1
|
Derakhshandeh N, Nazifi S, Mogheiseh A, Divar MR, Dadvand Z, Karimizadeh MS, Zeidabadi M. Oral nicotinic acid administration effect on lipids, thyroid hormones, and oxidative stress in intact adult dogs. BMC Vet Res 2025; 21:142. [PMID: 40038732 PMCID: PMC11881314 DOI: 10.1186/s12917-025-04597-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/11/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Nicotinic acid (niacin, Vitamin B3) is one of the most effective medicines for improving high-density lipoprotein concentrations. Obesity and related diseases are life-threatening to dogs. This study investigated the niacin effect on triglyceride, cholesterol, lipoproteins, thyroid hormones, oxidative stress, and lipid peroxidation in intact adult dogs. Blood samples were taken from seven healthy, intact adult dogs as a control group (day 0). Then, the animals received 1000 mg/dog of oral nicotinic acid tab daily for 42 days, and blood sampling was performed on days 14, 28, 42, and 56. RESULT The results showed an increasing trend in high-density lipoprotein (HDL) concentration. The highest HDL concentration (138.85 ± 43.72 mg/dl) was related to day 56; the HDL level followed a statistically significant increase between day 14 and 56. Unlike HDL, there was a decreasing trend in low-density lipoprotein (LDL) concentration. The lowest LDL concentration (21.85 ± 18.60 mg/dl) was related to day 56. The concentration of apolipoprotein A-I (apoA1) was significantly increased during the study. The highest concentration of apoA1 (1.66 ± 0.06 g/l) was on day 42. There was a significant increase in apoA1 concentrations between days 0 and 14, 42, and 56. The apoA1 was significantly increased between days 14 and 42 and 56. The apoA1 followed a statistically significant increase between days 28 and 42. Changes in thyroid hormone levels did not show any constant increasing or decreasing trend. On day 14, a decreasing trend in the concentrations of TT4, FT4, and T3 was observed. However, an increasing trend was detected in the concentrations of TT4, FT4, and T3 on days 28 and 42. However, the increase in the concentrations of TT4 and FT4 was less than that on day 0. After treatment (day 56), a decreasing trend was observed in thyroid hormone concentrations. The negative correlation was detected between apoA1 and triiodothyronine (T3), total thyroxine T4 (TT4)), and free T4 (FT4) concentrations on day 42. Furthermore, a significant negative relationship was observed between HDL and T4 on day 42. However, the relationship between triglyceride and T3 was statistically positive on day 14. There was an increasing trend in serum total antioxidant capacity (TAC). The highest TAC concentration (3.83 ± 0.62 µmol /l) was on day 56; however, the malondialdehyde (MDA) concentration was decreased during the study. The total antioxidant level followed a statistically significant increase between days 0 and 56 compared to days 14 and 42. CONCLUSION The study demonstrated the efficacy of nicotinic acid in improving serum HDL, apoA1, and TAC, as well as decreasing serum MDA and LDL concentrations.
Collapse
Affiliation(s)
- Nooshin Derakhshandeh
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, P.O.Box: 7144169115, Shiraz, Fars, Iran.
| | - Saeed Nazifi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, P.O.Box: 7144169115, Shiraz, Fars, Iran
| | - Asghar Mogheiseh
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, P.O.Box: 7144169115, Shiraz, Fars, Iran
| | - Mohammad Reza Divar
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, P.O.Box: 7144169115, Shiraz, Fars, Iran
| | - Zahra Dadvand
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, P.O.Box: 7144169115, Shiraz, Fars, Iran
| | - Mohammad Sadegh Karimizadeh
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, P.O.Box: 7144169115, Shiraz, Fars, Iran
| | - Mahboobeh Zeidabadi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, P.O.Box: 7144169115, Shiraz, Fars, Iran
| |
Collapse
|
2
|
Zhou J, Han J. Association of niacin intake and metabolic dysfunction-associated steatotic liver disease: findings from National Health and Nutrition Examination Survey. BMC Public Health 2024; 24:2742. [PMID: 39379884 PMCID: PMC11462762 DOI: 10.1186/s12889-024-20161-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/23/2024] [Indexed: 10/10/2024] Open
Abstract
AIM This study aims to explore the relationship between niacin intake and the prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) within a large, multi-ethnic cohort. METHODS A total of 2946 participants from the National Health and Nutrition Examination Survey (NHANES) were carefully selected based on strict inclusion and exclusion criteria. Participants meeting the eligibility criteria underwent two dietary recall interviews, and niacin intake was calculated using the USDA's Food and Nutrient Database for Dietary Studies (FNDDS). Liver steatosis was diagnosed using a Controlled Attenuation Parameter (CAP) of 248 dB/m, and MASLD diagnosis was based on metabolic indicators. Weighted multivariate logistic regression was utilized to analyze the correlation between niacin intake and MASLD prevalence, with potential nonlinear relationships explored through restricted cubic spline (RCS) regression. RESULTS Analysis of baseline data revealed that MASLD patients had lower niacin intake levels and poorer metabolic biomarker profiles. Both RCS analysis and multivariate logistic regression indicated a U-shaped association between niacin intake and MASLD prevalence. Specifically, there was a non-linear dose-response relationship, with the odds of MASLD gradually decreasing with increasing niacin intake until reaching a threshold of 23.6 mg, beyond which the odds of MASLD began to increase. CONCLUSION This study confirms a U-shaped nonlinear relationship between niacin intake and MASLD prevalence within the diverse American population.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Infectious Diseases, Affiliated Wuxi Fifth Hospital of Jiangnan University, The Fifth People's Hospital of Wuxi, Wuxi, 214065, China
| | - Jun Han
- Department of Infectious Diseases, Affiliated Wuxi Fifth Hospital of Jiangnan University, The Fifth People's Hospital of Wuxi, Wuxi, 214065, China.
| |
Collapse
|
3
|
Chen Q, Abudukeremu A, Li K, Zheng M, Li H, Huang T, Huang C, Wen K, Wang Y, Zhang Y. High-Density Lipoprotein Subclasses and Their Role in the Prevention and Treatment of Cardiovascular Disease: A Narrative Review. Int J Mol Sci 2024; 25:7856. [PMID: 39063097 PMCID: PMC11277419 DOI: 10.3390/ijms25147856] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The association between high-density lipoprotein cholesterol (HDL-C) and cardiovascular disease (CVD) is controversial. HDL-C is one content type of high-density lipoprotein (HDL). HDL consists of diverse proteins and lipids and can be classified into different subclasses based on size, shape, charge, and density, and can change dynamically in disease states. Therefore, HDL-C levels alone cannot represent HDLs' cardioprotective role. In this review, we summarized the methods for separating HDL subclasses, the studies on the association between HDL subclasses and cardiovascular risk (CVR), and the impact of lipid-modifying medications and nonpharmacological approaches (exercise training, dietary omega fatty acids, and low-density lipoprotein apheresis) on HDL subclasses. As HDL is a natural nanoplatform, recombinant HDLs (rHDLs) have been used as a delivery system in vivo by loading small interfering RNA, drugs, contrast agents, etc. Therefore, we further reviewed the HDL subclasses used in rHDLs and their advantages and disadvantages. This review would provide recommendations and guidance for future studies on HDL subclasses' cardioprotective roles.
Collapse
Affiliation(s)
- Qiaofei Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Ayiguli Abudukeremu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Kaiwen Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China;
| | - Minglong Zheng
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Hongwei Li
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Tongsheng Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Canxia Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Kexin Wen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Yue Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Yuling Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou 510080, China
| |
Collapse
|
4
|
Nacarelli GS, Fasolino T, Davis S. Dietary, macronutrient, micronutrient, and nutrigenetic factors impacting cardiovascular risk markers apolipoprotein B and apolipoprotein A1: a narrative review. Nutr Rev 2024; 82:949-962. [PMID: 37615981 DOI: 10.1093/nutrit/nuad102] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Genetic predisposition and dietary factors can impact cardiovascular disease (CVD) risk. Two important markers in assessing CVD risk are apolipoprotein (apo) B and apolipoprotein A1 plasma levels. These markers are measured as a ratio, with a high apoB:apoA1 ratio associated with increased CVD risk. Dietary and lifestyle recommendations are the cornerstone of managing primary and secondary CVD risk-mitigation strategies. One way to assess the impact of various dietary and lifestyle interventions on CVD risk is to evaluate the changes in CVD risk markers, such as apoB, apoA1, and apoB:apoA1 ratio. Various human studies have demonstrated the impact of dietary, macronutrient, and micronutrient interventions on apoB and apoA1 status. This review aims to elucidate dietary, macronutrient, micronutrient, and nutrigenetic considerations for impacting apoB and apoA1 levels. A low-carbohydrate, high-saturated-fat diet, low fiber intake, low vitamin and mineral intake, and zinc and iron deficiency are associated with an elevated apoB:apoA1 ratio. The Mediterranean diet, vegan diet, fermented dairy products, lower sugar intake, higher protein intake, higher polyunsaturated fat intake, and an omega-3-rich diet are associated with a decreased apoB:apoA1 ratio. Micronutrients associated with a decreased apoB:apoA1 ratio include vitamin D sufficiency, increased serum vitamin C, and magnesium. Variants in the APOE, APOA1, and FADS2 genes may alter the apoB:apoA1 ratio in response to various dietary interventions. When accounting for factors that may favorably alter the apoB:apoA1 ratio, researchers should consider a healthy diet sufficient in polyunsaturated fats, vitamins, minerals, trace minerals, and lower excess sugars.
Collapse
Affiliation(s)
| | - Tracy Fasolino
- Clemson School of Nursing, Clemson University, Clemson, South Carolina, USA
| | - Stephanie Davis
- Clemson School of Nursing, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
5
|
Lipoprotein(a) and Atherosclerotic Cardiovascular Disease, the Impact of Available Lipid-Lowering Medications on Lipoprotein(a): An Update on New Therapies. Endocr Pract 2022:S1530-891X(22)00901-6. [PMID: 36563785 DOI: 10.1016/j.eprac.2022.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/20/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To review evidence of existing and new pharmacological therapies for lowering lipoprotein(a) (Lp[a]) concentrations and their impact on clinically relevant outcomes. METHODS We searched for literature pertaining to Lp(a) and pharmacological treatments in PubMed. We reviewed articles published between 1963 and 2020. RESULTS We found that statins significantly increased Lp(a) concentrations. Therapies that demonstrated varying degrees of Lp(a) reduction included ezetimibe, niacin, proprotein convertase subtilisin/kexin type 9 inhibitors, lipoprotein apheresis, fibrates, aspirin, hormone replacement therapy, antisense oligonucleotide therapy, and small interfering RNA therapy. There was limited data from large observational studies and post hoc analyses showing the potential benefits of these therapies in improving cardiovascular outcomes. CONCLUSION There are multiple lipid-lowering agents currently being used to treat hyperlipidemia that also have a Lp(a)-lowering effect. Two RNA therapies specifically targeted to lower Lp(a) are being investigated in phase 3 clinical trials and, thus far, have shown promising results. However, evidence is lacking to determine the clinical relevance of reducing Lp(a). At present, there is a need for large-scale, randomized, controlled trials to evaluate cardiovascular outcomes associated with lowering Lp(a).
