1
|
Anderson JR, Phelan MM, Caamaño-Gutiérrez E, Clegg PD, Rubio-Martinez LM, Peffers MJ. Metabolomic and proteomic stratification of equine osteoarthritis. Equine Vet J 2025. [PMID: 39972657 DOI: 10.1111/evj.14490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Equine osteoarthritis (OA) is predominantly diagnosed through clinical examination and radiography, leading to detection only after significant joint pathology. The pathogenesis of OA remains unclear and while many medications modify the disease's inflammatory components, no curative or reversal treatments exist. Identifying differentially abundant metabolites and proteins correlated with osteoarthritis severity could improve early diagnosis, track disease progression, and evaluate responses to interventions. OBJECTIVES To identify molecular markers of osteoarthritis severity based on histological and macroscopic grading. STUDY DESIGN Cross-sectional study. METHODS Post-mortem synovial fluid was collected from 58 Thoroughbred racehorse joints and 83 joints from mixed breeds. Joints were histologically and macroscopically scored and categorised by OA and synovitis grade. Synovial fluid nuclear magnetic resonance metabolomic and mass spectrometry proteomic analyses were performed, individually and combined. RESULTS In Thoroughbreds, synovial fluid concentrations of metabolites 2-aminobutyrate, alanine and creatine were elevated for higher OA grades, while glutamate was reduced for both Thoroughbreds and mixed breeds. In mixed breeds, concentrations of three uncharacterised proteins, lipopolysaccharide binding protein and immunoglobulin kappa constant were lower for higher OA grades; concentrations of an uncharacterised protein were higher for OA grade 1 only, and apolipoprotein A1 concentrations were higher for OA grades 1 and 2 compared with lower grades. For Thoroughbreds, gelsolin concentrations were lower for higher OA grades, and afamin was lower at a higher synovitis grade. Correlation analyses of combined metabolomics and proteomics datasets revealed 58 and 32 significant variables for Thoroughbreds and mixed breeds, respectively, with correlations from -0.48 to 0.42 and -0.44 to 0.49. MAIN LIMITATIONS The study's reliance on post-mortem assessments limits correlation with clinical osteoarthritis severity. CONCLUSIONS Following stratification of equine OA severity through histological and macroscopic grading, synovial fluid metabolomic and proteomic profiling identified markers that may support earlier diagnosis and progression tracking. Further research is needed to correlate these markers with clinical osteoarthritis severity.
Collapse
Affiliation(s)
- James R Anderson
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Veterinary Anatomy, Physiology and Pathology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Marie M Phelan
- NMR Metabolomics Facility, Liverpool Shared Research Facilities (LivSRF) & Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| | - Eva Caamaño-Gutiérrez
- Computational Biology Facility, Technology Directorate & Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| | - Peter D Clegg
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Luis M Rubio-Martinez
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Equine Clinical Science, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK
- Sussex Equine Hospital, West Sussex, UK
| | - Mandy J Peffers
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
2
|
Futatsugi A, Tozuka M, Horiuchi Y, Ohkawa R, Kosho T. High-density lipoprotein functionality in cholesterol efflux in early childhood is related to the content ratio of triglyceride to cholesterol. Sci Rep 2024; 14:23323. [PMID: 39375444 PMCID: PMC11458590 DOI: 10.1038/s41598-024-74699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024] Open
Abstract
Cholesterol efflux capacity (CEC), commonly measured as a useful risk marker of atherosclerotic cardiovascular disease, depends on high-density lipoprotein (HDL) functionality and its concentration. We defined the relative HDL functionality in cholesterol efflux, not influenced by HDL concentration, as the ratio of measured CEC to standardized CEC (stCEC) based on HDL-cholesterol (HDL-C) of each individual using the curve regression equation obtained from the correlation. HDL-C, CEC, and CEC/stCEC levels in the < 28-day-old participants (neonates) were significantly low compared to those of the ≥ 28-day-old participants, indicating that the low CEC levels in the neonates depend on not only lower HDL-C but also lower HDL functionality. The low level of CEC/stCEC was remarkable in neonates born at < 34 weeks of gestation and did not improved to the reference level (1.000) until the infantile period. The relatively low or high CEC/stCEC ratios in neonates and infants were associated with lower or higher HDL-TG and HDL-TG/HDL-C ratio, respectively. However, no apparent effect of HDL-TG and HDL-TG/HDL-C ratio on CEC/stCEC was observed in the ≥ 1-year-old participants, indicating that HDL functionality in cholesterol efflux could be associated with the various HDL particles with various lipid compositions, but not just with HDL-TG and HDL-TG/HDL-C ratio.
Collapse
Affiliation(s)
- Akiko Futatsugi
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Clinical Laboratory, Nagano Children's Hospital, 3100 Toyoshina, Azumino, 399-8288, Japan
| | - Minoru Tozuka
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
- Life Science Research Center, Nagano Children's Hospital, 3100, Toyoshina, Azumino, 399-8288, Japan.
| | - Yuna Horiuchi
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, 6-8-1 Hinode, Urayasu, 279-0013, Japan
| | - Ryunosuke Ohkawa
- Department of Clinical Bioanalysis and Molecular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Tomoki Kosho
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Center for Medical Genetics, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Research Center for Supports to Advanced Science, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- BioBank Shinshu, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| |
Collapse
|
3
|
Environmental and Lifestyle Risk Factors in the Carcinogenesis of Gallbladder Cancer. J Pers Med 2022; 12:jpm12020234. [PMID: 35207722 PMCID: PMC8877116 DOI: 10.3390/jpm12020234] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/08/2021] [Accepted: 12/23/2021] [Indexed: 02/01/2023] Open
Abstract
Gallbladder cancer (GBC) is an aggressive neoplasm that in an early stage is generally asymptomatic and, in most cases, is diagnosed in advanced stages with a very low life expectancy because there is no curative treatment. Therefore, understanding the early carcinogenic mechanisms of this pathology is crucial to proposing preventive strategies for this cancer. The main risk factor is the presence of gallstones, which are associated with some environmental factors such as a sedentary lifestyle and a high-fat diet. Other risk factors such as autoimmune disorders and bacterial, parasitic and fungal infections have also been described. All these factors can generate a long-term inflammatory state characterized by the persistent activation of the immune system, the frequent release of pro-inflammatory cytokines, and the constant production of reactive oxygen species that result in a chronic damage/repair cycle, subsequently inducing the loss of the normal architecture of the gallbladder mucosa that leads to the development of GBC. This review addresses how the different risk factors could promote a chronic inflammatory state essential to the development of gallbladder carcinogenesis, which will make it possible to define some strategies such as anti-inflammatory drugs or public health proposals in the prevention of GBC.
Collapse
|
4
|
Zhu Z, Ju J, Zhang M, Yang H, Wei W, Zhang Y. Bisphenol A disturbs hepatic apolipoprotein A1 expression and cholesterol metabolism in rare minnow Gobiocypris rarus. Comp Biochem Physiol C Toxicol Pharmacol 2022; 252:109239. [PMID: 34748970 DOI: 10.1016/j.cbpc.2021.109239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/07/2021] [Accepted: 10/31/2021] [Indexed: 11/03/2022]
Abstract
Bisphenol A (BPA) is a well-known plasticizer, which is widely distributed in the aquatic environment. Lots of studies showed that BPA could lead to lipid metabolism disorder in fish, but few studies studied the mechanism from the perspective of lipid transport. Apolipoprotein A1 (ApoA1) is the main component of high-density lipoprotein (HDL), and plays important roles in reverse cholesterol transport (RCT). In this study, we investigated the effect and molecular mechanism of BPA on ApoA1 and its effect on cholesterol in adult male rare minnow. Results showed that BPA could disturb hepatic ApoA1 expression through regulating Esrrg recruitment and DNA methylation in its promoter region, and ultimately up-regulated ApoA1 protein levels. The increased hepatic ApoA1 improved HDL-C levels, enhanced RCT, and disrupted cholesterol levels. The present study reveals the effect and mechanism of BPA on fish cholesterol metabolism from the perspective of cholesterol transport.
Collapse
Affiliation(s)
- Zhu Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jian Ju
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Meng Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Hui Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Wenzhi Wei
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yingying Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
5
|
Kajani S, Curley S, O'Reilly ME, Yin X, Dillon ET, Guo W, Nilaweera KN, Brennan L, Roche HM, McGillicuddy FC. Sodium salicylate rewires hepatic metabolic pathways in obesity and attenuates IL-1β secretion from adipose tissue - implications for obesity-impaired reverse cholesterol transport. Mol Metab 2021; 56:101425. [PMID: 34954383 PMCID: PMC8762459 DOI: 10.1016/j.molmet.2021.101425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/13/2021] [Accepted: 12/21/2021] [Indexed: 11/26/2022] Open
Abstract
Introduction High-fat diet (HFD)-induced obesity impairs clearance of cholesterol through the Reverse Cholesterol Transport (RCT) pathway, with downregulation in hepatic expression of cholesterol and bile acid transporters, namely ABCG5/8 and ABCB11, and reduced high-density lipoprotein (HDL) cholesterol efflux capacity (CEC). In the current study, we hypothesized that the development of hepatosteatosis, secondary to adipose-tissue dysfunction, contributes to obesity-impaired RCT and that such effects could be mitigated using the anti-inflammatory drug sodium salicylate (NaS). Materials and methods C57BL/6J mice, fed HFD ± NaS or low-fat diet (LFD) for 24 weeks, underwent glucose and insulin tolerance testing. The 3H-cholesterol movement from macrophage-to-feces was assessed in vivo. HDL-CEC was determined ex vivo. Cytokine secretion from adipose-derived stromal vascular fraction (SVF) cells was measured ex vivo. Liver and HDL proteins were determined by mass spectrometry and analyzed using Ingenuity Pathway Analysis. Results NaS delayed HFD-induced weight gain, abrogated priming of pro-IL-1β in SVFs, attenuated insulin resistance, and prevented steatohepatitis (ectopic fat accumulation in the liver). Prevention of hepatosteatosis coincided with increased expression of PPAR-alpha/beta-oxidation proteins with NaS and reduced expression of LXR/RXR-induced proteins including apolipoproteins. The latter effects were mirrored within the HDL proteome in circulation. Despite remarkable protection shown against steatosis, HFD-induced hypercholesterolemia and repression of the liver-to-bile cholesterol transporter, ABCG5/8, could not be rescued with NaS. Discussions and conclusions The cardiometabolic health benefits of NaS may be attributed to the reprogramming of hepatic metabolic pathways to increase fatty acid utilization in the settings of nutritional overabundance. Reduced hepatic cholesterol levels, coupled with reduced LXR/RXR-induced proteins, may underlie the lack of rescue of ABCG5/8 expression with NaS. This remarkable protection against HFD-induced hepatosteatosis did not translate to improvements in cholesterol homeostasis. Sodium salicylate (NaS) initially delays weight-gain in mice fed high-fat diet (HFD) - catch-up evident in weeks 12–24. NaS prevents HFD-induced insulin resistance, hepatosteatosis and pro-IL-1β priming in adipose tissue even upon weight-gain. Hepatic expression of proteins involved in beta oxidation, oxidative phosphorylation and TCA cycle upregulated with NaS. Hepatic expression of LXR/RXR proteins eg. apolipoproteins reduced with NaS; these effects were mirrored in HDL proteome. NaS failed to improve HFD-impaired Reverse Cholesterol Transport or hypercholesterolemia despite preventing hepatosteatosis.
