1
|
Xu Z, Yang S, Cui L. Understanding the heterogeneity and dysfunction of HDL in chronic kidney disease: insights from recent reviews. BMC Nephrol 2024; 25:400. [PMID: 39511510 PMCID: PMC11542271 DOI: 10.1186/s12882-024-03808-3] [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: 06/25/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Chronic kidney disease (CKD) is a complex disease that affects the global population's health, with dyslipidemia being one of its major complications. High density lipoprotein (HDL) is regarded as the "hero" in the bloodstream due to its role in reverse cholesterol transport, which lowers cholesterol levels in the blood and prevents atherosclerosis. However, in the complex internal environment of CKD, even this "hero" may struggle to perform its beneficial functions and could potentially become harmful. This article reviews HDL heterogeneity, HDL subclasses, functional changes in HDL during the progression of CKD, and the application of HDL in CKD treatment. This review aims to deepen understanding of lipid metabolism abnormalities in CKD patients and provide a basis for new therapeutic strategies.
Collapse
Affiliation(s)
- Zhen Xu
- Peking University Third Hospital, Beijing, China
| | - Shuo Yang
- Peking University Third Hospital, Beijing, China
| | - Liyan Cui
- Peking University Third Hospital, Beijing, China.
| |
Collapse
|
2
|
Aydınyılmaz F, Özbeyaz NB, Guliyev İ, Algül E, Şahan HF, Kalkan K. Effect of Atherogenic Index of Plasma on Pre-Percutaneous Coronary Intervention Thrombolysis in Myocardial Infarction Flow in Patients With ST Elevation Myocardial Infarction. Angiology 2024; 75:841-848. [PMID: 37399526 DOI: 10.1177/00033197231185204] [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] [Indexed: 07/05/2023]
Abstract
Dyslipidemia is an important risk factor for cardiovascular morbidity and mortality. Although low-density lipoprotein (LDL) is primarily responsible, the importance of triglyceride (TG) and high-density lipoprotein (HDL) has also been recognized. The present study investigated the effect of the atherogenic index of plasma (AIP), in which atherogenic and protective lipoproteins were evaluated together, on the initial flow in patients with ST elevation myocardial infarction. AIP was calculated as log(TG/HDL-cholesterol). Patients included in the study (n = 1535) were divided into Thrombolysis in Myocardial Infarction (TIMI) flow grade 0 and >0. AIP was found to be significantly different between 2 groups (.55 ± .23 vs .67 ± .21; P < .001). AIP was an independent predictor for pre-intervention TIMI flow (Odds Ratio: 2.778). A moderate correlation was found between TIMI frame count measurements, calculated in patients with TIMI 2-3, and AIP (Pearson correlation coefficient: .63, P < .001). In the receiver operating characteristic analysis, AIP showed the highest area under curve (AUC) compared with other lipid parameters for predicting vascular patency. The AUC of AIP was .634, the cut-off value was .59, and the sensitivity and specificity were 67.6% and 68.4%, respectively (P < .001). In conclusion, AIP was found to be an important marker affecting pre-percutaneous coronary intervention TIMI flow.
Collapse
Affiliation(s)
- Faruk Aydınyılmaz
- Department of Cardiology, University of Health Sciences, Erzurum Bolge Training and Research Hospital, Erzurum, Turkey
| | | | - İlkin Guliyev
- Department of Cardiology, Gümüşhane State Hospital, Gümüşhane, Turkey
| | - Engin Algül
- Department of Cardiology, University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| | - Haluk Furkan Şahan
- Department of Cardiology, University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| | - Kamuran Kalkan
- Department of Cardiology, University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
3
|
Liang L, Deng Y, Ao Z, Liao C, Tian J, Li C, Yu X. Recent progress in biomimetic nanomedicines based on versatile targeting strategy for atherosclerosis therapy. J Drug Target 2024; 32:606-623. [PMID: 38656224 DOI: 10.1080/1061186x.2024.2347353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis (AS) is considered to be one of the major causes of cardiovascular disease. Its pathological microenvironment is characterised by increased production of reactive oxygen species, lipid oxides, and excessive inflammatory factors, which accumulate at the monolayer endothelial cells in the vascular wall to form AS plaques. Therefore, intervention in the pathological microenvironment would be beneficial in delaying AS. Researchers have designed biomimetic nanomedicines with excellent biocompatibility and the ability to avoid being cleared by the immune system through different therapeutic strategies to achieve better therapeutic effects for the characteristics of AS. Biomimetic nanomedicines can further enhance delivery efficiency and improve treatment efficacy due to their good biocompatibility and ability to evade clearance by the immune system. Biomimetic nanomedicines based on therapeutic strategies such as neutralising inflammatory factors, ROS scavengers, lipid clearance and integration of diagnosis and treatment are versatile approaches for effective treatment of AS. The review firstly summarises the targeting therapeutic strategy of biomimetic nanomedicine for AS in recent 5 years. Biomimetic nanomedicines using cell membranes, proteins, and extracellular vesicles as carriers have been developed for AS.
Collapse
Affiliation(s)
- Lijuan Liang
- Department of Pharmacy, Hejiang County People's Hospital, Luzhou, Sichuan, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Zuojin Ao
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Changli Liao
- Science and Technology Department, Southwest Medical University, Luzhou, Sichuan, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Zhang Y, Luo S, Gao Y, Tong W, Sun S. High-Density Lipoprotein Subfractions Remodeling: A Critical Process for the Treatment of Atherosclerotic Cardiovascular Diseases. Angiology 2024; 75:441-453. [PMID: 36788038 DOI: 10.1177/00033197231157473] [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] [Indexed: 02/16/2023]
Abstract
Numerous studies have shown that a low level of high-density lipoprotein cholesterol (HDL-C) is an independent biomarker of cardiovascular disease. High-density lipoprotein (HDL) is considered to be a protective factor for atherosclerosis (AS). Therefore, raising HDL-C has been widely recognized as a promising strategy to treat atherosclerotic cardiovascular diseases (ASCVD). However, several studies have found that increasing HDL-C levels does not necessarily reduce the risk of ASCVD. HDL particles are highly heterogeneous in structure, composition, and biological function. Moreover, HDL particles from atherosclerotic patients exhibit impaired anti-atherogenic functions and these dysfunctional HDL particles might even promote ASCVD. This makes it uncertain that HDL-raising therapy will prevent and treat ASCVD. It is necessary to comprehensively analyze the structure and function of HDL subfractions. We review current advances related to HDL subfractions remodeling and highlight how current lipid-modifying drugs such as niacin, statins, fibrates, and cholesteryl ester transfer protein inhibitors regulate cholesterol concentration of HDL and specific HDL subfractions.
Collapse
Affiliation(s)
- Yaling Zhang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Shiyu Luo
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Yi Gao
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, China
| | - Shaowei Sun
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
5
|
Kim HJ, Park SK, Park SH, Lee YG, Park JH, Hwang JT, Chung MY. Schisandrin A in Schisandra chinensis Upregulates the LDL Receptor by Inhibiting PCSK9 Protein Stabilization in Steatotic Model. J Microbiol Biotechnol 2024; 34:425-435. [PMID: 37997262 PMCID: PMC10940739 DOI: 10.4014/jmb.2306.06049] [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: 06/28/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/25/2023]
Abstract
Schisandra chinensis extract (SCE) protects against hypocholesterolemia by inhibiting proprotein convertase subtilisin/kexin 9 (PCSK9) protein stabilization. We hypothesized that the hypocholesterolemic activity of SCE can be attributable to upregulation of the PCSK9 inhibition-associated low-density lipoprotein receptor (LDLR). Male mice were fed a low-fat diet or a Western diet (WD) containing SCE at 1% for 12 weeks. WD increased final body weight and blood LDL cholesterol levels as well as alanine transaminase and aspartate aminotransferase expression. However, SCE supplementation significantly attenuated the increase in blood markers caused by WD. SCE also attenuated WD-mediated increases in hepatic LDLR protein expression in the obese mice. In addition, SCE increased LDLR protein expression and attenuated cellular PCSK9 levels in HepG2 cells supplemented with delipidated serum (DLPS). Non-toxic concentrations of schisandrin A (SA), one of the active components of SCE, significantly increased LDLR expression and tended to decrease PCSK9 protein levels in DLPS-treated HepG2 cells. High levels of SA-mediated PCSK9 attenuation was not attributable to reduced PCSK9 gene expression, but was associated with free PCSK9 protein degradation in this cell model. Our findings show that PCSK9 secretion can be significantly reduced by SA treatment, contributing to reductions in free cholesterol levels.
Collapse
Affiliation(s)
- Hyo-Jin Kim
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
- Department of Food Biotechnology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Seon Kyeong Park
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Soo Hyun Park
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Yu Geon Lee
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Jae-Ho Park
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Jin-Taek Hwang
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Min-Yu Chung
- Department of Food and Nutrition, Gangseo University, Seoul 07661, Republic of Korea
| |
Collapse
|
6
|
Rosú SA, Aguilar J, Urbano BF, Tarraga WA, Ramella NA, Longo GS, Finarelli GS, Sanchez Donoso SA, Tricerri MA. Interactions of variants of human apolipoprotein A-I with biopolymeric model matrices. Effect of collagen and heparin. Arch Biochem Biophys 2023; 750:109805. [PMID: 37913855 DOI: 10.1016/j.abb.2023.109805] [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: 08/10/2023] [Revised: 10/05/2023] [Accepted: 10/30/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND The extracellular matrix (ECM) is a complex tridimensional scaffold that actively participates in physiological and pathological events. The objective of this study was to test whether structural proteins of the ECM and glycosaminoglycans (GAGs) may favor the retention of human apolipoprotein A-I (apoA-I) variants associated with amyloidosis and atherosclerosis. METHODS Biopolymeric matrices containing collagen type I (Col, a main macromolecular component of the ECM) with or without heparin (Hep, a model of GAGs) were constructed and characterized, and used to compare the binding of apoA-I having the native sequence (Wt) or Arg173Pro, a natural variant inducing cardiac amyloidosis. Protein binding was observed by fluorescence microscopy and unbound proteins quantified by a colorimetric assay. RESULTS Both, Wt and Arg173Pro bound to the scaffolds containing Col, but the presence of Hep diminished the binding efficiency. Col-Hep matrices retained Arg173Pro more than the Wt. The retained protein was only partially removed from the matrices with saline solutions, indicating that electrostatic interactions may occur but are not the main driving force. Using in addition thermodynamic molecular simulations and size exclusion chromatography approaches, we suggest that the binding of apoA-I variants to the biopolymeric matrices is driven by many low affinity interactions. CONCLUSIONS Under this scenario Col-Hep scaffolds contribute to the binding of Arg173Pro, as a cooperative platform which could modify the native protein conformation affecting protein folding. GENERAL SIGNIFICANCE We show that the composition of the ECM is key to the protein retention, and well characterized biosynthetic matrices offer an invaluable in vitro model to mimic the hallmark of pathologies with interstitial infiltration such as cardiac amyloidosis.
Collapse
Affiliation(s)
- Silvana A Rosú
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET. Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Buenos Aires, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Joao Aguilar
- Laboratorio de Interacciones Macromoleculares (LIMM), Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Bruno F Urbano
- Laboratorio de Interacciones Macromoleculares (LIMM), Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Wilson A Tarraga
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Universidad Nacional de La Plata-CONICET, La Plata, Buenos Aires, Argentina
| | - Nahuel A Ramella
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET. Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Buenos Aires, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Gabriel S Longo
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Universidad Nacional de La Plata-CONICET, La Plata, Buenos Aires, Argentina
| | - Gabriela S Finarelli
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET. Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Buenos Aires, Argentina
| | - Susana A Sanchez Donoso
- Laboratorio de Interacciones Macromoleculares (LIMM), Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile.
| | - M Alejandra Tricerri
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET. Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Buenos Aires, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
7
|
Yaghmur A, Østergaard J, Mu H. Lipid nanoparticles for targeted delivery of anticancer therapeutics: Recent advances in development of siRNA and lipoprotein-mimicking nanocarriers. Adv Drug Deliv Rev 2023; 203:115136. [PMID: 37944644 DOI: 10.1016/j.addr.2023.115136] [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: 10/19/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
The limitations inherent in conventional cancer treatment methods have stimulated recent efforts towards the design of safe nanomedicines with high efficacy for combating cancer through various promising approaches. A plethora of nanoparticles has been introduced in the development of cancer nanomedicines. Among them, different lipid nanoparticles are attractive for use due to numerous advantages and unique opportunities, including biocompatibility and targeted drug delivery. However, a comprehensive understanding of nano-bio interactions is imperative to facilitate the translation of recent advancements in the development of cancer nanomedicines into clinical practice. In this contribution, we focus on lipoprotein-mimicking nanoparticles, which possess unique features and compositions facilitating drug transport through receptor binding mechanisms. Additionally, we describe potential applications of siRNA lipid nanoparticles in the future design of anticancer nanomedicines. Thus, this review highlights recent progress, challenges, and opportunities of lipid-based lipoprotein-mimicking nanoparticles and siRNA nanocarriers designed for the targeted delivery of anticancer therapeutic agents.