Collapse
|
6
|
Potential Therapeutic Agents That Target ATP Binding Cassette A1 (ABCA1) Gene Expression. Drugs 2022; 82:1055-1075. [PMID: 35861923 DOI: 10.1007/s40265-022-01743-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
The cholesterol efflux protein ATP binding cassette protein A1 (ABCA) and apolipoprotein A1 (apo A1) are key constituents in the process of reverse-cholesterol transport (RCT), whereby excess cholesterol in the periphery is transported to the liver where it can be converted primarily to bile acids for either use in digestion or excreted. Due to their essential roles in RCT, numerous studies have been conducted in cells, mice, and humans to more thoroughly understand the pathways that regulate their expression and activity with the goal of developing therapeutics that enhance RCT to reduce the risk of cardiovascular disease. Many of the drugs and natural compounds examined target several transcription factors critical for ABCA1 expression in both macrophages and the liver. Likewise, several miRNAs target not only ABCA1 but also the same transcription factors that are critical for its high expression. However, after years of research and many preclinical and clinical trials, only a few leads have proven beneficial in this regard. In this review we discuss the various transcription factors that serve as drug targets for ABCA1 and provide an update on some important leads.
Collapse
|
7
|
Effect of Siberian Ginseng Water Extract as a Dietary Additive on Growth Performance, Blood Biochemical Indexes, Lipid Metabolism, and Expression of PPARs Pathway-Related Genes in Genetically Improved Farmed Tilapia (Oreochromis niloticus). FISHES 2022. [DOI: 10.3390/fishes7040149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Overnutrition in high-density aquaculture can negatively affect the health of farmed fish. The Chinese herbal medicine Siberian ginseng (Acanthopanax senticosus, AS) can promote animal growth and immunity, and regulate lipid metabolism. Therefore, we conducted an 8-week experiment, in which Oreochromis niloticus was fed with a diet supplemented with different concentrations of AS water extract (ASW) (0‰, 0.1‰, 0.2‰, 0.4‰, 0.8‰, and 1.6‰). The ASW improved the growth performance and increased the specific growth rate (SGR). Linear regression analysis based on the SGR estimated that the optimal ASW amount was 0.74‰. Dietary supplementation with 0.4–0.8‰ ASW reduced the triglyceride and total cholesterol levels in the serum and liver, and regulated lipid transport by increasing the high-density lipoprotein cholesterol concentration and lowering the low-density lipoprotein cholesterol concentration. Dietary supplementation with ASW increased the activities of superoxide dismutase and catalase in the liver, thereby improving the antioxidant capacity. Moreover, ASW modulated the transcription of genes in the peroxisome proliferator-activated receptor signaling pathway in the liver (upregulation of PPARα, APOA1b, and FABP10a and downregulation of PPARγ), thereby regulating fatty acid synthesis and metabolism and slowing fat deposition. These results showed that 0.4–0.8‰ ASW can slow fat deposition and protected the liver from cell damage and abnormal lipid metabolism.
Collapse
|
8
|
Shaik A, Rosenson RS. Genetics of Triglyceride-Rich Lipoproteins Guide Identification of Pharmacotherapy for Cardiovascular Risk Reduction. Cardiovasc Drugs Ther 2021; 35:677-690. [PMID: 33710501 DOI: 10.1007/s10557-021-07168-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Despite aggressive reduction of low-density lipoprotein cholesterol (LDL-C), there is a residual risk of cardiovascular disease (CVD). Hypertriglyceridemia is known to be associated with increased CVD risk, independently of LDL-C. Triglycerides are one component of the heterogenous class of triglyceride-rich lipoproteins (TGRLs). METHODS/RESULTS Growing evidence from biology, epidemiology, and genetics supports the contribution of TGRLs to the development of CVD via a number of mechanisms, including through proinflammatory, proapoptotic, and procoagulant pathways. CONCLUSION New genetics-guided pharmacotherapies to reduce levels of triglycerides and TGRLs and thus reduce risk of CVD have been developed and will be discussed here.
Collapse
Affiliation(s)
- Aleesha Shaik
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert S Rosenson
- Cardiometabolics Unit, Zena and Michael A Wiener Cardiovascular Institute, Marie-Josee and Henry R Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Geisler CE, Miller KE, Ghimire S, Renquist BJ. The Role of GPR109a Signaling in Niacin Induced Effects on Fed and Fasted Hepatic Metabolism. Int J Mol Sci 2021; 22:4001. [PMID: 33924461 PMCID: PMC8069761 DOI: 10.3390/ijms22084001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/04/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Signaling through GPR109a, the putative receptor for the endogenous ligand β-OH butyrate, inhibits adipose tissue lipolysis. Niacin, an anti-atherosclerotic drug that can induce insulin resistance, activates GPR109a at nM concentrations. GPR109a is not essential for niacin to improve serum lipid profiles. To better understand the involvement of GPR109a signaling in regulating glucose and lipid metabolism, we treated GPR109a wild-type (+/+) and knockout (-/-) mice with repeated overnight injections of saline or niacin in physiological states characterized by low (ad libitum fed) or high (16 h fasted) concentrations of the endogenous ligand, β-OH butyrate. In the fed state, niacin increased expression of apolipoprotein-A1 mRNA and decreased sterol regulatory element-binding protein 1 mRNA independent of genotype, suggesting a possible GPR109a independent mechanism by which niacin increases high-density lipoprotein (HDL) production and limits transcriptional upregulation of lipogenic genes. Niacin decreased fasting serum non-esterified fatty acid concentrations in both GPR109a +/+ and -/- mice. Independent of GPR109a expression, niacin blunted fast-induced hepatic triglyceride accumulation and peroxisome proliferator-activated receptor α mRNA expression. Although unaffected by niacin treatment, fasting serum HDL concentrations were lower in GPR109a knockout mice. Surprisingly, GPR109a knockout did not affect glucose or lipid homeostasis or hepatic gene expression in either fed or fasted mice. In turn, GPR109a does not appear to be essential for the metabolic response to the fasting ketogenic state or the acute effects of niacin.
Collapse
Affiliation(s)
- Caroline E. Geisler
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA; (C.E.G.); (K.E.M.); (S.G.)
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kendra E. Miller
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA; (C.E.G.); (K.E.M.); (S.G.)
| | - Susma Ghimire
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA; (C.E.G.); (K.E.M.); (S.G.)
| | - Benjamin J. Renquist
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA; (C.E.G.); (K.E.M.); (S.G.)
| |
Collapse
|
10
|
Cesaro A, Schiavo A, Moscarella E, Coletta S, Conte M, Gragnano F, Fimiani F, Monda E, Caiazza M, Limongelli G, D'Erasmo L, Riccio C, Arca M, Calabrò P. Lipoprotein(a): a genetic marker for cardiovascular disease and target for emerging therapies. J Cardiovasc Med (Hagerstown) 2021; 22:151-161. [PMID: 32858625 DOI: 10.2459/jcm.0000000000001077] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lipoprotein(a) [Lp(a)] is an established cardiovascular risk factor, and growing evidence indicates its causal association with atherosclerotic disease because of the proatherogenic low-density lipoprotein (LDL)-like properties and the prothrombotic plasminogen-like activity of apolipoprotein(a) [apo(a)]. As genetics significantly influences its plasma concentration, Lp(a) is considered an inherited risk factor of atherosclerotic cardiovascular disease (ASCVD), especially in young individuals. Moreover, it has been suggested that elevated Lp(a) may significantly contribute to residual cardiovascular risk in patients with coronary artery disease and optimal LDL-C levels. Nonetheless, the fascinating hypothesis that lowering Lp(a) could reduce the risk of cardiovascular events - in primary or secondary prevention - still needs to be demonstrated by randomized clinical trials. To date, no specific Lp(a)-lowering agent has been approved for reducing the lipoprotein levels, and current lipid-lowering drugs have limited effects. In the future, emerging therapies targeting Lp(a) may offer the possibility to further investigate the relation between Lp(a) levels and cardiovascular outcomes in randomized controlled trials, ultimately leading to a new era in cardiovascular prevention. In this review, we aim to provide an updated overview of current evidence on Lp(a) as well as currently investigated therapeutic strategies that specifically address the reduction of the lipoprotein.
Collapse
Affiliation(s)
- Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Alessandra Schiavo
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Elisabetta Moscarella
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Silvio Coletta
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Matteo Conte
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Fabio Fimiani
- Division of Cardiology
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Monaldi Hospital, Naples
| | - Emanuele Monda
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
- Division of Cardiology
| | - Martina Caiazza
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Monaldi Hospital, Naples
| | - Giuseppe Limongelli
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Cardiology
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Monaldi Hospital, Naples
| | - Laura D'Erasmo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Carmine Riccio
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| |
Collapse
|
11
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
12
|
Hafiane A, Gasbarrino K, Daskalopoulou SS. The role of adiponectin in cholesterol efflux and HDL biogenesis and metabolism. Metabolism 2019; 100:153953. [PMID: 31377319 DOI: 10.1016/j.metabol.2019.153953] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/27/2022]
Abstract
Cholesterol efflux is the initial step in the reverse cholesterol transport pathway by which excess cholesterol in peripheral cells is exported and subsequently packaged into high-density lipoprotein (HDL) particles. Adiponectin is the most abundantly secreted adipokine that possesses anti-inflammatory and vasculoprotective properties via interaction with transmembrane receptors, AdipoR1 and AdipoR2. Evidence suggests that low levels of adiponectin may be a useful marker for atherosclerotic disease. A proposed anti-atherogenic mechanism of adiponectin involves its ability to promote cholesterol efflux. We performed a systematic review of the role of adiponectin in cholesterol efflux and HDL biogenesis, and of the proteins and receptors believed to be implicated in this process. Nineteen eligible studies (7 clinical, 11 fundamental, 1 clinical + fundamental) were identified through Ovid Medline, Ovid Embase, and Pubmed, that support the notion that adiponectin plays a key role in promoting ABCA1-dependent cholesterol efflux and in modulating HDL biogenesis via activation of the PPAR-γ/LXR-α signalling pathways in macrophages. AdipoR1 and AdipoR2 are suggested to also be implicated in this process, however the data are conflicting/insufficient to establish any firm conclusions. Once the exact mechanisms are unravelled, adiponectin may be critical in defining future treatment strategies directed towards increasing HDL functionality and ultimately reducing atherosclerotic disease.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| | - Karina Gasbarrino
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| | - Stella S Daskalopoulou
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
13
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
14
|
Olson EJ, Mahar KM, Haws TF, Fossler MJ, Gao F, de Gouville AC, Sprecher DL, Lepore JJ. A Randomized, Placebo-Controlled Trial to Assess the Effects of 8 Weeks of Administration of GSK256073, a Selective GPR109A Agonist, on High-Density Lipoprotein Cholesterol in Subjects With Dyslipidemia. Clin Pharmacol Drug Dev 2019; 8:871-883. [PMID: 31268250 DOI: 10.1002/cpdd.704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/07/2019] [Indexed: 11/11/2022]
Abstract
GPR109A (HM74A), a G-protein-coupled receptor, is hypothesized to mediate lipid and lipoprotein changes and dermal flushing associated with niacin administration. GSK256073 (8-chloro-3-pentyl-1H-purine-2,6[3H,7H]-dione) is a selective GPR109A agonist shown to suppress fatty acid levels and produce mild flushing in short-term clinical studies. This study evaluated the effects of GSK256073 on lipids in subjects with low high-density lipoprotein cholesterol (HDLc). Subjects (n = 80) were randomized (1:1:1:1) to receive GSK256073 5, 50, or 150 mg/day or matching placebo for 8 weeks. The primary end point was determining the GSK256073 exposure-response relationship for change from baseline in HDLc. No significant exposure response was observed between GSK256073 and HDLc levels. GSK256073 did not significantly alter HDLc levels versus placebo, but rather revealed a trend at the 150-mg dose for a nonsignificant decrease in HDLc (-6.31%; P = .12) and an increase in triglycerides (median, 24.4%; 95% confidence interval, 7.3%-41.6%). Flushing was reported in 21%, 25%, and 60% of subjects (5, 50, and 150 mg, respectively) versus 24% for placebo. Results indicated that selective activation of the GPR109A receptor with GSK256073 did not produce niacin-like lipid effects. These findings add to the increasing evidence that niacin-mediated lipoprotein changes occur predominantly via GPR109A-independent pathways.