Collapse
Affiliation(s)
- Sarina Kajani
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health
| | - Sean Curley
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health
| | - Marcella E O'Reilly
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health
| | - Xiaofei Yin
- UCD Conway Institute; UCD Institute of Food and Health; School of Agriculture and Food Science, University College Dublin, Dublin 4, Ireland
| | | | - Weili Guo
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health
| | - Kanishka N Nilaweera
- Teagasc Food Research Centre; VistaMilk Research Centre, Moorepark, Fermoy, Ireland
| | - Lorraine Brennan
- UCD Conway Institute; UCD Institute of Food and Health; School of Agriculture and Food Science, University College Dublin, Dublin 4, Ireland
| | - Helen M Roche
- Diabetes Complications Research Centre; UCD Conway Institute; Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science; UCD Institute of Food and Health
| | - Fiona C McGillicuddy
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health.
| |
Collapse
|
6
|
Zhang Y, Zhang M, Zhu Z, Yang H, Wei W, Li B. Bisphenol A regulates apolipoprotein A1 expression through estrogen receptors and DNA methlylation and leads to cholesterol disorder in rare minnow testis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 241:105999. [PMID: 34678657 DOI: 10.1016/j.aquatox.2021.105999] [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: 07/19/2021] [Revised: 10/02/2021] [Accepted: 10/08/2021] [Indexed: 06/13/2023]
Abstract
Bisphenol A (BPA) is a well-known plasticizer that widely distributed in the aquatic environment. BPA has many adverse effects on reproduction. However, few studies have investigated the mechanism of BPA affecting reproduction from the perspective of lipid metabolism. Apolipoprotein A1 (ApoA1) is the major component of high-density lipoprotein (HDL), and plays critical roles in reverse cholesterol transport (RCT). In this study, in order to investigate the effect and molecular mechanism of BPA on testicular ApoA1 and the role of ApoA1 in BPA induced abnormal spermatogenesis, adult male rare minnow Gobiocypris rarus were exposed to 15 μg/L of BPA for 1, 3 and 5 weeks. Results showed that BPA could significantly affect testicular ApoA1 mRNA and protein levels, testicular cholesterol levels, plasmatic sex hormone levels and the integrity of sperm head membrane. The main mechanism of BPA regulating ApoA1 expression is to alter Esr recruitment and CpG sites DNA methylation in ApoA1 promoter. The induced ApoA1 up-regulated high density lipoprotein cholesterol levels and enhanced RCT, and finally decreased the testicular free cholesterol levels. This is likely a key mechanism by which BPA induces sex hormone disorder and sperm head membrane damage. The present study reveals the mechanism by which BPA interferes with spermatogenesis from the perspective of cholesterol transport.
Collapse
Affiliation(s)
- Yingying Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Meng Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Zhu Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Hui Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Wenzhi Wei
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Bichun Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
7
|
Song SO, Hwang YC, Ryu HU, Kahn SE, Leonetti DL, Fujimoto WY, Boyko EJ. Lower High-Density Lipoprotein Cholesterol Concentration Is Independently Associated with Greater Future Accumulation of Intra-Abdominal Fat. Endocrinol Metab (Seoul) 2021; 36:835-844. [PMID: 34474518 PMCID: PMC8419607 DOI: 10.3803/enm.2021.1130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/15/2021] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Both intra-abdominal fat (IAF) and high-density lipoprotein cholesterol (HDL-C) are known to be associated with cardiometabolic health. We evaluated whether the accumulation of computed tomography (CT)-measured IAF over 5 years was related to baseline HDL-C concentration in a prospective cohort study. METHODS All participants were Japanese-Americans between the ages of 34 and 74 years. Plasma HDL-C concentration and CT measurements of IAF, abdominal subcutaneous fat (SCF), and thigh SCF cross-sectional areas were assessed at baseline and at 5-year follow-up visits. RESULTS A total of 397 subjects without diabetes were included. The mean±standard deviation HDL-C concentration was 51.6±13.0 mg/dL in men and 66.0±17.0 mg/dL in women, and the IAF was 91.9±48.4 cm2 in men and 63.1±39.5 cm2 in women. The baseline plasma concentration of HDL-C was inversely associated with the change in IAF over 5 years using multivariable regression analysis with adjustment for age, sex, family history of diabetes, weight change over 5 years, and baseline measurements of body mass index, IAF, abdominal SCF, abdominal circumference, thigh SCF, and homeostatic model assessment for insulin resistance. CONCLUSION These results demonstrate that HDL-C concentration significantly predicts future accumulation of IAF over 5 years independent of age, sex, insulin sensitivity, and body composition in Japanese-American men and women without diabetes.
Collapse
Affiliation(s)
- Sun Ok Song
- Epidemiologic Research and Information Center, VA Puget Sound Health Care System, Seattle, WA,
USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA,
USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang,
Korea
| | - You-Cheol Hwang
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul,
Korea
| | - Han Uk Ryu
- Department of Neurology, Jeonbuk National University Medical School, Jeonju,
Korea
| | - Steven E. Kahn
- Hospital and Specialty Medicine Service, VA Puget Sound Health Care System, Seattle, WA,
USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA,
USA
| | - Donna L. Leonetti
- Department of Anthropology, University of Washington, Seattle, WA,
USA
| | - Wilfred Y. Fujimoto
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA,
USA
| | - Edward J. Boyko
- Epidemiologic Research and Information Center, VA Puget Sound Health Care System, Seattle, WA,
USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA,
USA
| |
Collapse
|
8
|
Wang G, Deng J, Li J, Wu C, Dong H, Wu S, Zhong Y. The Role of High-Density Lipoprotein in COVID-19. Front Pharmacol 2021; 12:720283. [PMID: 34335279 PMCID: PMC8322438 DOI: 10.3389/fphar.2021.720283] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/06/2021] [Indexed: 01/08/2023] Open
Abstract
The current Coronavirus disease 2019 (COVID-19) pandemic has become a global challenge. Managing a large number of acutely ill patients in a short time, whilst reducing the fatality rate and dealing with complications, brings unique difficulties. The most striking pathophysiological features of patients with severe COVID-19 are dysregulated immune responses and abnormal coagulation function, which can result in multiple-organ failure and death. Normally metabolized high-density lipoprotein (HDL) performs several functions, including reverse cholesterol transport, direct binding to lipopolysaccharide (LPS) to neutralize LPS activity, regulation of inflammatory response, anti-thrombotic effects, antioxidant, and anti-apoptotic properties. Clinical data shows that significantly decreased HDL levels in patients with COVID-19 are correlated with both disease severity and mortality. However, the role of HDL in COVID-19 and its specific mechanism remain unclear. In this analysis, we review current evidence mainly in the following areas: firstly, the pathophysiological characteristics of COVID-19, secondly, the pleiotropic properties of HDL, thirdly, the changes and clinical significance of HDL in COVID-19, and fourthly the prospect of HDL-targeting therapy in COVID-19 to clarify the role of HDL in the pathogenesis of COVID-19 and discuss the potential of HDL therapy in COVID-19.
Collapse
Affiliation(s)
- Guyi Wang
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiayi Deng
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinxiu Li
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenfang Wu
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haiyun Dong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shangjie Wu
- Department of Respiratory, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanjun Zhong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Bekhit AEDA, Giteru SG, Holman BWB, Hopkins DL. Total volatile basic nitrogen and trimethylamine in muscle foods: Potential formation pathways and effects on human health. Compr Rev Food Sci Food Saf 2021; 20:3620-3666. [PMID: 34056832 DOI: 10.1111/1541-4337.12764] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/18/2022]
Abstract
The use of total volatile basic nitrogen (TVB-N) as a quality parameter for fish is rapidly growing to include other types of meat. Investigations of meat quality have recently focused on TVB-N as an index of freshness, but little is known on the biochemical pathways involved in its generation. Furthermore, TVB-N and methylated amines have been reported to exert deterimental health effects, but the relationship between these compounds and human health has not been critically reviewed. Here, literature on the formative pathways of TVB-N has been reviewed in depth. The association of methylated amines and human health has been critically evaluated. Interventions to mitigate the effects of TVB-N on human health are discussed. TVB-N levels in meat can be influenced by the diet of an animal, which calls for careful consideration when using TVB-N thresholds for regulatory purposes. Bacterial contamination and temperature abuse contribute to significant levels of post-mortem TVB-N increases. Therefore, controlling spoilage factors through a good level of hygiene during processing and preservation techniques may contribute to a substantial reduction of TVB-N. Trimethylamine (TMA) constitutes a significant part of TVB-N. TMA and trimethylamine oxide (TMA-N-O) have been related to the pathogenesis of noncommunicable diseases, including atherosclerosis, cancers, and diabetes. Proposed methods for mitigation of TMA and TMA-N-O accumulation are discussed, which include a reduction in their daily dietary intake, control of internal production pathways by targeting gut microbiota, and inhibition of flavin monooxygenase 3 enzymes. The levels of TMA and TMA-N-O have significant health effects, and this should, therefore, be considered when evaluating meat quality and acceptability. Agreed international values for TVB-N and TMA in meat products are required. The role of feed, gut microbiota, and translocation of methylated amines to muscles in farmed animals requires further investigation.
Collapse
Affiliation(s)
| | - Stephen G Giteru
- Department of Food Science, University of Otago, Dunedin, New Zealand.,Food & Bio-based Products, AgResearch Limited, Tennent Drive, Palmerston North, 4410, New Zealand
| | - Benjamin W B Holman
- Centre for Red Meat and Sheep Development, NSW Department of Primary Industries, Cowra, New South Wales, Australia
| | - David L Hopkins
- Centre for Red Meat and Sheep Development, NSW Department of Primary Industries, Cowra, New South Wales, Australia
| |
Collapse
|
10
|
Ahmed MO, Byrne RE, Pazderska A, Segurado R, Guo W, Gunness A, Frizelle I, Sherlock M, Ahmed KS, McGowan A, Moore K, Boran G, McGillicuddy FC, Gibney J. HDL particle size is increased and HDL-cholesterol efflux is enhanced in type 1 diabetes: a cross-sectional study. Diabetologia 2021; 64:656-667. [PMID: 33169205 DOI: 10.1007/s00125-020-05320-3] [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: 05/06/2020] [Accepted: 09/09/2020] [Indexed: 01/02/2023]
Abstract
AIMS/HYPOTHESIS The prevalence of atherosclerosis is increased in type 1 diabetes despite normal-to-high HDL-cholesterol levels. The cholesterol efflux capacity (CEC) of HDL is a better predictor of cardiovascular events than static HDL-cholesterol. This cross-sectional study addressed the hypothesis that impaired HDL function contributes to enhanced CVD risk within type 1 diabetes. METHODS We compared HDL particle size and concentration (by NMR), total CEC, ATP-binding cassette subfamily A, member 1 (ABCA1)-dependent CEC and ABCA1-independent CEC (by determining [3H]cholesterol efflux from J774-macrophages to ApoB-depleted serum), and carotid intima-media thickness (CIMT) in 100 individuals with type 1 diabetes (37.6 ± 1.2 years; BMI 26.9 ± 0.5 kg/m2) and 100 non-diabetic participants (37.7 ± 1.1 years; 27.1 ± 0.5 kg/m2). RESULTS Compared with non-diabetic participants, total HDL particle concentration was lower (mean ± SD 31.01 ± 8.66 vs 34.33 ± 8.04 μmol/l [mean difference (MD) -3.32 μmol/l]) in participants with type 1 diabetes. However, large HDL particle concentration was greater (9.36 ± 3.98 vs 6.99 ± 4.05 μmol/l [MD +2.37 μmol/l]), resulting in increased mean HDL particle size (9.82 ± 0.57 vs 9.44 ± 0.56 nm [MD +0.38 nm]) (p < 0.05 for all). Total CEC (14.57 ± 2.47%CEC/4 h vs 12.26 ± 3.81%CEC/4 h [MD +2.31%CEC/4 h]) was greater in participants with type 1 diabetes relative to non-diabetic participants. Increased HDL particle size was independently associated with increased total CEC; however, following adjustment for this in multivariable analysis, CEC remained greater in participants with type 1 diabetes. Both components of CEC, ABCA1-dependent (6.10 ± 2.41%CEC/4 h vs 5.22 ± 2.57%CEC/4 h [MD +0.88%CEC/4 h]) and ABCA1-independent (8.47 ± 1.79% CEC/4 h vs 7.05 ± 1.76% CEC/4 h [MD +1.42% CEC/4 h]) CEC, were greater in type 1 diabetes but the increase in ABCA1-dependent CEC was less marked and not statistically significant in multivariable analysis. CIMT was increased in participants with type 1 diabetes but in multivariable analysis it was only associated negatively with age and BMI. CONCLUSIONS/INTERPRETATION HDL particle size but not HDL-cholesterol level is independently associated with enhanced total CEC. HDL particle size is greater in individuals with type 1 diabetes but even after adjusting for this, total and ABCA1-independent CEC are enhanced in type 1 diabetes. Further studies are needed to understand the mechanisms underlying these effects, and whether they help attenuate progression of atherosclerosis in this high-risk group. Graphical abstract.