Collapse
Affiliation(s)
- Anan Yaghmur
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Jesper Østergaard
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Huiling Mu
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
8
|
Dastmalchi LN, German CA, Taub PR. High density lipoprotein: When to rethink too much of a good thing. Am J Prev Cardiol 2023; 15:100511. [PMID: 37434863 PMCID: PMC10331407 DOI: 10.1016/j.ajpc.2023.100511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023] Open
Abstract
High density lipoprotein cholesterol (HDL-C) is a known contributor to atherosclerotic cardiovascular disease (ASCVD) risk when HDL-C <40 mg/dL in men and <50 mg/dL in women. There has been much interest in the potential cardioprotective properties of HDL-C, as it removes cholesterol from the periphery to the liver for exertion and holds inherent anti-thrombotic and anti-inflammatory properties. However, clinical trials raising HDL-C pharmacologically have not shown to improve cardiovascular outcomes. In fact, observational studies have demonstrated an increased risk of non-cardiovascular mortality and infection when HDL-C >90 mg/dL and >70 mg/dL in women and men, respectively. The ability for the HDL particle to effectively transport cholesterol from the periphery for excretion in bile is more complex than illustrated on a standard cholesterol panel. There is variability in its function, size, density, subclass, reverse cholesterol transport, and cholesterol efflux capacity, which impact the particles ability to effectively reduce cardiovascular disease (CVD) risk. Research has shown that HDL particles are prone to have a reduction in its efficacy in response to infection, auto-immune disease, menopause and cardiometabolic conditions during pregnancy. Additionally, recent studies have shown that low HDL-C may not adequately influence ASCVD risk in Black adults. The purpose of this contemporary review is to highlight the utility of using HDL-C in assessing CVD risk.
Collapse
Affiliation(s)
- Lily N. Dastmalchi
- Section of Cardiology, Department of Medicine, Temple University Hospital, Philadelphia, PA, USA
| | - Charles A. German
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Pam R. Taub
- Section of Cardiology, Department of Medicine, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
9
|
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: 3] [Impact Index Per Article: 3.0] [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
|
10
|
Effects of Ethanolic and Aqueous Extracts of Garcinia gardneriana Leaves in an In Vivo Experimental Model Induced by a Hyperlipidic Diet. Nutrients 2023; 15:nu15061308. [PMID: 36986038 PMCID: PMC10051817 DOI: 10.3390/nu15061308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 03/09/2023] Open
Abstract
The study of medicinal plants, such as the genus Garcinia (Clusiaceae), in the treatment of non-communicable chronic diseases has aroused the interest of researchers. However, there are no studies in the literature that have investigated the effects of Garcinia gardneriana in experimental models of obesity for possible metabolic alterations. Swiss mice receiving a high-fat diet were supplemented with aqueous or ethanolic extract of G. gardneriana at doses of 200 or 400 mg/kg/day. It was found that there was a reduction in food consumption in experimental groups compared with the control groups, and the group supplemented with aqueous extract at a dose of 200 mg/kg/daydisplayed a reduction in weight. The results showed an increase in the values of high density lipoprotein (HDL-c), total cholesterol, triglycerides and fasting blood glucose. G. gardneriana did not protect against insulin resistance, and caused in an increase in monocyte chemoattractant protein-1 (MCP-1) concentrations and a reduction in interleukin 10 (IL-10). In addition, hepatic steatosis and microvesicular steatosis were indicated. It was revealed that, under the experimental conditions in the study, G. gardneriana did not prevent weight gain or comorbidities; that is, a different behavior was obtained from that described in the literature with regard to the medicinal potential of the Garcinia species, which is probably related to the phytochemical properties.
Collapse
|
11
|
Yuan W, Ernst K, Kuai R, Morin EE, Yu M, Sviridov DO, Tang J, Mei L, Li D, Ackermann R, Remaley AT, Schwendeman A. Systematic evaluation of the effect of different apolipoprotein A-I mimetic peptides on the performance of synthetic high-density lipoproteins in vitro and in vivo. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102646. [PMID: 36549559 DOI: 10.1016/j.nano.2022.102646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Synthetic high-density lipoproteins nanomedicine (sHDL) composed of apolipoprotein A-I (ApoA-I) mimetic peptides and lipids have shown very promising results for the treatment of various cardiovascular diseases. Numerous efforts have also been made to design different ApoA-I mimetic peptides to improve the potency of sHDL, especially the efficiency of reverse cholesterol transport. However, the way in which ApoA-I mimetic peptides affect the properties of sHDL, including stability, cholesterol efflux, cholesterol esterification, elimination in vivo, and the relationship of these properties, is still poorly understood. Revealing the effect of these factors on the potency of sHDL is important for the design of better ApoA-I mimetic peptides. In this study, three widely used ApoA-I mimetic peptides with different sequences, lengths, LCAT activation and lipid binding affinities were used for the preparation of sHDL and were evaluated in terms of physical/chemical properties, cholesterol efflux, cholesterol esterification, remodeling, and pharmacokinetics/pharmacodynamics. Our results showed that ApoA-I mimetic peptides with the highest cholesterol efflux and cholesterol esterification in vitro did not exhibit the highest cholesterol mobilization in vivo. Further analysis indicated that other factors, such as pharmacokinetics and remodeling of sHDL, need to be considered in order to predict the efficiency of cholesterol mobilization in vivo. Thus, our study highlights the importance of using the overall performance, rather than in vitro results alone, as the blueprint for the design and optimization of ApoA-I mimetic peptides.
Collapse
Affiliation(s)
- Wenmin Yuan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Kelsey Ernst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Rui Kuai
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Emily E Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Denis O Sviridov
- National Heart, Lung and Blood Institute, National Institutes of Health, Building 10 - 2C433, 10 Center Drive, MSC 1666, Bethesda, MD 20892, United States of America
| | - Jie Tang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Ling Mei
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Dan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Rose Ackermann
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America
| | - Alan T Remaley
- National Heart, Lung and Blood Institute, National Institutes of Health, Building 10 - 2C433, 10 Center Drive, MSC 1666, Bethesda, MD 20892, United States of America
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America; Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, United States of America; Biointerfaces Institute, University of Michigan, NCRC, 2800 Plymouth Road, Ann Arbor, MI 48109, United States of America.
| |
Collapse
|
12
|
Johansen NT, Tidemand FG, Pedersen MC, Arleth L. Travel light: Essential packing for membrane proteins with an active lifestyle. Biochimie 2023; 205:3-26. [PMID: 35963461 DOI: 10.1016/j.biochi.2022.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/29/2022] [Accepted: 07/23/2022] [Indexed: 11/02/2022]
Abstract
We review the considerable progress during the recent decade in the endeavours of designing, optimising, and utilising carrier particle systems for structural and functional studies of membrane proteins in near-native environments. New and improved systems are constantly emerging, novel studies push the perceived limits of a given carrier system, and specific carrier systems consolidate and entrench themselves as the system of choice for particular classes of target membrane protein systems. This review covers the most frequently used carrier systems for such studies and emphasises similarities and differences between these systems as well as current trends and future directions for the field. Particular interest is devoted to the biophysical properties and membrane mimicking ability of each system and the manner in which this may impact an embedded membrane protein and an eventual structural or functional study.
Collapse
Affiliation(s)
- Nicolai Tidemand Johansen
- Section for Transport Biology, Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, Frederiksberg C, 1871, Denmark.
| | - Frederik Grønbæk Tidemand
- Section for Transport Biology, Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, Frederiksberg C, 1871, Denmark
| | - Martin Cramer Pedersen
- Condensed Matter Physics, Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, Copenhagen E, 2100, Denmark
| | - Lise Arleth
- Condensed Matter Physics, Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, Copenhagen E, 2100, Denmark
| |
Collapse
|
13
|
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: 6.0] [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
|
14
|
Bond P, Smit DL, de Ronde W. Anabolic-androgenic steroids: How do they work and what are the risks? Front Endocrinol (Lausanne) 2022; 13:1059473. [PMID: 36644692 PMCID: PMC9837614 DOI: 10.3389/fendo.2022.1059473] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Anabolic-androgenic steroids (AAS) are a class of hormones that are widely abused for their muscle-building and strength-increasing properties in high, nontherapeutic, dosages. This review provides an up-to-date and comprehensive overview on how these hormones work and what side effects they might elicit. We discuss how AAS are absorbed into the circulation after intramuscular injection or oral ingestion and how they are subsequently transported to the tissues, where they will move into the extravascular compartment and diffuse into their target cells. Inside these cells, AAS can biotransform into different metabolites or bind to their cognate receptor: the androgen receptor. AAS and their metabolites can cause side effects such as acne vulgaris, hypertension, hepatotoxicity, dyslipidemia, testosterone deficiency, erectile dysfunction, gynecomastia, and cardiomyopathy. Where applicable, we mention treatment options and self-medication practices of AAS users to counteract these side effects. Clinicians may use this review as a guide for understanding how AAS use can impact health and to assist in patient education and, in some cases, the management of side effects.
Collapse
Affiliation(s)
| | - Diederik L. Smit
- Department of Internal Medicine, Elisabeth-TweeSteden Hospital, Tilburg, Netherlands
| | - Willem de Ronde
- Department of Internal Medicine, Spaarne Gasthuis, Haarlem, Netherlands
| |
Collapse
|
15
|
Gao P, Wen X, Ou Q, Zhang J. Which one of LDL-C /HDL-C ratio and non-HDL-C can better predict the severity of coronary artery disease in STEMI patients. BMC Cardiovasc Disord 2022; 22:318. [PMID: 35843962 PMCID: PMC9288699 DOI: 10.1186/s12872-022-02760-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
Background The increase of low-density lipoprotein cholesterol (LDL-C) is widely accepted as an important factor in the occurrence of atherosclerosis. In recent years, the guidelines have recommended non-high density lipoprotein cholesterol (non-HDL-C) as a secondary target for lipid-lowering therapy. But even as research on the relationship between LDL-C/HDL-C and atherosclerosis increases, it is still undetermined which index is most closely related to the severity of acute ST-segment elevation myocardial infarction (STEMI).
Methods 901 patients who received coronary angiography due to chest pain were selected. Among them, 772 patients with STEMI represented the test group, and 129 patients with basically normal coronary angiography represented the control group. Researchers measured fasting blood lipids and other indicators after admission, and determined the severity of coronary artery disease using the Gensini score. Results LDL-C/HDL-C and non-HDL-C indexes were statistically different between the two patient groups. In the test group, total cholesterol (TC), triglycerides (TG), LDL-C, high density lipoprotein cholesterol (HDL-C), non-HDL-C, arteriosclerosis index (AI), and LDL-C/HDL-C all correlated with the patients' Gensini score. After applying the stepwise method of multiple linear regression analysis (R2 = 0.423, β = 0.518, p < 0.05), LDL-C/HDL-C had the most correlation with the patient's Gensini score. ROC curve analysis suggested that LDL-C/HDL-C can predict whether patients with chest pain are STEMI (AUC: 0.880, 95% Cl: 0.847–0.912, p < 0.05). When cutoff value is 2.15, sensitivity is 0.845, and specificity is 0.202, LDL-C/HDL-C is an effective indicator for predicting whether patients with chest pain have STEMI. Conclusion Compared to ratios of non-HDL-C and LDL-C, the LDL-C/HDL-C ratio in patients with STEMI is more correlated with the severity of coronary artery disease. It can better evaluate the severity of coronary artery disease and better predict whether patients with chest pain are STEMI. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02760-0.