Collapse
Affiliation(s)
- Eric J Olson
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline, Collegeville, PA, USA
| | - Kelly M Mahar
- Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, Collegeville, PA, USA
| | - Thomas F Haws
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline, Collegeville, PA, USA
| | - Michael J Fossler
- Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, Collegeville, PA, USA
| | - Feng Gao
- Clinical Statistics, Metabolic Pathways and Cardiovascular Unit, GlaxoSmithKline, Collegeville, PA, USA
| | | | - Dennis L Sprecher
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline, Collegeville, PA, USA
| | - John J Lepore
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline, Collegeville, PA, USA
| |
Collapse
|
15
|
Scognamiglio M, Costa D, Sorriento A, Napoli C. Current Drugs and Nutraceuticals for the Treatment of Patients with Dyslipidemias. Curr Pharm Des 2019; 25:85-95. [DOI: 10.2174/1381612825666190130101108] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/20/2019] [Indexed: 02/05/2023]
Abstract
Coronary heart disease (CHD) remains the leading cause of disability and death in industrialized Countries.
Among many conditions, which contribute to the etiology and progression of CHD, the presence of high
low density lipoprotein-cholesterol (LDL-C) levels represents the major risk factor. Therefore, the reduction of
LDL-C levels plays a key role in the management of patients with high or very high cardiovascular risk. Although
statins represent the gold standard therapy for the reduction of cholesterol levels, these drugs do not allow to
achieve target levels of LDL-C in all patients. Indeed, a significant number of patients resulted intolerants, especially
when the dosage increased. The availability of new lipid-lowering drugs, such as ezetimibe and PCSK9
inhibitors, may represent an important alternative or complement to the conventional lipid-lowering therapies.
However, long-term studies are still needed to define both efficacy and safety of use of these latter new drugs.
Some nutraceuticals may become an adequate and effective support in the management of some patients. To date,
several nutraceuticals with different mechanism of actions that provide a good tolerability are available as lipidlowering
agents. In particular, the most investigated are red yeast rice, phytosterols, berberine, beta-glucans and
soy. The aim of this review was to report recent data on the efficacy and safety of principle hypocholesterolemic
drugs available and to evaluate the possible role of some nutraceuticals as support therapy in the management of
patients with dyslipidemias.
Collapse
Affiliation(s)
- Michele Scognamiglio
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences, University of Campania , Italy
| | - Dario Costa
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences, University of Campania , Italy
| | - Antonio Sorriento
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences, University of Campania , Italy
| | - Claudio Napoli
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences, University of Campania , Italy
| |
Collapse
|
16
|
Adiels M, Chapman MJ, Robillard P, Krempf M, Laville M, Borén J. Niacin action in the atherogenic mixed dyslipidemia of metabolic syndrome: Insights from metabolic biomarker profiling and network analysis. J Clin Lipidol 2018; 12:810-821.e1. [DOI: 10.1016/j.jacl.2018.03.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/19/2018] [Accepted: 03/22/2018] [Indexed: 01/26/2023]
|
17
|
Montserrat-de la Paz S, Lopez S, Bermudez B, Guerrero JM, Abia R, Muriana FJ. Effects of immediate-release niacin and dietary fatty acids on acute insulin and lipid status in individuals with metabolic syndrome. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:2194-2200. [PMID: 28960312 DOI: 10.1002/jsfa.8704] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/14/2017] [Accepted: 09/20/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND The nature of dietary fats profoundly affects postprandial hypertriglyceridemia and glucose homeostasis. Niacin is a potent lipid-lowering agent. However, limited data exist on postprandial triglycerides and glycemic control following co-administration of high-fat meals with a single dose of niacin in subjects with metabolic syndrome (MetS). The aim of the study was to explore whether a fat challenge containing predominantly saturated fatty acids (SFAs), monounsaturated fatty acids (MUFAs) or MUFAs plus omega-3 long-chain polyunsaturated (LCPUFAs) fatty acids together with a single dose of immediate-release niacin have a relevant role in postprandial insulin and lipid status in subjects with MetS. RESULTS In a randomized crossover within-subject design, 16 men with MetS were given a single dose of immediate-release niacin (2 g) and ∼15 cal kg-1 body weight meals containing either SFAs, MUFAs, MUFAs plus omega-3 LCPUFAs or no fat. At baseline and hourly over 6 h, plasma glucose, insulin, C-peptide, triglycerides, free fatty acids (FFAs), total cholesterol, and both high- and low-density lipoprotein cholesterol were assessed. Co-administered with niacin, high-fat meals significantly increased the postprandial concentrations of glucose, insulin, C-peptide, triglycerides, FFAs and postprandial indices of β-cell function. However, postprandial indices of insulin sensitivity were significantly decreased. These effects were significantly attenuated with MUFAs or MUFAs plus omega-3 LCPUFAs when compared with SFAs. CONCLUSION In the setting of niacin co-administration and compared to dietary SFAs, MUFAs limit the postprandial insulin, triglyceride and FFA excursions, and improve postprandial glucose homeostasis in MetS. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
| | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa (CSIC), Seville, Spain
| | - Beatriz Bermudez
- Department of Cell Biology, Faculty of Biology, University of Seville, Seville, Spain
| | - Juan M Guerrero
- Department of Clinical Biochemistry, University Hospital Virgen del Rocio, IBiS/CSIC/University of Seville, Seville, Spain
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa (CSIC), Seville, Spain
| | - Francisco Jg Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa (CSIC), Seville, Spain
| |
Collapse
|
18
|
Lamon-Fava S, Diffenderfer MR, Barrett PHR, Wan WY, Postfai B, Nartsupha C, Dolnikowski GG, Schaefer EJ. Differential Effects of Estrogen and Progestin on Apolipoprotein B100 and B48 Kinetics in Postmenopausal Women. Lipids 2018. [DOI: 10.1002/lipd.12011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University; Boston MA 02111 USA
| | - Margaret R. Diffenderfer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University; Boston MA 02111 USA
| | - P. Hugh R. Barrett
- School of Medicine and Pharmacology and Faculty of Engineering, Computing and Mathematics, The University of Western Australia; Perth WA 6009 Australia
| | - Wing Yee Wan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University; Boston MA 02111 USA
| | - Borbala Postfai
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University; Boston MA 02111 USA
| | - Chorthip Nartsupha
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University; Boston MA 02111 USA
| | - Gregory G. Dolnikowski
- Mass Spectrometry Core Unit; Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University; Boston MA 02111 USA
| | - Ernst J. Schaefer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University; Boston MA 02111 USA
| |
Collapse
|
19
|
Abstract
Nicotinic acid and nicotinamide, collectively referred to as niacin, are nutritional precursors of the bioactive molecules nicotinamide adenine dinucleotide (NAD) and nicotinamide adenine dinucleotide phosphate (NADP). NAD and NADP are important cofactors for most cellular redox reactions, and as such are essential to maintain cellular metabolism and respiration. NAD also serves as a cosubstrate for a large number of ADP-ribosylation enzymes with varied functions. Among the NAD-consuming enzymes identified to date are important genetic and epigenetic regulators, e.g., poly(ADP-ribose)polymerases and sirtuins. There is rapidly growing knowledge of the close connection between dietary niacin intake, NAD(P) availability, and the activity of NAD(P)-dependent epigenetic regulator enzymes. It points to an exciting role of dietary niacin intake as a central regulator of physiological processes, e.g., maintenance of genetic stability, and of epigenetic control mechanisms modulating metabolism and aging. Insight into the role of niacin and various NAD-related diseases ranging from cancer, aging, and metabolic diseases to cardiovascular problems has shifted our view of niacin as a vitamin to current views that explore its potential as a therapeutic.
Collapse
Affiliation(s)
- James B Kirkland
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | | |
Collapse
|
20
|
Mangat R, Borthwick F, Haase T, Jacome M, Nelson R, Kontush A, Vine DF, Proctor SD. Intestinal lymphatic HDL miR‐223 and ApoA‐I are reduced during insulin resistance and restored with niacin. FASEB J 2018; 32:1602-1612. [DOI: 10.1096/fj.201600298rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Rabban Mangat
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Faye Borthwick
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Tina Haase
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Miriam Jacome
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Randy Nelson
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Anatol Kontush
- National Institute for Health and Medical Research University of Pierre and Marie Curie, Salpétrière University Hospital Paris France
| | - Donna F. Vine
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Spencer D. Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| |
Collapse
|
21
|
Kudinov VA, Zakharova TS, Torkhovskaya TI, Ipatova OM, Archakov AI. [Pharmacological targets for dislipidemies correction. Opportunities and prospects of therapeutic usage]. BIOMEDITSINSKAIA KHIMIIA 2018; 64:66-83. [PMID: 29460837 DOI: 10.18097/pbmc20186401066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Literature data on influence of existing and new groups of drug preparations for dyslipidemias correction are systemized, and molecular mechanisms of their effects are reviewed. The results of experimental and clinical investigations aimed at revealing of new pharmacological targets of dyslipidemias correction were analyzed. The approaches for activation of high density lipoproteins functionality are described. The implementation of alternative preparations with new alternative mechanisms of action may be suggested to improve the effectiveness of traditional treatment in the future.
