1
|
Brunham LR. The role of high-density lipoproteins in sepsis. J Lipid Res 2025; 66:100728. [PMID: 39672330 PMCID: PMC11758940 DOI: 10.1016/j.jlr.2024.100728] [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: 07/23/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/15/2024] Open
Abstract
High density lipoproteins (HDLs) are best known for their role in atherosclerotic cardiovascular diseases. However, efforts to reduce cardiovascular risk by increasing the concentration of cholesterol in HDL have failed, raising the question of whether HDL may have other, more central aspects to its role in health and disease. In epidemiological studies, low levels of HDL cholesterol are strongly associated with risk of infectious diseases and death from sepsis and, during sepsis, a larger decline in HDL cholesterol predicts a worse outcome. Many preclinical studies have examined strategies to augment HDL genetically or pharmacologically and have shown that this leads to protection from sepsis in animal models. Data in humans are more limited, but suggest a clinically relevant role of HDL in mediating the response to pathogen-associated lipids and preventing excessive inflammation. Collectively, these data raise the intriguing possibility that a clinically important biological function of HDL is as a component of the innate immune system and suggest that pharmacological manipulation of HDL may be a strategy to reduce the organ dysfunction and death that results from uncontrolled inflammation during sepsis. This review article discusses recent advances in our understanding of the role of HDL in sepsis.
Collapse
Affiliation(s)
- Liam R Brunham
- Centre for Heart Lung Innovation, Department of Medicine, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
2
|
Costa GDO, Tavares Germano A, Bretanha LC, Micke GA, Siwe-Noundou X, Sandjo LP. GC-MS comparison of fatty acids profile of oils extracted from viscera of Tainha ( Mugil liza) and Tambaqui ( Colossoma macropomum). Nat Prod Res 2024; 38:2780-2785. [PMID: 37480346 DOI: 10.1080/14786419.2023.2236276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/19/2023] [Accepted: 07/07/2023] [Indexed: 07/24/2023]
Abstract
The commercial activity of the grey mullet (known as Tainha: TAI) and Tambaqui (TAM) generates tons of waste that can be turned into valuable resources. Therefore, this work aimed to chemically characterize and quantify the fatty acids profiles of the two fishes. GCMS quantification was performed by using calibration curves built from a standard that contains 19 FAME. The analysis revealed that visceral wastes from both fishes contain 16 fatty acids (FA) consisting of saturated (SFA), monounsaturated (MUFA) and polyunsaturated (PUFA). However, their compositions were different as FA side chains in TAI and TAM contain 12 to 20 and 13 to 22 carbon atoms, respectively. Also, the SFA amount in TAI was greater than in TAM. On the other hand, TAM is richer in MUFA and PUFA compared to TAI. Both have similar chemical compositions of ω-3 and ω-6 in PUFA and ω-5, ω-7, and ω-9 in MUFA.
Collapse
Affiliation(s)
| | - Amanda Tavares Germano
- Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Gustavo Amadeu Micke
- Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Xavier Siwe-Noundou
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - Louis Pergaud Sandjo
- Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
3
|
Aghasizadeh M, Ahmadi Hoseini A, Sahebi R, Kazemi T, Asadiyan‐Sohan P, Esmaily H, Samadi S, Avan A, Ferns GA, Khosravi S, Ghazizadeh H, Miri‐Moghaddam E, Ghayour‐Mobarhan M. Association of a genetic variant in angiopoietin-like 3 with serum HDL-C and risk of cardiovascular disease: A study of the MASHAD cohort over 6 years. Mol Genet Genomic Med 2024; 12:e2418. [PMID: 38634215 PMCID: PMC11024632 DOI: 10.1002/mgg3.2418] [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: 07/29/2023] [Revised: 01/17/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Loss-of-function (LOF) variants of the angiopoietin-like 3 (ANGPTL3) gene are reported to be associated with serum triglyceride (TG) and high-density lipoprotein cholesterol (HDL-C) concentrations and thereby affect the risk of cardiovascular disease (CVD). OBJECTIVE In the present study, we examined the association of rs10789117 in the ANGPTL 3 gene locus and the risk of CVD in the group of people who were part of the Mashhad-Stroke and Heart-Atherosclerotic-Disorders (MASHAD) cohort. METHODS One thousand and two healthy individuals enrolled in this study of whom 849 subjects were healthy and 153 subjects developed CVD outcomes after 6 years of follow-up. After a 12-h overnight fasting, 20 mL of blood samples were collected for the measurement of fasting blood glucose and lipid profile. DNA was extracted, and the Tetra-ARMS PCR (amplification refractory mutation system) was used for genotyping of rs10789117 in the ANGPTL3 gene. The genotype frequencies of the variant of rs10789117 in the ANGPTL3 gene were estimated using χ2 tests. Eventually, the statistical analysis was done by SPSS version 20. RESULTS Individuals with AC/CC genotypes (rs10789117) were found to have to greater risk of CVD events compared to AA genotype (OR = 1.43, 95%CI = 1.01-2.02, p = 0.041). There was a 1.3-fold increase in cardiovascular events in individuals carrying the C allele of rs10789117 variant compared to non-carriers (OR = 1.32, 95%CI = 1.06-1.72, p value = 0.038). There were significant differences between different genotypes for serum triglyceride levels within the control group, but this difference was not significant in the group with CVD. Moreover, there was a significant association between CC genotype and CVD risk in the individuals with a normal serum HDL-C. CONCLUSION We have found that a rs10789117 C>A in ANGPTL3 gene polymorphism was associated with incident CVD events, and this may be of value as a risk stratification biomarker in CVD in the Iranian population.
Collapse
Affiliation(s)
- Malihe Aghasizadeh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IranMashhad University of Medical SciencesMashhadIran
| | - Asieh Ahmadi Hoseini
- Department of Nutrition, Ghaem HospitalMashhad University of Medical SciencesMashhadIran
| | - Reza Sahebi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IranMashhad University of Medical SciencesMashhadIran
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Tooba Kazemi
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
- Razi Clinical Research Development Unit (RCRDU)Birjand University of Medical SciencesBirjandIran
| | - Parisa Asadiyan‐Sohan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IranMashhad University of Medical SciencesMashhadIran
| | - Habibollah Esmaily
- Social Determinants of Health Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Biostatistics, School of HealthMashhad University of Medical SciencesMashhadIran
| | - Sara Samadi
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Amir Avan
- Student Research Committee, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
| | - Gordon A. Ferns
- Division of Medical EducationBrighton & Sussex Medical SchoolBrightonUK
| | - Saeede Khosravi
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
| | - Hamideh Ghazizadeh
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Ebrahim Miri‐Moghaddam
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
| | - Majid Ghayour‐Mobarhan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IranMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
4
|
Luna-Castillo KP, López-Quintero A, Carrera-Quintanar L, Llamas-Covarrubias IM, Muñoz-Valle JF, Márquez-Sandoval F. The Influence of Lifestyle on High-Density Lipoprotein Concentration among Mexican Emerging Adults. Nutrients 2023; 15:4568. [PMID: 37960221 PMCID: PMC10649881 DOI: 10.3390/nu15214568] [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: 08/31/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Previous studies have highlighted the role of lifestyle on HDL-C concentrations in adults. To our knowledge, the health and nutritional status of emerging adults have been understudied. The present study aimed to explore the most important lifestyle factors, including micronutrient intake adequacy and the percentage of energy from food processing, according to HDL-C concentrations in emerging adults. In this context, a cross-sectional analysis was conducted on 261 Mexican emerging adults who were apparently healthy. Lifestyle factors were collected through a structured survey and the prevalence of micronutrient intake inadequacy was estimated using the estimated average requirement cut-point method. The percentage of energy from ultra-processed foods was assessed using the NOVA system. HDL-C was determined using the enzymatic colorimetric method. Statistical analyses were conducted in SPSS. The results revealed that lifestyle factors do not differ according to HDL-C status. The participants showed a poor nutritional diet that was energy-dense and micronutrient-inadequate. Nearly half of their energy came from processed and ultra-processed foods. Most participants did not meet the recommendations for key nutrients (ϖ3 fatty acids and phytosterols) that promote a healthy lipid status. In conclusion, regardless of their HDL-C levels, emerging adults exhibited lifestyle-related risk factors. The persistence of these findings over time could contribute to the development of metabolic disorders in the future. It is crucial to increase understanding and to develop effective nutritional interventions during this critical phase of life.
Collapse
Affiliation(s)
- Karla Paulina Luna-Castillo
- Doctorado en Ciencias de la Nutrición Traslacional (DCNT), Departamento de Clínicas de la Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico; (K.P.L.-C.); (L.C.-Q.); (I.M.L.-C.); (J.F.M.-V.)
- Instituto de Nutrigenética y Nutrigenómica Traslacional (INNUGET), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico
| | - Andres López-Quintero
- Doctorado en Ciencias de la Nutrición Traslacional (DCNT), Departamento de Clínicas de la Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico; (K.P.L.-C.); (L.C.-Q.); (I.M.L.-C.); (J.F.M.-V.)
- Instituto de Nutrigenética y Nutrigenómica Traslacional (INNUGET), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico
| | - Lucrecia Carrera-Quintanar
- Doctorado en Ciencias de la Nutrición Traslacional (DCNT), Departamento de Clínicas de la Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico; (K.P.L.-C.); (L.C.-Q.); (I.M.L.-C.); (J.F.M.-V.)
- Instituto de Investigación en Cáncer en la Infancia y Adolescencia (INICIA), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico
| | - Iris Monserrat Llamas-Covarrubias
- Doctorado en Ciencias de la Nutrición Traslacional (DCNT), Departamento de Clínicas de la Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico; (K.P.L.-C.); (L.C.-Q.); (I.M.L.-C.); (J.F.M.-V.)
- Instituto de Nutrigenética y Nutrigenómica Traslacional (INNUGET), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Doctorado en Ciencias de la Nutrición Traslacional (DCNT), Departamento de Clínicas de la Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico; (K.P.L.-C.); (L.C.-Q.); (I.M.L.-C.); (J.F.M.-V.)
