1
|
Moumou M, Tayebi A, Hadini A, Noman OM, Alhalmi A, Ahmoda H, Amrani S, Harnafi H. Combining In Vitro, In Vivo, and In Silico Approaches to Explore the Effect of Ceratonia siliqua and Ocimum basilicum Rich Phenolic Formula on Lipid Metabolism and Plasma Lipoprotein Oxidation in Mice Fed a High-Fat Diet: A Follow-Up Study. Metabolites 2025; 15:36. [PMID: 39852379 PMCID: PMC11767511 DOI: 10.3390/metabo15010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hyperlipidemia is a serious risk factor for cardiovascular diseases and liver steatosis. In this work, we explored the effect of an herbal formula (CBF) containing immature Ceratonia siliqua pods and Ocimum basilicum extracts on lipid metabolism disorders and lipoprotein-rich plasma (LRP) oxidation in mice. METHODS The phenolic composition was determined using HPLC-DAD analysis. The antioxidant activity was studied using various in vitro methods. Acute toxicity was evaluated in mice. Importantly, the effect of the CBF on lipid metabolism disorders was investigated in a high-fat diet (HFD) hyperlipidemia mouse model. An in silico study was carried out to predict underlying mechanisms. RESULTS The HPLC analysis revealed gallic acid, cinnamic acid, and naringenin as major phenolics of the carob pod aqueous extract. Concerning the basil hydro-ethanolic extract, rosmarinic, chicoric, caftaric, and caffeic acids were the main phenolics. Accordingly, the CBF prevented LRP oxidation in a concentration-dependent manner. This formula is not toxic in mice (LD50 > 2000 mg/kg body weight). Moreover, animals administered the CBF at 200 mg/kg/day presented a significant decline in their body weight gain, adipose tissue weight, plasma total cholesterol, low-density lipoprotein cholesterol (LDL-C) level, and glycaemia after 10 weeks' treatment. Accordingly, the CBF decreased the plasma atherogenic index and the LDL-C to HDL-C ratio and reduced the level of fats accumulated in the liver. The molecular docking study revealed that chicoric, rosmarinic, and caftaric acids, and naringenin bound particularly strongly to many proteins involved in the regulation of lipid and cholesterol metabolism. This includes the HMG-CoA reductase, PPARα/γ, PCSK9, Cyp7a1, and ATP-citrate lyase. CONCLUSIONS The CBF could be a good source of natural supplements, functional foods, and pharmaceuticals effective in managing hyperlipidemia and oxidative stress and preventing their related cardiovascular disorders.
Collapse
Affiliation(s)
- Mohammadine Moumou
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, University Mohammed First, Oujda 60000, Morocco; (M.M.); (A.T.); (A.H.); (S.A.)
| | - Amani Tayebi
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, University Mohammed First, Oujda 60000, Morocco; (M.M.); (A.T.); (A.H.); (S.A.)
| | - Abderrahmane Hadini
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, University Mohammed First, Oujda 60000, Morocco; (M.M.); (A.T.); (A.H.); (S.A.)
| | - Omar M. Noman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Abdulsalam Alhalmi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Hamza Ahmoda
- Universitätsklinik für Viszerale Chirurgie und Medizin, Universität Bern, 3010 Bern, Switzerland;
| | - Souliman Amrani
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, University Mohammed First, Oujda 60000, Morocco; (M.M.); (A.T.); (A.H.); (S.A.)
| | - Hicham Harnafi
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, University Mohammed First, Oujda 60000, Morocco; (M.M.); (A.T.); (A.H.); (S.A.)
| |
Collapse
|
2
|
Majethia GN, Haq W, Balendiran GK. A facile synthesis of 2-(4-((4-chlorophenyl)(hydroxy)methyl) phenoxy)-2-methylpropanoic acid: Metabolite of anti-hyperlipidemic drug Fenofibrate. RESULTS IN CHEMISTRY 2024; 7:101282. [PMID: 39086552 PMCID: PMC11290303 DOI: 10.1016/j.rechem.2023.101282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024] Open
Abstract
Synthesis and characterization of drug metabolites has emerged as an important area of research in consideration to the significant contribution of studies on metabolites in drug research. The present work comprises synthesis of 2-(4-((4-chlorophenyl)(hydroxy)methyl) phenoxy)-2-methylpropanoic acid, a metabolite of anti-hyperlipidemic drug fenofibrate. The desired compound was prepared by two different synthetic routes. The ketone group of fenofibric acid was reduced using sodium borohydride in one route whereas the hydrolysis of isopropyl ester of the reduced fenofibrate was achieved by the mild alkaline hydrolysis in the other path. Both the ways of synthesis furnished the desired compound in excellent yield and purity. The new synthetic congener was characterized by spectroscopic methods.
Collapse
Affiliation(s)
- Greesha N Majethia
- Department of Chemistry, Youngstown State University, One University Plaza, Youngstown, OH, USA
| | - Wahajul Haq
- Department of Chemistry, Youngstown State University, One University Plaza, Youngstown, OH, USA
| | | |
Collapse
|
3
|
Fuior EV, Zvintzou E, Filippatos T, Giannatou K, Mparnia V, Simionescu M, Gafencu AV, Kypreos KE. Peroxisome Proliferator-Activated Receptor α in Lipoprotein Metabolism and Atherosclerotic Cardiovascular Disease. Biomedicines 2023; 11:2696. [PMID: 37893070 PMCID: PMC10604751 DOI: 10.3390/biomedicines11102696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are a group of ligand-binding transcription factors with pivotal action in regulating pleiotropic signaling pathways of energetic metabolism, immune responses and cell proliferation and differentiation. A significant body of evidence indicates that the PPARα receptor is an important modulator of plasma lipid and lipoprotein metabolism, with pluripotent effects influencing the lipid and apolipoprotein cargo of both atherogenic and antiatherogenic lipoproteins and their functionality. Clinical evidence supports an important role of PPARα agonists (fibric acid derivatives) in the treatment of hypertriglyceridemia and/or low high-density lipoprotein (HDL) cholesterol levels, although the effects of clinical trials are contradictory and point to a reduction in the risk of nonfatal and fatal myocardial infarction events. In this manuscript, we provide an up-to-date critical review of the existing relevant literature.
Collapse
Affiliation(s)
- Elena Valeria Fuior
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Evangelia Zvintzou
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Theodosios Filippatos
- Internal Medicine Clinic, Department of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Katerina Giannatou
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Victoria Mparnia
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Anca Violeta Gafencu
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Kyriakos E. Kypreos
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
| |
Collapse
|
4
|
Thoduvayil S, Weerakkody JS, Sundaram RVK, Topper M, Bera M, Coleman J, Li X, Mariappan M, Ramakrishnan S. Rapid Quantification of First and Second Phase Insulin Secretion Dynamics using an In vitro Platform for Improving Insulin Therapy. Cell Calcium 2023; 113:102766. [PMID: 37295201 PMCID: PMC10450995 DOI: 10.1016/j.ceca.2023.102766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
High-throughput quantification of the first- and second-phase insulin secretion dynamics is intractable with current methods. The fact that independent secretion phases play distinct roles in metabolism necessitates partitioning them separately and performing high-throughput compound screening to target them individually. We developed an insulin-nanoluc luciferase reporter system to dissect the molecular and cellular pathways involved in the separate phases of insulin secretion. We validated this method through genetic studies, including knockdown and overexpression, as well as small-molecule screening and their effects on insulin secretion. Furthermore, we demonstrated that the results of this method are well correlated with those of single-vesicle exocytosis experiments conducted on live cells, providing a quantitative reference for the approach. Thus, we have developed a robust methodology for screening small molecules and cellular pathways that target specific phases of insulin secretion, resulting in a better understanding of insulin secretion, which in turn will result in a more effective insulin therapy through the stimulation of endogenous glucose-stimulated insulin secretion.
Collapse
Affiliation(s)
- Sikha Thoduvayil
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Jonathan S Weerakkody
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Ramalingam Venkat Kalyana Sundaram
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Mackenzie Topper
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA
| | - Manindra Bera
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Jeff Coleman
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Xia Li
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Malaiyalam Mariappan
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Sathish Ramakrishnan
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA.
| |
Collapse
|
5
|
Park MS, Youn JC, Kim EJ, Han KH, Lee SH, Kim SH, Kim BJ, Kwon SU, Ryu KH. Efficacy and Safety of Fenofibrate-Statin Combination Therapy in Patients With Inadequately Controlled Triglyceride Levels Despite Previous Statin Monotherapy: A Multicenter, Randomized, Double-blind, Phase IV Study. Clin Ther 2021; 43:1735-1747. [PMID: 34518033 DOI: 10.1016/j.clinthera.2021.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/21/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE Residual cardiovascular risk reduction by fenofibrate in patients with high serum triglyceride (TG) levels despite previous statin monotherapy is not well characterized. The purpose of this study was to evaluate the efficacy and safety of a combination of choline fenofibrate and statin in patients with inadequately controlled TG levels despite previous statin monotherapy. METHODS This prospective, multicenter, randomized, double-blind study was conducted in Korea. A total of 133 patients with controlled LDL-C but elevated TG levels, already receiving statin monotherapy, were enrolled in the study, which was conducted from July 2018 to December 2019. Patients were randomly assigned to receive combination therapy with choline fenofibrate and statin or statin monotherapy in a 1:1 ratio. After 8 weeks of treatment, the lipid profiles and safety parameters of the patients in the 2 groups were compared. FINDINGS The study included 127 patients (64 in the combination group and 63 in the control group) older than 19 years. After 8 weeks of therapy, mean serum TG levels significantly decreased from 269.8 to 145.5 mg/dL (P < 0.0001) in the combination therapy group, whereas no significant changes occurred in the statin monotherapy group (from 271.1 to 280.5 mg/dL). Contrarily, the mean serum HDLC levels significantly increased from 45.0 to 50.4 mg/dL (P = 0.0004) in the combination therapy group, whereas there were no significant changes in the monotherapy group (from 44.3 to 44.7 mg/dL). There were no additional serious adverse events in the combination therapy group compared with the statin monotherapy group. IMPLICATIONS The combination therapy using choline fenofibrate and statin was found to be effective in serum TG control and likely tolerable in patients with high TG levels despite statin monotherapy. A larger study, conducted for a longer duration, is needed to evaluate the effectiveness of this combination in reducing cardiovascular risk. ClinicalTrials.gov identifier: NCT03874260.