Collapse
Affiliation(s)
- Mohamad O Ahmed
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Rachel E Byrne
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Agnieszka Pazderska
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Ricardo Segurado
- School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Belfield, Dublin, Ireland
| | - Weili Guo
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Anjuli Gunness
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Isolda Frizelle
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Mark Sherlock
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Khalid S Ahmed
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Anne McGowan
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Kevin Moore
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Gerard Boran
- Department of Chemical Pathology, Tallaght University Hospital, Dublin, Ireland
| | - Fiona C McGillicuddy
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - James Gibney
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland.
| |
Collapse
|
11
|
Mesgari-Abbasi M, Mahmoudinezhad M, Farhangi MA. Soluble P-selectin, procalcitonin, transforming growth factor (TGF)-β and apo-proteins in association with the components of metabolic syndrome in obese individuals. Clin Nutr ESPEN 2021; 41:386-390. [PMID: 33487294 DOI: 10.1016/j.clnesp.2020.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/07/2020] [Accepted: 10/25/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND & AIMS Metabolic syndrome (MetS) is a clinical disorder with widespread prevalence. In the current study, we aimed to investigate the association between serum soluble P-selectin, procalcitonin, transforming growth factor (TGF)-β and apo-proteins with the components of metabolic syndrome in obese individuals. MATERIAL AND METHODS Sixty two obese patients with MetS and sixty five obese apparently healthy controls were participated in the current case-control study. The participants' anthropometric assessments and systolic and diastolic blood pressure (SBP and DBP) were measured. Serum lipids and the concentrations of ox-LDL, P-selectin, procalcitonin, TGF-β and apo-proteins were measured with commercial ELIZA kits. RESULTS Serum TG and TC were significantly higher in obese subjects with MetS; while TGF-β, procalcitonin, apoprotein B and insulin concentrations were higher in obese non- MetS group. In obese subjects with MetS, procalcitonin was in positive association with ox-LDL and apoprotein-B was in positive association with HDL. In obese subjects without MetS, apoprotein -B was in positive association with WC and HDL and WC. CONCLUSION The current study found several associations between serum lipids and PCT and serum apo-proteins in obese individuals either with or without MetS. Further studies with large sample size are warranted to better elucidate the observed relationships and underlying mechanism. TRIAL REGISTRATION Not Applicable.
Collapse
|
12
|
Curley S, Gall J, Byrne R, Yvan‐Charvet L, McGillicuddy FC. Metabolic Inflammation in Obesity—At the Crossroads between Fatty Acid and Cholesterol Metabolism. Mol Nutr Food Res 2020; 65:e1900482. [DOI: 10.1002/mnfr.201900482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/16/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Sean Curley
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| | - Julie Gall
- University of Nice Unité Mixte de Recherce (UMR) Institut National de la Santé et de la Recherche Médicale U1065 062104 Nice Cedex 3 France
| | - Rachel Byrne
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| | - Laurent Yvan‐Charvet
- University of Nice Unité Mixte de Recherce (UMR) Institut National de la Santé et de la Recherche Médicale U1065 062104 Nice Cedex 3 France
| | - Fiona C. McGillicuddy
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| |
Collapse
|
13
|
Zhang L, Chen B, Xie X, Zhao J, Wei J, Zhang Q, Fan Y, Zhang Y. Association of Waist-to-Height Ratio, Metabolic Syndrome, and Carotid Atherosclerosis in Individuals with a High Risk of Stroke: A Cross-Sectional Study of 9605 Study Participants. Metab Syndr Relat Disord 2020; 18:381-388. [PMID: 32589495 DOI: 10.1089/met.2020.0003] [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: 11/13/2022] Open
Abstract
Background: To determine the association of waist-to-height ratio (WHtR), metabolic syndrome (MetS), and carotid atherosclerosis (CAS) in individuals with a high risk of stroke. Methods: We performed a cross-sectional analysis of 9605 study responders from eight urban area communities in Northern China. Height, weight, waist circumference, blood pressure (BP), and blood lipid were measured. Information of population characteristics, smoking status, alcohol consumption, physical activity, and diet were determined by validated questionnaire. Results: A total of 9605 study responders were included in this study. The average age was 60 ± 9 years with 5911 (61.5%) patients being females. The prevalence of MetS and CAS was 26.2% and 75.1%, respectively. WHtR was significantly associated with CAS using our final adjusted model [odds ratio (OR): 1.233, 95% confidence interval (CI): 1.096-1.378]. The association of CAS with hypertension and hyperglycemia were statistically significant among factors that constitute MetS. Additional risk factors affecting the development of CAS included age, previous stroke, and smoking history (P < 0.05). Conclusion: WHtR was determined to perform better compared with other traditional indicators for correlating CAS. We believe that WHtR is a better indicator for the early identification of CAS in individuals with a high risk of stroke. This will facilitate the early detection and intervention of CAS.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurology, Dongfang Hospital, The Second Clinical Medical College of Beijing University of Chinese Medicine, Beijing, China
| | - Baoxin Chen
- Department of Neurology, Dongfang Hospital, The Second Clinical Medical College of Beijing University of Chinese Medicine, Beijing, China
| | - Xinran Xie
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jiayi Zhao
- Department of Rehabilitation, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingjing Wei
- Beijing University of Chinese Medicine, Beijing, China
| | - Qihui Zhang
- Department of Neurology, Dongfang Hospital, The Second Clinical Medical College of Beijing University of Chinese Medicine, Beijing, China
| | - Yue Fan
- Department of Neurology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yunling Zhang
- Department of Neurology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Sura SR, Germain-Lee EL. Treatment of rickets and dyslipidemia in twins with progressive familial intrahepatic cholestasis type 2. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2020; 2020:9. [PMID: 32508937 PMCID: PMC7249403 DOI: 10.1186/s13633-020-00079-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/24/2020] [Indexed: 12/01/2022]
Abstract
Background Progressive Familial Intrahepatic Cholestasis Type 2 (PFIC2) is a rare congenital cholestatic liver disease that progresses to end stage liver disease. It is associated with fat soluble vitamin D deficiency rickets and severe dyslipidemia; however, treatment of these secondary effects remains a challenge. Case presentation One year old twin males born to a mother with intrahepatic cholestasis during pregnancy presented with jaundice, pruritus and failure to thrive. Lab evaluation revealed significant transaminitis, direct hyperbilirubinemia and normal gamma glutamyl transferase (GGT). Genetic studies confirmed PFIC2. Further evaluation for fat soluble vitamin deficiencies revealed severe vitamin D deficiency rickets. High dose vitamin D replacement therapy using Ergocalciferol (Vitamin D2) 50,000 IU three times a week over 10 weeks led to the improvement of Vitamin D, 25-Hydroxy (25-OH) serum levels and resolution of rickets. Dyslipidemia with very low high density lipoprotein-cholesterol (HDL-C) and high triglycerides was more profound in our patients compared to what has been described in the literature thus far. The dyslipidemia improved 2 months after internal biliary diversion. Conclusions Higher doses of Vitamin D therapy are needed for treatment of rickets secondary to cholestasis. Extremely low HDL-C levels are characteristic of PFIC and improve with treatment of underlying cholestasis. Maternal intrahepatic cholestasis during pregnancy can be an early warning sign.
Collapse
Affiliation(s)
- Sunitha R Sura
- Connecticut Children's Medical Center, 505 Farmington Avenue, Farmington, CT 06032 USA.,Department of Pediatrics, University of Connecticut School of Medicine, 505 Farmington Avenue, Farmington, CT 06032 USA
| | - Emily L Germain-Lee
- Connecticut Children's Medical Center, 505 Farmington Avenue, Farmington, CT 06032 USA.,Department of Pediatrics, University of Connecticut School of Medicine, 505 Farmington Avenue, Farmington, CT 06032 USA
| |
Collapse
|
15
|
Song SO, Hwang YC, Kahn SE, Leonetti DL, Fujimoto WY, Boyko EJ. Intra-Abdominal Fat and High Density Lipoprotein Cholesterol Are Associated in a Non-Linear Pattern in Japanese-Americans. Diabetes Metab J 2020; 44:277-285. [PMID: 32174061 PMCID: PMC7188973 DOI: 10.4093/dmj.2019.0008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 07/04/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND We describe the association between high density lipoprotein cholesterol (HDL-C) concentration and computed tomography (CT)-measured fat depots. METHODS We examined the cross-sectional associations between HDL-C concentration and intra-abdominal (IAF), abdominal subcutaneous (SCF), and thigh fat (TF) areas in 641 Japanese-American men and women. IAF, SCF, and TF were measured by CT at the level of the umbilicus and mid-thigh. The associations between fat area measurements and HDL-C were examined using multivariate linear regression analysis adjusting for age, sex, diabetes family history, homeostasis model assessment of insulin resistance (HOMA-IR), and body mass index (BMI). Non-linearity was assessed using fractional polynomials. RESULTS Mean±standard deviation of HDL-C concentration and IAF in men and women were 1.30±0.34 mg/dL, 105±55.3 cm², and 1.67±0.43 mg/dL, 74.4±46.6 cm² and differed significantly by gender for both comparisons (P<0.001). In univariate analysis, HDL-C concentration was significantly associated with CT-measured fat depots. In multivariate analysis, IAF was significantly and non-linearly associated with HDL-C concentration adjusted for age, sex, BMI, HOMA-IR, SCF, and TF (IAF: β=-0.1012, P<0.001; IAF²: β=0.0008, P<0.001). SCF was also negatively and linearly associated with HDL-C (β=-0.4919, P=0.001). CONCLUSION HDL-C does not linearly decline with increasing IAF in Japanese-Americans. A more complex pattern better fits this association.
Collapse
Affiliation(s)
- Sun Ok Song
- Epidemiologic Research and Information Center, VA Puget Sound Health Care System, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, Korea.
| | - You Cheol Hwang
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul, Korea
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Hospital and Specialty Medicine Service, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Donna L Leonetti
- Department of Anthropology, University of Washington, Seattle, WA, USA
| | - Wilfred Y Fujimoto
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Edward J Boyko
- Epidemiologic Research and Information Center, VA Puget Sound Health Care System, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
16
|
O'Reilly ME, Lenighan YM, Dillon E, Kajani S, Curley S, Bruen R, Byrne R, Heslin AM, Moloney AP, Roche HM, McGillicuddy FC. Conjugated Linoleic Acid and Alpha Linolenic Acid Improve Cholesterol Homeostasis in Obesity by Modulating Distinct Hepatic Protein Pathways. Mol Nutr Food Res 2020; 64:e1900599. [DOI: 10.1002/mnfr.201900599] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 11/11/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Marcella E. O'Reilly
- Nutrigenomics Research Group, School of Public Health Physiotherapy and Sports ScienceUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
| | - Yvonne M. Lenighan
- Nutrigenomics Research Group, School of Public Health Physiotherapy and Sports ScienceUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
| | - Eugene Dillon
- Mass Spectrometry ResourceUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
| | - Sarina Kajani
- Diabetes Complications Research CentreUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
- UCD School of MedicineUniversity College Dublin Dublin 4 Ireland
| | - Sean Curley
- Diabetes Complications Research CentreUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
- UCD School of MedicineUniversity College Dublin Dublin 4 Ireland
| | - Robyn Bruen
- Diabetes Complications Research CentreUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
| | - Rachel Byrne
- Diabetes Complications Research CentreUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
- UCD School of MedicineUniversity College Dublin Dublin 4 Ireland
| | - Aoibhin Moore Heslin
- Nutrigenomics Research Group, School of Public Health Physiotherapy and Sports ScienceUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
| | - Aidan P. Moloney
- TeagascAnimal & Grassland Research and Innovation Centre Meath Ireland
| | - Helen M. Roche
- Nutrigenomics Research Group, School of Public Health Physiotherapy and Sports ScienceUniversity College Dublin Dublin 4 Ireland
- UCD Institute of Food and HealthUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
| | - Fiona C. McGillicuddy
- Diabetes Complications Research CentreUniversity College Dublin Dublin 4 Ireland
- UCD Conway InstituteUniversity College Dublin Dublin 4 Ireland
- UCD School of MedicineUniversity College Dublin Dublin 4 Ireland
| |
Collapse
|
17
|
Salek M, Clark CCT, Taghizadeh M, Jafarnejad S. N-3 fatty acids as preventive and therapeutic agents in attenuating PCOS complications. EXCLI JOURNAL 2019; 18:558-575. [PMID: 31611740 PMCID: PMC6785778 DOI: 10.17179/excli2019-1534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/23/2019] [Indexed: 12/20/2022]
Abstract
To our knowledge, in spite of several trials exploring the beneficial effect of n-3 polyunsaturated fatty acids (PUFA) on polycystic ovary syndrome (PCOS), no comprehensive evidence has investigated the effects of n-3 PUFA consumption on PCOS complications. Therefore, our aim was to conduct a review to investigate the possible effect and related mechanisms. A comprehensive systematic search was conducted in Embase, MEDLINE/PubMed, Google Scholar, and SCOPUS, to identify studies investigating n-3 fatty acids as a preventative or therapeutic agent for the attenuation of PCOS complications. Subsequently, the impact of omega-3 on PCOS, omega-3 and inflammation, omega-3 and insulin resistance, omega-3 and adipokines, omega-3 and lipid metabolism, omega-3 and endothelial function and omega-3 and hormonal factors were discussed. There are multiple mechanisms by which n-3 PUFAs may exert their beneficial effects on PCOS, including anti-obesity, glycemic and hormonal hemostasis, anti-inflammatory, regulation of adipokine production and enhancement of endothelial function.N-3 PUFAs are a promising agent in relieving complications associated with PCOS. Although most of the studies in patients with PCOS reported an improvement in most complications after administration of omega-3 supplements, there is a distinct dearth of studies investigating the dietary intake of these types of fatty acids. Moreover, favorable effects regarding the improvement of dyslipidemia, regulation of adipokines, regulation of hormonal factors and enhancement of endothelial function are limited. Therefore, more trials are warranted to investigate palatable mechanisms for clarifying the metabolic and hormonal effects of these agents in PCOS.