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico
| | - Fabiola Márquez-Sandoval
- Doctorado en Ciencias de la Nutrición Traslacional (DCNT), Departamento de Clínicas de la Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico; (K.P.L.-C.); (L.C.-Q.); (I.M.L.-C.); (J.F.M.-V.)
| |
Collapse
|
5
|
Sacher S, Mukherjee A, Ray A. Deciphering structural aspects of reverse cholesterol transport: mapping the knowns and unknowns. Biol Rev Camb Philos Soc 2023; 98:1160-1183. [PMID: 36880422 DOI: 10.1111/brv.12948] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 02/03/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023]
Abstract
Atherosclerosis is a major contributor to the onset and progression of cardiovascular disease (CVD). Cholesterol-loaded foam cells play a pivotal role in forming atherosclerotic plaques. Induction of cholesterol efflux from these cells may be a promising approach in treating CVD. The reverse cholesterol transport (RCT) pathway delivers cholesteryl ester (CE) packaged in high-density lipoproteins (HDL) from non-hepatic cells to the liver, thereby minimising cholesterol load of peripheral cells. RCT takes place via a well-organised interplay amongst apolipoprotein A1 (ApoA1), lecithin cholesterol acyltransferase (LCAT), ATP binding cassette transporter A1 (ABCA1), scavenger receptor-B1 (SR-B1), and the amount of free cholesterol. Unfortunately, modulation of RCT for treating atherosclerosis has failed in clinical trials owing to our lack of understanding of the relationship between HDL function and RCT. The fate of non-hepatic CEs in HDL is dependent on their access to proteins involved in remodelling and can be regulated at the structural level. An inadequate understanding of this inhibits the design of rational strategies for therapeutic interventions. Herein we extensively review the structure-function relationships that are essential for RCT. We also focus on genetic mutations that disturb the structural stability of proteins involved in RCT, rendering them partially or completely non-functional. Further studies are necessary for understanding the structural aspects of RCT pathway completely, and this review highlights alternative theories and unanswered questions.
Collapse
Affiliation(s)
- Sukriti Sacher
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase III, New Delhi, 110019, India
| | - Abhishek Mukherjee
- Dhiti Life Sciences Pvt Ltd, B-107, Okhla Phase I, New Delhi, 110020, India
| | - Arjun Ray
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase III, New Delhi, 110019, India
| |
Collapse
|
6
|
Chang HC, Nfor ON, Ho CC, Chen PH, Liaw YP. Variations in high density cholesterol levels based on apolipoprotein E variant and exercise type. Front Genet 2023; 14:1136483. [PMID: 37388939 PMCID: PMC10300272 DOI: 10.3389/fgene.2023.1136483] [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: 01/03/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
In various cross-sectional and longitudinal studies, exercise has been associated with cardiometabolic outcomes, including high-density lipoprotein (HDL) cholesterol. Exercise-induced changes in HDL cholesterol seem to be affected by genetic polymorphisms. In this study, we examined whether variant APOE rs7412 is involved in the association between HDL cholesterol and exercise. From adults assessed in Taiwan Biobank (TWB) between 2008 and 2019, we analyzed data from 57,638 normolipidemic subjects. To examine the association between exercise, APOE rs7412, and HDL cholesterol, a multiple linear regression model was used. A higher HDL was associated with both aerobic exercise (regression coefficient [mg/dL] beta- (β), 1.112; 95% confidence interval (CI); 0.903-1.322) and resistance exercise (β, 2.530; 95% CI, 2.093-2.966). In comparison with the APOE rs7412-CC genotype, the β was 2.589 (95% CI, 2.329-2.848) among those with the CT + TT genotype. Compared to adults who had the CC genotype and did not exercise (the CC/no exercise group), the β-coefficient determined for the different genotype and exercise groups was 1.135 (95% CI, 0.911-1.359) for the CC genotype and aerobic exercise group, 2.753 (95% CI, 2.283-3.322) for the CC genotype and resistance exercise group, 2.705 (95% CI, 2.390-3.020) for the CT + TT genotype and no exercise group, 3.682 (95% CI, 3.218-4.146) for the CT + TT genotype and aerobic exercise group, and 3.855 (95% CI, 2.727-4.982) for the CT + TT genotype and resistance exercise group, respectively. This study demonstrates that self-reported aerobic and resistance exercise both raised HDL levels, yet resistance exercise was associated with a greater increase, particularly among Taiwanese subjects carrying the APOE rs7412-CT+TT genotype.
Collapse
Affiliation(s)
- Huan-Cheng Chang
- Division of Family Medicine, Department of Community Medicine, Landseed International Hospital, Taoyuan City, Taiwan
- Department of Health Business Management Administration, Hungkuang University, Taichung City, Taiwan
| | - Oswald Ndi Nfor
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
| | - Chien-Chang Ho
- Department of Physical Education, Fu Jen Catholic University, New Taipei, Taiwan
- Research and Development Center for Physical Education, Health, and Information Technology, Fu Jen Catholic University, New Taipei, Taiwan
| | - Pei-Hsin Chen
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| |
Collapse
|
7
|
Bonilha I, Luchiari B, Nadruz W, Sposito AC. Very low HDL levels: clinical assessment and management. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:3-18. [PMID: 36651718 PMCID: PMC9983789 DOI: 10.20945/2359-3997000000585] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In individuals with very low high-density lipoprotein (HDL-C) cholesterol, such as Tangier disease, LCAT deficiency, and familial hypoalphalipoproteinemia, there is an increased risk of premature atherosclerosis. However, analyzes based on comparisons of populations with small variations in HDL-C mediated by polygenic alterations do not confirm these findings, suggesting that there is an indirect association or heterogeneity in the pathophysiological mechanisms related to the reduction of HDL-C. Trials that evaluated some of the HDL functions demonstrate a more robust degree of association between the HDL system and atherosclerotic risk, but as they were not designed to modify lipoprotein functionality, there is insufficient data to establish a causal relationship. We currently have randomized clinical trials of therapies that increase HDL-C concentration by various mechanisms, and this HDL-C elevation has not independently demonstrated a reduction in the risk of cardiovascular events. Therefore, this evidence shows that (a) measuring HDL-C as a way of estimating HDL-related atheroprotective system function is insufficient and (b) we still do not know how to increase cardiovascular protection with therapies aimed at modifying HDL metabolism. This leads us to a greater effort to understand the mechanisms of molecular action and cellular interaction of HDL, completely abandoning the traditional view focused on the plasma concentration of HDL-C. In this review, we will detail this new understanding and the new horizon for using the HDL system to mitigate residual atherosclerotic risk.
Collapse
Affiliation(s)
- Isabella Bonilha
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Beatriz Luchiari
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Wilson Nadruz
- Universidade de Campinas (Unicamp), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Andrei C Sposito
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil,
| |
Collapse
|
8
|
Desrochers GF, Filip R, Bastianelli M, Stern T, Pezacki JP. microRNA-27b regulates hepatic lipase enzyme LIPC and reduces triglyceride degradation during hepatitis C virus infection. J Biol Chem 2022; 298:101983. [PMID: 35483451 PMCID: PMC9163519 DOI: 10.1016/j.jbc.2022.101983] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 11/26/2022] Open
Abstract
miRNAs are short, noncoding RNAs that negatively and specifically regulate protein expression, the cumulative effects of which can result in broad changes to cell systems and architecture. The miRNA miR-27b is known to regulate lipid regulatory pathways in the human liver and is also induced by the hepatitis C virus (HCV). However, the functional targets of miR-27b are not well established. Herein, an activity-based protein profiling method using a serine hydrolase probe, coupled with stable isotope labeling and mass spectrometry identified direct and indirect targets of miR-27b. The hepatic lipase C (LIPC) stood out as both highly dependent on miR-27b and as a major modulator of lipid pathway misregulation. Modulation of miR-27b using both exogenous miRNA mimics and inhibitors demonstrated that transcription factors Jun, PPARα, and HNF4α, all of which also influence LIPC levels and activity, are regulated by miR-27b. LIPC was furthermore shown to affect the progress of the life cycle of HCV and to decrease levels of intracellular triglycerides, upon which HCV is known to depend. In summary, this work has demonstrated that miR-27b mediates HCV infection by downregulating LIPC, thereby reducing triglyceride degradation, which in turn increases cellular lipid levels.
Collapse
Affiliation(s)
| | - Roxana Filip
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
| | - Micheal Bastianelli
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Tiffany Stern
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
9
|
Coenzyme Q10 supplementation improves cholesterol efflux capacity and anti-inflammatory properties of HDL in Chinese adults with dyslipidemia. Nutrition 2022; 101:111703. [DOI: 10.1016/j.nut.2022.111703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/25/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022]
|
10
|
Zhalimov V, Gritsyna Y, Morenkov O. The effect of apolipoprotein A-I and perfluorocarbon emulsion on the production of specific antibodies in mice. Clin Exp Vaccine Res 2021; 10:259-263. [PMID: 34703809 PMCID: PMC8511586 DOI: 10.7774/cevr.2021.10.3.259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/15/2021] [Indexed: 11/15/2022] Open
Abstract
Adjuvants are often required to improve the potentially low immunogenicity of vaccines. In this study, it is proposed to use an emulsion based on fluorocarbons as an adjuvant. Since this emulsion adsorbs only a small range of proteins, apolipoprotein A-I (ApoAI) was used as an anchor. Antigen and ApoAI were combined by creating a fusion construct. Results showed that the combined use of a perfluorocarbon emulsion and ApoAI during immunization significantly increases the specific antibody titer in mice and in its effectiveness this system is close to the incomplete Freund's adjuvant.
Collapse
Affiliation(s)
- Vitaly Zhalimov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Yuliya Gritsyna
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Oleg Morenkov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
11
|
Kardassis D, Thymiakou E, Chroni A. Genetics and regulation of HDL metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1867:159060. [PMID: 34624513 DOI: 10.1016/j.bbalip.2021.159060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
The inverse association between plasma HDL cholesterol (HDL-C) levels and risk for cardiovascular disease (CVD) has been demonstrated by numerous epidemiological studies. However, efforts to reduce CVD risk by pharmaceutically manipulating HDL-C levels failed and refused the HDL hypothesis. HDL-C levels in the general population are highly heterogeneous and are determined by a combination of genetic and environmental factors. Insights into the causes of HDL-C heterogeneity came from the study of monogenic HDL deficiency syndromes but also from genome wide association and Μendelian randomization studies which revealed the contribution of a large number of loci to low or high HDL-C cases in the general or in restricted ethnic populations. Furthermore, HDL-C levels in the plasma are under the control of transcription factor families acting primarily in the liver including members of the hormone nuclear receptors (PPARs, LXRs, HNF-4) and forkhead box proteins (FOXO1-4) and activating transcription factors (ATFs). The effects of certain lipid lowering drugs used today are based on the modulation of the activity of specific members of these transcription factors. During the past decade, the roles of small or long non-coding RNAs acting post-transcriptionally on the expression of HDL genes have emerged and provided novel insights into HDL regulation and new opportunities for therapeutic interventions. In the present review we summarize recent progress made in the genetics and the regulation (transcriptional and post-transcriptional) of HDL metabolism.