Collapse
Affiliation(s)
- Myung Soo Park
- Department of Cardiology, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Jong-Chan Youn
- Division of Cardiology, Department of Internal Medicine, Seoul St Mary's Hospital, Cardiovascular Research Institute for Intractable Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eung Ju Kim
- Department of Cardiology, Korea University, Guro Hospital, Seoul, Korea
| | - Ki Hoon Han
- Department of Internal Medicine, Ulsan University, Asan Medical Center, Seoul, Korea
| | - Sang Hak Lee
- Department of Cardiology and Cardiovascular Research Institute, Yonsei University, Severance Hospital, Seoul, Korea
| | - Sung Hea Kim
- Department of Cardiology, Konkuk University Medical Center, Seoul, Korea
| | - Byung Jin Kim
- Department of Internal Medicine, Sungkyunkwan University, Kangbuk Samsung Hospital, Seoul, Korea
| | - Sung Uk Kwon
- Department of Cardiology, Inje University, Ilsan Paik Hospital, Goyang, Korea
| | - Kyu-Hyung Ryu
- Department of Cardiology, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Korea.
| |
Collapse
|
6
|
van den Hoek AM, Verschuren L, Caspers MPM, Worms N, Menke AL, Princen HMG. Beneficial effects of elafibranor on NASH in E3L.CETP mice and differences between mice and men. Sci Rep 2021; 11:5050. [PMID: 33658534 PMCID: PMC7930243 DOI: 10.1038/s41598-021-83974-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/02/2021] [Indexed: 11/25/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is the most rapidly growing liver disease that is nevertheless without approved pharmacological treatment. Despite great effort in developing novel NASH therapeutics, many have failed in clinical trials. This has raised questions on the adequacy of preclinical models. Elafibranor is one of the drugs currently in late stage development which had mixed results for phase 2/interim phase 3 trials. In the current study we investigated the response of elafibranor in APOE*3Leiden.CETP mice, a translational animal model that displays histopathological characteristics of NASH in the context of obesity, insulin resistance and hyperlipidemia. To induce NASH, mice were fed a high fat and cholesterol (HFC) diet for 15 weeks (HFC reference group) or 25 weeks (HFC control group) or the HFC diet supplemented with elafibranor (15 mg/kg/d) from week 15–25 (elafibranor group). The effects on plasma parameters and NASH histopathology were assessed and hepatic transcriptome analysis was used to investigate the underlying pathways affected by elafibranor. Elafibranor treatment significantly reduced steatosis and hepatic inflammation and precluded the progression of fibrosis. The underlying disease pathways of the model were compared with those of NASH patients and illustrated substantial similarity with molecular pathways involved, with 87% recapitulation of human pathways in mice. We compared the response of elafibranor in the mice to the response in human patients and discuss potential pitfalls when translating preclinical results of novel NASH therapeutics to human patients. When taking into account that due to species differences the response to some targets, like PPAR-α, may be overrepresented in animal models, we conclude that elafibranor may be particularly useful to reduce hepatic inflammation and could be a pharmacologically useful agent for human NASH, but probably in combination with other agents.
Collapse
Affiliation(s)
- Anita M van den Hoek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands.
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Martien P M Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Nicole Worms
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Aswin L Menke
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Hans M G Princen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
7
|
Dai J, Li Y, Kametani F, Cui X, Igarashi Y, Huo J, Miyahara H, Mori M, Higuchi K. Curcumin promotes AApoAII amyloidosis and peroxisome proliferation in mice by activating the PPARα signaling pathway. eLife 2021; 10:e63538. [PMID: 33496266 PMCID: PMC7880682 DOI: 10.7554/elife.63538] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/10/2021] [Indexed: 12/16/2022] Open
Abstract
Curcumin is a polyphenol compound that exhibits multiple physiological activities. To elucidate the mechanisms by which curcumin affects systemic amyloidosis, we investigated amyloid deposition and molecular changes in a mouse model of amyloid apolipoprotein A-II (AApoAII) amyloidosis, in which mice were fed a curcumin-supplemented diet. Curcumin supplementation for 12 weeks significantly increased AApoAII amyloid deposition relative to controls, especially in the liver and spleen. Liver weights and plasma ApoA-II and high-density lipoprotein concentrations were significantly elevated in curcumin-supplemented groups. RNA-sequence analysis revealed that curcumin intake affected hepatic lipid metabolism via the peroxisome proliferator-activated receptor (PPAR) pathway, especially PPARα activation, resulting in increased Apoa2 mRNA expression. The increase in liver weights was due to activation of PPARα and peroxisome proliferation. Taken together, these results demonstrate that curcumin is a PPARα activator and may affect expression levels of proteins involved in amyloid deposition to influence amyloidosis and metabolism in a complex manner.
Collapse
Affiliation(s)
- Jian Dai
- Department of Neuro-health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu UniversityMatsumotoJapan
- Department of Pathology, the Xiehe Hospital of TangshanTangshanChina
| | - Ying Li
- Aging Biology, Department of Biomedical Engineering, Graduate School of Medicine, Science and Technology Shinshu UniversityMatsumotoJapan
| | - Fuyuki Kametani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Xiaoran Cui
- Department of Aging Biology, Institute of Pathogenesis and Disease Prevention, Shinshu University Graduate School of MedicineMatsumotoJapan
| | - Yuichi Igarashi
- Department of Aging Biology, Institute of Pathogenesis and Disease Prevention, Shinshu University Graduate School of MedicineMatsumotoJapan
| | - Jia Huo
- Department of Orthopedic Surgery, the Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Hiroki Miyahara
- Department of Neuro-health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu UniversityMatsumotoJapan
- Department of Aging Biology, Shinshu University School of MedicineMatsumotoJapan
| | - Masayuki Mori
- Department of Neuro-health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu UniversityMatsumotoJapan
- Department of Aging Biology, Shinshu University School of MedicineMatsumotoJapan
| | - Keiichi Higuchi
- Department of Neuro-health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu UniversityMatsumotoJapan
- Department of Aging Biology, Shinshu University School of MedicineMatsumotoJapan
| |
Collapse
|
8
|
Ajima H, Kai Y, Fujimaki J, Akashi S, Morita A, Ezaki O, Kamei Y, Miura S. Effects of fenofibrate and its combination with lovastatin on the expression of genes involved in skeletal muscle atrophy, including FoxO1 and its targets. J Toxicol Sci 2021; 46:11-24. [PMID: 33408297 DOI: 10.2131/jts.46.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Fibrates and statins have been widely used to reduce triglyceride and cholesterol levels, respectively. Besides its lipid-lowering effect, the side effect of muscle atrophy after fibrate administration to humans has been demonstrated in some studies. Combination therapy with fibrates and statins also increases the risk of rhabdomyolysis. FoxO1, a member of the FoxO forkhead type transcription factor family, is markedly upregulated in skeletal muscle in energy-deprived states and induces muscle atrophy via the expression of E3-ubiquitine ligases. In this study, we investigated the changes in FoxO1 and its targets in murine skeletal muscle with fenofibrate treatment. High doses of fenofibrate (greater than 0.5% (wt/wt)) over one week increased the expression of FoxO1 and its targets in the skeletal muscles of mice and decreased skeletal muscle weight. These fenofibrate-induced changes were diminished in the PPARα knockout mice. When the effect of combination treatment with fenofibrate and lovastatin was investigated, a significant increase in FoxO1 protein levels was observed despite the lack of deterioration of muscle atrophy. Collectively, our findings suggest that a high dose of fenofibrate over one week causes skeletal muscle atrophy via enhancement of FoxO1, and combination treatment with fenofibrate and lovastatin may further increase FoxO1 protein level.