Collapse
|
16
|
Hayakawa S, Tamura A, Nikiforov N, Koike H, Kudo F, Cheng Y, Miyazaki T, Kubekina M, Kirichenko TV, Orekhov AN, Yui N, Manabe I, Oishi Y. Activated cholesterol metabolism is integral for innate macrophage responses by amplifying Myd88 signaling. JCI Insight 2022; 7:138539. [PMID: 36509286 DOI: 10.1172/jci.insight.138539] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/07/2022] [Indexed: 11/22/2022] Open
Abstract
Recent studies have shown that cellular metabolism is tightly linked to the regulation of immune cells. Here, we show that activation of cholesterol metabolism, involving cholesterol uptake, synthesis, and autophagy/lipophagy, is integral to innate immune responses in macrophages. In particular, cholesterol accumulation within endosomes and lysosomes is a hallmark of the cellular cholesterol dynamics elicited by Toll-like receptor 4 activation and is required for amplification of myeloid differentiation primary response 88 (Myd88) signaling. Mechanistically, Myd88 binds cholesterol via its CLR recognition/interaction amino acid consensus domain, which promotes the protein's self-oligomerization. Moreover, a novel supramolecular compound, polyrotaxane (PRX), inhibited Myd88‑dependent inflammatory macrophage activation by decreasing endolysosomal cholesterol via promotion of cholesterol trafficking and efflux. PRX activated liver X receptor, which led to upregulation of ATP binding cassette transporter A1, thereby promoting cholesterol efflux. PRX also inhibited atherogenesis in Ldlr-/- mice. In humans, cholesterol levels in circulating monocytes correlated positively with the severity of atherosclerosis. These findings demonstrate that dynamic changes in cholesterol metabolism are mechanistically linked to Myd88‑dependent inflammatory programs in macrophages and support the notion that cellular cholesterol metabolism is integral to innate activation of macrophages and is a potential therapeutic and diagnostic target for inflammatory diseases.
Collapse
Affiliation(s)
- Sumio Hayakawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Atsushi Tamura
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nikita Nikiforov
- National Medical Research Center of Cardiology, Institute of Experimental Cardiology, Moscow, Russia.,Institute of Gene Biology, Centre of Collective Usage, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Hiroyuki Koike
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Fujimi Kudo
- Department of Systems Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yinglan Cheng
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Tokyo, Japan
| | - Marina Kubekina
- Institute of Gene Biology, Centre of Collective Usage, Moscow, Russia
| | - Tatiana V Kirichenko
- National Medical Research Center of Cardiology, Institute of Experimental Cardiology, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia.,Institute for Atherosclerosis Research, Moscow, Russia
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Manabe
- Department of Systems Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yumiko Oishi
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
17
|
Hu PP, Luo SX, Fan XQ, Li D, Tong XY. Macrophage-targeted nanomedicine for the diagnosis and management of atherosclerosis. Front Pharmacol 2022; 13:1000316. [PMID: 36160452 PMCID: PMC9501673 DOI: 10.3389/fphar.2022.1000316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular diseases, such as myocardial infarction and stroke, which account for the highest death toll worldwide. Macrophage is the major contributor to atherosclerosis progression, and therefore, macrophage-associated pathological process is considered an extremely important target for the diagnosis and treatment of atherosclerosis. However, the existing clinical strategies still have many bottlenecks and challenges in atherosclerosis’s early detection and management. Nanomedicine, using various nanoparticles/nanocarriers for medical purposes, can effectively load therapeutic agents, significantly improve their stability and accurately deliver them to the atherosclerotic plaques. In this review, we summarized the latest progress of the macrophage-targeted nanomedicine in the diagnosis and treatment of atherosclerosis, and their potential applications and clinical benefits are also discussed.
Collapse
Affiliation(s)
- Ping Ping Hu
- Chongqing Engineering Research Center for Pharmacodynamics Evaluation, College of Pharmacy, Chongqing Medical University, Chongqing, China
- *Correspondence: Ping Ping Hu, ; Xiao Yong Tong,
| | - Shuang Xue Luo
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiao Qing Fan
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Di Li
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Yong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
- *Correspondence: Ping Ping Hu, ; Xiao Yong Tong,
| |
Collapse
|
18
|
Oberle R, Kührer K, Österreicher T, Weber F, Steinbauer S, Udonta F, Wroblewski M, Ben-Batalla I, Hassl I, Körbelin J, Unseld M, Jauhiainen M, Plochberger B, Röhrl C, Hengstschläger M, Loges S, Stangl H. The HDL particle composition determines its antitumor activity in pancreatic cancer. Life Sci Alliance 2022; 5:e202101317. [PMID: 35577388 PMCID: PMC9112193 DOI: 10.26508/lsa.202101317] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 12/03/2022] Open
Abstract
Despite enormous efforts to improve therapeutic options, pancreatic cancer remains a fatal disease and is expected to become the second leading cause of cancer-related deaths in the next decade. Previous research identified lipid metabolic pathways to be highly enriched in pancreatic ductal adenocarcinoma (PDAC) cells. Thereby, cholesterol uptake and synthesis promotes growth advantage to and chemotherapy resistance for PDAC tumor cells. Here, we demonstrate that high-density lipoprotein (HDL)-mediated efficient cholesterol removal from cancer cells results in PDAC cell growth reduction and induction of apoptosis in vitro. This effect is driven by an HDL particle composition-dependent interaction with SR-B1 and ABCA1 on cancer cells. AAV-mediated overexpression of APOA1 and rHDL injections decreased PDAC tumor development in vivo. Interestingly, plasma samples from pancreatic-cancer patients displayed a significantly reduced APOA1-to-SAA1 ratio and a reduced cholesterol efflux capacity compared with healthy donors. We conclude that efficient, HDL-mediated cholesterol depletion represents an interesting strategy to interfere with the aggressive growth characteristics of PDAC.
Collapse
Affiliation(s)
- Raimund Oberle
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Kristina Kührer
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Tamina Österreicher
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Florian Weber
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Linz, Austria
| | - Stefanie Steinbauer
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Wels, Austria
| | - Florian Udonta
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mark Wroblewski
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabel Ben-Batalla
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ingrid Hassl
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Jakob Körbelin
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Unseld
- Department of Medicine I, Division of Palliative Medicine, Medical University of Vienna, Vienna, Austria
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research and Finnish Institute for Health and Welfare, Genomics and Biobank Unit, Biomedicum 2U, Helsinki, Finland
| | - Birgit Plochberger
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Linz, Austria
| | - Clemens Röhrl
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Wels, Austria
| | - Markus Hengstschläger
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Sonja Loges
- Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Herbert Stangl
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Guan R, Li H, Jiao Y, Yu H. A nonrandomized controlled trial of individualized exercise prescription combined with remote exercise management in patients who are overweight or obese. BMC Sports Sci Med Rehabil 2022; 14:99. [PMID: 35655279 PMCID: PMC9161188 DOI: 10.1186/s13102-022-00479-1] [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: 06/27/2021] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background Between 2012 and 2020, the obesity rate increased among Chinese people aged 18 years and above, along with blood pressure, fasting blood glucose, serum total cholesterol, and triglycerides. Purpose Our objective was to compare the effects of a combined intervention, including individualized exercise prescription plus remote management versus individualized exercise prescription only, on cardiovascular risk factors in patients who are overweight or obese, with the aim of establishing a more effective remote model of health management than self-management. Methods This nonrandomized controlled trial (ChiCTR2100046307) studied patients who are overweight or obese at model labor health management centers from January 2019 to December 2019, including 55 people in the experimental group and 34 in the control group. The relevant indexes of all the research objects from both experimental group and control group were examined. Participants in the experimental group were given individualized exercise prescription combined with remote exercise management over a period of 3 months. The control group was prescribed exercise only at time of enrollment and taught about exercise once, followed by voluntary exercise and self-management for 3 months. Result After adjusting for baseline differences, the changes in weight (−2.72 ± 4.03 kg versus 0.32 ± 2.50 kg, P < 0.0001), body mass index (−0.99 ± 1.44 kg/m2 versus 0.11 ± 0.92 kg/m2, P < 0.0001), waist circumference (−2.98 ± 6.29 cm versus 0.60 ± 5.33 cm, P < 0.0001), visceral fat area (−9.75 ± 19.68 cm2 versus −1.31 ± 12.37 cm2, P = 0.028), body fat (− 2.65 ± 3.52 kg versus 0.54 ± 2.67 kg, P < 0.0001), body fat rate (−2.50 ± 3.32% versus 0.21 ± 3.30%, P < 0.0001), uric acid (−9.75 ± 19.68 µmol/L versus −1.31 ± 12.37 µmol/L, P = 0.028), serum total cholesterol (−0.11 ± 0.40 mmol/L versus −0.11 ± 0.59 mmol/L, P = 0.004), fasting insulin (− 2.36 ± 5.20 μU/mL versus 1.22 ± 7.34 μU/mL, P = 0.009), and homeostatic model assessment of insulin resistance (−0.62 ± 1.25 versus 0.14 ± 1.83, P = 0.022) were significantly better in the experimental group than in the control group after intervention. Conclusion Individualized exercise prescription combined with remote management in patients who are obese or overweight facilitated weight and fat loss, lowered blood pressure and serum total cholesterol, improved glucose metabolism and insulin resistance, and reduced cardiovascular risk factors. The intervention was superior to conventional education in terms of weight loss, fat reduction, total cholesterol reduction, fasting insulin reduction, and amelioration of insulin resistance.
Collapse
Affiliation(s)
- Rui Guan
- Department of Health Management, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu, Shijingshan District, Beijing, 100144, China
| | - Haijing Li
- Department of Health Management, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu, Shijingshan District, Beijing, 100144, China
| | - Yang Jiao
- Department of Health Management, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu, Shijingshan District, Beijing, 100144, China.
| | - Hong Yu
- Department of Health Management, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu, Shijingshan District, Beijing, 100144, China.
| |
Collapse
|
20
|
Ossoli A, Giorgio E, Cetti F, Ruscica M, Rabacchi C, Tarugi P, Parini P, Pedrelli M, Gomaraschi M. HDL-mediated reduction of cholesterol content inhibits the proliferation of prostate cancer cells induced by LDL: Role of ABCA1 and proteasome inhibition. Biofactors 2022; 48:707-717. [PMID: 35579277 PMCID: PMC9325382 DOI: 10.1002/biof.1845] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/26/2022] [Indexed: 12/24/2022]
Abstract
High-density lipoproteins (HDL) are well known for their atheroprotective function, mainly due to their ability to remove cell cholesterol and to exert antioxidant and anti-inflammatory activities. Through the same mechanisms HDL could also affect the development and progression of tumors. Cancer cells need cholesterol to proliferate, especially in hormone-dependent tumors, as prostate cancer (PCa). Aim of the study was to investigate the ability of HDL to modulate cholesterol content and metabolism in androgen receptor (AR)-positive and AR-null PCa cell lines and the consequences on cell proliferation. HDL inhibited colony formation of LNCaP and PC3 cells. HDL reduced cell cholesterol content and proliferation of LNCaP cells loaded with low-density lipoproteins but were not effective on PC3 cells. Here, the expression of the ATP-binding cassette transporter A1 (ABCA1) was markedly reduced due to proteasome degradation. Bortezomib, a proteasome inhibitor, restored ABCA1 expression and HDL ability to promote cholesterol removal from PC3; consequently, HDL inhibited the proliferation of PC3 cells induced by LDL only after bortezomib pre-treatment. In conclusion, the antiproliferative activity of HDL on AR-positive and AR-null PCa cells also rely on cholesterol removal, a process in which the ABCA1 transporter plays a key role.