Collapse
Affiliation(s)
- Dimitris Kardassis
- Laboratory of Biochemistry, Department of Basic Sciences, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece.
| | - Efstathia Thymiakou
- Laboratory of Biochemistry, Department of Basic Sciences, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| |
Collapse
|
12
|
Genome-Wide Association Study on Adiponectin-Mediated Suppression of HDL-C Levels in Taiwanese Individuals Identifies Functional Haplotypes in CDH13. Genes (Basel) 2021; 12:genes12101582. [PMID: 34680977 PMCID: PMC8535967 DOI: 10.3390/genes12101582] [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: 09/02/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/24/2022] Open
Abstract
CDH13 encodes T-cadherin, which is expressed in the vasculature and cardiac myocytes and is the receptor for hexameric and high-molecular-weight adiponectin. The CDH13 region is the most pivotal locus associated with adiponectin level. Mediation analysis is a method to explore the effect of a third variable, it is assumed that the magnitude of the relationship between the independent and dependent variables will be reduced by statistical adjustment for a third variable. In addition, mediation can further occur in the case when the mediator acts as a pathway-suppressor variable that means a suppression effect may be suggested if the statistical removal of a mediation effect could increase the magnitude of the relationship between the independent and dependent variables. Here, we aimed to explore the suppression effect in a genome-wide association study, and investigate possible mechanisms that may link adiponectin to CDH13 variants and high-density lipoprotein cholesterol (HDL-C). Genome-wide association data for adiponectin and HDL-C were accessible for 2349 Taiwan-biobank participants. The mediation analysis was conducted with the CDH13 lead single nucleotide polymorphism (SNP) rs4783244. The cloned constructs of CDH13 haplotypes (GG and TT) identified from the rs4783244 G/T and rs12051272 G/T SNPs were transiently expressed in HEK293T cells and investigated using the luciferase reporter assay. Genome-wide association analysis showed that HDL-C is significantly associated with variants in CDH13 after adjusting for the adiponectin level. The lead SNP rs4783244 was significantly associated with lower adiponectin levels and exhibited a suppression effect on HDL-C when adiponectin was included as a third factor in the mediation analysis. Luciferase reporter assay results further demonstrated that the GG haplotype increased enhancer activity, whereas the haplotype TT significantly reduced the activity of this enhancer. We present the first evidence of the suppressive role of adiponectin in the genome-wide association between CDH13 and HDL-C. CDH13 may increase the HDL-C levels, and its expression is suppressed by adiponectin.
Collapse
|
13
|
León-Mimila P, Villamil-Ramírez H, Macías-Kauffer LR, Jacobo-Albavera L, López-Contreras BE, Posadas-Sánchez R, Posadas-Romero C, Romero-Hidalgo S, Morán-Ramos S, Domínguez-Pérez M, Olivares-Arevalo M, López-Montoya P, Nieto-Guerra R, Acuña-Alonzo V, Macín-Pérez G, Barquera-Lozano R, Del-Río-Navarro BE, González-González I, Campos-Pérez F, Gómez-Pérez F, Valdés VJ, Sampieri A, Reyes-García JG, Carrasco-Portugal MDC, Flores-Murrieta FJ, Aguilar-Salinas CA, Vargas-Alarcón G, Shih D, Meikle PJ, Calkin AC, Drew BG, Vaca L, Lusis AJ, Huertas-Vazquez A, Villarreal-Molina T, Canizales-Quinteros S. Genome-Wide Association Study Identifies a Functional SIDT2 Variant Associated With HDL-C (High-Density Lipoprotein Cholesterol) Levels and Premature Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2021; 41:2494-2508. [PMID: 34233476 PMCID: PMC8664085 DOI: 10.1161/atvbaha.120.315391] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective Low HDL-C (high-density lipoprotein cholesterol) is the most frequent dyslipidemia in Mexicans, but few studies have examined the underlying genetic basis. Our purpose was to identify genetic variants associated with HDL-C levels and cardiovascular risk in the Mexican population. Approach and Results A genome-wide association studies for HDL-C levels in 2335 Mexicans, identified four loci associated with genome-wide significance: CETP, ABCA1, LIPC, and SIDT2. The SIDT2 missense Val636Ile variant was associated with HDL-C levels and was replicated in 3 independent cohorts (P=5.9×10−18 in the conjoint analysis). The SIDT2/Val636Ile variant is more frequent in Native American and derived populations than in other ethnic groups. This variant was also associated with increased ApoA1 and glycerophospholipid serum levels, decreased LDL-C (low-density lipoprotein cholesterol) and ApoB levels, and a lower risk of premature CAD. Because SIDT2 was previously identified as a protein involved in sterol transport, we tested whether the SIDT2/Ile636 protein affected this function using an in vitro site-directed mutagenesis approach. The SIDT2/Ile636 protein showed increased uptake of the cholesterol analog dehydroergosterol, suggesting this variant affects function. Finally, liver transcriptome data from humans and the Hybrid Mouse Diversity Panel are consistent with the involvement of SIDT2 in lipid and lipoprotein metabolism. Conclusions This is the first genome-wide association study for HDL-C levels seeking associations with coronary artery disease in the Mexican population. Our findings provide new insight into the genetic architecture of HDL-C and highlight SIDT2 as a new player in cholesterol and lipoprotein metabolism in humans.
Collapse
Affiliation(s)
- Paola León-Mimila
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
| | - Hugo Villamil-Ramírez
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
| | - Luis R Macías-Kauffer
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
- Dirección de Planeación, Enseñanza e Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Estado de México (L.R.M.-K.)
| | - Leonor Jacobo-Albavera
- Laboratorio de Enfermedades Cardiovasculares, INMEGEN, Mexico City (L.J.-A., M.D.-P., T.V.-M.)
| | - Blanca E López-Contreras
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
| | - Rosalinda Posadas-Sánchez
- Departamento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City (R.P.-S., C.P.-R.)
| | - Carlos Posadas-Romero
- Departamento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City (R.P.-S., C.P.-R.)
| | | | - Sofía Morán-Ramos
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Mexico City (S.M.-R.)
| | - Mayra Domínguez-Pérez
- Laboratorio de Enfermedades Cardiovasculares, INMEGEN, Mexico City (L.J.-A., M.D.-P., T.V.-M.)
| | - Marisol Olivares-Arevalo
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
| | - Priscilla López-Montoya
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
| | - Roberto Nieto-Guerra
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
| | | | - Gastón Macín-Pérez
- Escuela Nacional de Antropología e Historia, Mexico City (V.A.-A., G.M.-P.)
| | | | | | | | | | - Francisco Gómez-Pérez
- Unidad de Investigación en Enfermedades Metabólicas and Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (F.G.-P., C.A.A.-S.)
| | - Victor J Valdés
- Instituto de Fisiología Celular, UNAM, Mexico City (V.J.V., A.S., L.V.)
| | - Alicia Sampieri
- Instituto de Fisiología Celular, UNAM, Mexico City (V.J.V., A.S., L.V.)
| | - Juan G Reyes-García
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City (J.G.R.-G., F.J.F.-M.)
| | - Miriam Del C Carrasco-Portugal
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City (M.C.-P., F.J.F.-M.)
| | - Francisco J Flores-Murrieta
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City (J.G.R.-G., F.J.F.-M.)
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City (M.C.-P., F.J.F.-M.)
| | - Carlos A Aguilar-Salinas
- Unidad de Investigación en Enfermedades Metabólicas and Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (F.G.-P., C.A.A.-S.)
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, N.L. Mexico (C.A.A.-S.)
| | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City (G.V.-A.)
| | - Diana Shih
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (D.S., A.J.L., A.H.-V.)
| | - Peter J Meikle
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (P.J.M.)
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (A.C.C., B.G.D.)
- Central Clinical School, Monash University, Melbourne, VIC, Australia (A.C.C., B.G.D.)
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia (A.C.C., B.G.D.)
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (A.C.C., B.G.D.)
- Central Clinical School, Monash University, Melbourne, VIC, Australia (A.C.C., B.G.D.)
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia (A.C.C., B.G.D.)
| | - Luis Vaca
- Instituto de Fisiología Celular, UNAM, Mexico City (V.J.V., A.S., L.V.)
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (D.S., A.J.L., A.H.-V.)
| | - Adriana Huertas-Vazquez
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (D.S., A.J.L., A.H.-V.)
| | | | - Samuel Canizales-Quinteros
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México (UNAM)/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City (P.L.-M., H.V.-R., L.R.M.-K., B.E.L.-C., S.M.-R., M.O.-A., P.L.-M., R.N.-G., S.C.-Q.)
| |
Collapse
|
14
|
Schoch L, Badimon L, Vilahur G. Unraveling the Complexity of HDL Remodeling: On the Hunt to Restore HDL Quality. Biomedicines 2021; 9:805. [PMID: 34356869 PMCID: PMC8301317 DOI: 10.3390/biomedicines9070805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence has cast doubt over the HDL-cholesterol hypothesis. The complexity of the HDL particle and its proven susceptibility to remodel has paved the way for intense molecular investigation. This state-of-the-art review discusses the molecular changes in HDL particles that help to explain the failure of large clinical trials intending to interfere with HDL metabolism, and details the chemical modifications and compositional changes in HDL-forming components, as well as miRNA cargo, that render HDL particles ineffective. Finally, the paper discusses the challenges that need to be overcome to shed a light of hope on HDL-targeted approaches.