Collapse
Affiliation(s)
- Haruka Ajima
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka
| | - Yuko Kai
- Department of Nutritional Science, National Institute of Health and Nutrition
| | - Junya Fujimaki
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka
| | - Shiori Akashi
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka
| | - Akihito Morita
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka
| | - Osamu Ezaki
- Department of Nutritional Science, National Institute of Health and Nutrition
| | - Yasutomi Kamei
- Department of Nutritional Science, National Institute of Health and Nutrition
- Laboratory of Molecular Nutrition, Graduate School of Environmental and Life Science, Kyoto Prefectural University
| | - Shinji Miura
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka
- Department of Nutritional Science, National Institute of Health and Nutrition
| |
Collapse
|
9
|
Ramchoun M, Khouya T, Harnafi H, Alem C, Benlyas M, Simmet T, Ouguerram K, Amrani S. Effect of polyphenol, flavonoid, and saponin fractions from Thymus atlanticus on acute and chronic hyperlipidemia in mice. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2020. [DOI: 10.1186/s43094-020-00097-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Abstract
Background
Thymus atlanticus is an endemic plant of the Mediterranean region, which has been used in the Moroccan mountain area to treat several diseases. This study aimed to investigate the effect of polyphenol, flavonoid, and saponin fractions derived from this plant on acute and chronic hyperlipidemia in male albino mice.
Results
The results indicated that the injection of Triton WR-1339 (20 mg/100 g body weight (B.wt.)) and 6-week administration of a high-fat diet (which is an 81.8% standard diet supplemented with 2% cholesterol, 16% lard, and 0.2% cholic acid) significantly increased plasma total cholesterol, triglycerides and low-density lipoprotein cholesterol (LDL-C), but did not affect high-density lipoprotein cholesterol (HDL-C) levels in mice. Administration of a single dose (2 mg/kg B.wt.) of polyphenol, flavonoid, or saponin fractions significantly suppressed the effect of Triton injection on plasma total cholesterol, triglycerides, and LDL-C. In addition, the supplementation of the high-fat diet with polyphenol fraction (2 mg/kg B.wt./day) prevented the increase of total cholesterol, triglycerides, and LDL-C, and effectively increased HDL-C level when compared to mice feeding only the high-fat diet.
Conclusion
In conclusion, phenolic compounds from Thymus atlanticus possess a significant hypocholesterolemic and hypotriglyceridemic effects and, therefore, could have an important role in the management of dyslipidemia.
Collapse
|
10
|
Ihm SH, Chung WB, Lee JM, Hwang BH, Yoo KD, Her SH, Song WH, Chae IH, Park TH, Kim JH, Jeon DW, Cho BR, Kang SH, Park SD, Lee JB, Woo JT, Lee BW, Han KA, Won KH, Kim HS, Yu JM, Chung CH, Kim HJ, Cho HC, Seung KB. Efficacy and Tolerability of Pitavastatin Versus Pitavastatin/Fenofibrate in High-risk Korean Patients with Mixed Dyslipidemia: A Multicenter, Randomized, Double-blinded, Parallel, Therapeutic Confirmatory Clinical Trial. Clin Ther 2020; 42:2021-2035.e3. [PMID: 32891418 DOI: 10.1016/j.clinthera.2020.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/20/2020] [Accepted: 08/05/2020] [Indexed: 01/22/2023]
Abstract
PURPOSE Dyslipidemia is an important risk factor for cardiovascular disease (CVD). Statins are known to effectively reduce not only low-density lipoprotein cholesterol (LDL-C) level but also death and nonfatal myocardial infarction due to coronary heart disease. The risk for CVD from atherogenic dyslipidemia persists when elevated triglyceride (TG) and reduced high-density lipoprotein cholesterol (HDL-C) levels are not controlled with statin therapy. Therefore, statin/fenofibrate combination therapy is more effective in reducing CVD risk. Here, we assessed the efficacy and tolerability of pitavastatin/fenofibrate combination therapy in patients with mixed dyslipidemia and a high risk for CVD. METHODS This multicenter, randomized, double-blind, parallel-group, therapeutic-confirmatory clinical trial evaluated the efficacy and tolerability of fixed-dose combination therapy with pitavastatin/fenofibrate 2/160 mg in Korean patients with a high risk for CVD and a controlled LDL-C level (<100 mg/dL) and a TG level of 150-500 mg/dL after a run-in period with pitavastatin 2 mg alone. In the 8-week main study, 347 eligible patients were randomly assigned to receive pitavastatin 2 mg with or without fenofibrate 160 mg after a run-in period. In the extension study, patients with controlled LDL-C and non-HDL-C (<130 mg/dL) levels were included after the completion of the main study. All participants in the extension study received the pitavastatin/fenofibrate combination therapy for 16 weeks for the assessment of the tolerability of long-term treatment. FINDINGS The difference in the mean percentage change in non-HDL-C from baseline to week 8 between the combination therapy and monotherapy groups was -12.45% (95% CI, -17.18 to -7.72), and the combination therapy was associated with a greater reduction in non-HDL-C. The changes in lipid profile, including apolipoproteins, fibrinogen, and high-sensitivity C-reactive protein from baseline to weeks 4 and 8 were statistically significant with combination therapy compared to monotherapy at all time points. Furthermore, the rates of achievement of non-HDL-C and apolipoprotein B targets at week 8 in the combination therapy and monotherapy groups were 88.30% versus 77.98% (P = 0.0110) and 78.94% versus 68.45% (P = 0.0021), respectively. The combination therapy was well tolerated, with a safety profile similar to that of statin monotherapy. IMPLICATIONS In these Korean patients with mixed dyslipidemia and a high risk for CVD, combination therapy with pitavastatin/fenofibrate was associated with a greater reduction in non-HDL-C compared with that with pitavastatin monotherapy, and a significantly improvement in other lipid levels. Moreover, the combination therapy was well tolerated, with a safety profile similar to that of statin monotherapy. Therefore, pitavastatin/fenofibrate combination therapy could be effective and well tolerated in patients with mixed dyslipidemia. ClinicalTrials.gov identifier: NCT03618797.
Collapse
Affiliation(s)
- Sang-Hyun Ihm
- Department of Internal Medicine, Bucheon St. Mary's Hospital, Bucheon, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Woo-Baek Chung
- Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jong-Min Lee
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, Uijeongbu, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Byung-Hee Hwang
- Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ki-Dong Yoo
- Department of Internal Medicine, St. Vincent's Hospital, Suwon, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Ho Her
- Department of Internal Medicine, St. Vincent's Hospital, Suwon, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Woo-Hyuk Song
- Department of Internal Medicine, Ansan Hospital, Korea University, Ansan, South Korea
| | - In-Ho Chae
- Department of Internal Medicine, Bundang Hospital, College of Medicine, Seoul National University, Seongnam, South Korea
| | - Tae-Ho Park
- Department of Internal Medicine, Dong-A University Hospital, Busan, South Korea
| | - Ju-Han Kim
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju, South Korea
| | - Dong Woon Jeon
- Department of Internal Medicine, National Health Insurance Service, Ilsan Hospital, Goyang, South Korea
| | - Byung-Ryul Cho
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University, Chuncheon, South Korea
| | - Seung-Ho Kang
- Department of Internal Medicine, Cheju Halla General Hospital, Jeju, South Korea
| | - Sang-Don Park
- Department of Internal Medicine, Inha University Hospital, Incheon, South Korea
| | - Jin-Bae Lee
- Department of Internal Medicine, Daegu Catholic University Medical Center, Daegu, South Korea
| | - Jeong-Taek Woo
- Department of Internal Medicine, Kyunghee University Medical Center, Seoul, South Korea
| | - Byung-Wan Lee
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul, South Korea
| | - Kyung-Ah Han
- Department of Internal Medicine, Nowon Eulji Medical Center, Eulji University, Seoul, South Korea
| | - Kyung-Heon Won
- Department of Internal Medicine, Seoul Medical Center, Seoul, South Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jae-Myung Yu
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University, Seoul, South Korea
| | - Choon Hee Chung
- Department of Internal Medicine, Wonju Severance Christian Hospital, Wonju, South Korea
| | - Hae-Jin Kim
- Department of Internal Medicine, Ajou University Hospital, Suwon, South Korea
| | - Ho-Chan Cho
- Department of Internal Medicine, Dongsan Hospital, Keimyung University, Daegu, South Korea
| | - Ki-Bae Seung
- Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
11
|
Abdelmoneim D, El-Adl M, El-Sayed G, El-Sherbini ES. Protective effect of fenofibrate against high-fat-high-fructose diet induced non-obese NAFLD in rats. Fundam Clin Pharmacol 2020; 35:379-388. [PMID: 32757283 DOI: 10.1111/fcp.12597] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/13/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
The present study evaluated the protective effects of fenofibrate on liver function, oxidant-antioxidant balance, and insulin resistance (IR) in rats fed high-fat-high-fructose diet (HFFD). Twenty-four male Sprague-Dawley rats (110-130 gm) were allocated into four equal groups (n = 6). Rats in group I were fed a normal diet for 4 weeks. Rats in group II were fed a normal diet with fenofibrate at 50 mg/kg/day orally for four weeks. Rats in group III were fed a normal diet mixed with 25% palm oil and given 60% fructose solution orally for 4 weeks. Rats in group IV were fed a normal diet mixed with 25% palm oil, 60% oral fructose solution, and fenofibrate at 50 mg/kg/day orally for four weeks. After experimental induction, serum and liver tissue samples were collected to determine lipid profiles, glycemic status, antioxidant status, oxidative and stress markers, and histopathology of liver tissues. The results of the present study revealed that fenofibrate prevents the occurrence of fatty liver, enhancing glycemic status, decreasing oxidative stress, and improving antioxidant status. It can be concluded that fenofibrate has a lipotropic and antidiabetic role.