Collapse
Affiliation(s)
- V A Kudinov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | - O M Ipatova
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A I Archakov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
22
|
Schandelmaier S, Briel M, Saccilotto R, Olu KK, Arpagaus A, Hemkens LG, Nordmann AJ, Cochrane Heart Group. Niacin for primary and secondary prevention of cardiovascular events. Cochrane Database Syst Rev 2017; 6:CD009744. [PMID: 28616955 PMCID: PMC6481694 DOI: 10.1002/14651858.cd009744.pub2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Nicotinic acid (niacin) is known to decrease LDL-cholesterol, and triglycerides, and increase HDL-cholesterol levels. The evidence of benefits with niacin monotherapy or add-on to statin-based therapy is controversial. OBJECTIVES To assess the effectiveness of niacin therapy versus placebo, administered as monotherapy or add-on to statin-based therapy in people with or at risk of cardiovascular disease (CVD) in terms of mortality, CVD events, and side effects. SEARCH METHODS Two reviewers independently and in duplicate screened records and potentially eligible full texts identified through electronic searches of CENTRAL, MEDLINE, Embase, Web of Science, two trial registries, and reference lists of relevant articles (latest search in August 2016). SELECTION CRITERIA We included all randomised controlled trials (RCTs) that either compared niacin monotherapy to placebo/usual care or niacin in combination with other component versus other component alone. We considered RCTs that administered niacin for at least six months, reported a clinical outcome, and included adults with or without established CVD. DATA COLLECTION AND ANALYSIS Two reviewers used pre-piloted forms to independently and in duplicate extract trials characteristics, risk of bias items, and outcomes data. Disagreements were resolved by consensus or third party arbitration. We conducted random-effects meta-analyses, sensitivity analyses based on risk of bias and different assumptions for missing data, and used meta-regression analyses to investigate potential relationships between treatment effects and duration of treatment, proportion of participants with established coronary heart disease and proportion of participants receiving background statin therapy. We used GRADE to assess the quality of evidence. MAIN RESULTS We included 23 RCTs that were published between 1968 and 2015 and included 39,195 participants in total. The mean age ranged from 33 to 71 years. The median duration of treatment was 11.5 months, and the median dose of niacin was 2 g/day. The proportion of participants with prior myocardial infarction ranged from 0% (4 trials) to 100% (2 trials, median proportion 48%); the proportion of participants taking statin ranged from 0% (4 trials) to 100% (12 trials, median proportion 100%).Using available cases, niacin did not reduce overall mortality (risk ratio (RR) 1.05, 95% confidence interval (CI) 0.97 to 1.12; participants = 35,543; studies = 12; I2 = 0%; high-quality evidence), cardiovascular mortality (RR 1.02, 95% CI 0.93 to 1.12; participants = 32,966; studies = 5; I2 = 0%; moderate-quality evidence), non-cardiovascular mortality (RR 1.12, 95% CI 0.98 to 1.28; participants = 32,966; studies = 5; I2 = 0%; high-quality evidence), the number of fatal or non-fatal myocardial infarctions (RR 0.93, 95% CI 0.87 to 1.00; participants = 34,829; studies = 9; I2 = 0%; moderate-quality evidence), nor the number of fatal or non-fatal strokes (RR 0.95, 95% CI 0.74 to 1.22; participants = 33,661; studies = 7; I2 = 42%; low-quality evidence). Participants randomised to niacin were more likely to discontinue treatment due to side effects than participants randomised to control group (RR 2.17, 95% CI 1.70 to 2.77; participants = 33,539; studies = 17; I2 = 77%; moderate-quality evidence). The results were robust to sensitivity analyses using different assumptions for missing data. AUTHORS' CONCLUSIONS Moderate- to high-quality evidence suggests that niacin does not reduce mortality, cardiovascular mortality, non-cardiovascular mortality, the number of fatal or non-fatal myocardial infarctions, nor the number of fatal or non-fatal strokes but is associated with side effects. Benefits from niacin therapy in the prevention of cardiovascular disease events are unlikely.
Collapse
Affiliation(s)
- Stefan Schandelmaier
- McMaster UniversityDepartment of Health Research Methods, Evidence, and Impact1280 Main Street WestHamiltonONCanadaL8S4L8
| | - Matthias Briel
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Ramon Saccilotto
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Kelechi K Olu
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Armon Arpagaus
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Lars G Hemkens
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Alain J Nordmann
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | | |
Collapse
|
23
|
Sahebkar A, Reiner Ž, Simental-Mendía LE, Ferretti G, Cicero AFG. Effect of extended-release niacin on plasma lipoprotein(a) levels: A systematic review and meta-analysis of randomized placebo-controlled trials. Metabolism 2016; 65:1664-1678. [PMID: 27733255 DOI: 10.1016/j.metabol.2016.08.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 08/20/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023]
Abstract
AIM Lipoprotein(a) (Lp(a)) is a proatherogenic and prothrombotic lipoprotein. Our aim was to quantify the extended-release nicotinic acid Lp(a) reducing effect with a meta-analysis of the available randomized clinical trials. METHODS A meta-analysis and random-effects meta-regression were performed on data pooled from 14 randomized placebo-controlled clinical trials published between 1998 and 2015, comprising 17 treatment arms, which included 9013 subjects, with 5362 in the niacin arm. RESULTS The impact of ER niacin on plasma Lp(a) concentrations was reported in 17 treatment arms. Meta-analysis suggested a significant reduction of Lp(a) levels following ER niacin treatment (weighted mean difference - WMD: -22.90%, 95% CI: -27.32, -18.48, p<0.001). Results also remained similar when the meta-analysis was repeated with standardized mean difference as summary statistic (WMD: -0.66, 95% CI: -0.82, -0.50, p<0.001). When the studies were categorized according to the administered dose, there was a comparable effect between the subsets of studies with administered doses of <2000mg/day (WMD: -21.85%, 95% CI: -30.61, -13.10, p<0.001) and ≥2000mg/day (WMD: -23.21%, 95% CI: -28.41, -18.01, p<0.001). The results of the random-effects meta-regression did not suggest any significant association between the changes in plasma concentrations of Lp(a) with dose (slope: -0.0001; 95% CI: -0.01, 0.01; p=0.983), treatment duration (slope: -0.40; 95% CI: -0.97, 0.17; p=0.166), and percentage change in plasma HDL-C concentrations (slope: 0.44; 95% CI: -0.48, 1.36; p=0.350). CONCLUSION In this meta-analysis of randomized placebo-controlled clinical trials, treatment with nicotinic acid was associated with a significant reduction in Lp(a) levels.
Collapse
Affiliation(s)
- Amirhosssein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran; Metabolic Research Centre, Royal Perth Hospital, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Željko Reiner
- University Hospital Center Zagreb, Department of Internal medicine, Kišpatićeva 12, Zagreb, Croatia
| | | | - Gianna Ferretti
- Dipartimento di Scienze cliniche Specialistiche ed Odontostomatologiche (DISCO), Università Politecnica delle Marche, Italy
| | - Arrigo F G Cicero
- Medicine and Surgery Sciences Dept., Alma Mater Studiorum University of Bologna, Italy.
| |
Collapse
|
24
|
Abstract
Two cardiovascular outcome trials established niacin 3 g daily prevents hard cardiac events. However, as detailed in part I of this series, an extended-release (ER) alternative at only 2 g nightly demonstrated no comparable benefits in two outcome trials, implying the alternative is not equivalent to the established cardioprotective regimen. Since statins leave a significant treatment gap, this presents a major opportunity for developers. Importantly, the established regimen is cardioprotective, so the pathway is likely beneficial. Moreover, though effective, the established cardioprotective regimen is cumbersome, limiting clinical use. At the same time, the ER alternative has been thoroughly discredited as a viable substitute for the established cardioprotective regimen. Therefore, by exploiting the pathway and skillfully avoiding the problems with the established cardioprotective regimen and the ER alternative, developers could validate cardioprotective variations facing little meaningful competition from their predecessors. Thus, shrewd developers could effectively tap into a gold mine at the grave of the ER alternative. The GPR109A receptor was discovered a decade ago, leading to a large body of evidence commending the niacin pathway to a lower cardiovascular risk beyond statins. While mediating niacin's most prominent adverse effects, GPR109A also seems to mediate anti-lipolytic, anti-inflammatory, and anti-atherogenic effects of niacin. Several developers are investing heavily in novel strategies to exploit niacin's therapeutic pathways. These include selective GPR109A receptor agonists, niacin prodrugs, and a niacin metabolite, with encouraging early phase human data. In part II of this review, we summarize the accumulated results of these early phase studies of emerging niacin mimetics.
Collapse
|
25
|
Xiao C, Dash S, Morgantini C, Hegele RA, Lewis GF. Pharmacological Targeting of the Atherogenic Dyslipidemia Complex: The Next Frontier in CVD Prevention Beyond Lowering LDL Cholesterol. Diabetes 2016; 65:1767-78. [PMID: 27329952 DOI: 10.2337/db16-0046] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 03/23/2016] [Indexed: 11/13/2022]
Abstract
Notwithstanding the effectiveness of lowering LDL cholesterol, residual CVD risk remains in high-risk populations, including patients with diabetes, likely contributed to by non-LDL lipid abnormalities. In this Perspectives in Diabetes article, we emphasize that changing demographics and lifestyles over the past few decades have resulted in an epidemic of the "atherogenic dyslipidemia complex," the main features of which include hypertriglyceridemia, low HDL cholesterol levels, qualitative changes in LDL particles, accumulation of remnant lipoproteins, and postprandial hyperlipidemia. We briefly review the underlying pathophysiology of this form of dyslipidemia, in particular its association with insulin resistance, obesity, and type 2 diabetes, and the marked atherogenicity of this condition. We explain the failure of existing classes of therapeutic agents such as fibrates, niacin, and cholesteryl ester transfer protein inhibitors that are known to modify components of the atherogenic dyslipidemia complex. Finally, we discuss targeted repurposing of existing therapies and review promising new therapeutic strategies to modify the atherogenic dyslipidemia complex. We postulate that targeting the central abnormality of the atherogenic dyslipidemia complex, the elevation of triglyceride-rich lipoprotein particles, represents a new frontier in CVD prevention and is likely to prove the most effective strategy in correcting most aspects of the atherogenic dyslipidemia complex, thereby preventing CVD events.
Collapse
Affiliation(s)
- Changting Xiao
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Satya Dash
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Cecilia Morgantini
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Robert A Hegele
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Gary F Lewis
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Liu J, Hernandez-Ono A, Graham MJ, Galton VA, Ginsberg HN. Type 1 Deiodinase Regulates ApoA-I Gene Expression and ApoA-I Synthesis Independent of Thyroid Hormone Signaling. Arterioscler Thromb Vasc Biol 2016; 36:1356-66. [PMID: 27150392 DOI: 10.1161/atvbaha.116.307330] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/20/2016] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Plasma levels of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I (ApoA-I) are reduced in individuals with defective insulin signaling. Initial studies using liver-specific insulin receptor (InsR) knockout mice identified reduced expression of type 1 deiodinase (Dio1) as a potentially novel link between defective hepatic insulin signaling and reduced expression of the ApoA-I gene. Our objective was to examine the regulation of ApoA-I expression by Dio1. APPROACH AND RESULTS Acute inactivation of InsR by adenoviral delivery of Cre recombinase to InsR floxed mice reduced HDL-C and expression of both ApoA-I and Dio1. Overexpression of Dio1 in InsR knockout mice restored HDL-C and ApoA-I levels and increased the expression of ApoA-I. Dio1 knockout mice had low expression of ApoA-I and reduced serum levels of HDL-C and ApoA-I. Treatment of C57BL/6J mice with antisense to Dio1 reduced ApoA-I mRNA, HDL-C, and serum ApoA-I. Hepatic 3,5,3'-triiodothyronine content was normal or elevated in InsR knockout mice or Dio1 knockout mice. Knockdown of either InsR or Dio1 by siRNA in HepG2 cells decreased the expression of ApoA-I and ApoA-I synthesis and secretion. siRNA knockdown of InsR or Dio1 decreased activity of a region of the ApoA-I promoter lacking thyroid hormone response elements (region B). Electrophoretic mobility shift assay demonstrated that reduced Dio1 expression decreased the binding of nuclear proteins to region B. CONCLUSIONS Reductions in Dio1 expression reduce the expression of ApoA-I in a 3,5,3'-triiodothyronine-/thyroid hormone response element-independent manner.