Collapse
Affiliation(s)
- Leonie Schoch
- Cardiovascular Program, Institut de Recerca, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain; (L.S.); (L.B.)
- Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program, Institut de Recerca, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain; (L.S.); (L.B.)
- CiberCV, 08025 Barcelona, Spain
- Cardiovascular Research Chair, UAB, 08025 Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program, Institut de Recerca, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain; (L.S.); (L.B.)
- CiberCV, 08025 Barcelona, Spain
| |
Collapse
|
15
|
Gracia-Rubio I, Martín C, Civeira F, Cenarro A. SR-B1, a Key Receptor Involved in the Progression of Cardiovascular Disease: A Perspective from Mice and Human Genetic Studies. Biomedicines 2021; 9:biomedicines9060612. [PMID: 34072125 PMCID: PMC8229968 DOI: 10.3390/biomedicines9060612] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022] Open
Abstract
High plasma level of low-density lipoprotein (LDL) is the main driver of the initiation and progression of cardiovascular disease (CVD). Nevertheless, high-density lipoprotein (HDL) is considered an anti-atherogenic lipoprotein due to its role in reverse cholesterol transport and its ability to receive cholesterol that effluxes from macrophages in the artery wall. The scavenger receptor B class type 1 (SR-B1) was identified as the high-affinity HDL receptor, which facilitates the selective uptake of cholesterol ester (CE) into the liver via HDL and is also implicated in the plasma clearance of LDL, very low-density lipoprotein (VLDL) and lipoprotein(a) (Lp(a)). Thus, SR-B1 is a multifunctional receptor that plays a main role in the metabolism of different lipoproteins. The aim of this review is to highlight the association between SR-B1 and CVD risk through mice and human genetic studies.
Collapse
Affiliation(s)
- Irene Gracia-Rubio
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain; (F.C.); (A.C.)
- Correspondence: or ; Tel.: +34-976-765-500 (ext. 142895)
| | - César Martín
- Instituto Biofisika (UPV/EHU, CSIC) y Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco UPB/EHU, 48940 Bilbao, Spain;
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain; (F.C.); (A.C.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto Salud Carlos III, 28029 Madrid, Spain
- Departamento de Medicina, Psiquiatría y Dermatología, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Ana Cenarro
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain; (F.C.); (A.C.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto Salud Carlos III, 28029 Madrid, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| |
Collapse
|
16
|
Functional Haplotype of LIPC Induces Triglyceride-Mediated Suppression of HDL-C Levels According to Genome-Wide Association Studies. Genes (Basel) 2021; 12:genes12020148. [PMID: 33499410 PMCID: PMC7910859 DOI: 10.3390/genes12020148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023] Open
Abstract
Hepatic lipase (encoded by LIPC) is a glycoprotein in the triacylglycerol lipase family and mainly synthesized in and secreted from the liver. Previous studies demonstrated that hepatic lipase is crucial for reverse cholesterol transport and modulating metabolism and the plasma levels of several lipoproteins. This study was conducted to investigate the suppression effect of high-density lipoprotein cholesterol (HDL-C) levels in a genome-wide association study and explore the possible mechanisms linking triglyceride (TG) to LIPC variants and HDL-C. Genome-wide association data for TG and HDL-C were available for 4657 Taiwan-biobank participants. The prevalence of haplotypes in the LIPC promoter region and their effects were calculated. The cloned constructs of the haplotypes were expressed transiently in HepG2 cells and evaluated in a luciferase reporter assay. Genome-wide association analysis revealed that HDL-C was significantly associated with variations in LIPC after adjusting for TG. Three haplotypes (H1: TCG, H2: CTA and H3: CCA) in LIPC were identified. H2: CTA was significantly associated with HDL-C levels and H1: TCG suppressed HDL-C levels when a third factor, TG, was included in mediation analysis. The luciferase reporter assay further showed that the H2: CTA haplotype significantly inhibited luciferase activity compared with the H1: TCG haplotype. In conclusion, we identified a suppressive role for TG in the genome-wide association between LIPC and HDL-C. A functional haplotype of hepatic lipase may reduce HDL-C levels and is suppressed by TG.
Collapse
|
17
|
Ke J, Wang Y, Liu S, Li K, Xu Y, Yang L, Zhao D. Relationship of Para and Perirenal Fat and High-Density Lipoprotein and Its Function in Patients with Type 2 Diabetes Mellitus. Int J Endocrinol 2021; 2021:9286492. [PMID: 34976052 PMCID: PMC8716211 DOI: 10.1155/2021/9286492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/15/2021] [Accepted: 12/08/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Para and perirenal fat is a fat pad surrounding the kidneys. Recent studies showed the association between para and perirenal fat and cardiovascular diseases including atherosclerosis and hypertension. We aimed to assess the relationship between para-perirenal ultrasonographic fat thickness and serum high-density lipoprotein (HDL) level and cholesterol efflux capacity of HDL in patients with type 2 diabetes mellitus (T2DM). METHODS We recruited 58 subjects with T2DM and collected anthropometric indices including height, weight, waist circumference, and other clinical data. Para-perirenal ultrasonographic fat thickness (PUFT) was measured via ultrasound. Serum lipid profile and other metabolic indices were determined as well. Correlation analysis and regression analysis were performed to analyze the relationship between PUFT and HDL level and cholesterol efflux capacity in all patients and subgroups. RESULTS Patients with higher PUFT have lower serum HDL level but increased cholesterol efflux capacity. Further analysis showed that PUFT negatively correlated with the serum HDL level in all patients, with no difference in groups divided by body mass index (BMI). In addition, PUFT was positively correlated with cholesterol efflux capacity in all patients. Multiple stepwise regression analysis showed an independent association of PUFT and serum HDL level and cholesterol efflux capacity. CONCLUSIONS PUFT is closely correlated with the serum HDL level and cholesterol efflux capacity in patients with T2DM.
Collapse
Affiliation(s)
- Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Yan Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Simo Liu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Kun Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - YueChao Xu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Longyan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| |
Collapse
|
18
|
Helgadottir A, Sulem P, Thorgeirsson G, Gretarsdottir S, Thorleifsson G, Jensson BÖ, Arnadottir GA, Olafsson I, Eyjolfsson GI, Sigurdardottir O, Thorsteinsdottir U, Gudbjartsson DF, Holm H, Stefansson K. Rare SCARB1 mutations associate with high-density lipoprotein cholesterol but not with coronary artery disease. Eur Heart J 2019; 39:2172-2178. [PMID: 29596577 PMCID: PMC6001888 DOI: 10.1093/eurheartj/ehy169] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 03/14/2018] [Indexed: 12/16/2022] Open
Abstract
Aims Scavenger receptor Class B Type 1 (SR-BI) is a major receptor for high-density lipoprotein (HDL) that promotes hepatic uptake of cholesterol from HDL. A rare mutation p.P376L, in the gene encoding SR-BI, SCARB1, was recently reported to associate with elevated HDL cholesterol (HDL-C) and increased risk of coronary artery disease (CAD), suggesting that increased HDL-C caused by SR-BI impairment might be an independent marker of cardiovascular risk. We tested the hypothesis that alleles in or close to SCARB1 that associate with elevated levels of HDL-C also associate with increased risk of CAD in the relatively homogeneous population of Iceland. Methods and results Using a large resource of whole-genome sequenced Icelanders, we identified thirteen SCARB1 coding mutations that we examined for association with HDL-C (n = 136 672). Three rare SCARB1 mutations, encoding p.G319V, p.V111M, and p.V32M (combined allelic frequency = 0.2%) associate with elevated levels of HDL-C (p.G319V: β = 11.1 mg/dL, P = 8.0 × 10−7; p.V111M: β = 8.3 mg/dL, P = 1.1 × 10−6; p.V32M: β = 10.2 mg/dL, P = 8.1 × 10−4). These mutations do not associate with CAD (36 886 cases/306 268 controls) (odds ratio = 0.90, 95% confidence interval 0.67–1.22, P = 0.49), despite effects on HDL-C comparable to that reported for p.P376L, both in terms of direction and magnitude. Furthermore, HDL-C raising alleles of three common SCARB1 non-coding variants, including one previously unreported (rs61941676-C: β = 1.25 mg/dL, P = 1.7 × 10−18), and of one low frequency coding variant (p.V135I) that independently associate with higher HDL-C, do not confer increased risk of CAD. Conclusion Elevated HDL-C due to genetically compromised SR-BI function is not a marker of CAD risk. ![]()
Collapse
Affiliation(s)
- Anna Helgadottir
- deCODE Genetics/Amgen, Inc., Sturlugata 8, 101 Reykjavik, Iceland
| | - Patrick Sulem
- deCODE Genetics/Amgen, Inc., Sturlugata 8, 101 Reykjavik, Iceland
| | - Gudmundur Thorgeirsson
- deCODE Genetics/Amgen, Inc., Sturlugata 8, 101 Reykjavik, Iceland.,Faculty of Medicine, Department of Medicine, University of Iceland, Saemundargata 2, 101 Reykjavik, Iceland.,Division of Cardiology, Department of Internal Medicine, Landspitali, National University Hospital of Iceland, Hringbraut, 101 Reykjavik, Iceland
| | | | | | | | | | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali, National University Hospital, Hringbraut, 101 Reykjavik, Iceland
| | | | - Olof Sigurdardottir
- Department of Clinical Biochemistry, Akureyri Hospital, 600 Akureyri, Iceland
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Inc., Sturlugata 8, 101 Reykjavik, Iceland.,Faculty of Medicine, Department of Medicine, University of Iceland, Saemundargata 2, 101 Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Inc., Sturlugata 8, 101 Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, 101 Reykjavik, Iceland
| | - Hilma Holm
- deCODE Genetics/Amgen, Inc., Sturlugata 8, 101 Reykjavik, Iceland
| | - Kari Stefansson
- deCODE Genetics/Amgen, Inc., Sturlugata 8, 101 Reykjavik, Iceland.,Faculty of Medicine, Department of Medicine, University of Iceland, Saemundargata 2, 101 Reykjavik, Iceland
| |
Collapse
|
19
|
Rare P376L variant in the SR-BI gene associates with HDL dysfunction and risk of cardiovascular disease. Clin Biochem 2019; 73:44-49. [PMID: 31251897 DOI: 10.1016/j.clinbiochem.2019.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 06/22/2019] [Accepted: 06/22/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Scavenger receptor class B type 1 (SR-BI) encoded by SCARB1 gene serves as a multifunctional HDL receptor, facilitating the uptake of cholesteryl esters from HDL to the liver. Recent studies have identified the association between the P376L missense mutation of the SCARB1 gene with increased serum HDL-Cholesterol level. However, the contribution of this variant to the development of cardiovascular disease (CVD) remains unclear. OBJECTIVE We have investigated the association between the P376L polymorphism with the properties of HDL and CVD outcomes in a population sample recruited as part of the Mashhad-Stroke and Heart-Atherosclerotic-Disorders (MASHAD) cohort. METHODS Six hundred and fifteen individuals who had a median follow-up period of 7 years were recruited as part of the MASHAD cohort. Anthropometric, biochemical parameters and HDL lipid peroxidation (HDLox) were assessed. Genotyping was performed using TaqMan-real-time-PCR based method. The association of P376L-rs74830766 with cardiovascular-risk-factors and CVD events were evaluated. RESULTS Carriers of the P376L variant were significantly more likely than non-carriers to develop CVD using multivariate analyses adjusted for traditional CVD risk factors defined as: age, sex, BMI, presence of diabetes, or hypertension, positive smoking habit, and total cholesterol (OR: 3.75, 95%CI: 1.76-7.98, p = 0.001). In an adjusted model, there was a two fold increase in cardiovascular endpoints among individuals who were heterozygous for the P376L variant (hazard ratio, 2.08; 95% CI, 1.12-to 3.84, p = 0.02). Although there was no association between the presence of the P376L variant and HDL-C level, serum HDLox, measured as dysfunctional HDL, was 13% higher among carriers of the P376L variant than non-carriers. CONCLUSION We have found that carriers of the P376L variant possessed higher HDLox and were at increased risk of CVD in a representative population-based cohort, as compared to non-carriers.