Collapse
Affiliation(s)
- Alice Ossoli
- Centro Enrica Grossi Paoletti, Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanItaly
| | - Eleonora Giorgio
- Centro Enrica Grossi Paoletti, Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanItaly
| | - Federica Cetti
- Centro Enrica Grossi Paoletti, Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanItaly
| | - Claudio Rabacchi
- Department of Life SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Patrizia Tarugi
- Department of Life SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Paolo Parini
- Cardio Metabolic Unit, Department of Medicine and Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Matteo Pedrelli
- Cardio Metabolic Unit, Department of Medicine and Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Medicine Unit Endocrinology, Theme Inflammation and AgeingKarolinska University HospitalStockholmSweden
| | - Monica Gomaraschi
- Centro Enrica Grossi Paoletti, Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanItaly
| |
Collapse
|
21
|
Jarosz ŁS, Michalak K, Marek A, Hejdysz M, Ciszewski A, Kaczmarek S, Kwiecień M, Grądzki Z. The effect of feed supplementation with zinc glycine chelate and zinc sulphate on hepatic proteome profiles in chickens. Livest Sci 2022. [DOI: 10.1016/j.livsci.2022.104983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
22
|
Li Y, Bao Q, Yang S, Yang M, Mao C. Bionanoparticles in cancer imaging, diagnosis, and treatment. VIEW 2022. [DOI: 10.1002/viw.20200027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yan Li
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou Zhejiang China
| | - Qing Bao
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang China
| | - Shuxu Yang
- Department of Neurosurgery Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Mingying Yang
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou Zhejiang China
| | - Chuanbin Mao
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang China
- Department of Chemistry and Biochemistry Stephenson Life Science Research Center University of Oklahoma Norman Oklahoma USA
| |
Collapse
|
23
|
HDL Is Not Dead Yet. Biomedicines 2022; 10:biomedicines10010128. [PMID: 35052806 PMCID: PMC8773442 DOI: 10.3390/biomedicines10010128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/11/2022] Open
Abstract
High-density lipoprotein cholesterol (HDL-C) levels are inversely correlated with coronary heart disease (CHD) in multiple epidemiological studies, but whether HDL is causal or merely associated with CHD is unclear. Recent trials for HDL-raising drugs were either not effective in reducing CHD events or, if beneficial in reducing CHD events, were not conclusive as the findings could be attributed to the drugs’ LDL-reducing activity. Furthermore, the first large Mendelian randomization study did not causally relate HDL-C levels to decreased CHD. Thus, the hypothesis that HDL is protective against CHD has been rightfully challenged. However, subsequent Mendelian randomization studies found HDL characteristics that are causally related to decreased CHD. Many aspects of HDL structure and function, especially in reverse cholesterol transport, may be better indicators of HDL’s protective activity than simply measuring HDL-C. Cholesterol efflux capacity is associated with lower levels of prevalent and incident CHD, even after adjustment for HDL-C and apolipoprotein A-1 levels. Also, subjects with very high levels of HDL-C, including those with rare mutations that disrupt hepatic HDL uptake and reverse cholesterol transport, may be at higher risk for CHD than those with moderate levels. We describe here several cell-based and cell-free in vitro assays of HDL structure and function that may be used in clinical studies to determine which of HDL’s functions are best associated with protection against CHD. We conclude that the HDL hypothesis may need revision based on studies of HDL structure and function, but that the HDL hypothesis is not dead yet.
Collapse
|
24
|
Wang D, Yu B, Li Q, Guo Y, Koike T, Koike Y, Wu Q, Zhang J, Mao L, Tang X, Sun L, Lin X, Wu J, Chen YE, Peng D, Zeng R. OUP accepted manuscript. J Mol Cell Biol 2022; 14:6547772. [PMID: 35278086 PMCID: PMC9254886 DOI: 10.1093/jmcb/mjac004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 11/12/2022] Open
Abstract
Lipoprotein, especially high-density lipoprotein (HDL), particles are composed of multiple heterogeneous subgroups containing various proteins and lipids. The molecular distribution among these subgroups is closely related to cardiovascular disease (CVD). Here, we established high-resolution proteomics and lipidomics (HiPL) methods to depict the molecular profiles across lipoprotein (Lipo-HiPL) and HDL (HDL-HiPL) subgroups by optimizing the resolution of anion-exchange chromatography and comprehensive quantification of proteins and lipids on the omics level. Furthermore, based on the Pearson correlation coefficient analysis of molecular profiles across high-resolution subgroups, we achieved the relationship of proteome‒lipidome connectivity (PLC) for lipoprotein and HDL particles. By application of these methods to high-fat, high-cholesterol diet-fed rabbits and acute coronary syndrome (ACS) patients, we uncovered the delicate dynamics of the molecular profile and reconstruction of lipoprotein and HDL particles. Of note, the PLC features revealed by the HDL-HiPL method discriminated ACS from healthy individuals better than direct proteome and lipidome quantification or PLC features revealed by the Lipo-HiPL method, suggesting their potential in ACS diagnosis. Together, we established HiPL methods to trace the dynamics of the molecular profile and PLC of lipoprotein and even HDL during the development of CVD.
Collapse
Affiliation(s)
| | | | | | | | - Tomonari Koike
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yui Koike
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Qingqing Wu
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Ling Mao
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xiaoyu Tang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Liang Sun
- Key Laboratory of Nutrition and Metabolism, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiarui Wu
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | | | | | - Rong Zeng
- Correspondence to: Rong Zeng, E-mail:
| |
Collapse
|
25
|
Shulgin AA, Lebedev TD, Prassolov VS, Spirin PV. Plasmolipin and Its Role in Cell Processes. Mol Biol 2021; 55:773-785. [PMID: 34955555 PMCID: PMC8682038 DOI: 10.1134/s0026893321050113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/26/2021] [Accepted: 04/05/2021] [Indexed: 12/04/2022]
Abstract
The mechanisms involved in the origin and development of malignant and neurodegenerative diseases are an important area of modern biomedicine. A crucial task is to identify new molecular markers that are associated with rearrangements of intracellular signaling and can be used for prognosis and the development of effective treatment approaches. The proteolipid plasmolipin (PLLP) is a possible marker. PLLP is a main component of the myelin sheath and plays an important role in the development and normal function of the nervous system. PLLP is involved in intracellular transport, lipid raft formation, and Notch signaling. PLLP is presumably involved in various disorders, such as cancer, schizophrenia, Alzheimer's disease, and type 2 diabetes mellitus. PLLP and its homologs were identified as possible virus entry receptors. The review summarizes the data on the PLLP structure, normal functions, and role in diseases.
Collapse
Affiliation(s)
- A. A. Shulgin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Moscow oblast Russia
| | - T. D. Lebedev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - V. S. Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - P. V. Spirin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
26
|
Hancock-Cerutti W, Millar JS, Valentini S, Liu J, Billheimer JT, Rader DJ, Cuchel M. Assessing HDL Metabolism in Subjects with Elevated Levels of HDL Cholesterol and Coronary Artery Disease. Molecules 2021; 26:6862. [PMID: 34833954 PMCID: PMC8623898 DOI: 10.3390/molecules26226862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/27/2021] [Accepted: 11/06/2021] [Indexed: 12/26/2022] Open
Abstract
High-density lipoprotein cholesterol (HDL-C) is thought to be atheroprotective yet some patients with elevated HDL-C levels develop cardiovascular disease, possibly due to the presence of dysfunctional HDL. We aimed to assess the metabolic fate of circulating HDL particles in patients with high HDL-C with and without coronary artery disease (CAD) using in vivo dual labeling of its cholesterol and protein moieties. We measured HDL apolipoprotein (apo) A-I, apoA-II, free cholesterol (FC), and cholesteryl ester (CE) kinetics using stable isotope-labeled tracers (D3-leucine and 13C2-acetate) as well as ex vivo cholesterol efflux to HDL in subjects with (n = 6) and without (n = 6) CAD that had HDL-C levels >90th percentile. Healthy controls with HDL-C within the normal range (n = 6) who underwent the same procedures were used as the reference. Subjects with high HDL-C with and without CAD had similar plasma lipid levels and similar apoA-I, apoA-II, HDL FC, and CE pool sizes with no significant differences in fractional clearance rates (FCRs) or production rates (PRs) of these components between groups. Subjects with high HDL-C with and without CAD also had similar basal and cAMP-stimulated ex vivo cholesterol efflux to HDL. When all subjects were considered (n = 18), unstimulated non-ABCA1-mediated efflux (but not ABCA1-specific efflux) was correlated positively with apoA-I production (r = 0.552, p = 0.017) and HDL FC and CE pool sizes, and negatively with the fractional clearance rate of FC (r = -0.759, p = 4.1 × 10-4) and CE (r = -0.652, p = 4.57 × 10-3). Our data are consistent with the concept that ex vivo non-ABCA1 efflux capacity may correlate with slower in vivo turnover of HDL cholesterol moieties. The use of a dual labeling protocol provided for the first time the opportunity to assess the association of ex vivo cholesterol efflux capacity with in vivo HDL cholesterol metabolic parameters.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marina Cuchel
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104, USA; (W.H.-C.); (J.S.M.); (S.V.); (J.L.); (J.T.B.); (D.J.R.)
| |
Collapse
|
27
|
Zanotti I, Potì F, Cuchel M. HDL and reverse cholesterol transport in humans and animals: Lessons from pre-clinical models and clinical studies. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1867:159065. [PMID: 34637925 DOI: 10.1016/j.bbalip.2021.159065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/07/2021] [Accepted: 09/24/2021] [Indexed: 02/06/2023]
Abstract
The ability to accept cholesterol from cells and to promote reverse cholesterol transport (RCT) represents the best characterized antiatherogenic function of HDL. Studies carried out in animal models have unraveled the multiple mechanisms by which these lipoproteins drive cholesterol efflux from macrophages and cholesterol uptake to the liver. Moreover, the influence of HDL composition and the role of lipid transporters have been clarified by using suitable transgenic models or through experimental design employing pharmacological or nutritional interventions. Cholesterol efflux capacity (CEC), an in vitro assay developed to offer a measure of the first step of RCT, has been shown to associate with cardiovascular risk in several human cohorts, supporting the atheroprotective role of RCT in humans as well. However, negative data in other cohorts have raised concerns on the validity of this biomarker. In this review we will present the most relevant data documenting the role of HDL in RCT, as assessed in classical or innovative methodological approaches.
Collapse
Affiliation(s)
- Ilaria Zanotti
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy.
| | - Francesco Potì
- Dipartimento di Medicina e Chirurgia, Unità di Neuroscienze, Università di Parma, Via Volturno 39/F, 43125 Parma, Italy
| | - Marina Cuchel
- Division of Translational Medicine & Human Genetics, Perelman School of Medicine at the University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Kluck GEG, Yoo JA, Sakarya EH, Trigatti BL. Good Cholesterol Gone Bad? HDL and COVID-19. Int J Mol Sci 2021; 22:10182. [PMID: 34638523 PMCID: PMC8507803 DOI: 10.3390/ijms221910182] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
The transmissible respiratory disease COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has affected millions of people worldwide since its first reported outbreak in December of 2019 in Wuhan, China. Since then, multiple studies have shown an inverse correlation between the levels of high-density lipoprotein (HDL) particles and the severity of COVID-19, with low HDL levels being associated with an increased risk of severe outcomes. Some studies revealed that HDL binds to SARS-CoV-2 particles via the virus's spike protein and, under certain conditions, such as low HDL particle concentrations, it facilitates SARS-CoV-2 binding to angiotensin-converting enzyme 2 (ACE2) and infection of host cells. Other studies, however, reported that HDL suppressed SARS-CoV-2 infection. In both cases, the ability of HDL to enhance or suppress virus infection appears to be dependent on the expression of the HDL receptor, namely, the Scavenger Receptor Class B type 1 (SR-B1), in the target cells. SR-B1 and HDL represent crucial mediators of cholesterol metabolism. Herein, we review the complex role of HDL and SR-B1 in SARS-CoV-2-induced disease. We also review recent advances in our understanding of HDL structure, properties, and function during SARS-CoV-2 infection and the resulting COVID-19 disease.