Collapse
Affiliation(s)
- Jing Liu
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.).
| | - Antonio Hernandez-Ono
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.)
| | - Mark J Graham
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.)
| | - Valerie Anne Galton
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.)
| | - Henry N Ginsberg
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.).
| |
Collapse
|
27
|
Niacin Therapy, HDL Cholesterol, and Cardiovascular Disease: Is the HDL Hypothesis Defunct? Curr Atheroscler Rep 2016; 17:43. [PMID: 26048725 DOI: 10.1007/s11883-015-0521-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
High-density lipoprotein cholesterol (HDL-C) has been shown in epidemiologic studies to be associated with cardiovascular (CV) risk and thus significant efforts have been focused on HDL-C modulation. Multiple pharmaceutical agents have been developed with the goal of increasing HDL-C. Niacin, the most widely used medication to raise HDL-C, increases HDL-C by up to 25 % and was shown in multiple surrogate end point studies to reduce CV risk. However, two large randomized controlled trials of niacin, AIM-HIGH and HPS2-THRIVE, have shown that despite its effects on HDL-C, niacin does not decrease the incidence of CV events and may have significant adverse effects. Studies of other classes of agents such as cholesteryl ester transfer protein (CETP) inhibitors have also shown that even dramatic increases in HDL-C do not necessarily translate to reduction in clinical events. While these findings have cast doubt upon the importance of HDL-C modulation on CV risk, it is becoming increasingly clear that HDL function-related measures may be better targets for CV risk reduction. Increasing ApoA-I, the primary apolipoprotein associated with HDL, correlates with reduced risk of events, and HDL particle concentration (HDL-P) inversely associates with incident CV events adjusted for HDL-C and LDL particle measures. Cholesterol efflux, the mechanism by which macrophages in vessel walls secrete cholesterol outside cells, correlates with both surrogate end points and clinical events. The effects of niacin on these alternate measures of HDL have been conflicting. Further studies should determine if modulation of these HDL function markers translates to clinical benefits. Although the HDL cholesterol hypothesis may be defunct, the HDL function hypothesis is now poised to be rigorously tested.
Collapse
|
28
|
Diffenderfer MR, Lamon-Fava S, Marcovina SM, Barrett PHR, Lel J, Dolnikowski GG, Berglund L, Schaefer EJ. Distinct metabolism of apolipoproteins (a) and B-100 within plasma lipoprotein(a). Metabolism 2016; 65:381-90. [PMID: 26975530 PMCID: PMC4795479 DOI: 10.1016/j.metabol.2015.10.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/28/2015] [Accepted: 10/31/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Lipoprotein(a) [Lp(a)] is mainly similar in composition to LDL, but differs in having apolipoprotein (apo) (a) covalently linked to apoB-100. Our purpose was to examine the individual metabolism of apo(a) and apoB-100 within plasma Lp(a). MATERIALS AND METHODS The kinetics of apo(a) and apoB-100 in plasma Lp(a) were assessed in four men with dyslipidemia [Lp(a) concentration: 8.9-124.7nmol/L]. All subjects received a primed constant infusion of [5,5,5-(2)H3] L-leucine while in the constantly fed state. Lp(a) was immunoprecipitated directly from whole plasma; apo(a) and apoB-100 were separated by gel electrophoresis; and isotopic enrichment was determined by gas chromatography/mass spectrometry. RESULTS Multicompartmental modeling analysis indicated that the median fractional catabolic rates of apo(a) and apoB-100 within Lp(a) were significantly different at 0.104 and 0.263 pools/day, respectively (P=0.04). The median Lp(a) apo(a) production rate at 0.248nmol/kg·day(-1) was significantly lower than that of Lp(a) apoB-100 at 0.514nmol/kg·day(-1) (P=0.03). CONCLUSION Our data indicate that apo(a) has a plasma residence time (11days) that is more than twice as long as that of apoB-100 (4days) within Lp(a), supporting the concept that apo(a) and apoB-100 within plasma Lp(a) are not catabolized from the bloodstream as a unit in humans in the fed state.
Collapse
Affiliation(s)
- Margaret R Diffenderfer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | - Santica M Marcovina
- Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, 401 Queen Anne Avenue North, Seattle, WA 98109, USA.
| | - P Hugh R Barrett
- School of Medicine and Pharmacology and Faculty of Engineering, Computing and Mathematics, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Julian Lel
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | - Gregory G Dolnikowski
- Mass Spectrometry Unit, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | - Lars Berglund
- Clinical and Translational Science Center, University of California, Davis, 2921 Stockton Boulevard, Suite 1400, Sacramento, CA 95817, USA.
| | - Ernst J Schaefer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| |
Collapse
|
29
|
Pang J, Chan DC, Hamilton SJ, Tenneti VS, Watts GF, Barrett PHR. Effect of niacin on triglyceride-rich lipoprotein apolipoprotein B-48 kinetics in statin-treated patients with type 2 diabetes. Diabetes Obes Metab 2016; 18:384-91. [PMID: 26679079 DOI: 10.1111/dom.12622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/05/2015] [Accepted: 12/14/2015] [Indexed: 11/28/2022]
Abstract
AIM To investigate the effects of extended-release (ER) niacin on apolipoprotein B-48 (apoB-48) kinetics in statin-treated patients with type 2 diabetes (T2DM). METHODS A total of 12 men with T2DM were randomized to rosuvastatin or rosuvastatin plus ER niacin for 12 weeks and then crossed to the alternate therapy. Postprandial metabolic studies were performed at the end of each treatment period. D3-leucine tracer was administered as subjects consumed a high-fat liquid meal. ApoB-48 kinetics were determined using stable isotope tracer kinetics with fractional catabolic rates (FCRs) and secretion rates derived using a non-steady-state compartmental model. Area-under-the-curve (AUC) and incremental AUC (iAUC) for plasma triglyceride and apoB-48 were also calculated over the 10-h period after ingestion of the fat meal. RESULTS In statin-treated patients with T2DM, apoB-48 concentration was lower with ER niacin (8.24 ± 1.98 vs 5.48 ± 1.14 mg/l, p = 0.03) compared with statin alone. Postprandial triglyceride and apoB-48 AUC were also significantly lower on ER niacin treatment (-15 and -26%, respectively; p < 0.05), without any change to triglyceride and apoB-48 iAUC. ApoB-48 secretion rate in the basal state (3.21 ± 0.34 vs 2.50 ± 0.31 mg/kg/day; p = 0.04) and number of apoB-48-containing particles secreted in response to the fat load (1.35 ± 0.19 vs 0.84 ± 0.12 mg/kg; p = 0.02) were lower on ER niacin. ApoB-48 FCR was not altered with ER niacin (8.78 ± 1.04 vs 9.17 ± 1.26 pools/day; p = 0.79). CONCLUSIONS ER niacin reduces apoB-48 concentration by lowering fasting and postprandial apoB-48 secretion rate. This effect may be beneficial for lowering atherogenic postprandial lipoproteins and may provide cardiovascular disease risk benefit in patients with T2DM.
Collapse
Affiliation(s)
- J Pang
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - D C Chan
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - S J Hamilton
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
- Combined Universities Centre for Rural Health, University of Western Australia, Geraldton, Western Australia, Australia
| | - V S Tenneti
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - G F Watts
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - P H R Barrett
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
- Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
30
|
Liu L, Li C, Fu C, Li F. Dietary Niacin Supplementation Suppressed Hepatic Lipid Accumulation in Rabbits. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 29:1748-1755. [PMID: 27004817 PMCID: PMC5088423 DOI: 10.5713/ajas.15.0824] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/03/2016] [Accepted: 03/16/2016] [Indexed: 01/07/2023]
Abstract
An experiment was conducted to investigate the effect of niacin supplementation on hepatic lipid metabolism in rabbits. Rex Rabbits (90 d, n = 32) were allocated to two equal treatment groups: Fed basal diet (control) or fed basal diet with additional 200 mg/kg niacin supplementation (niacin). The results show that niacin significantly increased the levels of plasma adiponectin, hepatic apoprotein B and hepatic leptin receptors mRNA (p<0.05), but significantly decreased the hepatic fatty acid synthase activity and adiponectin receptor 2, insulin receptor and acetyl-CoA carboxylase mRNA levels (p<0.05). Plasma insulin had a decreasing tendency in the niacin treatment group compared with control (p = 0.067). Plasma very low density lipoproteins, leptin levels and the hepatic adiponectin receptor 1 and carnitine palmitoyl transferase 1 genes expression were not significantly altered with niacin addition to the diet (p>0.05). However, niacin treatment significantly inhibited the hepatocytes lipid accumulation compared with the control group (p<0.05). In conclusion, niacin treatment can decrease hepatic fatty acids synthesis, but does not alter fatty acids oxidation and triacylglycerol export. And this whole process attenuates lipid accumulation in liver. Besides, the hormones of insulin, leptin and adiponectin are associated with the regulation of niacin in hepatic lipid metabolism in rabbits.
Collapse
Affiliation(s)
- Lei Liu
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Chunyan Li
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Chunyan Fu
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Fuchang Li
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, China
| |
Collapse
|
31
|
Mooradian AD, Haas MJ. Targeting high-density lipoproteins: increasing de novo production versus decreasing clearance. Drugs 2016; 75:713-22. [PMID: 25895465 DOI: 10.1007/s40265-015-0390-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Although cardiovascular mortality has been decreasing in industrialized countries, there continues to be a substantial residual risk; thus, novel therapeutic agents and new targets of therapy have been sought. One highly plausible therapeutic target is high-density lipoprotein (HDL). HDL is a key player in reverse cholesterol transport and possesses a slew of other cardioprotective properties; however, recent trials with agents known to increase HDL levels have generally not shown any reduction in cardiovascular events. Further analysis of these trials suggest that fibrates have consistently reduced some cardiovascular outcomes, at least in the subgroup of patients with high serum triglycerides and low HDL cholesterol (HDLc) levels. Since fibrates, unlike niacin or cholesterol ester transfer protein inhibitors, increase HDLc level mostly through the stimulation of apolipoprotein A-I production, it is suggested that the quality and functionality of HDL are enhanced when de novo synthesis rather than inhibition of turnover is the mechanism of increasing HDL level. In this communication, the evidence for and against the cardioprotective properties of HDL is reviewed and the contemporary clinical trials are discussed.