Collapse
|
20
|
Analysis of differential gene expression by RNA-seq data in ABCG1 knockout mice. Gene 2018; 689:24-33. [PMID: 30528268 DOI: 10.1016/j.gene.2018.11.086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/05/2018] [Accepted: 11/22/2018] [Indexed: 12/20/2022]
Abstract
AIMS The previous studies on ABCG1 using genetically modified mice showed inconsistent results on atherosclerosis. The aim of this study was to determine whether accurate target knockout of ABCG1 would result in transcriptional changes of other atherosclerosis-related genes. METHODS ABCG1 knockout mouse model was obtained by precise gene targeting without affecting non-target DNA sequences in C57BL/6 background. The wildtype C57BL/6 mice were regarded as control group. 12-week-old male mice were used in current study. We performed whole transcriptome analysis on the peripheral blood mononuclear cells obtained from ABCG1 knockout mice (n = 3) and their wildtype controls (n = 3) by RNA-seq. RESULTS Compared with wildtype group, 605 genes were modified at the time of ABCG1 knockout and expressed differentially in knockout group, including 306 up-regulated genes and 299 down-regulated genes. 54 genes were associated with metabolism regulation, of which 13 were related to lipid metabolism. We also found some other modified genes in knockout mice involved in cell adhesion, leukocyte transendothelial migration and apoptosis, which might also play roles in the process of atherosclerosis. 7 significantly enriched GO terms and 19 significantly enriched KEGG pathways were identified, involving fatty acid biosynthesis, immune response and intracellular signal transduction. CONCLUSIONS ABCG1 knockout mice exhibited an altered expression of multiple genes related to many aspects of atherosclerosis, which might affect the further studies to insight into the effect of ABCG1 on atherosclerosis with this animal model.
Collapse
|
21
|
Zheng Z, Ren K, Peng X, Zhu X, Yi G. Lymphatic Vessels: A Potential Approach to the Treatment of Atherosclerosis? Lymphat Res Biol 2018; 16:498-506. [PMID: 30272526 DOI: 10.1089/lrb.2018.0015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Many basic and clinical studies have demonstrated that atherosclerosis is a chronic inflammatory disease. Although there are many factors affecting atherosclerosis, the role of lymphatic vessels in this disease has been neglected. Traditionally, lymphatic vessels have been considered to be passages for transporting interstitial fluid to the blood circulation. However, as early as the last century, researchers found that there are numerous lymphatic vessels surrounding sites of atherosclerosis; however, the relationship between lymphatic vessels and atherosclerosis is not clear. With further research, lymphatic vessels were determined to be involved in the induction and resolution of arterial inflammation and also to play a positive role in plaque cholesterol transport. There are abundant immune cells around atherosclerosis, and these immune cells not only have a significant impact on plaque formation but also affect local lymphangiogenesis (IAL). This promotion of IAL seems to relieve the progression of atherosclerosis. Therefore, research into the relationship between lymphatic vessels and atherosclerosis is of great importance for improving atherosclerosis treatment. This review highlights what is known about the relationship between lymphatic vessels and atherosclerosis, including the effect of immune cells on IAL, and reverse cholesterol transport. In addition, we present some of our views on the improvement of atherosclerosis treatment, which have significant clinical value in research.
Collapse
Affiliation(s)
- Zhi Zheng
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang City, China
| | - Kun Ren
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang City, China
| | - Xiaoshan Peng
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang City, China
| | - Xiao Zhu
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang City, China
| | - Guanghui Yi
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang City, China
| |
Collapse
|
22
|
Kim M, Kim M, Yoo HJ, Bang YJ, Lee SH, Lee JH. Apolipoprotein A5
gene variants are associated with decreased adiponectin levels and increased arterial stiffness in subjects with low high-density lipoprotein-cholesterol levels. Clin Genet 2018; 94:438-444. [DOI: 10.1111/cge.13439] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/26/2018] [Accepted: 08/18/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Minkyung Kim
- Research Center for Silver Science; Institute of Symbiotic Life-TECH, Yonsei University; Seoul Korea
| | - Minjoo Kim
- Research Center for Silver Science; Institute of Symbiotic Life-TECH, Yonsei University; Seoul Korea
| | - Hye Jin Yoo
- Department of Food and Nutrition; Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University; Seoul Korea
| | - Yung Ju Bang
- Department of Food and Nutrition; Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University; Seoul Korea
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition; College of Human Ecology, Yonsei University; Seoul Korea
| | - Sang-Hyun Lee
- Department of Family Practice; National Health Insurance Corporation, Ilsan Hospital; Goyang Korea
| | - Jong Ho Lee
- Research Center for Silver Science; Institute of Symbiotic Life-TECH, Yonsei University; Seoul Korea
- Department of Food and Nutrition; Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University; Seoul Korea
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition; College of Human Ecology, Yonsei University; Seoul Korea
| |
Collapse
|
23
|
Morais CLM, Lima KMG, Martin FL. Colourimetric Determination of High-Density Lipoprotein (HDL) Cholesterol Using Red–Green–Blue Digital Colour Imaging. ANAL LETT 2018. [DOI: 10.1080/00032719.2018.1453833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Camilo L. M. Morais
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, UK
| | - Kássio M. G. Lima
- Biological Chemistry and Chemometrics, Institute of Chemistry, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Francis L. Martin
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, UK
| |
Collapse
|
24
|
van Leeuwen EM, Emri E, Merle BMJ, Colijn JM, Kersten E, Cougnard-Gregoire A, Dammeier S, Meester-Smoor M, Pool FM, de Jong EK, Delcourt C, Rodrigez-Bocanegra E, Biarnés M, Luthert PJ, Ueffing M, Klaver CCW, Nogoceke E, den Hollander AI, Lengyel I. A new perspective on lipid research in age-related macular degeneration. Prog Retin Eye Res 2018; 67:56-86. [PMID: 29729972 DOI: 10.1016/j.preteyeres.2018.04.006] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/25/2018] [Accepted: 04/30/2018] [Indexed: 12/15/2022]
Abstract
There is an urgency to find new treatment strategies that could prevent or delay the onset or progression of AMD. Different classes of lipids and lipoproteins metabolism genes have been associated with AMD in a multiple ways, but despite the ever-increasing knowledge base, we still do not understand fully how circulating lipids or local lipid metabolism contribute to AMD. It is essential to clarify whether dietary lipids, systemic or local lipoprotein metabolismtrafficking of lipids in the retina should be targeted in the disease. In this article, we critically evaluate what has been reported in the literature and identify new directions needed to bring about a significant advance in our understanding of the role for lipids in AMD. This may help to develop potential new treatment strategies through targeting the lipid homeostasis.
Collapse
Affiliation(s)
- Elisabeth M van Leeuwen
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eszter Emri
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Benedicte M J Merle
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | - Johanna M Colijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eveline Kersten
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Audrey Cougnard-Gregoire
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | - Sascha Dammeier
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Magda Meester-Smoor
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Eiko K de Jong
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Cécile Delcourt
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | | | | | | | - Marius Ueffing
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Caroline C W Klaver
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Everson Nogoceke
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Imre Lengyel
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
25
|
Marateb HR, Mohebian MR, Javanmard SH, Tavallaei AA, Tajadini MH, Heidari-Beni M, Mañanas MA, Motlagh ME, Heshmat R, Mansourian M, Kelishadi R. Prediction of dyslipidemia using gene mutations, family history of diseases and anthropometric indicators in children and adolescents: The CASPIAN-III study. Comput Struct Biotechnol J 2018; 16:121-130. [PMID: 30026888 PMCID: PMC6050175 DOI: 10.1016/j.csbj.2018.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 12/12/2022] Open
Abstract
Dyslipidemia, the disorder of lipoprotein metabolism resulting in high lipid profile, is an important modifiable risk factor for coronary heart diseases. It is associated with more than four million worldwide deaths per year. Half of the children with dyslipidemia have hyperlipidemia during adulthood, and its prediction and screening are thus critical. We designed a new dyslipidemia diagnosis system. The sample size of 725 subjects (age 14.66 ± 2.61 years; 48% male; dyslipidemia prevalence of 42%) was selected by multistage random cluster sampling in Iran. Single nucleotide polymorphisms (rs1801177, rs708272, rs320, rs328, rs2066718, rs2230808, rs5880, rs5128, rs2893157, rs662799, and Apolipoprotein-E2/E3/E4), and anthropometric, life-style attributes, and family history of diseases were analyzed. A framework for classifying mixed-type data in imbalanced datasets was proposed. It included internal feature mapping and selection, re-sampling, optimized group method of data handling using convex and stochastic optimizations, a new cost function for imbalanced data and an internal validation. Its performance was assessed using hold-out and 4-foldcross-validation. Four other classifiers namely as supported vector machines, decision tree, and multilayer perceptron neural network and multiple logistic regression were also used. The average sensitivity, specificity, precision and accuracy of the proposed system were 93%, 94%, 94% and 92%, respectively in cross validation. It significantly outperformed the other classifiers and also showed excellent agreement and high correlation with the gold standard. A non-invasive economical version of the algorithm was also implemented suitable for low- and middle-income countries. It is thus a promising new tool for the prediction of dyslipidemia.