Collapse
Affiliation(s)
- Doaa Abdelmoneim
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, P.O. 35516, Mansoura, Egypt
| | - Mohamed El-Adl
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, P.O. 35516, Mansoura, Egypt
| | - Gehad El-Sayed
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, P.O. 35516, Mansoura, Egypt
| | - El Said El-Sherbini
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, P.O. 35516, Mansoura, Egypt
| |
Collapse
|
12
|
Pouwer MG, Pieterman EJ, Chang SC, Olsen GW, Caspers MPM, Verschuren L, Jukema JW, Princen HMG. Dose Effects of Ammonium Perfluorooctanoate on Lipoprotein Metabolism in APOE*3-Leiden.CETP Mice. Toxicol Sci 2020; 168:519-534. [PMID: 30657992 PMCID: PMC6432869 DOI: 10.1093/toxsci/kfz015] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Epidemiological studies have reported positive associations between serum perfluorooctanoic acid (PFOA) and total and non-high-density lipoprotein cholesterol (non-HDL-C) although the magnitude of effect of PFOA on cholesterol lacks consistency. The objectives of this study were to evaluate the effect of PFOA on plasma cholesterol and triglyceride metabolism at various plasma PFOA concentrations relevant to humans, and to elucidate the mechanisms using APOE*3-Leiden.CETP mice, a model with a human-like lipoprotein metabolism. APOE*3-Leiden.CETP mice were fed a Western-type diet with PFOA (10, 300, 30 000 ng/g/d) for 4-6 weeks. PFOA exposure did not alter plasma lipids in the 10 and 300 ng/g/d dietary PFOA dose groups. At 30 000 ng/g/d, PFOA decreased plasma triglycerides (TG), total cholesterol (TC), and non-HDL-C, whereas HDL-C was increased. The plasma lipid alterations could be explained by decreased very low-density lipoprotein (VLDL) production and increased VLDL clearance by the liver through increased lipoprotein lipase activity. The concomitant increase in HDL-C was mediated by decreased cholesteryl ester transfer activity and changes in gene expression of proteins involved in HDL metabolism. Hepatic gene expression and pathway analysis confirmed the changes in lipoprotein metabolism that were mediated for a major part through activation of the peroxisome proliferator-activated receptor (PPAR)α. Our data confirmed the findings from a phase 1 clinical trial in humans that demonstrated high serum or plasma PFOA levels resulted in lower cholesterol levels. The study findings do not show an increase in cholesterol at environmental or occupational levels of PFOA exposure, thereby indicating these findings are associative rather than causal.
Collapse
Affiliation(s)
- Marianne G Pouwer
- The Netherlands Organization of Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, 2333 CK, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Elsbet J Pieterman
- The Netherlands Organization of Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, 2333 CK, Leiden, The Netherlands
| | | | - Geary W Olsen
- Medical Department, 3M Company, Saint Paul, Minnesota 55144
| | - Martien P M Caspers
- The Netherlands Organization of Applied Scientific Research (TNO) - Microbiology and Systems Biology, 3704 HE Zeist, The Netherlands
| | - Lars Verschuren
- The Netherlands Organization of Applied Scientific Research (TNO) - Microbiology and Systems Biology, 3704 HE Zeist, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Hans M G Princen
- The Netherlands Organization of Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, 2333 CK, Leiden, The Netherlands
| |
Collapse
|
13
|
Najafipour M, Zareizadeh M, Khokhi MA, Najafipour F. Comparative study of the effect of atorvastatin and fenofibrate on high-density lipoprotein cholesterol levels in patients with type 2 diabetes. J Adv Pharm Technol Res 2019; 9:135-138. [PMID: 30637231 PMCID: PMC6302688 DOI: 10.4103/japtr.japtr_314_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Diabetes is the most common metabolic disease. Type 2 diabetes is a variable combination of insulin resistance and disorder in insulin secretion, leading to disorder of lipids and plasma lipoproteins. The most common pattern of dyslipidemia in diabetic is high triglyceride (TG) and low high-density lipoprotein cholesterol (HDL-C). This study was conducted to find a more effective drug to increase HDL-C. In this study, 80 patients (26 males and 54 females) with type 2 diabetes received fenofibrate in cross-sectional way for 2 months, and they did not take antilipid drugs for 2 month. Then, they underwent atorvastatin for 2 months and HDL-C was measured before and after taking drugs. Patients did not change their diet during this study. Effect of atorvastatin and fenofibrate on HDL-C levels in patients with type 2 diabetes was evaluated. The mean HDL-C and total cholesterol (TC) before and after taking drugs were 36.5 mg/dL and 174.56 mg/dL, respectively. After atorvastatin, the mean HDL-C and TC were 43.30 and 150.144 mg/dL, respectively, and after fenofibrate, 43.40 were mg/dL and 146.36 mg/dL, respectively. Atorvastatin caused increase in HDL-C by 18.44% and reduction in TC by 13.82% and fenofibrate increase in HDL-C by18.62% and reduction in TC by 16.05%. No difference was seen between atorvastatin and fenofibrate in terms of effect on the HDL-C excess (P = 0.449). In addition, no difference was seen between atorvastatin and fenofibrate in terms of effect on TC reduction (P = 0.992). In conclusion various factors are involved in increasing the HDL, such as race, sex, nutrition, physical activity and, of course, medications. The effect of medications is also different on races and genetics. The value of increase in HDL-C after Fenofibrate and Atorvastatin was associated with gender so that it caused more increase of HDL-C in females.
Collapse
Affiliation(s)
- Mostafa Najafipour
- Young Researchers and Elite Club, Ardabil Branch, Islamic Azad University, Ardabil, Iran.,Department of Endocrinology, Faculty of Medicine, Azad Ardabil University of Medical Sciences, Ardabil, Iran
| | - Masoumeh Zareizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farzad Najafipour
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Cholesteryl ester transfer protein: An enigmatic pharmacology – Antagonists and agonists. Atherosclerosis 2018; 278:286-298. [DOI: 10.1016/j.atherosclerosis.2018.09.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/04/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022]
|
15
|
Monteillet L, Gjorgjieva M, Silva M, Verzieux V, Imikirene L, Duchampt A, Guillou H, Mithieux G, Rajas F. Intracellular lipids are an independent cause of liver injury and chronic kidney disease in non alcoholic fatty liver disease-like context. Mol Metab 2018; 16:100-115. [PMID: 30100243 PMCID: PMC6157648 DOI: 10.1016/j.molmet.2018.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022] Open
Abstract
Objective Ectopic lipid accumulation in the liver and kidneys is a hallmark of metabolic diseases leading to non-alcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD). Moreover, recent data have highlighted a strong correlation between NAFLD and CKD incidences. In this study, we use two mouse models of hepatic steatosis or CKD, each initiated independently of the other upon the suppression of glucose production specifically in the liver or kidneys, to elucidate the mechanisms underlying the development of CKD in the context of NAFLD-like pathology. Methods Mice with a deletion of G6pc, encoding glucose-6 phosphatase catalytic subunit, specifically in the liver (L.G6pc−/− mice) or the kidneys (K.G6pc−/− mice), were fed with either a standard diet or a high fat/high sucrose (HF/HS) diet during 9 months. These mice represent two original models of a rare metabolic disease named Glycogen Storage Disease Type Ia (GSDIa) that is characterized by both NAFLD-like pathology and CKD. Two other groups of L.G6pc−/− and K.G6pc−/− mice were fed a standard diet for 6 months and then treated with fenofibrate for 3 months. Lipid and glucose metabolisms were characterized, and NAFLD-like and CKD damages were evaluated. Results Lipid depot exacerbation upon high-calorie diet strongly accelerated hepatic and renal pathologies induced by the G6pc-deficiency. In L.G6pc−/− mice, HF/HS diet increased liver injuries, characterized by higher levels of plasmatic transaminases and increased hepatic tumor incidence. In K.G6pc−/− mice, HF/HS diet increased urinary albumin and lipocalin 2 excretion and aggravated renal fibrosis. In both cases, the worsening of NAFLD-like injuries and CKD was independent of glycogen content. Furthermore, fenofibrate, via the activation of lipid oxidation significantly decreased the hepatic or renal lipid accumulations and prevented liver or kidney damages in L.G6pc−/− and K.G6pc−/− mice, respectively. Finally, we show that L.G6pc−/− mice and K.G6pc−/− mice developed NAFLD-like pathology and CKD independently. Conclusions This study highlights the crucial role that lipids play in the independent development of both NAFLD and CKD and demonstrates the importance of lipid-lowering treatments in various metabolic diseases featured by lipid load, from the “rare” GSDIa to the “epidemic” morbid obesity or type 2 diabetes. Exacerbating lipid accumulation aggravates liver/kidney injury in GSDI. Fenofibrate-mediated PPARα activation induces hepatic and renal lipid turnover. Increased lipid turnover prevents glycogen synthesis and accumulation. PPARα–mediated metabolic reprograming prevents hepatic and renal GSDI complications. NAFLD and CKD develop independently.