Collapse
Affiliation(s)
| | | | | | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute and Department of Biochemistry and Biomedical Sciences, McMaster University and Hamilton Health Sciences, Hamilton, ON L8L 2X2, Canada; (G.E.G.K.); (J.-A.Y.); (E.H.S.)
| |
Collapse
|
29
|
Patel S, Werstuck G. Characterizing the Role of Glycogen Synthase Kinase-3α/β in Macrophage Polarization and the Regulation of Pro-Atherogenic Pathways in Cultured Ldlr -/- Macrophages. Front Immunol 2021; 12:676752. [PMID: 34394077 PMCID: PMC8361494 DOI: 10.3389/fimmu.2021.676752] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/12/2021] [Indexed: 12/31/2022] Open
Abstract
The molecular and cellular mechanisms that link cardiovascular risk factors to the initiation and progression of atherosclerosis are not understood. Recent findings from our laboratory indicate that endoplasmic reticulum (ER) stress signaling through glycogen synthase kinase (GSK)-3α/β induces pro-atherosclerotic pathways. The objective of this study was to define the specific roles of GSK3α and GSK3β in the activation of pro-atherogenic processes in macrophages. Bone marrow derived macrophages (BMDM) were isolated from low-density lipoprotein receptor knockout (Ldlr-/-) mice and Ldlr-/- mice with myeloid deficiency of GSK3α and/or GSK3β. M1 and M2 macrophages were used to examine functions relevant to the development of atherosclerosis, including polarization, inflammatory response, cell viability, lipid accumulation, migration, and metabolism. GSK3α deficiency impairs M1 macrophage polarization, and reduces the inflammatory response and lipid accumulation, but increases macrophage mobility/migration. GSK3β deficiency promotes M1 macrophage polarization, which further increases the inflammatory response and lipid accumulation, but decreases macrophage migration. Macrophages deficient in both GSK3α and GSK3β exhibit increased cell viability, proliferation, and metabolism. These studies begin to delineate the specific roles of GSK3α and GSK3β in macrophage polarization and function. These data suggest that myeloid cell GSK3α signaling regulates M1 macrophage polarization and pro-atherogenic functions to promote atherosclerosis development.
Collapse
Affiliation(s)
- Sarvatit Patel
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada.,Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Geoff Werstuck
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada.,Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
30
|
Trp Fluorescence Redshift during HDL Apolipoprotein Denaturation Is Increased in Patients with Coronary Syndrome in Acute Phase: A New Assay to Evaluate HDL Stability. Int J Mol Sci 2021; 22:ijms22157819. [PMID: 34360583 PMCID: PMC8345965 DOI: 10.3390/ijms22157819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/30/2022] Open
Abstract
High-density lipoproteins' (HDL) stability is a determinant of their residence times in plasma and consequently an important parameter that influences the beneficial properties of these lipoproteins. Since there are no accessible procedures for this purpose, here, we describe the methodological conditions to assess the stability of the HDL based on the redshift of the fluorescence spectrum of tryptophans contained in the structure of HDL-apolipoproteins during incubation with urea 8M. Along the HDL denaturation kinetics, the main variations of fluorescence were observed at the wavelengths of 330, 344, and 365 nm at room temperature. Therefore, HDL denaturation was estimated using the tryptophan (Trp)-ratio of fluorescence intensity (rfi) at such wavelengths. By setting 100% of the measurable denaturation at 26 h, HDL reached 50% after 8 h of incubation with urea. Then, for further analyses we determined the percentage of HDL denaturation at 8 h as an estimation of the stability of these lipoproteins. To explore the potential usefulness of this test, we analyzed the stability of HDL isolated from the plasma of 24 patients diagnosed with acute coronary syndrome (ACS). These HDL presented significantly higher percentages of denaturation (64.9% (58.7-78.4)) than HDLs of healthy individuals (23.3% (20.3-27.0)). These results indicate that HDL in ACS are less stable than in control subjects. Moreover, the percentage of denaturation of HDL correlated with body mass index and aspartate transaminase plasma activity. Furthermore, apo-I, HDL-cholesterol, HDL-triglycerides, and apo A-I-to-triglycerides ratio correlated with the percentage of HDL denaturation, suggesting that the lipoprotein composition is a main determinant of HDL stability. Finally, the percentage of HDL denaturation is the parameter that predicted the presence of ACS as determined by a machine learning procedure and logistic regression analysis. In conclusion, we established the methodological conditions to assess the stability of HDL by a fluorescence-based method that merits exploration in prospective studies for evaluating the coronary artery disease risk.
Collapse
|
31
|
Joshita S, Yamashita Y, Okamoto T, Usami Y, Sugiura A, Yamazaki T, Kakino A, Ota M, Sawamura T, Umemura T. Quantitative and qualitative lipid improvement with chronic hepatitis C virus eradication using direct-acting antivirals. Hepatol Res 2021; 51:758-766. [PMID: 33982310 DOI: 10.1111/hepr.13666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/29/2021] [Accepted: 05/08/2021] [Indexed: 02/08/2023]
Abstract
AIM Direct-acting antivirals have revolutionized hepatitis C virus (HCV) therapy by providing a high sustained virological response (SVR) rate and subsequent favorable lipid increases. Proprotein convertase subtilisin-kexin like-9 (PCSK9) plays an important role in regulating quantitative lipid levels. This study examined the interactions between quantitative PCSK9 and lipid changes, as well as qualitative lipid changes in terms of lectin-like oxidized low-density lipoprotein (LDL) receptor-1 ligand containing apolipoprotein B (LAB) and high-density lipoprotein (HDL) cholesterol uptake capacity (HDL-CUC). METHODS Patients with chronic HCV infection (N = 231) who achieved an SVR by direct-acting antivirals without lipid-lowering therapy were included for comparisons of PCSK9, LAB, HDL-CUC, and other clinical indices between pretreatment and SVR12 time points. RESULTS LDL (LDL) cholesterol and HDL cholesterol levels were quantitatively increased at SVR12, along with higher PCSK9 (all p < 0.0001). PCSK9 was significantly correlated with LDL cholesterol (r = 0.244, p = 0.0003) and apolipoprotein B (r = 0.222, p = 0.0009) at SVR12. Regarding qualitative LDL changes, LAB was significantly decreased and LAB/LDL cholesterol and LAB/apolipoprotein B proportions were improved at SVR12 (all p < 0.0001). In terms of qualitative HDL changes, HDL-CUC was significantly ameliorated, along with HDL-CUC/HDL cholesterol, HDL-CUC/ apolipoprotein A1, and HDL-CUC/ apolipoprotein A2 at SVR12 (all p < 0.0001). CONCLUSIONS HCV eradication by direct-acting antivirals may produce quantitative lipid profile changes, along with PCSK9 production recovery in addition to qualitative lipid improvement, which possibly confers the additional secondary benefits of atherosclerosis improvement and cardiovascular disease event reduction.
Collapse
Affiliation(s)
- Satoru Joshita
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yuki Yamashita
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Toru Okamoto
- Institute for Advanced Co-Creation Studies Research, Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoko Usami
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Ayumi Sugiura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Tomoo Yamazaki
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Akemi Kakino
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Matsumoto, Japan.,Department of Life Innovation, Institute for Biomedical Sciences, Shinshu University, Matsumoto, Japan
| | - Masao Ota
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Matsumoto, Japan.,Department of Life Innovation, Institute for Biomedical Sciences, Shinshu University, Matsumoto, Japan
| | - Takeji Umemura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan.,Department of Life Innovation, Institute for Biomedical Sciences, Shinshu University, Matsumoto, Japan
| |
Collapse
|
32
|
Lavker RM, Kaplan N, McMahon KM, Calvert AE, Henrich SE, Onay UV, Lu KQ, Peng H, Thaxton CS. Synthetic high-density lipoprotein nanoparticles: Good things in small packages. Ocul Surf 2021; 21:19-26. [PMID: 33894397 PMCID: PMC8328934 DOI: 10.1016/j.jtos.2021.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/26/2021] [Accepted: 03/06/2021] [Indexed: 12/26/2022]
Abstract
Medicine has been a great beneficiary of the nanotechnology revolution. Nanotechnology involves the synthesis of functional materials with at least one size dimension between 1 and 100 nm. Advances in the field have enabled the synthesis of bio-nanoparticles that can interface with physiological systems to modulate fundamental cellular processes. One example of a diverse acting nanoparticle-based therapeutic is synthetic high-density lipoprotein (HDL) nanoparticles (NP), which have great potential for treating diseases of the ocular surface. Our group has developed a spherical HDL NP using a gold nanoparticle core. HDL NPs: (i) closely mimic the physical and chemical features of natural HDLs; (ii) contain apoA-I; (iii) bind with high-affinity to SR-B1, which is the major receptor through which HDL modulates cell cholesterol metabolism and controls the selective uptake of HDL cargo into cells; (iv) are non-toxic to cells and tissues; and (v) can be chemically engineered to display nearly any surface or core composition desired. With respect to the ocular surface, topical application of HDL NPs accelerates re-epithelization of the cornea following wounding, attenuates inflammation resulting from chemical burns and/or other stresses, and effectively delivers microRNAs with biological activity to corneal cells and tissues. HDL NPs will be the foundation of a new class of topical eye drops with great translational potential and exemplify the impact that nanoparticles can have in medicine.
Collapse
Affiliation(s)
- Robert M Lavker
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Nihal Kaplan
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kaylin M McMahon
- Department of Dermatology Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Andrea E Calvert
- Department of Dermatology Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Stephen E Henrich
- Department of Dermatology Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ummiye V Onay
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kurt Q Lu
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Han Peng
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C Shad Thaxton
- Department of Dermatology Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
33
|
Trakaki A, Wolf P, Weger W, Eichmann TO, Scharnagl H, Stadler JT, Salmhofer W, Knuplez E, Holzer M, Marsche G. Biological anti-psoriatic therapy profoundly affects high-density lipoprotein function. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158943. [PMID: 33862237 DOI: 10.1016/j.bbalip.2021.158943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/24/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022]
Abstract
Psoriasis is a common chronic inflammatory skin disease linked to increased cardiovascular risk. Functional impairment of high-density lipoprotein (HDL) may contribute to excessive cardiovascular mortality in psoriasis patients. Anti-cytokine therapies with biologics have been efficiently used for the management of psoriasis, however little data is available on the effects of biologic anti-psoriatic therapies on the composition and functionality of HDL. Blood samples were taken from 17 healthy volunteers and from 27 real-world psoriasis patients at baseline (no therapy with biologics) and after short-term (3 to 6 months) and intermediate-term (1 to 2 years) therapy. The biologics used included anti-interleukin (IL)-12/23p40 (ustekinumab), anti-IL17A (secukinumab) or anti-tumor necrosis factor-α (etanercept or adalimumab) antibodies. We observed that in psoriasis patients at baseline, metrics of HDL function including cholesterol efflux capacity of apolipoprotein B-depleted serum (p = 0.021), paraoxonase (p < 0.001) and lecithin-cholesterol acyltransferase (p < 0.001) activities were impaired, when compared to controls. Unexpectedly, we observed that short- and especially intermediate-term therapy with biologics markedly reduced HDL cholesterol efflux capacity (p < 0.001) and rendered HDL pro-inflammatory (p < 0.001), but increased paraoxonase (p = 0.009) and lecithin-cholesterol acyltransferase (p = 0.019) activities. All biologics caused similar changes in HDL composition, subclass distribution and cholesterol efflux capacity. Our results provide evidence that anti-psoriatic therapy with biologic agents is associated with changes in HDL functionality, particle composition and subclass distribution.
Collapse
Affiliation(s)
- Athina Trakaki
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Peter Wolf
- Department of Dermatology and Venereology, Auenbruggerplatz 8, 8036 Graz, Austria.
| | - Wolfgang Weger
- Department of Dermatology and Venereology, Auenbruggerplatz 8, 8036 Graz, Austria
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Center for Explorative Lipidomics, BioTechMed-Graz, Graz, Austria
| | - Hubert Scharnagl
- Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Wolfgang Salmhofer
- Department of Dermatology and Venereology, Auenbruggerplatz 8, 8036 Graz, Austria
| | - Eva Knuplez
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Michael Holzer
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria; BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria.
| |
Collapse
|
34
|
Bonizzi A, Piuri G, Corsi F, Cazzola R, Mazzucchelli S. HDL Dysfunctionality: Clinical Relevance of Quality Rather Than Quantity. Biomedicines 2021; 9:729. [PMID: 34202201 PMCID: PMC8301425 DOI: 10.3390/biomedicines9070729] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 11/28/2022] Open
Abstract
High-density lipoproteins (HDLs) represent a class of lipoproteins very heterogeneous in structure, composition, and biological functions, which carry out reverse cholesterol transport, antioxidant, anti-inflammatory, antithrombotic, and vasodilator actions. Despite the evidence suggesting a clear inverse relationship between HDL cholesterol (HDL-c) concentration and the risk for cardiovascular disease, plasma HDL cholesterol levels do not predict the functionality and composition of HDLs. The importance of defining both the amount of cholesterol transported and lipoprotein functionality has recently been highlighted. Indeed, different clinical conditions such as obesity, diabetes mellitus type 2 (T2DM), and cardiovascular disease (CVD) can alter the HDL functionality, converting normal HDLs into dysfunctional ones, undergoing structural changes, and exhibiting proinflammatory, pro-oxidant, prothrombotic, and proapoptotic properties. The aim of the current review is to summarize the actual knowledge concerning the physical-chemical alteration of HDLs related to their functions, which have been found to be relevant in several pathological conditions associated with systemic inflammation and oxidative stress.