Collapse
Affiliation(s)
- Arshag D Mooradian
- Department of Medicine, University of Florida College of Medicine, 655 West 11th Street, Jacksonville, FL, 32209, USA,
| | | |
Collapse
|
32
|
Abstract
The armamentarium for the treatment of dyslipidemia today comprises six different modes of action with overall around 24 different drugs. The treatment of lipid disorders was revolutionized with the introduction of statins which have become the most important therapeutic option available today to reduce and prevent atherosclerosis and its detrimental consequences like cardiovascular diseases and stroke. With and optimized reduction of elevated LDL levels with statins, the risk for cardiovascular diseases (CVD) can be reduced by 30%, indicating a residual remaining risk of 70% for the development and progression of CVD notifying still a high medical need for more effective antilipidemic drugs. Consequently, the search for novel lipid-modifying drugs is still one of the most active areas in research and development in the pharmaceutical industry. Major focus lies on approaches to LDL-lowering drugs superior to statins with regard to efficacy, safety, and patient compliance and on approaches modifying plasma levels and functionality of HDL particles based on the clinically validated inverse relationship between high-plasma HDL levels and the risk for CVD. The available drugs today for the treatment of dyslipidemia are small organic molecules or nonabsorbable polymers for binding of bile acids to be applied orally. Besides small molecules for novel targets, biological drugs such as monoclonal antibodies, antisense or gene-silencing oligonucleotides, peptidomimetics, reconstituted synthetic HDL particles and therapeutic proteins are novel approaches in clinical development are which have to be applied by injection or infusion. The promising clinical results of several novel drug candidates, particularly for LDL cholesterol lowering with monoclonal antibodies raised against PCSK9, may indicate more than a decade after the statins, the entrance of new breakthrough therapies to treat lipid disorders.
Collapse
Affiliation(s)
- Werner Kramer
- Institute of Biochemistry, Biocenter, Goethe-Universität Frankfurt, Max-von-Laue-Str. 9, Frankfurt, Germany.
| |
Collapse
|
33
|
Bell DA, Watts GF. Contemporary and Novel Therapeutic Options for Hypertriglyceridemia. Clin Ther 2015; 37:2732-50. [DOI: 10.1016/j.clinthera.2015.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/16/2022]
|
34
|
Titov VN, Rozhkova TA, Aripovsky AV. [Consecutive formation of the functions of high-, low-density and very-low-density lipoproteins during phylogenesis. Unique algorithm of the effects of lipid-lowering drugs]. TERAPEVT ARKH 2015; 87:123-131. [PMID: 26591564 DOI: 10.17116/terarkh2015879123-131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
During phylogenesis, all fatty acids (FA) were initially transported to cells by apoA-I high-density lipoproteins (HDL) in polar lipids. Later, active cellular uptake of saturated, monoenoic and unsaturated FA occurred via triglycerides (TG) in low-density lipoproteins (LDL). Active uptake of polyenoic FA (PUFA) required the following: a) PUFA re-esterified from polar phospholipids into nonpolar cholesteryl polyesters (poly-CLE), b) a novel protein, cholesteryl ester transfer protein (CETP), initiated poly-CLE transformation from HDL to LDL. CETP formed blood HDL-CETP-LDL complexes in which poly-CLE spontaneously came from polar lipids of TG in HDL to nonpolar TG in LDL. Then ligand LDLs formed and the cells actively absorbed PUFA via apoB-100 endocytosis. Some animal species (rats, mice, dogs) developed a spontaneous CETP-minus mutation followed by population death from atherosclerosis. However, there was another active CETP-independent uptake formed during phylogenesis; the cells internalized poly-CLE in HDL. Since apoA-I had no domain-ligand, another apoE/A-I ligand formed; the cells began synthesizing apoE/A-1 receptors. In cells of rabbits and primates absorbed cells PUFA consecutively: HDL-->LDL-->apoB-100 endocytosis; those of rats and dogs did HDL directly: HDL-->anoE/A-I endocytosis. In the rabbits, CETP was high, apoE in HDL was low, and the animals were sensitive to exogenous hypercholesterolemia. In the rats, CETP was low and ApoE in HDL-was high, and the animals were resistant to hypercholesterolemia. Reduced bioavailability of PUFA during their consecutive cellular uptake and develdpment of intercellular PUFA deficiency are fundamental to the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- V N Titov
- Russian Cardiology Research-and-Production Center, Ministry of Health of Russia, Moscow, Russia
| | - T A Rozhkova
- Russian Cardiology Research-and-Production Center, Ministry of Health of Russia, Moscow, Russia
| | - A V Aripovsky
- Russian Cardiology Research-and-Production Center, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
35
|
Mendivil CO, Furtado J, Morton AM, Wang L, Sacks FM. Novel Pathways of Apolipoprotein A-I Metabolism in High-Density Lipoprotein of Different Sizes in Humans. Arterioscler Thromb Vasc Biol 2015; 36:156-65. [PMID: 26543096 DOI: 10.1161/atvbaha.115.306138] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/21/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE A prevailing concept is that high-density lipoprotein (HDL) is secreted into the systemic circulation as a small mainly discoidal particle, which expands progressively and becomes spherical by uptake and esterification of cellular cholesterol and then contracts by cholesterol ester delivery to the liver, a process known as reverse cholesterol transport, thought to be impaired in people with low HDL cholesterol (HDLc). This metabolic framework has not been established in humans. APPROACH AND RESULTS We studied the metabolism of apolipoprotein A-I in 4 standard HDL sizes by endogenous isotopic labeling in 6 overweight adults with low HDLc and in 6 adults with normal body weight with high plasma HDLc. Contrary to expectation, HDL was secreted into the circulation in its entire size distribution from very small to very large similarly in both groups. Very small (prebeta) HDL comprised only 8% of total apolipoprotein A-I secretion. Each HDL subfraction circulated mostly within its secreted size range for 1 to 4 days and then was cleared. Enlargement of very small and medium to large and very large HDL and generation of very small from medium HDL were minor metabolic pathways. Prebeta HDL was cleared slower, whereas medium, large, and very large HDL were cleared faster in the low HDLc group. CONCLUSIONS A new model is proposed from these results in which HDL is metabolized in plasma mainly within several discrete, stable sizes across the common range of HDLc concentrations.
Collapse
Affiliation(s)
- Carlos O Mendivil
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.)
| | - Jeremy Furtado
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.)
| | - Allyson M Morton
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.)
| | - Liyun Wang
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.)
| | - Frank M Sacks
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.).
| |
Collapse
|
36
|
Ooi EM, Watts GF, Chan DC, Pang J, Tenneti VS, Hamilton SJ, McCormick SP, Marcovina SM, Barrett PHR. Effects of extended-release niacin on the postprandial metabolism of Lp(a) and ApoB-100-containing lipoproteins in statin-treated men with type 2 diabetes mellitus. Arterioscler Thromb Vasc Biol 2015; 35:2686-93. [PMID: 26515419 DOI: 10.1161/atvbaha.115.306136] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/20/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The effects of extended-release niacin (ERN; 1-2 g/d) on the metabolism of lipoprotein(a) (Lp(a)) and apolipoprotein (apo) B-100-containing lipoproteins were investigated in 11 statin-treated white men with type 2 diabetes mellitus in a randomized, crossover trial of 12-weeks duration. APPROACH AND RESULTS The kinetics of Lp(a) and very low-density lipoprotein (VLDL), intermediate-density lipoprotein, and low-density lipoprotein (LDL) apoB-100 were determined following a standardized oral fat load (87% fat) using intravenous administration of D3-leucine, gas chromatography-mass spectrometry, and compartmental modeling. ERN significantly decreased fasting plasma total cholesterol, LDL cholesterol, and triglyceride concentrations. These effects were achieved without significant changes in body weight or insulin resistance. ERN significantly decreased plasma Lp(a) concentration (-26.5%) and the production rates of apo(a) (-41.5%) and Lp(a)-apoB-100 (-32.1%); the effect was greater in individuals with elevated Lp(a) concentration. ERN significantly decreased VLDL (-58.7%), intermediate-density lipoprotein (-33.6%), and LDL (-18.3%) apoB-100 concentrations and the corresponding production rates (VLDL, -49.8%; intermediate-density lipoprotein, -44.7%; LDL, -46.1%). The number of VLDL apoB-100 particles secreted increased in response to the oral fat load. Despite this, total VLDL apoB-100 production over the 10-hour postprandial period was significantly decreased with ERN (-21.9%). CONCLUSIONS In statin-treated men with type 2 diabetes mellitus, ERN decreased plasma Lp(a) concentrations by decreasing the production of apo(a) and Lp(a)-apoB-100. ERN also decreased the concentrations of apoB-100-containing lipoproteins by decreasing VLDL production and the transport of these particles down the VLDL to LDL cascade. Our study provides further mechanistic insights into the lipid-regulating effects of ERN.
Collapse
Affiliation(s)
- Esther M Ooi
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.)
| | - Gerald F Watts
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.)
| | - Dick C Chan
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.)
| | - Jing Pang
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.)
| | - Vijay S Tenneti
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.)
| | - Sandra J Hamilton
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.)
| | - Sally P McCormick
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.)
| | - Santica M Marcovina
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.)
| | - P Hugh R Barrett
- From the Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (E.M.O., G.F.W., D.C.C., J.P., V.S.T., P.H.R.B.); Lipid Disorders Clinic, Cardiometabolic Service, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia (G.F.W.); Western Australian Centre for Rural Health, University of Western Australia, Geraldton, Australia (S.J.H.); Department of Biochemistry, University of Otago, Dunedin, New Zealand (S.P.M.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); and Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia (P.H.R.B.).
| |
Collapse
|
37
|
Yadav R, Liu Y, Kwok S, Hama S, France M, Eatough R, Pemberton P, Schofield J, Siahmansur TJ, Malik R, Ammori BA, Issa B, Younis N, Donn R, Stevens A, Durrington P, Soran H. Effect of Extended-Release Niacin on High-Density Lipoprotein (HDL) Functionality, Lipoprotein Metabolism, and Mediators of Vascular Inflammation in Statin-Treated Patients. J Am Heart Assoc 2015; 4:e001508. [PMID: 26374297 PMCID: PMC4599486 DOI: 10.1161/jaha.114.001508] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background The aim of this study was to explore the influence of extended-release niacin/laropiprant (ERN/LRP) versus placebo on high-density lipoprotein (HDL) antioxidant function, cholesterol efflux, apolipoprotein B100 (apoB)-containing lipoproteins, and mediators of vascular inflammation associated with 15% increase in high-density lipoprotein cholesterol (HDL-C). Study patients had persistent dyslipidemia despite receiving high-dose statin treatment. Methods and Results In a randomized double-blind, placebo-controlled, crossover trial, we compared the effect of ERN/LRP with placebo in 27 statin-treated dyslipidemic patients who had not achieved National Cholesterol Education Program-ATP III targets for low-density lipoprotein cholesterol (LDL-C). We measured fasting lipid profile, apolipoproteins, cholesteryl ester transfer protein (CETP) activity, paraoxonase 1 (PON1) activity, small dense LDL apoB (sdLDL-apoB), oxidized LDL (oxLDL), glycated apoB (glyc-apoB), lipoprotein phospholipase A2 (Lp-PLA2), lysophosphatidyl choline (lyso-PC), macrophage chemoattractant protein (MCP1), serum amyloid A (SAA) and myeloperoxidase (MPO). We also examined the capacity of HDL to protect LDL from in vitro oxidation and the percentage cholesterol efflux mediated by apoB depleted serum. ERN/LRP was associated with an 18% increase in HDL-C levels compared to placebo (1.55 versus 1.31 mmol/L, P<0.0001). There were significant reductions in total cholesterol, triglycerides, LDL cholesterol, total serum apoB, lipoprotein (a), CETP activity, oxLDL, Lp-PLA2, lyso-PC, MCP1, and SAA, but no significant changes in glyc-apoB or sdLDL-apoB concentration. There was a modest increase in cholesterol efflux function of HDL (19.5%, P=0.045), but no change in the antioxidant capacity of HDL in vitro or PON1 activity. Conclusions ERN/LRP reduces LDL-associated mediators of vascular inflammation, but has varied effects on HDL functionality and LDL quality, which may counter its HDL-C-raising effect. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT01054508.