Collapse
Affiliation(s)
- Mina Salek
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Cain C T Clark
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Sadegh Jafarnejad
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
18
|
Horiuchi Y, Ohkawa R, Lai SJ, Yamazaki A, Ikoma H, Yano K, Kameda T, Tozuka M. Characterization of the cholesterol efflux of apolipoprotein E-containing high-density lipoprotein in THP-1 cells. Biol Chem 2019; 400:209-218. [PMID: 30210053 DOI: 10.1515/hsz-2018-0284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/02/2018] [Indexed: 01/02/2023]
Abstract
High-density lipoprotein (HDL), also known as antiatherogenic lipoprotein, consists of heterogeneous particles in terms of size, density and composition, suggesting differences among HDL subclasses in characteristics and functions. We investigated the role of apolipoprotein E (apoE)-containing HDL, a minor HDL subclass, in the cholesterol efflux capacity (CEC) of HDL, which is its predominant atheroprotective function. The CEC of apoE-containing HDL was similar to that of apoE-deficient HDL, but the former exhibited a greater rate increase (1.48-fold) compared to that of the latter (1.10-fold) by the stimulation of THP-1 macrophages with the Liver X Receptor (LXR) agonist. No difference in CEC was observed without the LXR agonist between apoA-I, the main apolipoprotein in HDL, and apoE, whereas the increase in CEC in response to treatment with the LXR agonist was greater for apoA-I (4.25-fold) than for apoE (2.22-fold). Furthermore, the increase in the CEC of apoE-containing HDL induced by the LXR agonist was significantly reduced by treatment with glyburide, an inhibitor of ATP-binding cassette transporter A1 (ABCA1). These results suggest that apoE-containing HDL, unlike apoE-deficient HDL, is involved in cholesterol efflux via ABCA1.
Collapse
Affiliation(s)
- Yuna Horiuchi
- Department of Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryunosuke Ohkawa
- Department of Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shao-Jui Lai
- Department of Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Azusa Yamazaki
- Department of Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hayato Ikoma
- Clinical Laboratory, Hamamatsu University Hospital, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kouji Yano
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Takahiro Kameda
- Department of Medical Technology, School of Health Sciences, Tokyo University of Technology, 5-23-22 Nishikamata, Ota-ku, Tokyo 144-8535, Japan
| | - Minoru Tozuka
- Department of Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
19
|
Tian X, Tang Z. A comparison of fractional flow reserve determination and coronary angiography results in patients with unstable angina and analysis of related factors. J Thorac Dis 2019; 11:549-556. [PMID: 30962999 PMCID: PMC6409283 DOI: 10.21037/jtd.2019.01.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/05/2019] [Indexed: 11/06/2022]
Abstract
BACKGROUND Coronary angiography (CAG) and fractional flow reserve (FFR) are currently used to identify the lesions and guide the treatment of unstable angina (UA) patients. This study aims to compare the two methods and investigate factors affecting FFR value. METHODS A total of 284 UA patients (296 coronary artery lesions) were enrolled from the Emergency Department of Anzhen Hospital Affiliated to Capital Medical University from January 2017 to December 2017. CAG and FFR determination were performed in all patients, and the roles of these two methods in guiding the treatment of UA were compared and analyzed. The subjects were divided into FFR ≤0.8 group and FFR >0.8 group. The general data and laboratory findings were compared between these two groups, and the possible influential factors were analyzed. The statistical analysis of t-test or chi square test was done with SPSS 20.0 software. RESULTS Of 296 UA lesions, 160 (54.1%) had ≥75% angiographic stenosis and 136 (45.9%) had <75% angiographic stenosis; 168 (56.8%) had an FFR value of ≤0.8 and 128 (43.2%) had an FFR value >0.8. There was no significant difference between these two examination methods (P=0.508, χ2=0.438). Further analysis showed that 43 (26.9%) of the 160 lesions with ≥75% stenosis had an FFR value of >0.8 and did not require PCI; 49 (38.3%) of the 128 lesions with 50-70% stenosis had an FFR value of ≤0.8 and needed PCI; 2 of 8 patients with <50% stenosis had an FFR value of ≤0.8 and needed PCI. If FFR was used as the "gold standard" of PCI, the sensitivity, specificity, positive predictive value, and negative predictive value of CAG in guiding PCI for UA were 69.6%, 66.4%, 73.1%, and 62.5%, respectively. Multivariate analysis with Logistic regression revealed low high-density lipoprotein (HDL) and hypertension were independent risk factors of FFR <0.8 in UA patients. CONCLUSIONS CAG and FFR readings could be different. A combination of CAG and FFR may help to achieve more accurate and tailored treatment of UA. The history of hypertension is an independent risk factor for FFR in UA patients, and HDL is an independent protective factor.
Collapse
Affiliation(s)
- Xue Tian
- Emergency Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Zhe Tang
- Emergency Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
20
|
Coelho MS, Soares-Freitas RAM, Arêas JAG, Gandra EA, Salas-Mellado MDLM. Peptides from Chia Present Antibacterial Activity and Inhibit Cholesterol Synthesis. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2018; 73:101-107. [PMID: 29679358 DOI: 10.1007/s11130-018-0668-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In previous studies, it has not been reported that protein isolated from chia interferes favorably with antibacterial activity, and reduces cholesterol synthesis. The objective of this study was to determine whether commonly used commercial microbial proteases can be utilized to generate chia protein-based antibacterial and hypocholesterolemic hydrolysates/peptides, considering the effects of protein extraction method. Alcalase, Flavourzyme and sequential Alcalase-Flavourzyme were used to produce hydrolysates from chia protein (CF), protein-rich fraction (PRF) and chia protein concentrates (CPC1 and CPC2). These hydrolysates were evaluated for their antimicrobial activity against Gram-positive (G+) and Gram-negative (G-) microorganisms. The protein hydrolysates were purified by ultrafiltration through a membrane with 3 kDa nominal molecular weight, for evaluation of hypocholesterolemic activity. An inhibition zone was observed when the hydrolysate was tested against S. aureus, and minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) values were obtained. Peptides from chia protein with molecular mass lower than 3 kDa reduced up to 80.7% of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase) enzymatic reaction velocity. It was also observed that, independent of the method used to obtain chia proteins, the fractions showed relevant bioactivity. Moreover, the intensity of the bioactivity varied with the method for obtaining the protein and with the enzyme used in the hydrolysis process. This is the first report to demonstrate that chia peptides are able to inhibit cholesterol homeostasis.
Collapse
Affiliation(s)
- Michele Silveira Coelho
- Laboratory of Food Technology, School of Chemistry and Food, Federal University of Rio Grande, Av. Italy 8 km, Carreiros, Rio Grande, RS, 96203-900, Brazil.
| | | | - José Alfredo Gomes Arêas
- Faculty of Public Health, University of São Paulo, Av. Dr. Arnaldo, 715, São Paulo, SP, 01246-904, Brazil
| | - Eliezer Avila Gandra
- Center of Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas, Campus do Capão do Leão, Pelotas, RS, 96010-900, Brazil
| | - Myriam de Las Mercedes Salas-Mellado
- Laboratory of Food Technology, School of Chemistry and Food, Federal University of Rio Grande, Av. Italy 8 km, Carreiros, Rio Grande, RS, 96203-900, Brazil
| |
Collapse
|
21
|
Validation and application of a novel cholesterol efflux assay using immobilized liposomes as a substitute for cultured cells. Biosci Rep 2018; 38:BSR20180144. [PMID: 29545317 PMCID: PMC5897742 DOI: 10.1042/bsr20180144] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/08/2018] [Accepted: 03/12/2018] [Indexed: 02/02/2023] Open
Abstract
Estimation of the function as well as the amount of high-density lipoprotein (HDL) is required to predict the risk of cardiovascular disease development. Cholesterol efflux capacity (CEC) is the key metric for determining the antiatherosclerotic function of HDL. However, the assay methods currently used to calculate CEC are not ideal for clinical use as they require the culture of cells. In the present study, we developed a novel CEC assay using immobilized liposome-bound gel beads (ILGs), containing fluorescently labeled cholesterol, as a substitute for cultured cells. When apolipoprotein B-100 depleted serum, obtained by polyethylene glycol precipitation, was used as the cholesterol acceptors, the basic properties of this method, such as the available range of HDL-cholesterol, efflux temperature and time, and normalization parameters, indicate that this method is sufficient to estimate CEC. Furthermore, the CEC values obtained with this ILG method were also correlated with those obtained with a conventional method using THP-1 macrophages derived foam cells and 3H-cholesterol as a tracer (r = 0.932). Overall, this novel cholesterol efflux assay method is a realistic and effective alternative to current methods in the field while also being easier to use in clinical laboratories as neither cell culture, radioisotope nor ultracentrifugation is required.
Collapse
|
22
|
Pizzini A, Lunger L, Demetz E, Hilbe R, Weiss G, Ebenbichler C, Tancevski I. The Role of Omega-3 Fatty Acids in Reverse Cholesterol Transport: A Review. Nutrients 2017; 9:nu9101099. [PMID: 28984832 PMCID: PMC5691715 DOI: 10.3390/nu9101099] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/22/2017] [Accepted: 09/28/2017] [Indexed: 01/31/2023] Open
Abstract
The beneficial effects of omega-3 polyunsaturated fatty acids (n-3 PUFAs) on cardiovascular disease have been studied extensively. However, it remains unclear to what extent n-3 PUFAs may impact Reverse Cholesterol Transport (RCT). RCT describes a mechanism by which excess cholesterol from peripheral tissues is transported to the liver for hepatobiliary excretion, thereby inhibiting foam cell formation and the development of atherosclerosis. The aim of this review is to summarize the literature and to provide an updated overview of the effects of n-3 PUFAs on key players in RCT, including apoliprotein AI (apoA-I), ATP-binding cassette transporter A1 (ABCA1), ABCG1, apoE, scavenger receptor class B type I (SR-BI), cholesteryl ester transfer protein (CETP), low-density lipoprotein receptor (LDLr), cholesterol 7 alpha-hydroxylase (CYP7A1) and ABCG5/G8. Based on current knowledge, we conclude that n-3 PUFAs may beneficially affect RCT, mainly by influencing high-density lipoprotein (HDL) remodeling and by promoting hepatobiliary sterol excretion.