Collapse
Affiliation(s)
- Arianna Bonizzi
- Department of Biomedical and Clinical Sciences "L. Sacco", Università di Milano, 20157 Milan, Italy
| | - Gabriele Piuri
- Department of Biomedical and Clinical Sciences "L. Sacco", Università di Milano, 20157 Milan, Italy
| | - Fabio Corsi
- Department of Biomedical and Clinical Sciences "L. Sacco", Università di Milano, 20157 Milan, Italy
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Roberta Cazzola
- Department of Biomedical and Clinical Sciences "L. Sacco", Università di Milano, 20157 Milan, Italy
| | - Serena Mazzucchelli
- Department of Biomedical and Clinical Sciences "L. Sacco", Università di Milano, 20157 Milan, Italy
| |
Collapse
|
35
|
Yang J, Wang H, Du H, Fang H, Han M, Xu L, Liu S, Yi J, Chen Y, Jiang Q, He G. Serum perfluoroalkyl substances in relation to lipid metabolism in Chinese pregnant women. CHEMOSPHERE 2021; 273:128566. [PMID: 33097232 DOI: 10.1016/j.chemosphere.2020.128566] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 06/11/2023]
Abstract
Laboratory and epidemiologic studies suggested that exposure to perfluoroalkyl substances (PFASs) could affect lipid metabolisms, but data remain limited for pregnant women. A total of 436 pregnant women were selected in Tangshan City, North China. Serum levels of 11 PFASs were determined in the early term of pregnancy. Four lipids (total cholesterol (TC), triglyceride (TG), high-density lipoprotein (HDL), and low-density lipoprotein (LDL)) were measured in the late term of pregnancy. Of 11 PFASs, seven had a detection rate of greater than 70%. After adjusting for potential confounders, natural log-transformed perfluororohexanesulfonic acid (ln PFHxS) was positively associated with TC (β: 0.184, 95% CI: 0.045-0.321), HDL (β: 0.040, 95% CI: 0.001-0.083), and LDL (β: 0.091, 95% CI: 0.001-0.185). Ln perfluoroundecanoic acid (PFUdA) was positively associated with HDL (β: 0.021, 95% CI: 0.001-0.044), while Ln perfluorodecanoic acid (PFDA) was negatively associated with LDL (β: -0.053, 95% CI: -0.098∼-0.009) and ln perfluorootanoic acid (PFOA) was negatively associated with LDL/HDL (β: -0.042, 95% CI: -0.075∼-0.009). In principal component analysis, the component with a large loading of 31.3% for PFOA, perfluorononanoic acid (PFNA), PFDA and PFUdA showed a negative association with LDL/HDL. After serum concentrations of PFASs were categorized into quartiles, a higher level of TC was seen in the second quartile of PFOA or PFNA than the first quartile, but a lower LDL/HDL ratio was seen in the fourth quartile of PFOA, PFUdA or PFDA. These results suggested that exposure to PFASs has a potential to influence lipid metabolisms in pregnant women.
Collapse
Affiliation(s)
- Jiaqi Yang
- School of Public Health, Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Hexing Wang
- School of Public Health, Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Hongyi Du
- Institute of Reproductive and Child Health, School of Public Health, Peking University Health Science Center, Beijing, 100083, China
| | - Hongji Fang
- School of Public Health, Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Minghui Han
- School of Public Health, Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Linji Xu
- Maternal and Child Health Care Hospital, Tangshan Municipality, No. 14 South Jianshe Road, Tangshan, 063000, Hebei province, China
| | - Shuping Liu
- Maternal and Child Health Care Hospital, Tangshan Municipality, No. 14 South Jianshe Road, Tangshan, 063000, Hebei province, China
| | - Jianping Yi
- Maternal and Child Health Care Hospital, Tangshan Municipality, No. 14 South Jianshe Road, Tangshan, 063000, Hebei province, China
| | - Yue Chen
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1G 5Z3, Canada
| | - Qingwu Jiang
- School of Public Health, Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Gengsheng He
- School of Public Health, Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
36
|
Balder Y, Vignoli A, Tenori L, Luchinat C, Saccenti E. Exploration of Blood Lipoprotein and Lipid Fraction Profiles in Healthy Subjects through Integrated Univariate, Multivariate, and Network Analysis Reveals Association of Lipase Activity and Cholesterol Esterification with Sex and Age. Metabolites 2021; 11:metabo11050326. [PMID: 34070169 PMCID: PMC8158518 DOI: 10.3390/metabo11050326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023] Open
Abstract
In this study, we investigated blood lipoprotein and lipid fraction profiles, quantified using nuclear magnetic resonance, in a cohort of 844 healthy blood donors, integrating standard univariate and multivariate analysis with predictive modeling and network analysis. We observed a strong association of lipoprotein and lipid main fraction profiles with sex and age. Our results suggest an age-dependent remodulation of lipase lipoprotein activity in men and a change in the mechanisms controlling the ratio between esterified and non-esterified cholesterol in both men and women.
Collapse
Affiliation(s)
- Yasmijn Balder
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands;
| | - Alessia Vignoli
- Magnetic Resonance Center (CERM) and Department of Chemistry “Ugo Schiff”, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy; (A.V.); (L.T.); (C.L.)
- Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Leonardo Tenori
- Magnetic Resonance Center (CERM) and Department of Chemistry “Ugo Schiff”, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy; (A.V.); (L.T.); (C.L.)
- Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM) and Department of Chemistry “Ugo Schiff”, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy; (A.V.); (L.T.); (C.L.)
- Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands;
- Correspondence:
| |
Collapse
|
37
|
Mechanism of Huo-Xue-Qu-Yu Formula in Treating Nonalcoholic Hepatic Steatosis by Regulating Lipid Metabolism and Oxidative Stress in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6026319. [PMID: 34007294 PMCID: PMC8102110 DOI: 10.1155/2021/6026319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 01/11/2021] [Accepted: 04/15/2021] [Indexed: 11/17/2022]
Abstract
Huo-Xue-Qu-Yu formula (HXQYF) is a prescription consisting of Ginkgo biloba leaf and Paeonia lactiflora Pall. for treating hyperlipidemia and NAFLD in China. Here, we investigated the hepatic and renal function, oxidative stress and lipid metabolism, and potential mechanisms of HXQYF on nonalcoholic fatty liver disease (NAFLD) rat models. NAFLD rat models were induced with high-fat diet (HFD) and 10% fructose water for 18 weeks and orally administered with or without HXQYF simultaneously. The results showed that HXQYF (22.5, 45, 90 mg/kg) significantly improved blood lipid levels via reducing serum TC, TG, LDL-C, and APOB values and elevating HDL-C and APOA1 levels in NAFLD rats. The higher levels of ALT, AST, CR, and BUN in serum induced by HFD were reduced by HXQYF. HE staining showed that HXQYF (90 mg/kg) reduced the accumulation of fat droplets and alleviated inflammatory response in liver cells. Three doses of HXQYF exhibited notable antioxidant effects by elevating SOD, GSH, and CAT activities and decreasing MDA and OH-1 levels in the liver. Furthermore, abnormal lipid metabolism caused by HFD was alleviated by HXQYF, which was associated with the upregulation of PPAR-α, AdipoR2, and CPT1 mRNAs as well as the downregulation of CYP2E1 and SREBP-1c mRNAs in liver tissue. In conclusion, our work verified that HXQYF could reduce the degree of hepatic steatosis, suppress oxidative stress, and attenuate lipid metabolism, thus preventing NAFLD.
Collapse
|
38
|
Tereshkina YA, Kostryukova LV, Torkhovskaya TI, Khudoklinova YY, Tikhonova EG. [Plasma high density lipoproteins phospholipds as an indirect indicator of their cholesterol efflux capacity - new suspected atherosclerosis risk factor]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:119-129. [PMID: 33860768 DOI: 10.18097/pbmc20216702119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
High density lipoproteins (HDL) are a unique natural structure, protecting the body from the development of atherosclerotic vascular lesions and cardiovascular diseases due to this ability to remove cholesterol from cells. Plasma HDL level estimated by their cholesterol content, is a common lipid parameter, and its decrease is considered as an established atherosclerosis risk factor. However, a number of studies have shown the absence of positive clinical effects after drug-induced increase in HDL cholesterol. There is increasing evidence that not only HDL concentration, but also HDL properties, considered in this review are important. Many studies showed the decrease of HDL cholesterol efflux capacity in patients with coronary heart diseases and its association with disease severity. Some authors consider a decrease of this HDL capacity as a new additional risk factor of atherosclerosis. The review summarizes existing information on various protein and lipid components of HDL with a primary emphasis on the HDL. Special attention is paid to correlation between the HDL cholesterol efflux capacity and HDL phospholipids and the ratio "phospholipids/free cholesterol". The accumulated information indicates importance of evaluation in the HDL fraction not only in terms of their cholesterol, but also phospholipids. In addition to the traditionally used lipid criteria, this would provide more comprehensive information about the activity of the reverse cholesterol transport process in the body and could contribute to the targeted correction of the detected disorders.
Collapse
|
39
|
Stadler JT, Wadsack C, Marsche G. Fetal High-Density Lipoproteins: Current Knowledge on Particle Metabolism, Composition and Function in Health and Disease. Biomedicines 2021; 9:biomedicines9040349. [PMID: 33808220 PMCID: PMC8067099 DOI: 10.3390/biomedicines9040349] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
Cholesterol and other lipids carried by lipoproteins play an indispensable role in fetal development. Recent evidence suggests that maternally derived high-density lipoprotein (HDL) differs from fetal HDL with respect to its proteome, size, and function. Compared to the HDL of adults, fetal HDL is the major carrier of cholesterol and has a unique composition that implies other physiological functions. Fetal HDL is enriched in apolipoprotein E, which binds with high affinity to the low-density lipoprotein receptor. Thus, it appears that a primary function of fetal HDL is the transport of cholesterol to tissues as is accomplished by low-density lipoproteins in adults. The fetal HDL-associated bioactive sphingolipid sphingosine-1-phosphate shows strong vasoprotective effects at the fetoplacental vasculature. Moreover, lipoprotein-associated phospholipase A2 carried by fetal-HDL exerts anti-oxidative and athero-protective functions on the fetoplacental endothelium. Notably, the mass and activity of HDL-associated paraoxonase 1 are about 5-fold lower in the fetus, accompanied by an attenuation of anti-oxidative activity of fetal HDL. Cholesteryl ester transfer protein activity is reduced in fetal circulation despite similar amounts of the enzyme in maternal and fetal serum. This review summarizes the current knowledge on fetal HDL as a potential vasoprotective lipoprotein during fetal development. We also provide an overview of whether and how the protective functionalities of HDL are impaired in pregnancy-related syndromes such as pre-eclampsia or gestational diabetes mellitus.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
- Correspondence: (J.T.S.); (G.M.); Tel.: +43-316-385-74115 (J.T.S.); +43-316-385-74128 (G.M.)