Collapse
Affiliation(s)
- Hamid R Marateb
- Department of Biomedical Engineering, Facultyof Engineering, University of Isfahan, Isfahan, Iran.,Department of Automatic Control, Biomedical Engineering Research Center, Universitat Politècnica de Catalunya, BarcelonaTech (UPC), Barcelona, Spain
| | - Mohammad Reza Mohebian
- Department of Biomedical Engineering, Facultyof Engineering, University of Isfahan, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied physiology researchcenter, Isfahan cardiovascular research institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Ali Tavallaei
- Department of Biomedical Engineering, Facultyof Engineering, University of Isfahan, Isfahan, Iran
| | | | - Motahar Heidari-Beni
- Nutrition Department, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease,Isfahan University of Medical Sciences, Isfahan, Iran
| | - Miguel Angel Mañanas
- Department of Automatic Control, Biomedical Engineering Research Center, Universitat Politècnica de Catalunya, BarcelonaTech (UPC), Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterialsand Nanomedicine (CIBER-BBN), Barcelona, Spain
| | | | - Ramin Heshmat
- Department of Epidemiology, Chronic Diseases Research Center, Endocrinology and MetabolismPopulation Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marjan Mansourian
- Applied physiology researchcenter, Isfahan cardiovascular research institute, Isfahan University of Medical Sciences, Isfahan, Iran.,Biostatistics and Epidemiology Department, Faculty of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Kelishadi
- Pediatrics Department, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
26
|
Mangat R, Borthwick F, Haase T, Jacome M, Nelson R, Kontush A, Vine DF, Proctor SD. Intestinal lymphatic HDL miR‐223 and ApoA‐I are reduced during insulin resistance and restored with niacin. FASEB J 2018; 32:1602-1612. [DOI: 10.1096/fj.201600298rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Rabban Mangat
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Faye Borthwick
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Tina Haase
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Miriam Jacome
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Randy Nelson
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Anatol Kontush
- National Institute for Health and Medical Research University of Pierre and Marie Curie, Salpétrière University Hospital Paris France
| | - Donna F. Vine
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Spencer D. Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| |
Collapse
|
27
|
Storti F, Raphael G, Griesser V, Klee K, Drawnel F, Willburger C, Scholz R, Langmann T, von Eckardstein A, Fingerle J, Grimm C, Maugeais C. Regulated efflux of photoreceptor outer segment-derived cholesterol by human RPE cells. Exp Eye Res 2017; 165:65-77. [PMID: 28943268 DOI: 10.1016/j.exer.2017.09.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/18/2017] [Indexed: 12/19/2022]
Abstract
Genetic studies have linked age-related macular degeneration (AMD) to genes involved in high-density lipoprotein (HDL) metabolism, including ATP-binding cassette transporter A1 (ABCA1). The retinal pigment epithelium (RPE) handles large amounts of lipids, among others cholesterol, partially derived from internalized photoreceptor outer segments (OS) and lipids physiologically accumulate in the aging eye. To analyze the potential function of ABCA1 in the eye, we measured cholesterol efflux, the first step of HDL generation, in RPE cells. We show the expression of selected genes related to HDL metabolism in mouse and human eyecups as well as in ARPE-19 and human primary RPE cells. Immunofluorescence staining revealed localization of ABCA1 on both sides of polarized RPE cells. This was functionally confirmed by directional efflux to apolipoprotein AI (ApoA-I) of 3H-labeled cholesterol given to the cells via serum or via OS. ABCA1 expression and activity was modulated using a liver-X-receptor (LXR) agonist and an ABCA1 neutralizing antibody, demonstrating that the efflux was ABCA1-dependent. We concluded that the ABCA1-mediated lipid efflux pathway, and hence HDL biosynthesis, is functional in RPE cells towards both the basal (choroidal) and apical (subretinal) space. Impaired activity of the pathway might cause age-related perturbations of lipid homeostasis in the outer retina and thus may contribute to disease development and/or progression.
Collapse
Affiliation(s)
- Federica Storti
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland
| | - Gabriele Raphael
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Vera Griesser
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Katrin Klee
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Faye Drawnel
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Carolin Willburger
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Rebecca Scholz
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | | | - Jürgen Fingerle
- Natural and Medical Sciences Institute, University of Tübingen, Tübingen, Germany
| | - Christian Grimm
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland.
| | - Cyrille Maugeais
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
28
|
Abstract
High-density lipoprotein cholesterol (HDL-C) levels are inversely related to risk of atherosclerotic cardiovascular disease (ASCVD). However, the simplistic assumption that HDL-C levels directly and causally impact atherogenesis has been challenged in recent years. The purpose of this article is to review the current state of knowledge regarding genetically determined HDL-C levels and ASCVD risk and determine what insight these studies provide into the causal relationship between HDL and atherosclerosis.
Collapse
Affiliation(s)
- Liam R Brunham
- Department of Medicine, University of British Columbia, Vancouver, Canada. .,Centre for Heart Lung Innovation, Providence Health Care Research Institute, University of British Columbia, St. Paul's Hospital, Room 166-1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada. .,Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
29
|
|
30
|
Miele EM, Headley SA, Germain M, Joubert J, Herrick S, Milch C, Evans E, Cornelius A, Brewer B, Taylor B, Wood RJ. High-density lipoprotein particle pattern and overall lipid responses to a short-term moderate-intensity aerobic exercise training intervention in patients with chronic kidney disease. Clin Kidney J 2017; 10:524-531. [PMID: 28852492 PMCID: PMC5570090 DOI: 10.1093/ckj/sfx006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/17/2017] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is associated with abnormal lipid profiles and altered high-density lipoprotein (HDL) particle size patterns. Lower levels of the larger, cardioprotective HDL particles found in CKD may play a role in the increased risk for cardiovascular disease in these patients. The current study was designed to assess the effects of short-term moderate-intensity aerobic exercise training on the HDL particle pattern and overall lipid profiles in stage 3 CKD patients. METHODS Forty-six men and women with stage 3 CKD were randomized to either exercise (EX, n = 25) or control (CON, n = 21). Those in the EX group completed 16 weeks of supervised moderate-intensity aerobic exercise three times per week. Serum total cholesterol, HDL cholesterol (HDL-C), triglycerides (TGs), low-density lipoprotein cholesterol (LDL-C), HDL particle size, estimated glomerular filtration rate (eGFR), body composition and peak oxygen uptake (VO2peak) were assessed at baseline and week 16. RESULTS The rate of compliance in the EX group was 97 ± 7.2%. No change was observed in eGFR over time in either group. There was an 8.2% improvement in VO2peak in the EX group (P = 0.05), while VO2peak decreased in the CON group. HDL-C, TGs, HDL particle size and body composition remained unchanged in both groups. A trend was found for lower total cholesterol (TC) (P = 0.051) and LDL-C (P = 0.07) in the CON group. CONCLUSION Our findings indicate that a short-term aerobic exercise training intervention in stage 3 CKD patients does not induce changes in HDL particle size or favorable lipid profile modifications.
Collapse
Affiliation(s)
| | | | - Michael Germain
- Renal and Transplant Associates of New England, Springfield, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Moghadasi M, Kelishadi R, Marateb HR, Haghjooy Javanmard S, Mansourian M, Heshmat R, Esmaeil Motlagh M. Logic Regression Analysis of Gene Polymorphisms and HDL Levels in a Nationally Representative Sample of Iranian Adolescents: The CASPIAN-III Study. Int J Endocrinol Metab 2017; 15:e14037. [PMID: 30805016 PMCID: PMC6372018 DOI: 10.5812/ijem.14037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 07/01/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND To investigate the associations of genetic polymorphism with high-density lipoprotein-cholesterol (HDL-C) levels in Iranian adolescents. METHODS This multicentre study was conducted on 10 - 18 year-old students from 27 provinces in Iran. Logic regression approach was used to determine the main effects and interactions of polymorphisms related to HDL-C levels. RESULTS The rs708272 polymorphism was significantly related to HDL-C levels. Moreover, rs708272 increased HDL-C levels and had a protective effect on HDL-C. The interaction of rs2230808 and rs5880 polymorphisms as well as the interaction of rs320 and rs708272 polymorphisms were associated with lower HDL-C levels. Furthermore, the interaction of rs320 and rs1801177 polymorphisms was associated with lower HDL-C levels. CONCLUSIONS We found that not only single SNPs, but also interactions of several SNPs affect HDL-C levels. Given the high prevalence of low HDL-C in Middle Eastern populations, further genetic studies are required for detailed analysis.