Collapse
Affiliation(s)
- Alex Pizzini
- Department of Internal Medicine II, Infectious Diseases, Pneumology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Lukas Lunger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Egon Demetz
- Department of Internal Medicine II, Infectious Diseases, Pneumology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Richard Hilbe
- Department of Internal Medicine II, Infectious Diseases, Pneumology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Guenter Weiss
- Department of Internal Medicine II, Infectious Diseases, Pneumology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Christoph Ebenbichler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Ivan Tancevski
- Department of Internal Medicine II, Infectious Diseases, Pneumology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
23
|
Nutritional modulation of metabolic inflammation. Biochem Soc Trans 2017; 45:979-985. [PMID: 28710289 DOI: 10.1042/bst20160465] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/11/2017] [Accepted: 05/16/2017] [Indexed: 01/22/2023]
Abstract
Metabolic inflammation is a very topical area of research, wherein aberrations in metabolic and inflammatory pathways probably contribute to atherosclerosis, insulin resistance (IR) and type 2 diabetes. Metabolic insults arising from obesity promote inflammation, which in turn impedes insulin signalling and reverse cholesterol transport (RCT). Key cells in the process are metabolically activated macrophages, which up-regulate both pro- and anti-inflammatory pathways in response to lipid spillover from adipocytes. Peroxisome proliferator-activated receptors and AMP-activated protein kinase (AMPK) are regulators of cellular homeostasis that influence both inflammatory and metabolic pathways. Dietary fats, such as saturated fatty acids (SFAs), can differentially modulate metabolic inflammation. Palmitic acid, in particular, is a well-characterized nutrient that promotes metabolic inflammation via the NLRP3 (the nod-like receptor containing a pyrin domain) inflammasome, which is partly attributable to AMPK inhibition. Conversely, some unsaturated fatty acids are less potent agonists of metabolic inflammation. For example, monounsaturated fatty acid does not reduce AMPK as potently as SFA and n-3 polyunsaturated fatty acids actively resolve inflammation via resolvins and protectins. Nevertheless, the full extent to which nutritional state modulates metabolic inflammation requires greater clarification.
Collapse
|
24
|
Ahmadi Y, Ghorbanihaghjo A, Argani H. The effect of statins on the organs: similar or contradictory? J Cardiovasc Thorac Res 2017; 9:64-70. [PMID: 28740624 PMCID: PMC5516053 DOI: 10.15171/jcvtr.2017.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 06/09/2017] [Indexed: 12/19/2022] Open
Abstract
Hydroxy-Methyl-Glutaryl-CoA reductase (HMGCR) – the main enzyme of the cholesterol biosynthesis pathway – is mostly inhibited by statins in hepatocytes. In spite of the other tissues, liver utilizes cholesterol in different ways such as the synthesis of bile acids, excretion in to the intestine and synthesis of lipoproteins. Therefore, statins theoretically alter these pathways; although, there have not been such effects. In this review, we aim to show the roles of extra-hepatic tissues, in particular intestine, adipose and cutaneous tissues in providing the cholesterol after reduction of the whole body cholesterol content by statins.
Collapse
Affiliation(s)
- Yasin Ahmadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghorbanihaghjo
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Argani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
25
|
Sukenik S, Dunsky S, Barnoy A, Shumilin I, Harries D. TMAO mediates effective attraction between lipid membranes by partitioning unevenly between bulk and lipid domains. Phys Chem Chem Phys 2017; 19:29862-29871. [DOI: 10.1039/c7cp04603k] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
TMAO induces an attractive force between lipid bilayers. The force is traced to the preferential repulsion of the osmolyte from lipid.
Collapse
Affiliation(s)
- Shahar Sukenik
- Institute of Chemistry and the Fritz Haber Research Center
- The Hebrew University
- Jerusalem 91904
- Israel
| | - Shaked Dunsky
- Institute of Chemistry and the Fritz Haber Research Center
- The Hebrew University
- Jerusalem 91904
- Israel
| | - Avishai Barnoy
- Institute of Chemistry and the Fritz Haber Research Center
- The Hebrew University
- Jerusalem 91904
- Israel
| | - Ilan Shumilin
- Institute of Chemistry and the Fritz Haber Research Center
- The Hebrew University
- Jerusalem 91904
- Israel
| | - Daniel Harries
- Institute of Chemistry and the Fritz Haber Research Center
- The Hebrew University
- Jerusalem 91904
- Israel
| |
Collapse
|
26
|
Labadzhyan A, Cui J, Péterfy M, Guo X, Chen YDI, Hsueh WA, Rotter JI, Goodarzi MO. Insulin Clearance Is Associated with Hepatic Lipase Activity and Lipid and Adiposity Traits in Mexican Americans. PLoS One 2016; 11:e0166263. [PMID: 27846285 PMCID: PMC5112869 DOI: 10.1371/journal.pone.0166263] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/25/2016] [Indexed: 12/18/2022] Open
Abstract
Reduction in insulin clearance plays an important role in the compensatory response to insulin resistance. Given the importance of this trait to the pathogenesis of diabetes, a deeper understanding of its regulation is warranted. Our goal was to identify metabolic and cardiovascular traits that are independently associated with metabolic clearance rate of insulin (MCRI). We conducted a cross-sectional analysis of metabolic and cardiovascular traits in 765 participants from the Mexican-American Coronary Artery Disease (MACAD) project who had undergone blood sampling, oral glucose tolerance test, euglycemic-hyperinsulinemic clamp, dual-energy X-ray absorptiometry, and carotid ultrasound. We assessed correlations of MCRI with traits from seven domains, including anthropometry, biomarkers, cardiovascular, glucose homeostasis, lipase activity, lipid profile, and liver function tests. We found inverse independent correlations between MCRI and hepatic lipase (P = 0.0004), insulin secretion (P = 0.0002), alanine aminotransferase (P = 0.0045), total fat mass (P = 0.014), and diabetes (P = 0.03). MCRI and apolipoprotein A-I exhibited a positive independent correlation (P = 0.035). These results generate a hypothesis that lipid and adiposity associated traits related to liver function may play a role in insulin clearance.
Collapse
Affiliation(s)
- Artak Labadzhyan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Jinrui Cui
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Miklós Péterfy
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Yii-Der I. Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Willa A. Hsueh
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
27
|
Mondal JA. Effect of Trimethylamine N-Oxide on Interfacial Electrostatics at Phospholipid Monolayer-Water Interfaces and Its Relevance to Cardiovascular Disease. J Phys Chem Lett 2016; 7:1704-1708. [PMID: 27096306 DOI: 10.1021/acs.jpclett.6b00613] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Trimethylamine N-oxide (TMAO), a metabolite of choline containing dietary nutrients which are abundant in red meat, egg, and other animal foods, increases the risk of cardiovascular disease (e.g., atherosclerosis) by boosted accumulation of fatty deposits on artery wall. Hence, for the molecular level elucidation of the pathogenesis of atherosclerosis, it is important to understand the effect of TMAO at the endothelial cell membrane-blood interface (artery wall). Heterodyne-detected vibrational sum frequency generation (HD-VSFG) study of a zwitterionic phosphatidylcholine (PC) lipid monolayer-water interface (mimic of endothelial membrane-blood interface) shows that the interfacial water becomes increasingly H-up oriented in the presence of TMAO in the aqueous phase, revealing a dramatic change in the interfacial electrostatics. Examinations of charged lipid interfaces show that TMAO screens anionic phosphate less effectively than cationic choline, which confirms that TMAO increases the relative influence of the anionic phosphate by preferential screening of the cationic choline at the zwitterionic PC lipid interface where the phosphate and choline groups are simultaneously present. Together, it is conceivable that at an elevated TMAO level in serum would modify the electrostatics at the endothelial cell membrane-blood interface (artery wall), which may affect the influx/efflux of fatty deposits on artery wall, setting the stage for atherosclerosis.
Collapse
Affiliation(s)
- Jahur A Mondal
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre , Trombay, Mumbai 400085, India
| |
Collapse
|
28
|
O'Reilly M, Dillon E, Guo W, Finucane O, McMorrow A, Murphy A, Lyons C, Jones D, Ryan M, Gibney M, Gibney E, Brennan L, de la Llera Moya M, Reilly MP, Roche HM, McGillicuddy FC. High-Density Lipoprotein Proteomic Composition, and not Efflux Capacity, Reflects Differential Modulation of Reverse Cholesterol Transport by Saturated and Monounsaturated Fat Diets. Circulation 2016; 133:1838-50. [PMID: 27081117 DOI: 10.1161/circulationaha.115.020278] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 03/18/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Acute inflammation impairs reverse cholesterol transport (RCT) and reduces high-density lipoprotein (HDL) function in vivo. This study hypothesized that obesity-induced inflammation impedes RCT and alters HDL composition, and investigated if dietary replacement of saturated (SFA) for monounsaturated (MUFA) fatty acids modulates RCT. METHODS AND RESULTS Macrophage-to-feces RCT, HDL efflux capacity, and HDL proteomic profiling was determined in C57BL/6j mice following 24 weeks on SFA- or MUFA-enriched high-fat diets (HFDs) or low-fat diet. The impact of dietary SFA consumption and insulin resistance on HDL efflux function was also assessed in humans. Both HFDs increased plasma (3)H-cholesterol counts during RCT in vivo and ATP-binding cassette, subfamily A, member 1-independent efflux to plasma ex vivo, effects that were attributable to elevated HDL cholesterol. By contrast, ATP-binding cassette, subfamily A, member 1-dependent efflux was reduced after both HFDs, an effect that was also observed with insulin resistance and high SFA consumption in humans. SFA-HFD impaired liver-to-feces RCT, increased hepatic inflammation, and reduced ABC subfamily G member 5/8 and ABC subfamily B member 11 transporter expression in comparison with low-fat diet, whereas liver-to-feces RCT was preserved after MUFA-HFD. HDL particles were enriched with acute-phase proteins (serum amyloid A, haptoglobin, and hemopexin) and depleted of paraoxonase-1 after SFA-HFD in comparison with MUFA-HFD. CONCLUSIONS Ex vivo efflux assays validated increased macrophage-to-plasma RCT in vivo after both HFDs but failed to capture differential modulation of hepatic cholesterol trafficking. By contrast, proteomics revealed the association of hepatic-derived inflammatory proteins on HDL after SFA-HFD in comparison with MUFA-HFD, which reflected differential hepatic cholesterol trafficking between groups. Acute-phase protein levels on HDL may serve as novel biomarkers of impaired liver-to-feces RCT in vivo.
Collapse
Affiliation(s)
- Marcella O'Reilly
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Eugene Dillon
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Weili Guo
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Orla Finucane
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Aoibheann McMorrow
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Aoife Murphy
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Claire Lyons
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Daniel Jones
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Miriam Ryan
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Michael Gibney
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Eileen Gibney
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Lorraine Brennan
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Margarita de la Llera Moya
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Muredach P Reilly
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Helen M Roche
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia
| | - Fiona C McGillicuddy
- From Nutrigenomics Research Group (M.O., E.D., W.G., O.F., A. McMorrow, A. Murphy, C.L., D.J., H.M.R., F.C.M.), UCD Institute of Food and Health (M.R., M.G., E.G., L.B., H.M.R., F.C.M.), Diabetes Complications Research Centre (F.C.M.), UCD Conway Institute and School of Medicine, University College Dublin, Ireland; Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, PA (M.d.l.L.M.); and Cardiovascular Institute (M.P.R.) and Institute for Translational Medicine and Therapeutics (M.P.R.), University of Pennsylvania School of Medicine, Philadelphia.
| |
Collapse
|
29
|
Cameron SJ, Morrell CN, Bao C, Swaim AF, Rodriguez A, Lowenstein CJ. A Novel Anti-Inflammatory Effect for High Density Lipoprotein. PLoS One 2015; 10:e0144372. [PMID: 26680360 PMCID: PMC4683005 DOI: 10.1371/journal.pone.0144372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 11/17/2015] [Indexed: 11/21/2022] Open
Abstract
High density lipoprotein has anti-inflammatory effects in addition to mediating reverse cholesterol transport. While many of the chronic anti-inflammatory effects of high density lipoprotein (HDL) are attributed to changes in cell adhesion molecules, little is known about acute signal transduction events elicited by HDL in endothelial cells. We now show that high density lipoprotein decreases endothelial cell exocytosis, the first step in leukocyte trafficking. ApoA-I, a major apolipoprotein of HDL, mediates inhibition of endothelial cell exocytosis by interacting with endothelial scavenger receptor-BI which triggers an intracellular protective signaling cascade involving protein kinase C (PKC). Other apolipoproteins within the HDL particle have only modest effects upon endothelial exocytosis. Using a human primary culture of endothelial cells and murine apo-AI knockout mice, we show that apo-AI prevents endothelial cell exocytosis which limits leukocyte recruitment. These data suggest that high density lipoprotein may inhibit diseases associated with vascular inflammation in part by blocking endothelial exocytosis.