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria;
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
- Correspondence: (J.T.S.); (G.M.); Tel.: +43-316-385-74115 (J.T.S.); +43-316-385-74128 (G.M.)
| |
Collapse
|
40
|
Cariello M, Salvia R, Härdfeldt J, Piglionica M, Rutigliano D, Caldarola P, Ossoli A, Vacca M, Graziano G, Battaglia S, Zerlotin R, Arconzo M, Crudele L, Sabbà C, Calabresi L, Moschetta A. Intracoronary monocyte expression pattern and HDL subfractions after non-ST elevation myocardial infarction. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166116. [PMID: 33667626 DOI: 10.1016/j.bbadis.2021.166116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/04/2021] [Accepted: 02/24/2021] [Indexed: 10/22/2022]
Abstract
AIMS Coronary artery disease (CAD) is described as a range of clinical conditions including myocardial infarction (MI) and unstable angina. Lipid and apolipoprotein profiles together with the study of cholesterol deposit and efflux serve to identify novel pre and post infarct scenarios for the treatment of these patients. In (non-ST elevation myocardial infarction) NSTEMI patients, we analysed both systemic and intracoronary serum ability to accept cholesterol as well as cholesterol efflux capacity (CEC) of monocytes in terms of expression of genes involved in the reverse cholesterol transport (RCT). METHODS AND RESULTS While HDL-C quantity was similar between systemic and coronary arterial blood, in 21 NSTEMI patients we observed a significant reduction of the preβ-HDL fraction and the levels of Apolipoproteins AI, AII, B and E in coronary versus systemic serum. These data are complemented with the observed reduction of CEC. On the contrary, compared to systemic arterial monocytes, in coronary microenvironment of NSTEMI patients after myocardial infarction, the monocytes exhibited a higher mRNA expression of nuclear receptor LXRα and its targets ABCA1 and APOE, which drive cholesterol efflux capacity. CONCLUSION In this cross-sectional study we observe that in the immediate post infarction period, there is a spontaneous bona fide ligand-induced activation of the LXR driven cholesterol efflux capacity of intracoronary monocytes to overcome the reduced serum ability to accept cholesterol and to inhibit the post-infarction pro-inflammatory local microenvironment.
Collapse
Affiliation(s)
- Marica Cariello
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari, Italy
| | - Roberto Salvia
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari, Italy
| | - Jennifer Härdfeldt
- INBB, National Institute for Biostructures and Biosystems, 00136 Rome, Italy
| | - Marilidia Piglionica
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari, Italy
| | | | | | - Alice Ossoli
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Michele Vacca
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Giusi Graziano
- INBB, National Institute for Biostructures and Biosystems, 00136 Rome, Italy
| | - Stefano Battaglia
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari, Italy; Department of Tissues and Organs Transplantation and Cellular Therapies, "Aldo Moro" University of Bari, Bari, Italy
| | - Roberta Zerlotin
- INBB, National Institute for Biostructures and Biosystems, 00136 Rome, Italy
| | - Maria Arconzo
- INBB, National Institute for Biostructures and Biosystems, 00136 Rome, Italy
| | - Lucilla Crudele
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari, Italy
| | - Carlo Sabbà
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari, Italy
| | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari, Italy; INBB, National Institute for Biostructures and Biosystems, 00136 Rome, Italy.
| |
Collapse
|
41
|
Gupta A, Sharma R, Kuche K, Jain S. Exploring the therapeutic potential of the bioinspired reconstituted high density lipoprotein nanostructures. Int J Pharm 2021; 596:120272. [DOI: 10.1016/j.ijpharm.2021.120272] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/20/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022]
|
42
|
Malajczuk CJ, Gandhi NS, Mancera RL. Structure and intermolecular interactions in spheroidal high-density lipoprotein subpopulations. JOURNAL OF STRUCTURAL BIOLOGY-X 2020; 5:100042. [PMID: 33437963 PMCID: PMC7788233 DOI: 10.1016/j.yjsbx.2020.100042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/13/2022]
Abstract
High-density lipoprotein subpopulations have unique surface profiles and dynamics. Relative hydrophobic surface area decreases with increasing lipoprotein size. Core lipid exposure at the lipoprotein surface decreases with increasing size. Cholesterol molecules localise near apolipoprotein A-I central helices. Lipid and protein interactions stabilise multifoil models of apolipoprotein A-I.
Human serum high-density lipoproteins (HDLs) are a population of small, dense protein-lipid aggregates that are crucial for intravascular lipid trafficking and are protective against cardiovascular disease. The spheroidal HDL subfraction can be separated by size and density into five major subpopulations with distinct molecular compositions and unique biological functionalities: HDL3c, HDL3b, HDL3a, HDL2a and HDL2b. Representative molecular models of these five subpopulations were developed and characterised for the first time in the presence of multiple copies of its primary protein component apolipoprotein A-I (apoA-I) using coarse-grained molecular dynamics simulations. Each HDL model exhibited size, morphological and compositional profiles consistent with experimental observables. With increasing particle size the separation of core and surface molecules became progressively more defined, resulting in enhanced core lipid mixing, reduced core lipid exposure at the surface, and the formation of an interstitial region between core and surface molecules in HDL2b. Cholesterol molecules tended to localise around the central helix-5 of apoA-I, whilst triglyceride molecules predominantly interacted with aromatic, hydrophobic residues located within the terminal helix-10 across all subpopulation models. The three intermediate HDL models exhibited similar surface profiles despite having distinct molecular compositions. ApoA-I in trefoil, quatrefoil and pentafoil arrangements across the surface of HDL particles exhibited significant warping and twisting, but largely retained intermolecular contacts between adjacent apoA-I chains. Representative HDL subpopulations differed in particle size, morphology, intermolecular interaction profiles and lipid and protein dynamics. These findings reveal how different HDL subpopulations might exhibit distinct functional associations depending on particle size, form and composition.
Collapse
Affiliation(s)
- Chris J Malajczuk
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| | - Neha S Gandhi
- School of Mathematical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Ricardo L Mancera
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| |
Collapse
|
43
|
El Khoudary ( سمر رياض الخضري ) SR, Chen (陈曦润) X, Nasr ( ألكسس نصر ) A, Billheimer J, Brooks MM, McConnell D, Orchard TJ, Crawford SL, Matthews KA, Rader DJ. HDL (High-Density Lipoprotein) Subclasses, Lipid Content, and Function Trajectories Across the Menopause Transition: SWAN-HDL Study. Arterioscler Thromb Vasc Biol 2020; 41:951-961. [PMID: 33267661 DOI: 10.1161/atvbaha.120.315355] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The cardioprotective capacity of HDL (high-density lipoprotein) cholesterol postmenopause has been challenged. HDL subclasses, lipid contents, and function might be better predictors of cardiovascular risk than HDL cholesterol. Changes in these measures have not been characterized over the menopause transition (MT) with respect to timing relative to the final menstrual period. Approach and Results: Four hundred seventy-one women with HDL particle (HDL-P) subclasses (nuclear magnetic resonance spectroscopy total, large, medium, and small HDL-P and HDL size), HDL lipid content (HDL phospholipids and triglycerides), and HDL function (cholesterol efflux capacity [HDL-CEC]) measured for a maximum of 5 time points across the MT were included. HDL cholesterol and total HDL-P increased across the MT. Within the 1 to 2 years bracketing the final menstrual period, large HDL-P and HDL size declined while small HDL-P and HDL-triglyceride increased. Although overall HDL-CEC increased across the MT, HDL-CEC per HDL-P declined. Higher concentrations of total, large, and medium HDL-P and greater HDL size were associated with greater HDL-CEC while of small HDL-P were associated with lower HDL-CEC. Associations of large HDL-P and HDL size with HDL-CEC varied significantly across the MT such that higher large HDL-P concentrations and greater HDL size were associated with lower HDL-CEC within the 1 to 2 years around the final menstrual period. CONCLUSIONS Although HDL cholesterol increased over the MT, HDL subclasses and lipid content showed adverse changes. While overall HDL-CEC increased, HDL-CEC per HDL-P declined, consistent with reduced function per particle. Large HDL-P may become less efficient in promoting HDL-CEC during the MT.
Collapse
Affiliation(s)
| | - Xirun Chen (陈曦润)
- Graduate School of Public Health (S.R.E., X.C., A.N., M.M.B., T.J.O.), University of Pittsburgh, PA
| | | | - Jeff Billheimer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia (J.B., D.J.R.)
| | - Maria Mori Brooks
- Graduate School of Public Health (S.R.E., X.C., A.N., M.M.B., T.J.O.), University of Pittsburgh, PA
| | - Dan McConnell
- Central Ligand Assay Satellite Services Laboratories, University of Michigan, Ann Arbor (D.M.)
| | - Trevor J Orchard
- Graduate School of Public Health (S.R.E., X.C., A.N., M.M.B., T.J.O.), University of Pittsburgh, PA
| | - Sybil L Crawford
- Graduate School of Nursing, University of Massachusetts Medical School, Worcester (S.L.C.)
| | | | - Daniel J Rader
- Perelman School of Medicine, University of Pennsylvania, Philadelphia (J.B., D.J.R.)
| |
Collapse
|
44
|
Tassew NG, Laing ST, Shatz W, Crowell SR, Loyet KM, Schuetz C, Blanchette C. Fab-Nanolipoprotein Conjugate Causes Vitreous Opacity and Cataracts Following a Single Intravitreal Administration in New Zealand White Rabbits. Toxicol Pathol 2020; 49:647-655. [PMID: 33733956 DOI: 10.1177/0192623320969671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One strategy employed to prolong the ocular half-life of large molecule therapeutics is via covalent attachment to a carrier, resulting in an increase in size thereby slowing their clearance from the eye. Rabbit antigen-binding fragment conjugated to nanolipoprotein (RabFab-NLP) is a novel conjugate intended to prolong ocular half-life through an increase in hydrodynamic radius compared to Fab alone (∼12 vs ∼3 nm). Nanolipoproteins are mimetics of endogenous high-density lipoproteins and consist of lipids and apolipoproteins (ApoE422k), both biologically derived materials. The objective of this study was to evaluate the ocular toxicity and toxicokinetics of RabFab-NLP after a single intravitreal administration in New Zealand White rabbits. Serum toxicokinetic data suggested a significant increase in ocular residence time of RabFab-NLP compared to RabFab alone. Ophthalmic examinations showed that RabFab-NLP caused vitreous and lens opacities as early as day 3 and day 8 postdose, respectively, which persisted for the entire study duration to day 30. The RabFab-NLP-related microscopic findings were present in the lens, vitreous cavity, and/or optic nerve head. Based on the observed ocular toxicity, a single intravitreal dose of 1.3 mg/eye RabFab-NLP was not tolerated and caused vitreous opacity and cataracts in rabbit eyes.
Collapse
Affiliation(s)
- Nardos G Tassew
- 7412Genentech Inc, Department of Safety Assessment, South San Francisco, CA, USA
| | - Steven T Laing
- 7412Genentech Inc, Department of Safety Assessment, South San Francisco, CA, USA
| | - Whitney Shatz
- 7412Genentech Inc, Protein Chemistry, South San Francisco, CA, USA
| | - Susan R Crowell
- 7412Genentech Inc, Department of Preclinical and Translational Pharmacokinetics & Pharmacodynamics, South San Francisco, CA, USA
| | - Kelly M Loyet
- 7412Genentech Inc, Department of Biochemical Cellular Pharmacology, South San Francisco, CA, USA
| | - Chris Schuetz
- 7412Genentech Inc, Department of Safety Assessment, South San Francisco, CA, USA
| | - Craig Blanchette
- 7412Genentech Inc, Protein Chemistry, South San Francisco, CA, USA
| |
Collapse
|
45
|
Wang Q, Xu X, Zeng Z, Hylkema MN, Cai Z, Huo X. PAH exposure is associated with enhanced risk for pediatric dyslipidemia through serum SOD reduction. ENVIRONMENT INTERNATIONAL 2020; 145:106132. [PMID: 32979814 DOI: 10.1016/j.envint.2020.106132] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/23/2020] [Accepted: 09/09/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Exposure to polycyclic aromatic hydrocarbons (PAHs) is linked to abnormal lipid metabolism, but evidence regarding PAHs as risk factors for dyslipidemia is lacking. OBJECTIVE To investigate the respective role and interaction of PAH exposure and antioxidant consumption in the risk for pediatric dyslipidemia. METHODS We measured the concentrations of serum lipids, superoxide dismutase (SOD) and urinary hydroxylated PAHs (OH-PAHs) in 403 children, of which 203 were from an e-waste-exposed area (Guiyu) and 200 were from a reference area (Haojiang). Biological interactions were calculated by additive models. RESULTS Guiyu children had higher serum triglyceride concentration and dyslipidemia incidence, and lower serum concentration of high-density lipoprotein (HDL) than Haojiang children. Elevated OH-PAH concentration, and concomitant SOD reduction, were both associated with lower HDL concentration and higher hypo-HDL risk (∑3OH-Phes: B for lgHDL = -0.048, P < 0.01; OR for hypo-HDL = 3.708, 95% CI: 1.200, 11.453; SOD: BT3 for lgHDL = 0.061, P < 0.01; ORT3 for hypo-HDL = 0.168, 95% CI: 0.030, 0.941; all were adjusted for confounders). Biological interaction between phenanthrol exposure and SOD reduction was linked to dyslipidemia risk (RERI = 2.783, AP = 0.498, S = 2.537). Children with both risk factors (higher ∑3OH-Phes and lower SOD) had 5.594-times (95% CI: 1.119, 27.958) the dyslipidemia risk than children with neither risk factors (lower ∑3OH-Phes and higher SOD). CONCLUSION High PAH exposure combined with SOD reduction is recommended for predicting elevated risk for pediatric dyslipidemia. Risk assessment of PAH-related dyslipidemia should take antioxidant concentration into consideration.