Collapse
Affiliation(s)
- Mehri Moghadasi
- Student Research Center, Biostatistics and Epidemiology Department, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Kelishadi
- Pediatrics Department, Child Growth and Development Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Reza Marateb
- Biomedical Engineering Department, Faculty of Engineering, University of Isfahan, Isfahan, Iran
- Department of Automatic Control, Biomedical Engineering Research Center, Universitat Politècnica de Catalunya, BarcelonaTech (UPC), Barcelona, Spain
| | | | - Marjan Mansourian
- Pediatrics Department, Child Growth and Development Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Physiology Department, Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Biostatistics and Epidemiology Department, Faculty of Health, Isfahan University of Medical Sciences, Isfahan, Iran
- Corresponding author: Marjan Mansourian, Department of Biostatistics and Epidemiology, School of Public Health, Isfahan University of Medical Sciences, Hezarjarib St, Isfahan, Iran. Tel: +98-3137923256, Fax: +98-3136687898, E-mail:
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
32
|
Pikó P, Fiatal S, Kósa Z, Sándor J, Ádány R. Genetic factors exist behind the high prevalence of reduced high-density lipoprotein cholesterol levels in the Roma population. Atherosclerosis 2017. [PMID: 28624686 DOI: 10.1016/j.atherosclerosis.2017.05.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND AIMS Previous findings showed that reduced plasma high-density lipoprotein cholesterol (HDL-C) levels are more frequent in all age groups of the Hungarian Roma compared to the general population. It suggests that genetic factors may exist behind this phenomenon. Our present study was designed to test this hypothesis, i.e., to define whether genetic factors contribute to the higher prevalence of reduced HDL-C among Roma. Single nucleotide polymorphisms (N = 21) contributing to the variation in plasma HDL-C concentrations were analysed in the Hungarian Roma (N = 646) and general (N = 1542) populations. METHODS Genetic risk scores, unweighted (GRS) and weighted (wGRS), were computed and compared. Associations between the GRSs and the prevalence of reduced HDL-C levels were analysed. RESULTS The GRS and wGRS were significantly higher in the Roma compared to the general population (GRS: 22.2 ± 3.2 vs. 21.5 ± 3.3; wGRS: 0.57 ± 0.1 vs. 0.53 ± 0.1; p<0.001). One half per cent of Roma subjects were in the bottom fifth of the wGRS (wGRS≤ 0.3) compared with 1.8% of those in the general population (p=0.025), while 5% of the Roma subjects were in the top fifth of the wGRS (wGRS≥ 0.75) compared with 2.6% of those in the general population (p=0.004). The GRS showed similar correlation with reduced plasma HDL-C levels in the two populations, whilst the wGRS showed stronger correlation with the trait among Roma after controlling for confounders. CONCLUSIONS These results strongly suggest that genetic factors contribute to the higher prevalence of reduced HDL-C levels among Roma, so interventions aiming to improve Roma health status need to consider their increased genetic susceptibility.
Collapse
Affiliation(s)
- Péter Pikó
- MTA-DE Public Health Research Group of the Hungarian Academy of Sciences, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary
| | - Szilvia Fiatal
- Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; WHO Collaborating Centre on Vulnerability and Health, Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary
| | - Zsigmond Kósa
- Department of Health Visitor Methodology and Public Health, Faculty of Health, University of Debrecen, Nyíregyháza 4400, Hungary
| | - János Sándor
- Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; WHO Collaborating Centre on Vulnerability and Health, Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary
| | - Róza Ádány
- MTA-DE Public Health Research Group of the Hungarian Academy of Sciences, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; WHO Collaborating Centre on Vulnerability and Health, Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary.
| |
Collapse
|
33
|
Desgagné V, Bouchard L, Guérin R. microRNAs in lipoprotein and lipid metabolism: from biological function to clinical application. Clin Chem Lab Med 2017; 55:667-686. [PMID: 27987357 DOI: 10.1515/cclm-2016-0575] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022]
Abstract
microRNAs (miRNAs) are short (~22 nucleotides), non-coding, single-stranded RNA molecules that regulate the expression of target genes by partial sequence-specific base-pairing to the targeted mRNA 3'UTR, blocking its translation, and promoting its degradation or its sequestration into processing bodies. miRNAs are important regulators of several physiological processes including developmental and metabolic functions, but their concentration in circulation has also been reported to be altered in many pathological conditions such as familial hypercholesterolemia, cardiovascular diseases, obesity, type 2 diabetes, and cancers. In this review, we focus on the role of miRNAs in lipoprotein and lipid metabolism, with special attention to the well-characterized miR-33a/b, and on the huge potential of miRNAs for clinical application as biomarkers and therapeutics in the context of cardiometabolic diseases.
Collapse
Affiliation(s)
| | - Luigi Bouchard
- Département de biochimie, Université de Sherbrooke, Sherbrooke, Québec
| | - Renée Guérin
- Département de biochimie, Université de Sherbrooke, Sherbrooke, Québec
| |
Collapse
|
34
|
DiMarco DM, Norris GH, Millar CL, Blesso CN, Fernandez ML. Intake of up to 3 Eggs per Day Is Associated with Changes in HDL Function and Increased Plasma Antioxidants in Healthy, Young Adults. J Nutr 2017; 147:323-329. [DOI: 10.3945/jn.116.241877] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/12/2016] [Accepted: 12/12/2016] [Indexed: 11/14/2022] Open
|
35
|
Manjunatha S, Distelmaier K, Dasari S, Carter RE, Kudva YC, Nair KS. Functional and proteomic alterations of plasma high density lipoproteins in type 1 diabetes mellitus. Metabolism 2016; 65:1421-31. [PMID: 27506748 DOI: 10.1016/j.metabol.2016.06.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/01/2016] [Accepted: 06/24/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Higher HDL-cholesterol (HDL-C) is linked to lower cardiovascular risk but individuals with type 1 diabetes mellitus (T1DM) with normal or high HDL-C have higher cardiovascular events compared to age matched non-diabetic controls (ND). We determined whether altered HDL functions despite having normal HDL-C concentration may explain increased cardiovascular risk in T1DM individuals. We also determined whether irreversible posttranslational modifications (PTMs) of HDL bound proteins occur in T1DM individuals with altered HDL functions. METHODS T1DM with poor glycemic control (T1D-PC, HbA1c≥8.5%, n=15) and T1DM with good glycemic control (T1D-GC, HbA1c≤6.6%, n=15) were compared with equal numbers of NDs, ND-PC and ND-GC respectively, matched for age, sex and body mass index (BMI). We measured cholesterol efflux capacity (CEC) of HDL in the serum using J774 macrophages, antioxidant function of HDL as the ability to reverse the oxidative damage of LDL and PON1 activity using commercially available kit. For proteomic analysis, HDL was isolated by density gradient ultracentrifugation and was analyzed by mass spectrometry and shotgun proteomics method. RESULTS Plasma HDL-C concentrations in both T1DM groups were similar to their ND. However, CEC (%) of T1D-PC (16.9±0.8) and T1D-GC (17.1±1) were lower than their respective ND (17.9±1, p=0.01 and 18.2±1.4, p=0.02). HDL antioxidative function also was lower (p<0.05). The abundance of oxidative PTMs of apolipoproteins involved in CEC and antioxidative functions of HDL were higher in T1D-PC (ApoA4, p=0.041) and T1D-GC (ApoA4, p=0.025 and ApoE, p=0.041) in comparison with ND. Both T1D-PC and T1D-GC groups had higher abundance of amadori modification of ApoD (p=0.002 and p=0.041 respectively) and deamidation modification of ApoA4 was higher in T1D-PC (p=0.025). CONCLUSIONS Compromised functions of HDL particles in T1DM individuals, irrespective of glycemic control, could be explained by higher abundance of irreversible PTMs of HDL proteins. These results lend mechanistic support to the hypothesis that HDL quality rather than quantity determines HDL function in T1DM and suggest that measurements of concentrations of HbA1c and HDL-C are not sufficient as biomarkers of effective treatment to lower cardiovascular risk in T1DM individuals.
Collapse
Affiliation(s)
| | - Klaus Distelmaier
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Rickey E Carter
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Yogish C Kudva
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - K Sreekumaran Nair
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
36
|
Wang HY, Quan C, Hu C, Xie B, Du Y, Chen L, Yang W, Yang L, Chen Q, Shen B, Hu B, Zheng Z, Zhu H, Huang X, Xu G, Chen S. A lipidomics study reveals hepatic lipid signatures associating with deficiency of the LDL receptor in a rat model. Biol Open 2016; 5:979-86. [PMID: 27378433 PMCID: PMC4958281 DOI: 10.1242/bio.019802] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The low-density lipoprotein receptor (LDLR) plays a critical role in the liver for the clearance of plasma low-density lipoprotein (LDL). Its deficiency causes hypercholesterolemia in many models. To facilitate the usage of rats as animal models for the discovery of cholesterol-lowering drugs, we took a genetic approach to delete the LDLR in rats aiming to increase plasma LDL cholesterol (LDL-C). An LDLR knockout rat was generated via zinc-finger nuclease technology, which harbors a 19-basepair deletion in the seventh exon of the ldlr gene. As expected, deletion of the LDLR elevated total cholesterol and total triglyceride in the plasma, and caused a tenfold increase of plasma LDL-C and a fourfold increase of plasma very low-density lipoprotein (VLDL-C). A lipidomics analysis revealed that deletion of the LDLR affected hepatic lipid metabolism, particularly lysophosphatidylcholines, free fatty acids and sphingolipids in the liver. Cholesterol ester (CE) 20:4 also displayed a significant increase in the LDLR knockout rats. Taken together, the LDLR knockout rat offers a new model of hypercholesterolemia, and the lipidomics analysis reveals hepatic lipid signatures associating with deficiency of the LDL receptor. Summary: An LDL receptor knockout rat model was generated which offers a new hypercholesterolemia model. A lipidomics analysis reveals hepatic lipid signatures associating with LDLR deficiency in rats.
Collapse
Affiliation(s)
- Hong Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China Collaborative Innovation Center of Genetics and Development, Shanghai 200438, China
| | - Chao Quan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Chunxiu Hu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Bingxian Xie
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Yinan Du
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Liang Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Wei Yang
- Laboratory Animal Center, China Medical University, Shenyang 110001, China
| | - Liu Yang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Bin Shen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Bian Hu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Zhihong Zheng
- Laboratory Animal Center, China Medical University, Shenyang 110001, China
| | - Haibo Zhu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xingxu Huang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China Collaborative Innovation Center of Genetics and Development, Shanghai 200438, China
| |
Collapse
|
37
|
Meriwether D, Sulaiman D, Wagner A, Grijalva V, Kaji I, Williams KJ, Yu L, Fogelman S, Volpe C, Bensinger SJ, Anantharamaiah GM, Shechter I, Fogelman AM, Reddy ST. Transintestinal transport of the anti-inflammatory drug 4F and the modulation of transintestinal cholesterol efflux. J Lipid Res 2016; 57:1175-93. [PMID: 27199144 DOI: 10.1194/jlr.m067025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Indexed: 01/28/2023] Open
Abstract
The site and mechanism of action of the apoA-I mimetic peptide 4F are incompletely understood. Transintestinal cholesterol efflux (TICE) is a process involved in the clearance of excess cholesterol from the body. While TICE is responsible for at least 30% of the clearance of neutral sterols from the circulation into the intestinal lumen, few pharmacological agents have been identified that modulate this pathway. We show first that circulating 4F selectively targets the small intestine (SI) and that it is predominantly transported into the intestinal lumen. This transport of 4F into the SI lumen is transintestinal in nature, and it is modulated by TICE. We also show that circulating 4F increases reverse cholesterol transport from macrophages and cholesterol efflux from lipoproteins via the TICE pathway. We identify the cause of this modulation of TICE either as 4F being a cholesterol acceptor with respect to enterocytes, from which 4F enhances cholesterol efflux, or as 4F being an intestinal chaperone with respect to TICE. Our results assign a novel role for 4F as a modulator of the TICE pathway and suggest that the anti-inflammatory functions of 4F may be a partial consequence of the codependent intestinal transport of both 4F and cholesterol.
