII in relation to inflammation and autoimmunity in in vitro and in vivo studies of T1D. The aim is to highlight the need for exploring this field as we still are only seeing the top of the iceberg.
Collapse
|