Collapse
Affiliation(s)
- Qihua Wang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Zhijun Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China
| | - Machteld N Hylkema
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China.
| |
Collapse
|
46
|
Stadler JT, Marsche G. Obesity-Related Changes in High-Density Lipoprotein Metabolism and Function. Int J Mol Sci 2020; 21:E8985. [PMID: 33256096 PMCID: PMC7731239 DOI: 10.3390/ijms21238985] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
In obese individuals, atherogenic dyslipidemia is a very common and important factor in the increased risk of cardiovascular disease. Adiposity-associated dyslipidemia is characterized by low high-density lipoprotein cholesterol (HDL-C) levels and an increase in triglyceride-rich lipoproteins. Several factors and mechanisms are involved in lowering HDL-C levels in the obese state and HDL quantity and quality is closely related to adiponectin levels and the bioactive lipid sphingosine-1-phosphate. Recent studies have shown that obesity profoundly alters HDL metabolism, resulting in altered HDL subclass distribution, composition, and function. Importantly, weight loss through gastric bypass surgery and Mediterranean diet, especially when enriched with virgin olive oil, is associated with increased HDL-C levels and significantly improved metrics of HDL function. A thorough understanding of the underlying mechanisms is crucial for a better understanding of the impact of obesity on lipoprotein metabolism and for the development of appropriate therapeutic approaches. The objective of this review article was to summarize the newly identified changes in the metabolism, composition, and function of HDL in obesity and to discuss possible pathophysiological consequences.
Collapse
Affiliation(s)
- Julia T. Stadler
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
47
|
Gliozzi M, Musolino V, Bosco F, Scicchitano M, Scarano F, Nucera S, Zito MC, Ruga S, Carresi C, Macrì R, Guarnieri L, Maiuolo J, Tavernese A, Coppoletta AR, Nicita C, Mollace R, Palma E, Muscoli C, Belzung C, Mollace V. Cholesterol homeostasis: Researching a dialogue between the brain and peripheral tissues. Pharmacol Res 2020; 163:105215. [PMID: 33007421 DOI: 10.1016/j.phrs.2020.105215] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
Cholesterol homeostasis is a highly regulated process in human body because of its several functions underlying the biology of cell membranes, the synthesis of all steroid hormones and bile acids and the need of trafficking lipids destined to cell metabolism. In particular, it has been recognized that peripheral and central nervous system cholesterol metabolism are separated by the blood brain barrier and are regulated independently; indeed, peripherally, it depends on the balance between dietary intake and hepatic synthesis on one hand and its degradation on the other, whereas in central nervous system it is synthetized de novo to ensure brain physiology. In view of this complex metabolism and its relevant functions in mammalian, impaired levels of cholesterol can induce severe cellular dysfunction leading to metabolic, cardiovascular and neurodegenerative diseases. The aim of this review is to clarify the role of cholesterol homeostasis in health and disease highlighting new intriguing aspects of the cross talk between its central and peripheral metabolism.
Collapse
Affiliation(s)
- Micaela Gliozzi
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Francesca Bosco
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Federica Scarano
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Saverio Nucera
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Stefano Ruga
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Cristina Carresi
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Roberta Macrì
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Lorenza Guarnieri
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Annamaria Tavernese
- Division of Cardiology, University Hospital Policlinico Tor Vergata, Rome, Italy.
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Caterina Nicita
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Rocco Mollace
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Ernesto Palma
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Carolina Muscoli
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy.
| | | | - Vincenzo Mollace
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy.
| |
Collapse
|
48
|
van der Boom T, Jia C, Lefrandt JD, Connelly MA, Links TP, Tietge UJF, Dullaart RPF. HDL Cholesterol Efflux Capacity is Impaired in Severe Short-Term Hypothyroidism Despite Increased HDL Cholesterol. J Clin Endocrinol Metab 2020; 105:5881625. [PMID: 32761088 PMCID: PMC7947992 DOI: 10.1210/clinem/dgaa411] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/28/2020] [Indexed: 01/01/2023]
Abstract
CONTEXT Severe hypothyroidism has profound effects on lipoprotein metabolism including high-density lipoprotein (HDL) cholesterol elevations but effects on HDL function metrics are unknown. OBJECTIVE To determine the impact of severe short-term hypothyroidism on HDL particle characteristics, HDL cholesterol efflux capacity (CEC), and HDL antioxidative capacity. DESIGN Observational study with variables measured during severe short-term hypothyroidism (median TSH 81 mU/L) and after 20 weeks of thyroid hormone supplementation (median TSH 0.03 mU/L) (Netherlands Trial Registry ID 7228). SETTING University hospital setting in The Netherlands. PATIENTS Seventeen patients who had undergone a total thyroidectomy for differentiated thyroid carcinoma. MAIN OUTCOME MEASURES HDL particle characteristics (nuclear magnetic resonance spectrometry), CEC (human THP-1-derived macrophage foam cells and apolipoprotein B-depleted plasma), and HDL anti-oxidative capacity (inhibition of low-density lipoprotein oxidation). RESULTS During hypothyroidism plasma total cholesterol, HDL cholesterol and apolipoprotein A-I were increased (P ≤ 0.001). HDL particle concentration was unchanged, but there was a shift in HDL subclasses toward larger HDL particles (P < 0.001). CEC was decreased (P = 0.035), also when corrected for HDL cholesterol (P < 0.001) or HDL particle concentration (P = 0.011). HDL antioxidative capacity did not change. CONCLUSION During severe short-term hypothyroidism CEC, an important antiatherogenic metric of HDL function, is impaired. HDL cholesterol and larger HDL particles are increased but HDL particle concentration is unchanged. Combined, these findings suggest that HDL quality and quantity are not improved, reflecting dysfunctional HDL in hypothyroidism.
Collapse
Affiliation(s)
- Trynke van der Boom
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
- Department of Vascular Medicine, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| | - Congzhuo Jia
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, RB Groningen, The Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joop D Lefrandt
- Department of Vascular Medicine, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, North Carolina
| | - Thera P Links
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
- Correspondence and Reprint Requests: T.P. Links, MD, PhD, University Medical Center Groningen, Department of Endocrinology, PO Box 30.001, 9700 RB Groningen, The Netherlands. E-mail:
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, RB Groningen, The Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| |
Collapse
|
49
|
Santos LP, Umpierre D. Exercise, Cardiovascular Health, and Risk Factors for Atherosclerosis: A Narrative Review on These Complex Relationships and Caveats of Literature. Front Physiol 2020; 11:840. [PMID: 32848823 PMCID: PMC7411151 DOI: 10.3389/fphys.2020.00840] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 06/22/2020] [Indexed: 12/20/2022] Open
Abstract
The following narrative review addresses the relationship between physical activity and exercise with cardiovascular health, focusing primarily on the following risk factors for atherosclerosis: hypertension, dyslipidemia, and vascular function. Cardiovascular diseases are intimately associated with mortality and morbidity, and current societal organization contributes to the incidence of cardiovascular events. A worldwide epidemiological transition to cardiovascular deaths was observed in the last century, with important decrements in physical activity and diet quality. An atherogenic environment started to be the new normal, with risk factors such as dyslipidemia, hypertension, and endothelial dysfunction observed in great portions of the population. Exercise is an important tool to improve overall health. For hypertension, a great amount of evidence now puts exercise as an effective therapeutic tool in the treatment of this condition. The effects of exercise in modifying blood lipid-lipoprotein are less clear. Despite the rationale remaining solid, methodological difficulties impair the interpretation of possible effects in these variables. Vascular function, as assessed by flow-mediated dilatation, is a good measure of overall vascular health and is consistently improved by exercise in many populations. However, in individuals with hypertension, the exercise literature still needs a further description of possible effects on vascular function variables. Physical activity and exercise are associated with improved cardiovascular health, especially with reduced blood pressure, and should be encouraged on the individual and population level. Evidence regarding its effects on blood lipids and flow-mediated dilatation still need solid landmark studies to guide clinical practice.
Collapse
Affiliation(s)
- Lucas P. Santos
- Exercise Pathophysiology Laboratory, Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- National Institute of Science and Technology for Health Technology Assessment (IATS/HCPA), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Health Sciences, Cardiology and Cardiovascular Sciences, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Daniel Umpierre
- Exercise Pathophysiology Laboratory, Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- National Institute of Science and Technology for Health Technology Assessment (IATS/HCPA), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Health Sciences, Cardiology and Cardiovascular Sciences, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
50
|
Gao Q, Wei A, Chen F, Chen X, Ding W, Ding Z, Wu Z, Du R, Cao W. Enhancing PPARγ by HDAC inhibition reduces foam cell formation and atherosclerosis in ApoE deficient mice. Pharmacol Res 2020; 160:105059. [PMID: 32621955 DOI: 10.1016/j.phrs.2020.105059] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022]
Abstract
Atherosclerosis (AS) is a risky cardiovascular disease with limited treatment options. Various pan or type-selective histone deacetylase (HDAC) inhibitors are reportedly atheroprotective against atherosclerosis (AS); however, the key effectors and the main cellular processes that mediate the protective effects remain poorly defined. Here, we report that PPARγ (Peroxisome proliferator-activated receptor gamma), a transcription factor actively involved in lipid metabolism with strong tissue protective and anti-inflammation properties, is a critical mediator of the anti-AS effects by HDAC inhibition. We showed that a well-known pan-HDAC inhibitor TSA (Trichostatin A) reduced foam cell formation of macrophages that is accompanied by a marked elevation of PPARγ and its downstream cholesterol efflux transporter ABCA1 (ATP-binding membrane cassette transport protein A1) and ABCG1. In an AS model of ApoE-/- mice fed on high-fat diet, TSA treatment alleviated AS lesions, similarly increased PPARγ and the downstream cholesterol transporters and mitigated the induction of inflammatory cytokine TNFα and IL-1β. Exploring the potential cause of PPARγ elevation revealed that TSA induced the acetylation of C/EBPα (CCAAT enhancer binding protein alpha), the upstream regulator of PPARγ, through which it increased PPARγ transactivation. More importantly, we generated a strain of PPARγ/ApoE double knockout mice and demonstrated that lack of PPARγ abrogated the protective effects of TSA on foam cell formation of peritoneal macrophages and the AS pathogenesis. Taken together, these results unravel that C/EBPα and PPARγ are the HDAC-sensitive components of an epigenetic signaling pathway mediating foam cell formation and AS development, and suggest that targeting C/EBPα/PPARγ axis by HDAC inhibitors possesses therapeutic potentials in retarding the progression of AS and the related cardiovascular diseases.
Collapse
Affiliation(s)
- Qi Gao
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Ai Wei
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Fang Chen
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Xingren Chen
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Wenwen Ding
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Zhiquan Ding
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Zhiwei Wu
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Ronghui Du
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China.
| | - Wangsen Cao
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China.
| |
Collapse
|