Collapse
Affiliation(s)
- Laure Monteillet
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Marine Silva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Vincent Verzieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Linda Imikirene
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Adeline Duchampt
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Hervé Guillou
- Toxalim, Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, 31027, France.
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| |
Collapse
|
16
|
Pouwer MG, Pieterman EJ, Verschuren L, Caspers MPM, Kluft C, Garcia RA, Aman J, Jukema JW, Princen HMG. The BCR-ABL1 Inhibitors Imatinib and Ponatinib Decrease Plasma Cholesterol and Atherosclerosis, and Nilotinib and Ponatinib Activate Coagulation in a Translational Mouse Model. Front Cardiovasc Med 2018; 5:55. [PMID: 29946549 PMCID: PMC6005845 DOI: 10.3389/fcvm.2018.00055] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/10/2018] [Indexed: 01/16/2023] Open
Abstract
Treatment with the second and third generation BCR-ABL1 tyrosine kinase inhibitors (TKIs) increases cardiovascular risk in chronic myeloid leukemia (CML) patients. We investigated the vascular adverse effects of three generations of TKIs in a translational model for atherosclerosis, the APOE*3Leiden.CETP mouse. Mice were treated for sixteen weeks with imatinib (150 mg/kg BID), nilotinib (10 and 30 mg/kg QD) or ponatinib (3 and 10 mg/kg QD), giving similar drug exposures as in CML-patients. Cardiovascular risk factors were analyzed longitudinally, and histopathological analysis of atherosclerosis and transcriptome analysis of the liver was performed. Imatinib and ponatinib decreased plasma cholesterol (imatinib, −69%, p < 0.001; ponatinib 3 mg/kg, −37%, p < 0.001; ponatinib 10 mg/kg−44%, p < 0.001) and atherosclerotic lesion area (imatinib, −78%, p < 0.001; ponatinib 3 mg/kg, −52%, p = 0.002; ponatinib 10 mg/kg, −48%, p = 0.006), which were not affected by nilotinib. In addition, imatinib increased plaque stability. Gene expression and pathway analysis demonstrated that ponatinib enhanced the mRNA expression of coagulation factors of both the contact activation (intrinsic) and tissue factor (extrinsic) pathways. In line with this, ponatinib enhanced plasma levels of FVII, whereas nilotinib increased plasma FVIIa activity. While imatinib showed a beneficial cardiovascular risk profile, nilotinib and ponatinib increased the cardiovascular risk through induction of a pro-thrombotic state.
Collapse
Affiliation(s)
- Marianne G Pouwer
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elsbet J Pieterman
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, Netherlands
| | - Lars Verschuren
- Microbiology and Systems Biology, The Netherlands Organization of Applied Scientific Research (TNO), Zeist, Netherlands
| | - Martien P M Caspers
- Microbiology and Systems Biology, The Netherlands Organization of Applied Scientific Research (TNO), Zeist, Netherlands
| | | | - Ricardo A Garcia
- Cardiovascular Drug Discovery, Bristol-Meyers Squibb, New York, United States
| | - Jurjan Aman
- Departments of Physiology and Pulmonary Diseases, VU University Medical Center, Amsterdam, Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Hans M G Princen
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, Netherlands
| |
Collapse
|
17
|
Results, meta-analysis and a first evaluation of U NOxR, the urinary nitrate-to-nitrite molar ratio, as a measure of nitrite reabsorption in experimental and clinical settings. Amino Acids 2018; 50:799-821. [PMID: 29728915 DOI: 10.1007/s00726-018-2573-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 04/19/2018] [Indexed: 02/07/2023]
Abstract
We recently found that renal carbonic anhydrase (CA) is involved in the reabsorption of inorganic nitrite (NO2-), an abundant reservoir of nitric oxide (NO) in tissues and cells. Impaired NO synthesis in the endothelium and decreased NO bioavailability in the circulation are considered major contributors to the development and progression of renal and cardiovascular diseases in different conditions including diabetes. Isolated human and bovine erythrocytic CAII and CAIV can convert nitrite to nitrous acid (HONO) and its anhydride N2O3 which, in the presence of thiols (RSH), are further converted to S-nitrosothiols (RSNO) and NO. Thus, CA may be responsible both for the homeostasis of nitrite and for its bioactivation to RSNO/NO. We hypothesized that enhanced excretion of nitrite in the urine may contribute to NO-related dysfunctions in the renal and cardiovascular systems, and proposed the urinary nitrate-to-nitrite molar ratio, i.e., UNOxR, as a measure of renal CA-dependent excretion of nitrite. Based on results from clinical and experimental animal studies, here, we report on a first evaluation of UNOxR. We determined UNOxR values in preterm neonates, healthy children, and adults, in children suffering from type 1 diabetes mellitus (T1DM) or Duchenne muscular dystrophy (DMD), in elderly subjects suffering from chronic rheumatic diseases, type 2 diabetes mellitus (T2DM), coronary artery disease (CAD), or peripheral arterial occlusive disease (PAOD). We also determined UNOxR values in healthy young men who ingested isosorbide dinitrate (ISDN), pentaerythrityl tetranitrate (PETN), or inorganic nitrate. In addition, we tested the utility of UNOxR in two animal models, i.e., the LEW.1AR1-iddm rat, an animal model of human T1DM, and the APOE*3-Leiden.CETP mice, a model of human dyslipidemia. Mean UNOxR values were lower in adult patients with rheumatic diseases (187) and in T2DM patients of the DALI study (74) as compared to healthy elderly adults (660) and healthy young men (1500). The intra- and inter-variabilities of UNOxR were of the order of 50% in young and elderly healthy subjects. UNOxR values were lower in black compared to white boys (314 vs. 483, P = 0.007), which is in line with reported lower NO bioavailability in black ethnicity. Mean UNOxR values were lower in DMD (424) compared to healthy (730) children, but they were higher in T1DM children (1192). ISDN (3 × 30 mg) decreased stronger UNOxR compared to PETN (3 × 80 mg) after 1 day (P = 0.046) and after 5 days (P = 0.0016) of oral administration of therapeutically equivalent doses. In healthy young men who ingested NaNO3 (0.1 mmol/kg/d), UNOxR was higher than in those who ingested the same dose of NaCl (1709 vs. 369). In LEW.1AR1-iddm rats, mean UNOxR values were lower than in healthy rats (198 vs. 308) and comparable to those in APOE*3-Leiden.CETP mice (151).
Collapse
|
18
|
Ramakrishnan SK, Russo L, Ghanem SS, Patel PR, Oyarce AM, Heinrich G, Najjar SM. Fenofibrate Decreases Insulin Clearance and Insulin Secretion to Maintain Insulin Sensitivity. J Biol Chem 2016; 291:23915-23924. [PMID: 27662905 DOI: 10.1074/jbc.m116.745778] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/22/2016] [Indexed: 01/18/2023] Open
Abstract
High fat diet reduces the expression of CEACAM1 (carcinoembryonic antigen-related cell adhesion molecule 1), a transmembrane glycoprotein that promotes insulin clearance and down-regulates fatty acid synthase activity in the liver upon its phosphorylation by the insulin receptor. Because peroxisome proliferator-activated receptor α (PPARα) transcriptionally suppresses CEACAM1 expression, we herein examined whether high fat down-regulates CEACAM1 expression in a PPARα-dependent mechanism. By activating PPARα, the lipid-lowering drug fenofibrate reverses dyslipidemia and improves insulin sensitivity in type 2 diabetes in part by promoting fatty acid oxidation. Despite reducing glucose-stimulated insulin secretion, fenofibrate treatment does not result in insulin insufficiency. To examine whether this is mediated by a parallel decrease in CEACAM1-dependent hepatic insulin clearance pathways, we fed wild-type and Pparα-/- null mice a high fat diet supplemented with either fenofibrate or Wy14643, a selective PPARα agonist, and examined their effect on insulin metabolism and action. We demonstrated that the decrease in insulin secretion by fenofibrate and Wy14643 is accompanied by reduction in insulin clearance in wild-type but not Pparα-/- mice, thereby maintaining normoinsulinemia and insulin sensitivity despite continuous high fat intake. Intact insulin secretion in L-CC1 mice with protected hepatic insulin clearance and CEACAM1 levels provides in vivo evidence that insulin secretion responds to changes in insulin clearance to maintain physiologic insulin and glucose homeostasis. These results also emphasize the relevant role of hepatic insulin extraction in regulating insulin sensitivity.