Collapse
Affiliation(s)
- Rahul Yadav
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.) Cardiovascular Trials Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom (R.Y., S.K., M.F., R.E., J.S., H.S.)
| | - Yifen Liu
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.)
| | - See Kwok
- Cardiovascular Trials Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom (R.Y., S.K., M.F., R.E., J.S., H.S.)
| | - Salam Hama
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.)
| | - Michael France
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.) Cardiovascular Trials Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom (R.Y., S.K., M.F., R.E., J.S., H.S.) The Institute of Inflammation & Repair at the University of Manchester, United Kingdom (M.F.)
| | - Ruth Eatough
- Cardiovascular Trials Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom (R.Y., S.K., M.F., R.E., J.S., H.S.)
| | - Phil Pemberton
- Department of Biochemistry, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom (P.P.)
| | - Jonathan Schofield
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.) Cardiovascular Trials Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom (R.Y., S.K., M.F., R.E., J.S., H.S.)
| | - Tarza J Siahmansur
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.)
| | - Rayaz Malik
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.)
| | - Basil A Ammori
- Department of Surgery, Salford Royal NHS Foundation Trust, Salford, United Kingdom (B.A.A.)
| | - Basil Issa
- Department of Diabetes and Endocrinology, University Hospital of South Manchester, United Kingdom (B.I., N.Y.)
| | - Naveed Younis
- Department of Diabetes and Endocrinology, University Hospital of South Manchester, United Kingdom (B.I., N.Y.)
| | - Rachelle Donn
- Complex Disease Genetics, Centre for Musculoskeletal Research, University of Manchester, United Kingdom (R.D.)
| | - Adam Stevens
- Royal Manchester Children's Hospital, Manchester, United Kingdom (A.S.)
| | - Paul Durrington
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.)
| | - Handrean Soran
- Cardiovascular Research Group, Core Technologies Facility, University of Manchester, United Kingdom (R.Y., Y.L., S.H., M.F., J.S., T.J.S., R.M., P.D., H.S.) Cardiovascular Trials Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom (R.Y., S.K., M.F., R.E., J.S., H.S.)
| |
Collapse
|
38
|
Nicotinic Acid Accelerates HDL Cholesteryl Ester Turnover in Obese Insulin-Resistant Dogs. PLoS One 2015; 10:e0136934. [PMID: 26366727 PMCID: PMC4569091 DOI: 10.1371/journal.pone.0136934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 08/10/2015] [Indexed: 11/19/2022] Open
Abstract
AIM Nicotinic acid (NA) treatment decreases plasma triglycerides and increases HDL cholesterol, but the mechanisms involved in these change are not fully understood. A reduction in cholesteryl ester transfer protein (CETP) activity has been advanced to explain most lipid-modulating effects of NA. However, due to the central role of CETP in reverse cholesterol transport in humans, other effects of NA may have been hidden. As dogs have no CETP activity, we conducted this study to examine the specific effects of extended-release niacin (NA) on lipids and high-density lipoprotein (HDL) cholesteryl ester (CE) turnover in obese Insulin-Resistant dogs with increase plasma triglycerides. METHODS HDL kinetics were assessed in fasting dogs before and four weeks after NA treatment through endogenous labeling of cholesterol and apolipoprotein AI by simultaneous infusion of [1,2 13C2] acetate and [5,5,5 2H3] leucine for 8 h. Kinetic data were analyzed by compartmental modeling. In vitro cell cholesterol efflux of serum from NA-treated dogs was also measured. RESULTS NA reduced plasma total cholesterol, low-density lipoprotein cholesterol, HDL cholesterol, triglycerides (TG), and very-low-density lipoprotein TG concentrations (p < 0.05). The kinetic study also showed a higher cholesterol esterification rate (p < 0.05). HDL-CE turnover was accelerated (p < 0.05) via HDL removal through endocytosis and selective CE uptake (p < 0.05). We measured an elevated in vitro cell cholesterol efflux (p < 0.05) with NA treatment in accordance with a higher cholesterol esterification. CONCLUSION NA decreased HDL cholesterol but promoted cholesterol efflux and esterification, leading to improved reverse cholesterol transport. These results highlight the CETP-independent effects of NA in changes of plasma lipid profile.
Collapse
|
39
|
Croyal M, Ouguerram K, Passard M, Ferchaud-Roucher V, Chétiveaux M, Billon-Crossouard S, de Gouville AC, Lambert G, Krempf M, Nobécourt E. Effects of Extended-Release Nicotinic Acid on Apolipoprotein (a) Kinetics in Hypertriglyceridemic Patients. Arterioscler Thromb Vasc Biol 2015; 35:2042-7. [PMID: 26160958 DOI: 10.1161/atvbaha.115.305835] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/24/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine the mechanisms by which extended-release nicotinic acid reduces circulating lipoprotein (a) concentrations in hypertriglyceridemic patients. APPROACH AND RESULTS Eight nondiabetic, obese male subjects (aged 48±12 years; body mass index, 31.2±1.8 kg/m(2)) with hypertriglyceridemia (triglycerides, 226±78 mg/dL) were enrolled in an 8 week, double blind, placebo-controlled cross-over study. At the end of each treatment phase, fasted subjects received a 10 µmol/L per kg bolus injection of [5,5,5-(2)H3]-l-Leucine immediately followed by constant infusion of [5,5,5-(2)H3]-l-Leucine (10 µmol L(-1) kg(-1) h(-1)) for 14 hours, and blood samples were collected. A liquid chromatography-tandem mass spectrometry method was used to study apolipoprotein (a) (Apo(a)) kinetics. The fractional catabolic rate of Apo(a) was calculated with a single compartmental model using the apolipoprotein B100 (ApoB100) containing very low density lipoprotein tracer enrichment as a precursor pool. Extended-release nicotinic acid decreased plasma triglycerides (-46%; P=0.023), raised high-density lipoprotein cholesterol (+20%; P=0.008), and decreased Apo(a) plasma concentrations (-20%; P=0.008). Extended-release nicotinic acid also decreased ApoB100 (22%; P=0.008) and proprotein convertase subtilisin/kexin type 9 (PCSK9, -29%; P=0.008) plasma concentrations. Apo(a) fractional catabolic rate and production rates were decreased by 37% (0.58±0.28 versus 0.36±0.19 pool/d; P=0.008) and 50% (1.4±0.8 versus 0.7±0.4 nmol/kg per day; P=0.008), respectively. CONCLUSIONS Extended-release nicotinic acid treatment decreased Apo(a) plasma concentrations by 20%, production rates by 50%, and catabolism by 37%. ApoB100 and PCSK9 concentrations were also decreased by treatment, but no correlation was found with Apo(a) kinetic parameters.
Collapse
Affiliation(s)
- Mikaël Croyal
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Khadija Ouguerram
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Maxime Passard
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Véronique Ferchaud-Roucher
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Maud Chétiveaux
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Stéphanie Billon-Crossouard
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Anne-Charlotte de Gouville
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Gilles Lambert
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Michel Krempf
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.).
| | - Estelle Nobécourt
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| |
Collapse
|
40
|
Gomaraschi M, Adorni MP, Banach M, Bernini F, Franceschini G, Calabresi L. Effects of established hypolipidemic drugs on HDL concentration, subclass distribution, and function. Handb Exp Pharmacol 2015; 224:593-615. [PMID: 25523003 DOI: 10.1007/978-3-319-09665-0_19] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The knowledge of an inverse relationship between plasma high-density lipoprotein cholesterol (HDL-C) concentrations and rates of cardiovascular disease has led to the concept that increasing plasma HDL-C levels would be protective against cardiovascular events. Therapeutic interventions presently available to correct the plasma lipid profile have not been designed to specifically act on HDL, but have modest to moderate effects on plasma HDL-C concentrations. Statins, the first-line lipid-lowering drug therapy in primary and secondary cardiovascular prevention, have quite modest effects on plasma HDL-C concentrations (2-10%). Fibrates, primarily used to reduce plasma triglyceride levels, also moderately increase HDL-C levels (5-15%). Niacin is the most potent available drug in increasing HDL-C levels (up to 30%), but its use is limited by side effects, especially flushing.The present chapter reviews the effects of established hypolipidemic drugs (statins, fibrates, and niacin) on plasma HDL-C levels and HDL subclass distribution, and on HDL functions, including cholesterol efflux capacity, endothelial protection, and antioxidant properties.
Collapse
Affiliation(s)
- Monica Gomaraschi
- Center E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133, Milan, Italy,
| | | | | | | | | | | |
Collapse
|
41
|
Wierzbicki AS, Viljoen A. Fibrates and niacin: is there a place for them in clinical practice? Expert Opin Pharmacother 2014; 15:2673-80. [DOI: 10.1517/14656566.2014.972365] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
42
|
Ding Y, Li Y, Wen A. Effect of niacin on lipids and glucose in patients with type 2 diabetes: A meta-analysis of randomized, controlled clinical trials. Clin Nutr 2014; 34:838-44. [PMID: 25306426 DOI: 10.1016/j.clnu.2014.09.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/16/2014] [Accepted: 09/18/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS This study aims to conduct a meta-analysis to evaluate the effects of niacin on serum lipids and glucose in patients with type 2 diabetes mellitus (T2DM). METHODS A comprehensive literature search in Medline, Scopus, AMED, Cochrane and Clinical trial registry databases was performed to identify randomized controlled trials investigating the effect of niacin on serum HDL cholesterol (HDL-c), LDL cholesterol (LDL-c), triglycerides (TG) and fasting plasma glucose (FPG). Pooled effects were measured by weighted mean difference (WMD) using fixed-effects or random-effects models. Quality assessment, and subgroup, meta-regression and sensitivity analyses were conducted using standard methods. Inter-study heterogeneity was assessed and quantified. RESULTS The estimated pooled mean changes (95% confidence interval) with niacin were 0.27 (95% CI: 0.24 to 0.30; P < 0.001) mmol/L for HDL-c, -0.250 (95% CI: -0.47 to -0.03; P < 0.05) mmol/L for LDL-c and -0.39 (95% CI: -0.43 to -0.34; P < 0.001) mmol/L for TG compared with controls. There was a significant heterogeneity for the impact of niacin on LDL-c and FPG. Subgroup analyses revealed a significant increase in FPG 0.085 (95% CI: 0.029 to 0.141; P < 0.05) mmol/L compared with controls in patients with long term treatment. Our analysis also showed the absence of publication bias and any dose-response relations between niacin and effect size. CONCLUSIONS Analysis of the results showed that niacin alone or in combination significantly improved lipid abnormalities in patients with TDM, but requires monitoring of glucose in long term treatment.