Collapse
Affiliation(s)
- David Meriwether
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Dawoud Sulaiman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, Los Angeles, CA
| | - Alan Wagner
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Victor Grijalva
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Izumi Kaji
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Kevin J Williams
- Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Liqing Yu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD
| | - Spencer Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Carmen Volpe
- Division of Laboratory Animal Medicine, University of California Los Angeles, Los Angeles, CA
| | - Steven J Bensinger
- Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Ishaiahu Shechter
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Alan M Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
38
|
Variants with large effects on blood lipids and the role of cholesterol and triglycerides in coronary disease. Nat Genet 2016; 48:634-9. [PMID: 27135400 DOI: 10.1038/ng.3561] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 04/06/2016] [Indexed: 11/08/2022]
Abstract
Sequence variants affecting blood lipids and coronary artery disease (CAD) may enhance understanding of the atherogenicity of lipid fractions. Using a large resource of whole-genome sequence data, we examined rare and low-frequency variants for association with non-HDL cholesterol, HDL cholesterol, LDL cholesterol, and triglycerides in up to 119,146 Icelanders. We discovered 13 variants with large effects (within ANGPTL3, APOB, ABCA1, NR1H3, APOA1, LIPC, CETP, LDLR, and APOC1) and replicated 14 variants. Five variants within PCSK9, APOA1, ANGPTL4, and LDLR associate with CAD (33,090 cases and 236,254 controls). We used genetic risk scores for the lipid fractions to examine their causal relationship with CAD. The non-HDL cholesterol genetic risk score associates most strongly with CAD (P = 2.7 × 10(-28)), and no other genetic risk score associates with CAD after accounting for non-HDL cholesterol. The genetic risk score for non-HDL cholesterol confers CAD risk beyond that of LDL cholesterol (P = 5.5 × 10(-8)), suggesting that targeting atherogenic remnant cholesterol may reduce cardiovascular risk.
Collapse
|
39
|
Lee-Rueckert M, Escola-Gil JC, Kovanen PT. HDL functionality in reverse cholesterol transport--Challenges in translating data emerging from mouse models to human disease. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:566-83. [PMID: 26968096 DOI: 10.1016/j.bbalip.2016.03.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
Abstract
Whereas LDL-derived cholesterol accumulates in atherosclerotic lesions, HDL particles are thought to facilitate removal of cholesterol from the lesions back to the liver thereby promoting its fecal excretion from the body. Because generation of cholesterol-loaded macrophages is inherent to atherogenesis, studies on the mechanisms stimulating the release of cholesterol from these cells and its ultimate excretion into feces are crucial to learn how to prevent lesion development or even induce lesion regression. Modulation of this key anti-atherogenic pathway, known as the macrophage-specific reverse cholesterol transport, has been extensively studied in several mouse models with the ultimate aim of applying the emerging knowledge to humans. The present review provides a detailed comparison and critical analysis of the various steps of reverse cholesterol transport in mouse and man. We attempt to translate this in vivo complex scenario into practical concepts, which could serve as valuable tools when developing novel HDL-targeted therapies.
Collapse
|
40
|
Lisik MZ, Gutmajster E, Sieroń AL. Low Levels of HDL in Fragile X Syndrome Patients. Lipids 2016; 51:189-92. [PMID: 26712713 PMCID: PMC4735238 DOI: 10.1007/s11745-015-4109-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/28/2015] [Indexed: 01/08/2023]
Abstract
Fragile X syndrome (FXS) is the most common form of familial mental retardation and one of the leading known causes of autism. The mutation responsible for FXS is a large expansion of the CGG repeats in the promoter region of the FMR1 gene resulting in the transcriptional silencing of the gene in the pathophysiology of Fragile X syndrome was hypothesized. 23 male patients affected by Fragile X syndrome (full mutation in the FMR1 gene) and 24 controls were included in the study. The serum levels of HDL-C were lower in FXS patients (p < 0.001). The serum levels triacylglycerols were higher in FXS patients (p = 0.007) Further study involving larger samples are necessary to confirm the results and define the health implications for abnormal lipid levels in FXS patients.
Collapse
Affiliation(s)
- Małgorzata Z Lisik
- Department of Molecular Biology and Genetics, School of Medicine in Katowice ul., Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland.
| | - Ewa Gutmajster
- Department of Molecular Biology and Genetics, School of Medicine in Katowice ul., Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Aleksander L Sieroń
- Department of Molecular Biology and Genetics, School of Medicine in Katowice ul., Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| |
Collapse
|
41
|
Liu D, Zhang M, Xie W, Lan G, Cheng HP, Gong D, Huang C, Lv YC, Yao F, Tan YL, Li L, Zheng XL, Tang CK. MiR-486 regulates cholesterol efflux by targeting HAT1. Biochem Biophys Res Commun 2015; 472:418-24. [PMID: 26654953 DOI: 10.1016/j.bbrc.2015.11.128] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/27/2015] [Indexed: 12/21/2022]
Abstract
RATIONALE Excessive cholesterol accumulation in macrophages is a major factor of foam cell formation and development of atherosclerosis. Previous studies suggested that miR-486 plays an important role in cardiovascular diseases, but the underlying mechanism is still unknown. OBJECTIVE The purpose of this study is to determine whether miR-486 regulates ATP-binding cassette transporter A1 (ABCA1) mediated cholesterol efflux, and also explore the underlying mechanism. METHODS AND RESULTS Based on bioinformatics analysis and luciferase reporter assay, we transfected miR-486 mimic and miR-486 inhibitor into THP-1 macrophage-derived foam cells, and found that miR-486 directly bound to histone acetyltransferase-1 (HAT1) 3'UTR, and downregulated its mRNA and protein expression. In addition, our studies through transfection with wildtype HAT1 or shHAT1 (short hairpin HAT1) revealed that HAT1 could promote the expression of ABCA1 at both mRNA and protein levels. At the same time, the acetylation levels of the lysines 5 and 12 of histone H4 were upregulated after overexpression with HAT1. Meanwhile, the results of liquid scintillation counter and high performance liquid chromatography (HPLC) showed that miR-486 promoted cholesterol accumulation in THP-1 macrophages. CONCLUSION These data indicated that miR-486 aggravate the cholesterol accumulation in THP-1 cells by targeting HAT1.
Collapse
Affiliation(s)
- Dan Liu
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Min Zhang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Wei Xie
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Gang Lan
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Hai-Peng Cheng
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Duo Gong
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Chong Huang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Yun-Cheng Lv
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Feng Yao
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Yu-Lin Tan
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Liang Li
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary T2N 4N1, Alberta, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
42
|
Gomez-Delgado F, Garcia-Rios A, Alcala-Diaz JF, Rangel-Zuñiga O, Delgado-Lista J, Yubero-Serrano EM, Lopez-Moreno J, Tinahones FJ, Ordovas JM, Garaulet M, Lopez-Miranda J, Perez-Martinez P. Chronic consumption of a low-fat diet improves cardiometabolic risk factors according to theCLOCKgene in patients with coronary heart disease. Mol Nutr Food Res 2015; 59:2556-64. [DOI: 10.1002/mnfr.201500375] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 08/14/2015] [Accepted: 08/18/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Francisco Gomez-Delgado
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Antonio Garcia-Rios
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Juan Francisco Alcala-Diaz
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Oriol Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Elena M. Yubero-Serrano
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Javier Lopez-Moreno
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Francisco Jose Tinahones
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
- Biomedical Research Laboratory; Endocrinology Department; Hospital Virgen de la Victoria; Malaga Spain
| | - Jose M. Ordovas
- Nutrition and Genomics Laboratory; J.M.-US Department of Agriculture Human Nutrition Research Center on Aging at Tufts University; Boston MA USA
| | - Marta Garaulet
- Department of physiology, University of Murcia, IMIB; Murcia Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, IMIBIC; Reina Sofia University Hospital; University of Cordoba; Cordoba Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| |
Collapse
|
43
|
Huang LH, Elvington A, Randolph GJ. The role of the lymphatic system in cholesterol transport. Front Pharmacol 2015; 6:182. [PMID: 26388772 PMCID: PMC4557107 DOI: 10.3389/fphar.2015.00182] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/12/2015] [Indexed: 11/13/2022] Open
Abstract
Reverse cholesterol transport (RCT) is the pathway for removal of peripheral tissue cholesterol and involves transport of cholesterol back to liver for excretion, starting from cellular cholesterol efflux facilitated by lipid-free apolipoprotein A1 (ApoA1) or other lipidated high-density lipoprotein (HDL) particles within the interstitial space. Extracellular cholesterol then is picked up and transported through the lymphatic vasculature before entering into bloodstream. There is increasing evidence supporting a role for enhanced macrophage cholesterol efflux and RCT in ameliorating atherosclerosis, and recent data suggest that these processes may serve as better diagnostic biomarkers than plasma HDL levels. Hence, it is important to better understand the processes governing ApoA1 and HDL influx into peripheral tissues from the bloodstream, modification and facilitation of cellular cholesterol removal within the interstitial space, and transport through the lymphatic vasculature. New findings will complement therapeutic strategies for the treatment of atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Li-Hao Huang
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO, USA
| | - Andrew Elvington
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO, USA
| |
Collapse
|