Collapse
Affiliation(s)
- Sadeesh K Ramakrishnan
- From the Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio 43614
| | - Lucia Russo
- From the Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio 43614
| | - Simona S Ghanem
- From the Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio 43614
| | - Payal R Patel
- From the Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio 43614
| | - Ana Maria Oyarce
- From the Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio 43614.,the Department of Pharmacology and Experimental Therapeutics College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, Ohio 43614, and
| | - Garrett Heinrich
- the Department of Pharmacology and Experimental Therapeutics College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, Ohio 43614, and
| | - Sonia M Najjar
- From the Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio 43614, .,the Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| |
Collapse
|
19
|
Sohn M, Kim K, Uddin MJ, Lee G, Hwang I, Kang H, Kim H, Lee JH, Ha H. Delayed treatment with fenofibrate protects against high-fat diet-induced kidney injury in mice: the possible role of AMPK autophagy. Am J Physiol Renal Physiol 2016; 312:F323-F334. [PMID: 27465995 DOI: 10.1152/ajprenal.00596.2015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/22/2016] [Indexed: 11/22/2022] Open
Abstract
Fenofibrate activates not only peroxisome proliferator-activated receptor-α (PPARα) but also adenosine monophosphate-activated protein kinase (AMPK). AMPK-mediated cellular responses protect kidney from high-fat diet (HFD)-induced injury, and autophagy resulting from AMPK activation has been regarded as a stress-response mechanism. Thus the present study examined the role of AMPK and autophagy in the renotherapeutic effects of fenofibrate. C57BL/6J mice were divided into three groups: normal diet (ND), HFD, and HFD + fenofibrate (HFD + FF). Fenofibrate was administered 4 wk after the initiation of the HFD when renal injury was initiated. Mouse proximal tubule cells (mProx24) were used to clarify the role of AMPK. Feeding mice with HFD for 12 wk induced insulin resistance and kidney injury such as albuminuria, glomerulosclerosis, tubular injury, and inflammation, which were effectively inhibited by fenofibrate. In addition, fenofibrate treatment resulted in the activation of renal AMPK, upregulation of fatty acid oxidation (FAO) enzymes and antioxidants, and induction of autophagy in the HFD mice. In mProx24 cells, fenofibrate activated AMPK in a concentration-dependent manner, upregulated FAO enzymes and antioxidants, and induced autophagy, all of which were inhibited by treatment of compound C, an AMPK inhibitor. Fenofibrate-induced autophagy was also significantly blocked by AMPKα1 siRNA but not by PPARα siRNA. Collectively, these results demonstrate that delayed treatment with fenofibrate has a therapeutic effect on HFD-induced kidney injury, at least in part, through the activation of AMPK and induction of subsequent downstream effectors: autophagy, FAO enzymes, and antioxidants.
Collapse
Affiliation(s)
- Minji Sohn
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Keumji Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Md Jamal Uddin
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Gayoung Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Inah Hwang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hyeji Kang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hyunji Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Jung Hwa Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| |
Collapse
|
20
|
Serum CETP concentration is not associated with measures of body fat: The NEO study. Atherosclerosis 2016; 246:267-73. [DOI: 10.1016/j.atherosclerosis.2016.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/18/2016] [Accepted: 01/18/2016] [Indexed: 01/22/2023]
|
21
|
Princen HMG, Pouwer MG, Pieterman EJ. Comment on "Hypercholesterolemia with consumption of PFOA-laced Western diets is dependent on strain and sex of mice" by Rebholz S.L. et al. Toxicol. Rep. 2016 (3) 46-54. Toxicol Rep 2016; 3:306-309. [PMID: 28959551 PMCID: PMC5615825 DOI: 10.1016/j.toxrep.2016.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/07/2016] [Indexed: 01/01/2023] Open
Affiliation(s)
- Hans M G Princen
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Marianne G Pouwer
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Elsbet J Pieterman
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| |
Collapse
|
22
|
Wang Y, van der Tuin S, Tjeerdema N, van Dam AD, Rensen SS, Hendrikx T, Berbée JFP, Atanasovska B, Fu J, Hoekstra M, Bekkering S, Riksen NP, Buurman WA, Greve JW, Hofker MH, Shiri-Sverdlov R, Meijer OC, Smit JWA, Havekes LM, van Dijk KW, Rensen PCN. Plasma cholesteryl ester transfer protein is predominantly derived from Kupffer cells. Hepatology 2015; 62:1710-22. [PMID: 26174697 DOI: 10.1002/hep.27985] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 07/10/2015] [Indexed: 02/06/2023]
Abstract
UNLABELLED The role of Kupffer cells (KCs) in the pathophysiology of the liver has been firmly established. Nevertheless, KCs have been underexplored as a target for diagnosis and treatment of liver diseases owing to the lack of noninvasive diagnostic tests. We addressed the hypothesis that cholesteryl ester transfer protein (CETP) is mainly derived from KCs and may predict KC content. Microarray analysis of liver and adipose tissue biopsies, obtained from 93 obese subjects who underwent elective bariatric surgery, showed that expression of CETP is markedly higher in liver than adipose tissue. Hepatic expression of CETP correlated strongly with that of KC markers, and CETP messenger RNA and protein colocalized specifically with KCs in human liver sections. Hepatic KC content as well as hepatic CETP expression correlated strongly with plasma CETP concentration. Mechanistic and intervention studies on the role of KCs in determining the plasma CETP concentration were performed in a transgenic (Tg) mouse model expressing human CETP. Selective elimination of KCs from the liver in CETP Tg mice virtually abolished hepatic CETP expression and largely reduced plasma CETP concentration, consequently improving the lipoprotein profile. Conversely, augmentation of KCs after Bacille-Calemette-Guérin vaccination largely increased hepatic CETP expression and plasma CETP. Also, lipid-lowering drugs fenofibrate and niacin reduced liver KC content, accompanied by reduced plasma CETP concentration. CONCLUSIONS Plasma CETP is predominantly derived from KCs, and plasma CETP level predicts hepatic KC content in humans.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sam van der Tuin
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nathanja Tjeerdema
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrea D van Dam
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sander S Rensen
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Tim Hendrikx
- Department of Molecular Genetics, Maastricht University, Maastricht, The Netherlands
| | - Jimmy F P Berbée
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Biljana Atanasovska
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Menno Hoekstra
- Department of Biopharmaceutics, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The, Netherlands
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The, Netherlands
| | - Wim A Buurman
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jan Willem Greve
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marten H Hofker
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, Maastricht University, Maastricht, The Netherlands
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes W A Smit
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The, Netherlands
| | - Louis M Havekes
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ko Willems van Dijk
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
23
|
Liang W, Verschuren L, Mulder P, van der Hoorn JWA, Verheij J, van Dam AD, Boon MR, Princen HMG, Havekes LM, Kleemann R, van den Hoek AM. Salsalate attenuates diet induced non-alcoholic steatohepatitis in mice by decreasing lipogenic and inflammatory processes. Br J Pharmacol 2015; 172:5293-305. [PMID: 26292849 DOI: 10.1111/bph.13315] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 08/10/2015] [Accepted: 08/13/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Salsalate (salicylsalicylic acid) is an anti-inflammatory drug that was recently found to exert beneficial metabolic effects on glucose and lipid metabolism. Although its utility in the prevention and management of a wide range of vascular disorders, including type 2 diabetes and metabolic syndrome has been suggested before, the potential of salsalate to protect against non-alcoholic steatohepatitis (NASH) remains unclear. The aim of the present study was therefore to ascertain the effects of salsalate on the development of NASH. EXPERIMENTAL APPROACH Transgenic APOE*3Leiden.CETP mice were fed a high-fat and high-cholesterol diet with or without salsalate for 12 and 20 weeks. The effects on body weight, plasma biochemical variables, liver histology and hepatic gene expression were assessed. KEY RESULTS Salsalate prevented weight gain, improved dyslipidemia and insulin resistance and ameliorated diet-induced NASH, as shown by decreased hepatic microvesicular and macrovesicular steatosis, reduced hepatic inflammation and reduced development of fibrosis. Salsalate affected lipid metabolism by increasing β-oxidation and decreasing lipogenesis, as shown by the activation of PPAR-α, PPAR-γ co-activator 1β, RXR-α and inhibition of genes controlled by the transcription factor MLXIPL/ChREBP. Inflammation was reduced by down-regulation of the NF-κB pathway, and fibrosis development was prevented by down-regulation of TGF-β signalling. CONCLUSIONS AND IMPLICATIONS Salsalate exerted a preventive effect on the development of NASH and progression to fibrosis. These data suggest a clinical application of salsalate in preventing NASH.
Collapse
Affiliation(s)
- Wen Liang
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands.,Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, TNO, Zeist, The Netherlands
| | - Petra Mulder
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - José W A van der Hoorn
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Andrea D van Dam
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariette R Boon
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans M G Princen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Louis M Havekes
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands.,Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Anita M van den Hoek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
24
|
Das M, Irvin MR, Sha J, Aslibekyan S, Hidalgo B, Perry RT, Zhi D, Tiwari HK, Absher D, Ordovas JM, Arnett DK. Lipid changes due to fenofibrate treatment are not associated with changes in DNA methylation patterns in the GOLDN study. Front Genet 2015; 6:304. [PMID: 26483836 PMCID: PMC4586504 DOI: 10.3389/fgene.2015.00304] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/12/2015] [Indexed: 11/15/2022] Open
Abstract
Fenofibrate lowers triglycerides (TG) and raises high density lipoprotein cholesterol (HDLc) in dyslipidemic individuals. Several studies have shown genetic variability in lipid responses to fenofibrate treatment. It is, however, not known whether epigenetic patterns are also correlated with the changes in lipids due to fenofibrate treatment. The present study was therefore undertaken to examine the changes in DNA methylation among the participants of Genetics of Lipid Lowering Drugs and Diet Network (GOLDN) study. A total of 443 individuals were studied for epigenome-wide changes in DNA methylation, assessed using the Illumina Infinium HumanMethylation450 array, before and after a 3-week daily treatment with 160 mg of fenofibrate. The association between the change in DNA methylation and changes in TG, HDLc, and low-density lipoprotein cholesterol (LDLc) were assessed using linear mixed models adjusted for age, sex, baseline lipids, and study center as fixed effects and family as a random effect. Changes in DNA methylation were not significantly associated with changes in TG, HDLc, or LDLc after 3 weeks of fenofibrate for any CpG. CpG changes in genes known to be involved in fenofibrate response, e.g., PPAR-α, APOA1, LPL, APOA5, APOC3, CETP, and APOB, also did not show evidence of association. In conclusion, changes in lipids in response to 3-week treatment with fenofibrate were not associated with changes in DNA methylation. Studies of longer duration may be required to detect treatment-induced changes in methylation.