Collapse
Affiliation(s)
- Scott J. Cameron
- Departments of Medicine, Division of Cardiology, University of Rochester School of Medicine, Box 679, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
- * E-mail:
| | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
- Department of Comparative Medicine, The Johns Hopkins University School of Medicine 733 N. Broadway, MRB 827, Baltimore, MD, 21205, United States of America
| | - Clare Bao
- Department of Medicine, The Johns Hopkins University School of Medicine, 950 Ross Building, 720 Rutland Ave, Baltimore, MD, 21205, United States of America
| | - AnneMarie F. Swaim
- Department of Comparative Medicine, The Johns Hopkins University School of Medicine 733 N. Broadway, MRB 827, Baltimore, MD, 21205, United States of America
| | - Annabelle Rodriguez
- Department of Cell Biology, University of Connecticut School of Medicine, E5050, 263 Farmington Avenue, Farmington, CT, 06030, United States of America
| | - Charles J. Lowenstein
- Departments of Medicine, Division of Cardiology, University of Rochester School of Medicine, Box 679, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
| |
Collapse
|
30
|
Reina SA, Llabre MM, Allison MA, Wilkins JT, Mendez AJ, Arnan MK, Schneiderman N, Sacco RL, Carnethon M, Delaney JAC. HDL cholesterol and stroke risk: The Multi-Ethnic Study of Atherosclerosis. Atherosclerosis 2015; 243:314-9. [PMID: 26425994 PMCID: PMC4609625 DOI: 10.1016/j.atherosclerosis.2015.09.031] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 09/02/2015] [Accepted: 09/21/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND PURPOSE Accurate identification of risk factors for stroke is important for public health promotion and disease prevention. HDL cholesterol is a potential risk factor, yet its role in stroke risk is unclear, as is whether HDL cholesterol content or particle number might be a better indicator of stroke risk. Furthermore, the degree to which ethnicity moderates the risk is unknown. As such, the current study examines the associations between incident stroke and both HDL cholesterol concentration and particle number, and assesses the moderating role of race and ethnicity. METHODS The sample is a racially diverse cohort of US adults between the ages of 45-84 years enrolled in the Multi-Ethnic Study of Atherosclerosis between 2000 and 2002 and followed until December 2011. The associations among cholesterol content and stroke risk, particle number and stroke risk, and the interaction with race were explored. RESULTS The incidence of stroke was 2.6%. HDL cholesterol concentration (mmol/L) (Hazard Ratio (HR) = .56; 95% Confidence Interval (CI): .312-.988) and number of large HDL particles (μmol/L) (HR = .52, CI: .278-.956) were associated with lower stroke risk. When interactions with race were evaluated, the relationship between both HDL variables and stroke were significant in Blacks, but not other races. CONCLUSIONS Higher HDL cholesterol and a higher concentration of large particles are associated with lower risk of stroke in Blacks. Further research is needed to elucidate the mechanisms by which HDL subfractions may differentially affect stroke outcome in different races/ethnicities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ralph L Sacco
- University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | |
Collapse
|
31
|
Lin SH, Huang KJ, Weng CF, Shiuan D. Exploration of natural product ingredients as inhibitors of human HMG-CoA reductase through structure-based virtual screening. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3313-24. [PMID: 26170618 PMCID: PMC4492635 DOI: 10.2147/dddt.s84641] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cholesterol plays an important role in living cells. However, a very high level of cholesterol may lead to atherosclerosis. HMG-CoA (3-hydroxy-3-methylglutaryl coenzyme A) reductase is the key enzyme in the cholesterol biosynthesis pathway, and the statin-like drugs are inhibitors of human HMG-CoA reductase (hHMGR). The present study aimed to virtually screen for potential hHMGR inhibitors from natural product to discover hypolipidemic drug candidates with fewer side effects and lesser toxicities. We used the 3D structure 1HWK from the PDB (Protein Data Bank) database of hHMGR as the target to screen for the strongly bound compounds from the traditional Chinese medicine database. Many interesting molecules including polyphenolic compounds, polisubstituted heterocyclics, and linear lipophilic alcohols were identified and their ADMET (absorption, disrtibution, metabolism, excretion, toxicity) properties were predicted. Finally, four compounds were obtained for the in vitro validation experiments. The results indicated that curcumin and salvianolic acid C can effectively inhibit hHMGR, with IC50 (half maximal inhibitory concentration) values of 4.3 µM and 8 µM, respectively. The present study also demonstrated the feasibility of discovering new drug candidates through structure-based virtual screening.
Collapse
Affiliation(s)
- Shih-Hung Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Kao-Jean Huang
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China ; Development Center of Biotechnology, Taipei, Taiwan, Republic of China
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China
| | - David Shiuan
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China
| |
Collapse
|
32
|
Afolabi OK, Wusu AD, Ogunrinola OO, Abam EO, Babayemi DO, Dosumu OA, Onunkwor OB, Balogun EA, Odukoya OO, Ademuyiwa O. Arsenic-induced dyslipidemia in male albino rats: comparison between trivalent and pentavalent inorganic arsenic in drinking water. BMC Pharmacol Toxicol 2015; 16:15. [PMID: 26044777 PMCID: PMC4455335 DOI: 10.1186/s40360-015-0015-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2015] [Indexed: 01/05/2023] Open
Abstract
Background Recent epidemiological evidences indicate close association between inorganic arsenic exposure via drinking water and cardiovascular diseases. However, the exact mechanism of this arsenic-mediated increase in cardiovascular risk factors remains enigmatic. Methods In order to investigate the effects of inorganic arsenic exposure on lipid metabolism, male albino rats were exposed to 50, 100 and 150 ppm arsenic as sodium arsenite and 100, 150 and 200 ppm arsenic as sodium arsenate respectively in their drinking water for 12 weeks. Results Dyslipidemia induced by the two arsenicals exhibited different patterns. Hypocholesterolemia characterised the effect of arsenite at all the doses, but arsenate induced hypercholesterolemia at the 150 ppm As dose. Hypertriglyceridemia was the hallmark of arsenate effect whereas plasma free fatty acids (FFAs) was increased by the two arsenicals. Reverse cholesterol transport was inhibited by the two arsenicals as evidenced by decreased HDL cholesterol concentrations whereas hepatic cholesterol was increased by arsenite (100 ppm As), but decreased by arsenite (150 ppm As) and arsenate (100 ppm As) respectively. Brain cholesterol and triglyceride were decreased by the two arsenicals; arsenate decreased the renal content of cholesterol, but increased renal content of triglyceride. Arsenite, on the other hand, increased the renal contents of the two lipids. The two arsenicals induced phospholipidosis in the spleen. Arsenite (150 ppm As) and arsenate (100 ppm As) inhibited hepatic HMG CoA reductase. At other doses of the two arsenicals, hepatic activity of the enzyme was up-regulated. The two arsenicals however up-regulated the activity of the brain enzyme. We observed positive associations between tissue arsenic levels and plasma FFA and negative associations between tissue arsenic levels and HDL cholesterol. Conclusion Our findings indicate that even though sub-chronic exposure to arsenite and arsenate through drinking water produced different patterns of dyslipidemia, our study identified two common denominators of dyslipidemia namely: inhibition of reverse cholesterol transport and increase in plasma FFA. These two denominators (in addition to other individual perturbations of lipid metabolism induced by each arsenical), suggest that in contrast to strengthening a dose-dependent effect phenomenon, the two forms of inorganic arsenic induced lipotoxic and non-lipotoxic dyslipidemia at “low” or “medium” doses and these might be responsible for the cardiovascular and other disease endpoints of inorganic arsenic exposure through drinking water.
Collapse
Affiliation(s)
- Olusegun K Afolabi
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria. .,Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Nigeria.
| | - Adedoja D Wusu
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria. .,Department of Biochemistry, Lagos State University, Ojoo, Lagos, Nigeria.
| | - Olabisi O Ogunrinola
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria. .,Department of Biochemistry, Lagos State University, Ojoo, Lagos, Nigeria.
| | - Esther O Abam
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria. .,Biochemistry Unit, Department of Chemical Sciences, Bells University of Technology, Ota, Nigeria.
| | - David O Babayemi
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria.
| | - Oluwatosin A Dosumu
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria.
| | - Okechukwu B Onunkwor
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria.
| | - Elizabeth A Balogun
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria. .,Department of Biochemistry, University of Ilorin, Ilorin, Nigeria.
| | - Olusegun O Odukoya
- Department of Chemistry, Federal University of Agriculture, Abeokuta, Nigeria.
| | - Oladipo Ademuyiwa
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria.
| |
Collapse
|
33
|
Peptide inhibitors of human HMG-CoA reductase as potential hypocholesterolemia agents. Biochem Biophys Res Commun 2014; 456:104-9. [PMID: 25446106 DOI: 10.1016/j.bbrc.2014.11.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 11/14/2014] [Indexed: 11/22/2022]
Abstract
Hypercholesterolemia may lead to obesity and cardiovascular diseases. To prevent hypercholesterolemia, many drugs have been developed while searching for better drugs to treat hypercholesterolemia has never been ended. Other than small molecule drugs, peptide drugs are gaining more visibilities in many therapeutic areas. In the present study, we employed phage-display techniques to screen peptide inhibitors against human HMG-CoA reductase. The results indicate that the tetrapeptide PMAS inhibits hHMGR effectively (IC50=68 μM), could be a lead compound to develop hypocholesterolemic agents.
Collapse
|
34
|
SCHMIEDTOVA M, HECZKOVA M, KOVAR J, KRALOVA LESNA I, POLEDNE R. Reverse Transport of Cholesterol Is the Reason for Resistance to Development of Atherosclerosis in Prague Hereditary Hypercholesterolemic (PHHC) Rat. Physiol Res 2014; 63:591-6. [DOI: 10.33549/physiolres.932680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The Prague Hereditary Hypercholesterolemic (PHHC) rat is a model of hypercholesterolemia. In previous experiments, it was found to be completely resistant to the development of atherosclerosis. It was assumed that the reverse transport of cholesterol (RCT) might be the reason for this resistance. In this study, RCT was measured in vivo by cholesterol efflux from macrophages to plasma, using previously established methods for RCT in mice (Rader 2003), optimized for measurements in rats. Primary cell culture of macrophages was labeled with 14C-cholesterol and then injected intraperitoneally into rats. Plasma and feces were collected at 24 and 48 h. The plasma 14C-cholesterol levels at both 24 and 48 h were significantly higher in male PHHC rats compared to control Wistar rats. The PHHC rats excreted less 14C-cholesterol in feces in 24 and 48 h compared to Wistar rats. The largest pool of 14C-cholesterol was found in the adipose tissue of PHHC rats and in contrast lower levels of 14C-cholesterol were measured in the liver and muscle tissues of PHHC rats compared with Wistar rats. Increasing release of 14C-cholesterol efflux from macrophages demonstrates accelerated RTC and leads to prevention of atherogenesis in PHHC rats.
Collapse
Affiliation(s)
| | | | | | | | - R. POLEDNE
- Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
35
|
Babič M, Schmiedtová M, Poledne R, Herynek V, Horák D. In vivo monitoring of rat macrophages labeled with poly(l-lysine)-iron oxide nanoparticles. J Biomed Mater Res B Appl Biomater 2014; 103:1141-8. [PMID: 25283523 DOI: 10.1002/jbm.b.33292] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/18/2014] [Accepted: 09/12/2014] [Indexed: 12/23/2022]
Abstract
Coprecipitation of FeCl2 and FeCl3 with aqueous ammonia was used to prepare iron oxide nanoparticles dispersible in aqueous medium. Oxidation of the particles with sodium hypochlorite then yielded maghemite (γ-Fe2 O3 ) nanoparticles which were coated with two types of coating -d-mannose or poly(l-lysine) (PLL) as confirmed by FTIR analysis. The particles were <10 nm according to transmission electron microscopy. Their hydrodynamic particle size was ∼180 nm (by dynamic light scattering). The d-mannose-, PLL-coated, and neat γ-Fe2 O3 particles as well as commercial Resovist® were used to label rat macrophages. The viability and contrast properties of labeled macrophages were compared. PLL-coated γ-Fe2 O3 nanoparticles were found optimal. The labeled macrophages were injected to rats monitored in vivo by magnetic resonance imaging up to 48 h. Transport of macrophages labeled with PLL-γ-Fe2 O3 nanoparticles in rats was confirmed. Tracking of macrophages using the developed particles can be used for monitoring of inflammations and cell migration in cell therapy.