Collapse
Affiliation(s)
- Yi Ding
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - YuWen Li
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - AiDong Wen
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
43
|
Taylor JK, Plaisance EP, Mahurin AJ, Mestek ML, Moncada-Jimenez J, Grandjean PW. Paraoxonase responses to exercise and niacin therapy in men with metabolic syndrome. Redox Rep 2014; 20:42-8. [PMID: 25180827 DOI: 10.1179/1351000214y.0000000103] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Our purpose was to characterize changes in paraoxonase 1 (PON1) activity and concentration after single aerobic exercise sessions conducted before and after 6 weeks of niacin therapy in men with metabolic syndrome (MetS). Twelve men with MetS expended 500 kcal by walking at 65% of VO2max before and after a 6-week regimen of niacin. Niacin doses were titrated by 500 mg/week from 500 to 1500 mg/day and maintained at 1500 mg/day for the last 4 weeks. Fasting blood samples were collected before and 24 hours after each exercise session and analyzed for PON1 activity, PON1 concentration, myeloperoxidase (MPO), apolipoprotein A1, oxidized low-density lipoprotein (oLDL), lipoprotein particle sizes and concentrations. PON1 activity, PON1 concentration, MPO, and oLDL were unaltered following the independent effects of exercise and niacin (P > 0.05 for all). High-density lipoprotein particle size decreased by 3% (P = 0.040) and concentrations of small very low-density lipoprotein increased (P = 0.016) following exercise. PON1 activity increased 6.1% (P = 0.037) and PON1 concentrations increased 11.3% (P = 0.015) with the combination of exercise and niacin. Exercise and niacin works synergistically to increase PON1 activity and concentration with little or no changes in lipoproteins or markers of lipid oxidation.
Collapse
|
44
|
Sahebkar A, Chew GT, Watts GF. Recent advances in pharmacotherapy for hypertriglyceridemia. Prog Lipid Res 2014; 56:47-66. [PMID: 25083925 DOI: 10.1016/j.plipres.2014.07.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/10/2014] [Accepted: 07/18/2014] [Indexed: 12/20/2022]
Abstract
Elevated plasma triglyceride (TG) concentrations are associated with an increased risk of atherosclerotic cardiovascular disease (CVD), hepatic steatosis and pancreatitis. Existing pharmacotherapies, such as fibrates, n-3 polyunsaturated fatty acids (PUFAs) and niacin, are partially efficacious in correcting elevated plasma TG. However, several new TG-lowering agents are in development that can regulate the transport of triglyceride-rich lipoproteins (TRLs) by modulating key enzymes, receptors or ligands involved in their metabolism. Balanced dual peroxisome proliferator-activated receptor (PPAR) α/γ agonists, inhibitors of microsomal triglyceride transfer protein (MTTP) and acyl-CoA:diacylglycerol acyltransferase-1 (DGAT-1), incretin mimetics, and apolipoprotein (apo) B-targeted antisense oligonucleotides (ASOs) can all decrease the production and secretion of TRLs; inhibitors of cholesteryl ester transfer protein (CETP) and angiopoietin-like proteins (ANGPTLs) 3 and 4, monoclonal antibodies (Mabs) against proprotein convertase subtilisin/kexin type 9 (PCSK9), apoC-III-targeted ASOs, selective peroxisome proliferator-activated receptor modulators (SPPARMs), and lipoprotein lipase (LPL) gene replacement therapy (alipogene tiparvovec) enhance the catabolism and clearance of TRLs; dual PPAR-α/δ agonists and n-3 polyunsaturated fatty acids can lower plasma TG by regulating both TRL secretion and catabolism. Varying degrees of TG reduction have been reported with the use of these therapies, and for some agents such as CETP inhibitors and PCSK9 Mabs findings have not been consistent. Whether they reduce CVD events has not been established. Trials investigating the effect of CETP inhibitors (anacetrapib and evacetrapib) and PCSK9 Mabs (AMG-145 and REGN727/SAR236553) on CVD outcomes are currently in progress, although these agents also regulate LDL metabolism and, in the case of CETP inhibitors, HDL metabolism. Further to CVD risk reduction, these new treatments might also have a potential role in the management of diabetes and non-alcoholic fatty liver disease owing to their insulin-sensitizing action (PPAR-α/γ agonists) and potential capacity to decrease hepatic TG accumulation (PPAR-α/δ agonists and DGAT-1 inhibitors), but this needs to be tested in future trials. We summarize the clinical trial findings regarding the efficacy and safety of these novel therapies for hypertriglyceridemia.
Collapse
Affiliation(s)
- Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Gerard T Chew
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Gerald F Watts
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia.
| |
Collapse
|
45
|
Abstract
High-density lipoproteins (HDL) are a target for drug development because of their proposed anti-atherogenic properties. In this review, we will briefly discuss the currently established drugs for increasing HDL-C, namely niacin and fibrates, and some of their limitations. Next, we will focus on novel alternative therapies that are currently being developed for raising HDL-C, such as CETP inhibitors. Finally, we will conclude with a review of novel drugs that are being developed for modulating the function of HDL based on HDL mimetics. Gaps in our knowledge and the challenges that will have to be overcome for these new HDL based therapies will also be discussed.
Collapse
Affiliation(s)
- Alan T Remaley
- National Heart, Lung and Blood Institute, NIH, 10 Center Drive, Bldg. 10, Rm. 2C-433, Bethesda, MD, USA
| | - Giuseppe D Norata
- Department of Pharmacological Sciences, Università degli Studi di Milano, Milano, Italy Center for the Study of Atherosclerosis, Società Italiana Studio Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, Italy The Blizard Institute, Centre for Diabetes, Barts and The London School of Medicine & Dentistry, Queen Mary University, London, UK
| | - Alberico L Catapano
- Department of Pharmacological Sciences, Università degli Studi di Milano, Milano, Italy IRCCS Multimedica, Milan, Italy
| |
Collapse
|
46
|
|
47
|
Chan DC, Barrett PHR, Watts GF. The metabolic and pharmacologic bases for treating atherogenic dyslipidaemia. Best Pract Res Clin Endocrinol Metab 2014; 28:369-85. [PMID: 24840265 DOI: 10.1016/j.beem.2013.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Dyslipoproteinaemia is a cardinal feature of the metabolic syndrome that accelerates atherosclerosis. It is characterized by high plasma concentrations of triglyceride-rich and apolipoprotein (apo) B-containing lipoproteins, with depressed high-density lipoprotein (HDL) and increased small dense low-density lipoprotein (LDL) particle concentrations. Dysregulation of lipoprotein metabolism in the metabolic syndrome may be due to a combination of overproduction of very-low density lipoprotein (VLDL) apoB, decreased catabolism of apoB-containing particles, and increased catabolism of HDL apoA-I particles. These abnormalities are due to a global metabolic effect of insulin resistance and visceral obesity. Lifestyle modifications (dietary restriction and increased exercise) and pharmacological treatments favourably alter lipoprotein transport by decreasing the hepatic secretion of VLDL-apoB and the catabolism of HDL apoA-I, as well as by increasing the clearance of LDL-apoB. The safety and tolerability of combination drug therapy based on statins is important and merits further investigation. There are several pipeline therapies for correcting triglyceride-rich lipoprotein and HDL metabolism. However, their clinical efficacy, safety and cost-effectiveness remain to be demonstrated.
Collapse
Affiliation(s)
- Dick C Chan
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - P Hugh R Barrett
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia; Faculty of Engineering, Computing and Mathematics, University of Western Australia, Perth, Australia
| | - Gerald F Watts
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Royal Perth Hospital, Perth, Australia.
| |
Collapse
|
48
|
Kingwell BA, Chapman MJ, Kontush A, Miller NE. HDL-targeted therapies: progress, failures and future. Nat Rev Drug Discov 2014; 13:445-64. [DOI: 10.1038/nrd4279] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Wang H, Blumberg JB, Chen CYO, Choi SW, Corcoran MP, Harris SS, Jacques PF, Kristo AS, Lai CQ, Lamon-Fava S, Matthan NR, McKay DL, Meydani M, Parnell LD, Prokopy MP, Scott TM, Lichtenstein AH. Dietary modulators of statin efficacy in cardiovascular disease and cognition. Mol Aspects Med 2014; 38:1-53. [PMID: 24813475 DOI: 10.1016/j.mam.2014.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/14/2014] [Accepted: 04/14/2014] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease remains the leading cause of morbidity and mortality in the United States and other developed countries, and is fast growing in developing countries, particularly as life expectancy in all parts of the world increases. Current recommendations for the prevention of cardiovascular disease issued jointly from the American Academy of Cardiology and American Heart Association emphasize that lifestyle modification should be incorporated into any treatment plan, including those on statin drugs. However, there is a dearth of data on the interaction between diet and statins with respect to additive, complementary or antagonistic effects. This review collates the available data on the interaction of statins and dietary patterns, cognition, genetics and individual nutrients, including vitamin D, niacin, omega-3 fatty acids, fiber, phytochemicals (polyphenols and stanols) and alcohol. Of note, although the available data is summarized, the scope is limited, conflicting and disparate. In some cases it is likely there is unrecognized synergism. Virtually no data are available describing the interactions of statins with dietary components or dietary pattern in subgroups of the population, particularly those who may benefit most were positive effects identified. Hence, it is virtually impossible to draw any firm conclusions at this time. Nevertheless, this area is important because were the effects of statins and diet additive or synergistic harnessing the effect could potentially lead to the use of a lower intensity statin or dose.
Collapse
Affiliation(s)
- Huifen Wang
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Jeffrey B Blumberg
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - C-Y Oliver Chen
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Sang-Woon Choi
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA.
| | - Michael P Corcoran
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Susan S Harris
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Paul F Jacques
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Aleksandra S Kristo
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Chao-Qiang Lai
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Stefania Lamon-Fava
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Nirupa R Matthan
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Diane L McKay
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Mohsen Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Laurence D Parnell
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Max P Prokopy
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Tammy M Scott
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Alice H Lichtenstein
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| |
Collapse
|
50
|
Niacin activates the G protein estrogen receptor (GPER)-mediated signalling. Cell Signal 2014; 26:1466-75. [PMID: 24662263 DOI: 10.1016/j.cellsig.2014.03.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/17/2014] [Accepted: 03/17/2014] [Indexed: 01/13/2023]
Abstract
Nicotinic acid, also known as niacin, is the water soluble vitamin B3 used for decades for the treatment of dyslipidemic diseases. Its action is mainly mediated by the G protein-coupled receptor (GPR) 109A; however, certain regulatory effects on lipid levels occur in a GPR109A-independent manner. The amide form of nicotinic acid, named nicotinamide, acts as a vitamin although neither activates the GPR109A nor exhibits the pharmacological properties of nicotinic acid. In the present study, we demonstrate for the first time that nicotinic acid and nicotinamide bind to and activate the GPER-mediated signalling in breast cancer cells and cancer-associated fibroblasts (CAFs). In particular, we show that both molecules are able to promote the up-regulation of well established GPER target genes through the EGFR/ERK transduction pathway. As a biological counterpart, nicotinic acid and nicotinamide induce proliferative and migratory effects in breast cancer cells and CAFs in a GPER-dependent fashion. Moreover, nicotinic acid prevents the up-regulation of ICAM-1 triggered by the pro-inflammatory cytokine TNF-α and stimulates the formation of endothelial tubes through GPER in HUVECs. Together, our findings concerning the agonist activity for GPER displayed by both nicotinic acid and nicotinamide broaden the mechanisms involved in the biological action of these molecules and further support the potential of a ligand to induce different responses mediated in a promiscuous manner by distinct GPCRs.
Collapse
|