Collapse
Affiliation(s)
- Mithun Das
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - M Ryan Irvin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Jin Sha
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Stella Aslibekyan
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Rodney T Perry
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Degui Zhi
- Department of Biostatistics, Section on Statistical Genetics, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Hemant K Tiwari
- Department of Biostatistics, Section on Statistical Genetics, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Devin Absher
- Absher Laboratory, HudsonAlpha Institute of Biotechnology Huntsville, AL, USA
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University Boston, MA, USA
| | - Donna K Arnett
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| |
Collapse
|
25
|
van der Tuin SJL, Kühnast S, Berbée JFP, Verschuren L, Pieterman EJ, Havekes LM, van der Hoorn JWA, Rensen PCN, Jukema JW, Princen HMG, Willems van Dijk K, Wang Y. Anacetrapib reduces (V)LDL cholesterol by inhibition of CETP activity and reduction of plasma PCSK9. J Lipid Res 2015; 56:2085-93. [PMID: 26342106 DOI: 10.1194/jlr.m057794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Indexed: 01/14/2023] Open
Abstract
Recently, we showed in APOE*3-Leiden cholesteryl ester transfer protein (E3L.CETP) mice that anacetrapib attenuated atherosclerosis development by reducing (V)LDL cholesterol [(V)LDL-C] rather than by raising HDL cholesterol. Here, we investigated the mechanism by which anacetrapib reduces (V)LDL-C and whether this effect was dependent on the inhibition of CETP. E3L.CETP mice were fed a Western-type diet alone or supplemented with anacetrapib (30 mg/kg body weight per day). Microarray analyses of livers revealed downregulation of the cholesterol biosynthesis pathway (P < 0.001) and predicted downregulation of pathways controlled by sterol regulatory element-binding proteins 1 and 2 (z-scores -2.56 and -2.90, respectively; both P < 0.001). These data suggest increased supply of cholesterol to the liver. We found that hepatic proprotein convertase subtilisin/kexin type 9 (Pcsk9) expression was decreased (-28%, P < 0.01), accompanied by decreased plasma PCSK9 levels (-47%, P < 0.001) and increased hepatic LDL receptor (LDLr) content (+64%, P < 0.01). Consistent with this, anacetrapib increased the clearance and hepatic uptake (+25%, P < 0.001) of [(14)C]cholesteryl oleate-labeled VLDL-mimicking particles. In E3L mice that do not express CETP, anacetrapib still decreased (V)LDL-C and plasma PCSK9 levels, indicating that these effects were independent of CETP inhibition. We conclude that anacetrapib reduces (V)LDL-C by two mechanisms: 1) inhibition of CETP activity, resulting in remodeled VLDL particles that are more susceptible to hepatic uptake; and 2) a CETP-independent reduction of plasma PCSK9 levels that has the potential to increase LDLr-mediated hepatic remnant clearance.
Collapse
Affiliation(s)
- Sam J L van der Tuin
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Susan Kühnast
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands The Netherlands Organization for Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Jimmy F P Berbée
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lars Verschuren
- TNO, Microbiology and Systems Biology, Zeist, The Netherlands
| | - Elsbet J Pieterman
- The Netherlands Organization for Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Louis M Havekes
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands The Netherlands Organization for Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - José W A van der Hoorn
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands The Netherlands Organization for Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans M G Princen
- The Netherlands Organization for Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Ko Willems van Dijk
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Yanan Wang
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
26
|
Kühnast S, Fiocco M, van der Hoorn JWA, Princen HMG, Jukema JW. Innovative pharmaceutical interventions in cardiovascular disease: Focusing on the contribution of non-HDL-C/LDL-C-lowering versus HDL-C-raising: A systematic review and meta-analysis of relevant preclinical studies and clinical trials. Eur J Pharmacol 2015; 763:48-63. [PMID: 25989133 DOI: 10.1016/j.ejphar.2015.03.089] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/27/2015] [Accepted: 03/05/2015] [Indexed: 12/25/2022]
Abstract
Non-HDL-cholesterol is well recognised as a primary causal risk factor in cardiovascular disease. However, despite consistent epidemiological evidence for an inverse association between HDL-C and coronary heart disease, clinical trials aimed at raising HDL-C (AIM-HIGH, HPS2-THRIVE, dal-OUTCOMES) failed to meet their primary goals. This systematic review and meta-analysis investigated the effects of established and novel treatment strategies, specifically targeting HDL, on inhibition of atherosclerosis in cholesteryl ester transfer protein-expressing animals, and the prevention of clinical events in randomised controlled trials. Linear regression analyses using data from preclinical studies revealed associations for TC and non-HDL-C and lesion area (R(2)=0.258, P=0.045; R(2)=0.760, P<0.001), but not for HDL-C (R(2)=0.030, P=0.556). In clinical trials, non-fatal myocardial infarction risk was significantly less in the treatment group with pooled odd ratios of 0.87 [0.81; 0.94] for all trials and 0.85 [0.78; 0.93] after excluding some trials due to off-target adverse events, whereas all-cause mortality was not affected (OR 1.05 [0.99-1.10]). Meta-regression analyses revealed a trend towards an association between between-group differences in absolute change from baseline in LDL-C and non-fatal myocardial infarction (P=0.066), whereas no correlation was found for HDL-C (P=0.955). We conclude that the protective role of lowering LDL-C and non-HDL-C is well-established. The contribution of raising HDL-C on inhibition of atherosclerosis and the prevention of cardiovascular disease remains undefined and may be dependent on the mode of action of HDL-C-modification. Nonetheless, treatment strategies aimed at improving HDL function and raising apolipoprotein A-I may be worth exploring.
Collapse
Affiliation(s)
- Susan Kühnast
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands; Department of Cardiology, LUMC, Leiden, The Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Marta Fiocco
- Department of Medical Statistics and Bioinformatics, LUMC, Leiden, The Netherlands; Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - José W A van der Hoorn
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands; Department of Cardiology, LUMC, Leiden, The Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Hans M G Princen
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands.
| | | |
Collapse
|
27
|
van Dam AD, Nahon KJ, Kooijman S, van den Berg SM, Kanhai AA, Kikuchi T, Heemskerk MM, van Harmelen V, Lombès M, van den Hoek AM, de Winther MPJ, Lutgens E, Guigas B, Rensen PCN, Boon MR. Salsalate activates brown adipose tissue in mice. Diabetes 2015; 64:1544-54. [PMID: 25475439 DOI: 10.2337/db14-1125] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/24/2014] [Indexed: 11/13/2022]
Abstract
Salsalate improves glucose intolerance and dyslipidemia in type 2 diabetes patients, but the mechanism is still unknown. The aim of the current study was to unravel the molecular mechanisms involved in these beneficial metabolic effects of salsalate by treating mice with salsalate during and after development of high-fat diet-induced obesity. We found that salsalate attenuated and reversed high-fat diet-induced weight gain, in particular fat mass accumulation, improved glucose tolerance, and lowered plasma triglyceride levels. Mechanistically, salsalate selectively promoted the uptake of fatty acids from glycerol tri[(3)H]oleate-labeled lipoprotein-like emulsion particles by brown adipose tissue (BAT), decreased the intracellular lipid content in BAT, and increased rectal temperature, all pointing to more active BAT. The treatment of differentiated T37i brown adipocytes with salsalate increased uncoupled respiration. Moreover, salsalate upregulated Ucp1 expression and enhanced glycerol release, a dual effect that was abolished by the inhibition of cAMP-dependent protein kinase (PKA). In conclusion, salsalate activates BAT, presumably by directly activating brown adipocytes via the PKA pathway, suggesting a novel mechanism that may explain its beneficial metabolic effects in type 2 diabetes patients.