Collapse
Affiliation(s)
- Michal Babič
- Institute of Macromolecular Chemistry, Department of Polymer Particles, Academy of Sciences of the Czech Republic, 162 06, Prague 6, Czech Republic
| | - Martina Schmiedtová
- Institute for Clinical and Experimental Medicine, Laboratory for Atherosclerosis Research, 140 21, Prague 4, Czech Republic
| | - Rudolf Poledne
- Institute for Clinical and Experimental Medicine, Laboratory for Atherosclerosis Research, 140 21, Prague 4, Czech Republic
| | - Vít Herynek
- Institute for Clinical and Experimental Medicine, Laboratory for Atherosclerosis Research, 140 21, Prague 4, Czech Republic.,Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská, 1083, Czech Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Department of Polymer Particles, Academy of Sciences of the Czech Republic, 162 06, Prague 6, Czech Republic
| |
Collapse
|
36
|
Ćwiklińska A, Kortas-Stempak B, Gliwińska A, Pacanis A, Kuchta A, Wróblewska M. Interaction between VLDL and phosphatidylcholine liposomes generates new γ-LpE-like particles. Lipids 2014; 49:143-53. [PMID: 24234844 PMCID: PMC3902082 DOI: 10.1007/s11745-013-3861-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/28/2013] [Indexed: 01/08/2023]
Abstract
One of the subfractions of HDL involved in reverse cholesterol transport is γ-LpE. It has been assumed that, like preβ-LpAI, it can be generated during the interaction between phosphatidylcholine liposomes and lipoproteins and can contribute to more efficient cholesterol efflux after the introduction of liposomes to plasma. However, there has been no evidence concerning what the sources of these particles in plasma might be. Here, we determined whether the interaction of phosphatidylcholine liposomes with VLDL and the subsequent conversions of particles could be a source of new γ-LpE particles. We found that the interaction between liposomes and VLDL affected its lipid and protein composition. The content of phospholipids increased (~96 %) while the content of free cholesterol and apolipoprotein E decreased in VLDL during the reaction with liposomes (~100 and ~24 %, respectively). New particles which did not contain apolipoprotein B were generated. Heterogeneous HDL-sized populations of particles were generated, containing phospholipids and apolipoprotein E as the sole apolipoprotein, with densities from 1.063 to 1.21 g/ml, either with γ-mobility on agarose gel and Stokes diameters from 8.58 to 22.07 nm or with preβ-mobility and Stokes diameters from 9.9 to 21.08 nm. The obtained results contribute to the understanding of changes in lipoproteins under the influence of phosphatidylcholine liposomes, showing the formation of new (γ-LpE)-like and (preβ-LpE)-like particles, similar in mobility and size to plasma HDL-LpE. These newly generated particles can claim a share of the antiatherogenic effects of liposomes, observed in studies both in vitro and in vivo.
Collapse
|
37
|
Sphingosine 1-phosphate induced anti-atherogenic and atheroprotective M2 macrophage polarization through IL-4. Cell Signal 2014; 26:2249-58. [PMID: 25035231 DOI: 10.1016/j.cellsig.2014.07.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 07/09/2014] [Indexed: 11/23/2022]
Abstract
Sphingosine 1-phosphate (S1P) has been implicated in anti-atherogenic properties of high-density lipoproteins. However, the roles and signaling of S1P in macrophages, the main contributor to atherosclerosis, have not been well studied. Furthermore, pro-inflammatory M1 and anti-inflammatory M2 macrophage phenotypes may influence the development of atherosclerosis. Therefore, we investigated the effects of S1P on macrophage phenotypes, especially on M2 polarization and its signaling in relation to the anti-atherogenic properties of S1P. It was found that S1P induced anti-inflammatory M2 polarization via IL-4 secretion and its signaling, and induced IL-4Rα and IL-2Rγ. In addition, down-stream signalings, such as, stat-6 phosphorylation, SOCS1 induction, and SOCS3 suppression were also observed in macrophages in response to S1P. Furthermore, S1P-induced ERK activation, and the inhibitions of p38 MAPK and JNK were found to be key signals for IL-4 induction. Moreover, the anti-atherogenic effect of S1P in HDL was confirmed by the observation that oxidized LDL-induced lipid accumulation was attenuated in S1P-treated M2 macrophages. Furthermore, the atheroprotective effect of S1P was demonstrated by its anti-apoptotic effect on S1P-treated macrophages. The present study shows that S1P-induced M2 polarization of macrophages could be mediated via IL-4 signaling, and suggests that M2 polarization by S1P is responsible for the anti-atherogenic and atheroprotective properties of high-density lipoproteins in vivo.
Collapse
|
38
|
Roe A, Hillman J, Butts S, Smith M, Rader D, Playford M, Mehta NN, Dokras A. Decreased cholesterol efflux capacity and atherogenic lipid profile in young women with PCOS. J Clin Endocrinol Metab 2014; 99:E841-7. [PMID: 24512495 PMCID: PMC4010695 DOI: 10.1210/jc.2013-3918] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
CONTEXT Women with polycystic ovary syndrome (PCOS) have a high prevalence of cardiovascular disease (CVD) risk factors including dyslipidemia. Lipoproteins are heterogeneous, and measurement of serum lipids provides only the size of the pool and does not predict their function or composition. Recently, high-density lipoprotein cholesterol (HDL-C) function, as determined by cholesterol efflux capacity from macrophages, has been shown to be an independent predictor of subclinical CVD. OBJECTIVE The aim of the study was to comprehensively evaluate lipoprotein profile including lipid particle size and number and cholesterol efflux capacity in PCOS to better define CVD risk. DESIGN AND SETTING A case control study was performed at an academic PCOS center. PATIENTS Women with PCOS (n = 124) and geographically matched controls (n = 67) were included in the study. MAIN OUTCOME MEASURES The primary outcome was to measure HDL-C efflux capacity by an ex vivo system involving the incubation of macrophages with apolipoprotein (Apo) B-depleted serum from subjects, and the secondary outcome was to measure lipid particle size and number using nuclear magnetic resonance spectroscopy. RESULTS Women with PCOS had significantly higher body mass index and blood pressure but similar HDL-C and low-density lipoprotein cholesterol levels compared to controls. The mean ApoA1 levels were lower, and the ApoB/ApoA1 ratio was higher in PCOS subjects compared to controls (P < .01). There were no differences in ApoB levels. Women with PCOS had an 7% decrease in normalized cholesterol efflux capacity compared to controls (P < .003). Cholesterol efflux capacity in PCOS correlated with body mass index, ApoA1, HDL-C, and the presence of metabolic syndrome. In a multivariable regression model, PCOS was significantly associated with diminished cholesterol efflux. PCOS was also associated with an atherogenic profile including an increase in large very low-density lipoprotein particles, very low-density lipoprotein (VLDL) size, and small low-density lipoprotein cholesterol particles (P < .01). CONCLUSIONS Our novel findings of decreased cholesterol efflux and an atherogenic lipid particle number and size pattern in women with PCOS, independent of obesity, further substantiate the increased risk of CVD in this population.
Collapse
Affiliation(s)
- Andrea Roe
- Department of Obstetrics and Gynecology (A.R., J.H., S.B., M.S., A.D.), Division of Reproductive Endocrinology, University of Pennsylvania, Philadelphia, Pennsylvania 19104; Division of Translational Medicine and Human Genetics (D.R.), 11-125 Translational Research Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104; and Section of Inflammation and Cardiometabolic Disease (M.P., N.N.M.), National Heart, Lung and Blood Institute, Bethesda, Maryland 20892
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Zhang Q, Ma AZS, Song ZY, Wang C, Fu XD. Nifedipine enhances cholesterol efflux in RAW264.7 macrophages. Cardiovasc Drugs Ther 2014; 27:425-31. [PMID: 23812592 DOI: 10.1007/s10557-013-6472-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE Studies have shown that nifedipine protects against atherosclerotic progression, but its underlying mechanisms remain unclear. In this study, we examined if nifedipine increases macrophage cholesterol efflux, a pathway known to inhibit atherogenesis. METHODS We evaluated the ability of different doses of nifedipine to affect cholesterol efflux in RAW264.7 macrophages and its relationship with mRNA and protein levels of several well-characterized proteins involved in cholesterol efflux, including ABCA1, ABCG1, SR-BI and LXRα, using quantitative real-time PCR, Western blotting, and siRNA techniques. RESULTS Nifedipne at 1, 10, and 100 nmol/L increased apoA-I-mediated cholesterol efflux from 2.55 % to 5.65 %, 6.20 %, and 6.10 %, as well as HDL-mediated cholesterol efflux from 31.0 % to 42.5 %, 46.0 %, and 43.5 %, respectively, in RAW264.7 macrophages (p < 0.05), which was associated with increased mRNA expression levels of ABCA1, ABCG1, SR-BI, and LXRα (405 %, 381 %, 336 %; 890 %, 960 %, 1002 %; 285 %, 325 %, 336 %; 482 %, 445 %, 405 %, respectively, p < 0.05), and with increased protein levels of ABCA1, ABCG1, SR-BI, and LXRα (428 %, 492 %, 361 %; 288 %, 331 %, 365 %; 283 %, 320 %, 505 %; 581 %, 678 %, 608 %, respectively, p < 0.05). SiRNA-mediated silencing of LXRα revealed that LXRα was involved in these increases and the enhanced cholesterol efflux. CONCLUSION Nifedipine may protect against atherosclerosis partly by promoting macrophage cholesterol efflux through the stimulation of LXRα-dependent expression of ABCA1, ABCG1, and SR-BI.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
40
|
Kulkarni H, Meikle PJ, Mamtani M, Weir JM, Almeida M, Diego V, Peralta JM, Barlow CK, Bellis C, Dyer TD, Almasy L, Mahaney MC, Comuzzie AG, Göring HHH, Curran JE, Blangero J. Plasma lipidome is independently associated with variability in metabolic syndrome in Mexican American families. J Lipid Res 2014; 55:939-46. [PMID: 24627127 DOI: 10.1194/jlr.m044065] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Plasma lipidome is now increasingly recognized as a potentially important marker of chronic diseases, but the exact extent of its contribution to the interindividual phenotypic variability in family studies is unknown. Here, we used the rich data from the ongoing San Antonio Family Heart Study (SAFHS) and developed a novel statistical approach to quantify the independent and additive value of the plasma lipidome in explaining metabolic syndrome (MS) variability in Mexican American families recruited in the SAFHS. Our analytical approach included two preprocessing steps: principal components analysis of the high-resolution plasma lipidomics data and construction of a subject-subject lipidomic similarity matrix. We then used the Sequential Oligogenic Linkage Analysis Routines software to model the complex family relationships, lipidomic similarities, and other important covariates in a variance components framework. Our results suggested that even after accounting for the shared genetic influences, indicators of lipemic status (total serum cholesterol, TGs, and HDL cholesterol), and obesity, the plasma lipidome independently explained 22% of variability in the homeostatic model of assessment-insulin resistance trait and 16% to 22% variability in glucose, insulin, and waist circumference. Our results demonstrate that plasma lipidomic studies can additively contribute to an understanding of the interindividual variability in MS.
Collapse
Affiliation(s)
- Hemant Kulkarni
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Despite the critical importance of plasma lipoproteins in the development of atherosclerosis, varying degrees of evidence surround the causal associations of lipoproteins with coronary artery disease (CAD). These causal contributions can be assessed by employing genetic variants as unbiased proxies for lipid levels. A relatively large number of low-density lipoprotein cholesterol (LDL-C) variants strongly associate with CAD, confirming the causal impact of this lipoprotein on atherosclerosis. Although not as firmly established, genetic evidence supporting a causal role of triglycerides (TG) in CAD is growing. Conversely, high-density lipoprotein cholesterol (HDL-C) variants not associated with LDL-C or TG have not yet been shown to be convincingly associated with CAD, raising questions about the causality of HDL-C in atherosclerosis. Finally, genetic variants at the LPA locus associated with lipoprotein(a) [Lp(a)] are decisively linked to CAD, indicating a causal role for Lp(a). Translational investigation of CAD-associated lipid variants may identify novel regulatory pathways with therapeutic potential to alter CAD risk.
Collapse
|
42
|
Chauke CG, Arieff Z, Kaur M, Seier JV. Effects of short-term niacin treatment on plasma lipoprotein concentrations in African green monkeys (Chlorocebus aethiops). Lab Anim (NY) 2014; 43:58-62. [PMID: 24451360 DOI: 10.1038/laban.424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 10/01/2013] [Indexed: 11/09/2022]
Abstract
Niacin is the most effective drug available for raising levels of high-density lipoprotein (HDL) cholesterol. To evaluate its effects on plasma lipid concentrations, the authors administered a low dose of niacin to healthy, adult, female African green monkeys for 3 months. In the treated monkeys, low-density lipoprotein cholesterol concentrations decreased by 43% from baseline, whereas concentrations of HDL cholesterol and apolipoprotein A-I increased by 49% and 34%, respectively. The results suggest that in this primate model, a low dose of niacin can effectively increase concentrations of HDL cholesterol.
Collapse
Affiliation(s)
- Chesa G Chauke
- Medical Research Council Primate Unit, Tygerberg, South Africa
| | - Zainunisha Arieff
- Biotechnology Department, University of the Western Cape, Cape Town, South Africa
| | - Mandeep Kaur
- 1] Biotechnology Department, University of the Western Cape, Cape Town, South Africa. [2] Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Jurgen V Seier
- Medical Research Council Primate Unit, Tygerberg, South Africa
| |
Collapse
|
43
|
Kalvinsh IY. NEW PATHOGENETIC FACTORS OF ATHEROSCLEROSIS DEVELOPMENT AND POTENTIAL THERAPEUTIC APPROACHES. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2013. [DOI: 10.15829/1728-8800-2013-5-87-90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
It has been demonstrated that one of the factors in the pathogenesis of atherosclerosis, with the subsequent development of myocardial infarction (MI), stroke (S), or sudden death (SD), is trimethylaminoxide (TMAO), the end-product of dietary choline, betaine, or carnitine metabolism by intestinal microflora. Simultaneously elevated levels of TMAO and carnitine are associated with a doubled or even tripled risk of MI, S, and SD. Therefore, dietary intake of choline-rich phosphatidylcholine fats and red meats and carnitine-rich dairy is an important risk factor (RF) of atherosclerosis. At the moment, there is no universally accepted therapeutic approach which reduces TMAO and carnitine levels. The only agent which can simultaneously reduce the levels of these two atherosclerosis-associated RFs is a well-known cardio- and cytoprotector Mildronate. Experimental and pilot clinical studies of Mildronate effectiveness in patients with obliterating atherosclerosis suggest the need for further, more detailed clinical trials of Mildronate, for estimation of its efficacy and safety.
Collapse
|
44
|
D'Amore S, Vacca M, Graziano G, D'Orazio A, Cariello M, Martelli N, Di Tullio G, Salvia R, Grandaliano G, Belfiore A, Pellegrini F, Palasciano G, Moschetta A. Nuclear receptors expression chart in peripheral blood mononuclear cells identifies patients with Metabolic Syndrome. Biochim Biophys Acta Mol Basis Dis 2013; 1832:2289-301. [PMID: 24060638 DOI: 10.1016/j.bbadis.2013.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/11/2013] [Accepted: 09/15/2013] [Indexed: 01/27/2023]
Abstract
BACKGROUND Nuclear receptors are a class of 48 ligand-activated transcription factors identified as key players of metabolic and developmental processes. Most of these receptors are potential targets for pharmacological strategies in the Metabolic Syndrome. In the present study, we analyzed changes in the mRNA expression of nuclear receptors in the peripheral blood mononuclear cells of patients with Metabolic Syndrome, in order to identify novel biomarkers of disease and candidate targets for putative therapeutical approaches. METHODS AND RESULTS We enrolled thirty healthy controls (14 M:16 F) and thirty naïve patients (16 M: 14 F; >3 criteria for Metabolic Syndrome upon Adult Treatment Panel III) without organ damage. Using quantitative real-time PCR, we assessed the expression patterns of nuclear receptors in peripheral blood mononuclear cells. 33/48 nuclear receptors were expressed in peripheral blood mononuclear cells. In patients with Metabolic Syndrome, we found a significant down-regulation of the entire PPAR, NR4A and RAR families, together with a repression of RXRα, VDR, and Rev-Erbα. Furthermore, we performed a novel statistical analysis with classification trees, which allowed us to depict a predictive core of nuclear receptor expression patterns characterizing subjects with Metabolic Syndrome. Random Forest Analysis identified NOR1 and PPARδ, which were both reduced in peripheral blood mononuclear cells and specifically in CD14(+) cells (mostly monocytes), as classifiers of Metabolic Syndrome, with high specificity and sensitivity. CONCLUSIONS Our results point to the use of PPAR and NR4A mRNA levels in the overall peripheral blood mononuclear cells as biomarkers of Metabolic Syndrome and bona fide putative targets of pharmacological therapy.
Collapse
Affiliation(s)
- Simona D'Amore
- Clinica Medica "A. Murri", "Aldo Moro" University of Bari, Italy; National Cancer Research Center, IRCCS Oncologico Giovanni Paolo II, Bari, Italy; Laboratory of Lipid Metabolism and Cancer, Consorzio Mario Negri Sud, Santa Maria Imbaro (Chieti), Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Vincent PE, Weinberg PD. Flow-dependent concentration polarization and the endothelial glycocalyx layer: multi-scale aspects of arterial mass transport and their implications for atherosclerosis. Biomech Model Mechanobiol 2013; 13:313-26. [DOI: 10.1007/s10237-013-0512-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 06/26/2013] [Indexed: 10/26/2022]
|
46
|
Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WHW, Bushman FD, Lusis AJ, Hazen SL. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 2013; 19:576-85. [PMID: 23563705 PMCID: PMC3650111 DOI: 10.1038/nm.3145] [Citation(s) in RCA: 3092] [Impact Index Per Article: 257.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 02/27/2013] [Indexed: 02/07/2023]
Abstract
Intestinal microbiota metabolism of choline and phosphatidylcholine produces trimethylamine (TMA), which is further metabolized to a proatherogenic species, trimethylamine-N-oxide (TMAO). We demonstrate here that metabolism by intestinal microbiota of dietary L-carnitine, a trimethylamine abundant in red meat, also produces TMAO and accelerates atherosclerosis in mice. Omnivorous human subjects produced more TMAO than did vegans or vegetarians following ingestion of L-carnitine through a microbiota-dependent mechanism. The presence of specific bacterial taxa in human feces was associated with both plasma TMAO concentration and dietary status. Plasma L-carnitine levels in subjects undergoing cardiac evaluation (n = 2,595) predicted increased risks for both prevalent cardiovascular disease (CVD) and incident major adverse cardiac events (myocardial infarction, stroke or death), but only among subjects with concurrently high TMAO levels. Chronic dietary L-carnitine supplementation in mice altered cecal microbial composition, markedly enhanced synthesis of TMA and TMAO, and increased atherosclerosis, but this did not occur if intestinal microbiota was concurrently suppressed. In mice with an intact intestinal microbiota, dietary supplementation with TMAO or either carnitine or choline reduced in vivo reverse cholesterol transport. Intestinal microbiota may thus contribute to the well-established link between high levels of red meat consumption and CVD risk.
Collapse
Affiliation(s)
- Robert A. Koeth
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Zeneng Wang
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Bruce S. Levison
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Jennifer A. Buffa
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Elin Org
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles 90095, USA
| | - Brendan T. Sheehy
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Earl B. Britt
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Xiaoming Fu
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Yuping Wu
- Department of Mathematics, Cleveland State University, Cleveland, Ohio 44115, USA
| | - Lin Li
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Jonathan D. Smith
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Joseph A. DiDonato
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Jun Chen
- Department of Microbiology, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongzhe Li
- Department of Microbiology, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gary D. Wu
- Division of Gastroenterology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James D. Lewis
- Department of Microbiology, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Manya Warrier
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - J. Mark Brown
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Ronald M. Krauss
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - W. H. Wilson Tang
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Frederic D. Bushman
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Aldons J. Lusis
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles 90095, USA
| | - Stanley L. Hazen
- Department of Cellular & Molecular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
47
|
Abstract
In addition to its role in reverse cholesterol transport, high-density lipoprotein (HDL) cholesterol has direct action on numerous cell types that influence cardiovascular and metabolic health. Cellular responses to HDL entail its capacity to invoke cholesterol efflux that causes signal initiation via scavenger receptor class B, type I, and plasma membrane receptor activation by HDL cargo molecules. In endothelial cells and their progenitors, HDL attenuates apoptosis and stimulates proliferation and migration. HDL also has diverse anti-inflammatory actions in both endothelial cells and leukocytes. In vascular smooth muscles, HDL tempers proinflammatory, promigratory, and degradative processes, and through actions on endothelium and platelets HDL is antithrombotic. There are additional actions of HDL of potential cardiovascular consequence that are indirect, including the capacities to promote pancreatic β-cell insulin secretion, to protect pancreatic β cells from apoptosis, and to enhance glucose uptake by skeletal muscle myocytes. Furthermore, HDL decreases white adipose tissue mass, increases energy expenditure, and promotes the production of adipose-derived cytokine adiponectin that has its own vascular-protective properties. Many of these numerous actions of HDL have been observed not only in cell culture and animal models but also in human studies, and assessments of these functions are now being applied to patient populations to better-elucidate which actions of HDL may contribute to its cardioprotective potential and how they can be quantified and targeted. Further work on the many mechanisms of HDL action promises to reveal new prophylactic and therapeutic strategies to optimize both cardiovascular and metabolic health.
Collapse
Affiliation(s)
- Chieko Mineo
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
48
|
Abstract
In addition to its role in reverse cholesterol transport, high-density lipoprotein (HDL) cholesterol has direct action on numerous cell types that influence cardiovascular and metabolic health. Cellular responses to HDL entail its capacity to invoke cholesterol efflux that causes signal initiation via scavenger receptor class B, type I, and plasma membrane receptor activation by HDL cargo molecules. In endothelial cells and their progenitors, HDL attenuates apoptosis and stimulates proliferation and migration. HDL also has diverse anti-inflammatory actions in both endothelial cells and leukocytes. In vascular smooth muscles, HDL tempers proinflammatory, promigratory, and degradative processes, and through actions on endothelium and platelets HDL is antithrombotic. There are additional actions of HDL of potential cardiovascular consequence that are indirect, including the capacities to promote pancreatic β-cell insulin secretion, to protect pancreatic β cells from apoptosis, and to enhance glucose uptake by skeletal muscle myocytes. Furthermore, HDL decreases white adipose tissue mass, increases energy expenditure, and promotes the production of adipose-derived cytokine adiponectin that has its own vascular-protective properties. Many of these numerous actions of HDL have been observed not only in cell culture and animal models but also in human studies, and assessments of these functions are now being applied to patient populations to better-elucidate which actions of HDL may contribute to its cardioprotective potential and how they can be quantified and targeted. Further work on the many mechanisms of HDL action promises to reveal new prophylactic and therapeutic strategies to optimize both cardiovascular and metabolic health.
Collapse
Affiliation(s)
- Chieko Mineo
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
49
|
Tung CL, Lin ST, Chou HC, Chen YW, Lin HC, Tung CL, Huang KJ, Chen YJ, Lee YR, Chan HL. Proteomics-based identification of plasma biomarkers in oral squamous cell carcinoma. J Pharm Biomed Anal 2012; 75:7-17. [PMID: 23312379 DOI: 10.1016/j.jpba.2012.11.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 11/09/2012] [Accepted: 11/10/2012] [Indexed: 12/20/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is an aggressive cancer and its occurrence is closely related to betel nut chewing in Taiwan. However, there are few prognostic and diagnostic biomarkers for this disease especially for its association with betel nut chewing. Recent progresses in quantitative proteomics have offered opportunities to discover plasma proteins as biomarkers for tracking the progression and for understanding the molecular mechanisms of OSCC. In present study, plasma samples from OSCC patients with at least 5-year history of betel nut chewing and healthy donors were analyzed by fluorescence 2D-DIGE-based proteomic analysis. Totally, 38 proteins have been firmly identified representing 13 unique gene products. These proteins mainly function in inflammatory responses (such as fibrinogen gamma chain) and transport (Apolipoprotein A-I). Additionally, the current quantitative proteomic approach has identified numerous OSCC biomarkers including fibrinogen (alpha/beta/gamma) chain, haptoglobin, leucine-rich alpha-2-glycoprotein and ribosomal protein S6 kinase alpha-3 (RSK2) which have not been reported and may be associated with the progression and development of the disease. In summary, this study reports a comprehensive patient-based proteomic approach for the identification of potential plasma biomarkers in OSCC. The potential of utilizing these markers for screening and treating OSCC warrants further investigations.
Collapse
Affiliation(s)
- Chun-Liang Tung
- Department of Pathology, Chiayi Christian Hospital, Chiayi, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Pak VV, Kwon DY, Yun LM, Yili A, Aisa HA, Shakhidoyatov KM. Conformational analysis of yvae peptide derivatives. Chem Nat Compd 2012. [DOI: 10.1007/s10600-012-0399-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|