Collapse
Affiliation(s)
- Andrea D van Dam
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - Kimberly J Nahon
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - Sander Kooijman
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - Susan M van den Berg
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, the Netherlands
| | - Anish A Kanhai
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - Takuya Kikuchi
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - Mattijs M Heemskerk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Vanessa van Harmelen
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Marc Lombès
- Institut National de la Santé et de la Recherche Médicale, Unité 693, Le Kremlin-Bicêtre, France
| | - Anita M van den Hoek
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research, Leiden, the Netherlands
| | - Menno P J de Winther
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, the Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, the Netherlands Institute for Cardiovascular Prevention, Ludwig Maximilian's University Munich, Munich, Germany
| | - Bruno Guigas
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - Mariëtte R Boon
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| |
Collapse
|
28
|
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
|
29
|
Abstract
High-density lipoprotein (HDL) is considered to be an anti-atherogenic lipoprotein moiety. Generation of genetically modified (total body and tissue-specific knockout) mouse models has significantly contributed to our understanding of HDL function. Here we will review data from knockout mouse studies on the importance of HDL's major alipoprotein apoA-I, the ABC transporters A1 and G1, lecithin:cholesterol acyltransferase, phospholipid transfer protein, and scavenger receptor BI for HDL's metabolism and its protection against atherosclerosis in mice. The initial generation and maturation of HDL particles as well as the selective delivery of its cholesterol to the liver are essential parameters in the life cycle of HDL. Detrimental atherosclerosis effects observed in response to HDL deficiency in mice cannot be solely attributed to the low HDL levels per se, as the low HDL levels are in most models paralleled by changes in non-HDL-cholesterol levels. However, the cholesterol efflux function of HDL is of critical importance to overcome foam cell formation and the development of atherosclerotic lesions in mice. Although HDL is predominantly studied for its atheroprotective action, the mouse data also suggest an essential role for HDL as cholesterol donor for steroidogenic tissues, including the adrenals and ovaries. Furthermore, it appears that a relevant interaction exists between HDL-mediated cellular cholesterol efflux and the susceptibility to inflammation, which (1) provides strong support for the novel concept that inflammation and metabolism are intertwining biological processes and (2) identifies the efflux function of HDL as putative therapeutic target also in other inflammatory diseases than atherosclerosis.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of Biopharmaceutics, Gorlaeus Laboratories, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands,
| | | |
Collapse
|
30
|
Ason B, van der Hoorn JWA, Chan J, Lee E, Pieterman EJ, Nguyen KK, Di M, Shetterly S, Tang J, Yeh WC, Schwarz M, Jukema JW, Scott R, Wasserman SM, Princen HMG, Jackson S. PCSK9 inhibition fails to alter hepatic LDLR, circulating cholesterol, and atherosclerosis in the absence of ApoE. J Lipid Res 2014; 55:2370-9. [PMID: 25258384 DOI: 10.1194/jlr.m053207] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
LDL cholesterol (LDL-C) contributes to coronary heart disease. Proprotein convertase subtilisin/kexin type 9 (PCSK9) increases LDL-C by inhibiting LDL-C clearance. The therapeutic potential for PCSK9 inhibitors is highlighted by the fact that PCSK9 loss-of-function carriers exhibit 15-30% lower circulating LDL-C and a disproportionately lower risk (47-88%) of experiencing a cardiovascular event. Here, we utilized pcsk9(-/-) mice and an anti-PCSK9 antibody to study the role of the LDL receptor (LDLR) and ApoE in PCSK9-mediated regulation of plasma cholesterol and atherosclerotic lesion development. We found that circulating cholesterol and atherosclerotic lesions were minimally modified in pcsk9(-/-) mice on either an LDLR- or ApoE-deficient background. Acute administration of an anti-PCSK9 antibody did not reduce circulating cholesterol in an ApoE-deficient background, but did reduce circulating cholesterol (-45%) and TGs (-36%) in APOE*3Leiden.cholesteryl ester transfer protein (CETP) mice, which contain mouse ApoE, human mutant APOE3*Leiden, and a functional LDLR. Chronic anti-PCSK9 antibody treatment in APOE*3Leiden.CETP mice resulted in a significant reduction in atherosclerotic lesion area (-91%) and reduced lesion complexity. Taken together, these results indicate that both LDLR and ApoE are required for PCSK9 inhibitor-mediated reductions in atherosclerosis, as both are needed to increase hepatic LDLR expression.
Collapse
Affiliation(s)
- Brandon Ason
- Metabolic Disorders Amgen, Inc., South San Francisco, CA
| | | | - Joyce Chan
- Metabolic Disorders Amgen, Inc., South San Francisco, CA
| | - Edward Lee
- Metabolic Disorders Amgen, Inc., South San Francisco, CA
| | - Elsbet J Pieterman
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | | | - Mei Di
- Metabolic Disorders Amgen, Inc., South San Francisco, CA
| | | | - Jie Tang
- Protein Technologies, Amgen, Inc., South San Francisco, CA
| | - Wen-Chen Yeh
- Metabolic Disorders Amgen, Inc., South San Francisco, CA
| | | | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob Scott
- Cardiovascular, Amgen Inc., Thousand Oaks, CA
| | | | - Hans M G Princen
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Simon Jackson
- Metabolic Disorders Amgen, Inc., South San Francisco, CA
| |
Collapse
|
31
|
Boon MR, Kooijman S, van Dam AD, Pelgrom LR, Berbée JFP, Visseren CAR, van Aggele RC, van den Hoek AM, Sips HCM, Lombès M, Havekes LM, Tamsma JT, Guigas B, Meijer OC, Jukema JW, Rensen PCN. Peripheral cannabinoid 1 receptor blockade activates brown adipose tissue and diminishes dyslipidemia and obesity. FASEB J 2014; 28:5361-75. [PMID: 25154875 DOI: 10.1096/fj.13-247643] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The endocannabinoid system is an important player in energy metabolism by regulating appetite, lipolysis, and energy expenditure. Chronic blockade of the cannabinoid 1 receptor (CB1R) leads to long-term maintenance of weight loss and reduction of dyslipidemia in experimental and human obesity. The molecular mechanism by which CB1R blockade reverses dyslipidemia in obesity has not yet been clarified. In this study, we showed that CB1R blockade with the systemic CB1R blocker rimonabant enhanced whole-body energy expenditure and activated brown adipose tissue (BAT), indicated by increased expression of genes involved in BAT thermogenesis and decreased lipid droplet size in BAT. This was accompanied by selectively increased triglyceride (TG) uptake by BAT and lower plasma TG levels. Interestingly, the effects on BAT activation were still present at thermoneutrality and could be recapitulated by using the strictly peripheral CB1R antagonist AM6545, indicating direct peripheral activation of BAT. Indeed, CB1R blockade directly activated T37i brown adipocytes, resulting in enhanced uncoupled respiration, most likely via enhancing cAMP/PKA signaling via the adrenergic receptor pathway. Our data indicate that selective targeting of the peripheral CB1R in BAT has therapeutic potential in attenuating dyslipidemia and obesity.
Collapse
Affiliation(s)
- Mariëtte R Boon
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands;
| | - Sander Kooijman
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Andrea D van Dam
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Leonard R Pelgrom
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Jimmy F P Berbée
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Cheryl A R Visseren
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Robin C van Aggele
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | | | - Hetty C M Sips
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Marc Lombès
- Institut National de la Santé et de la Recherche Médicale, Unité 693, Le Kremlin-Bicêtre, France
| | - Louis M Havekes
- Department of Endocrinology and Metabolic Diseases, Department of Cardiology, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands; TNO Biosciences, Leiden, The Netherlands; and
| | | | - Bruno Guigas
- Department of Molecular Cell Biology, and Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Onno C Meijer
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | | | - Patrick C N Rensen
- Department of Endocrinology and Metabolic Diseases, Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| |
Collapse
|
32
|
Kühnast S, van der Tuin SJL, van der Hoorn JWA, van Klinken JB, Simic B, Pieterman E, Havekes LM, Landmesser U, Lüscher TF, Willems van Dijk K, Rensen PCN, Jukema JW, Princen HMG. Anacetrapib reduces progression of atherosclerosis, mainly by reducing non-HDL-cholesterol, improves lesion stability and adds to the beneficial effects of atorvastatin. Eur Heart J 2014; 36:39-48. [PMID: 25142968 PMCID: PMC4286319 DOI: 10.1093/eurheartj/ehu319] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The residual risk that remains after statin treatment supports the addition of other LDL-C-lowering agents and has stimulated the search for secondary treatment targets. Epidemiological studies propose HDL-C as a possible candidate. Cholesteryl ester transfer protein (CETP) transfers cholesteryl esters from atheroprotective HDL to atherogenic (V)LDL. The CETP inhibitor anacetrapib decreases (V)LDL-C by ∼15-40% and increases HDL-C by ∼40-140% in clinical trials. We evaluated the effects of a broad dose range of anacetrapib on atherosclerosis and HDL function, and examined possible additive/synergistic effects of anacetrapib on top of atorvastatin in APOE*3Leiden.CETP mice. METHODS AND RESULTS Mice were fed a diet without or with ascending dosages of anacetrapib (0.03; 0.3; 3; 30 mg/kg/day), atorvastatin (2.4 mg/kg/day) alone or in combination with anacetrapib (0.3 mg/kg/day) for 21 weeks. Anacetrapib dose-dependently reduced CETP activity (-59 to -100%, P < 0.001), thereby decreasing non-HDL-C (-24 to -45%, P < 0.001) and increasing HDL-C (+30 to +86%, P < 0.001). Anacetrapib dose-dependently reduced the atherosclerotic lesion area (-41 to -92%, P < 0.01) and severity, increased plaque stability index and added to the effects of atorvastatin by further decreasing lesion size (-95%, P < 0.001) and severity. Analysis of covariance showed that both anacetrapib (P < 0.05) and non-HDL-C (P < 0.001), but not HDL-C (P = 0.76), independently determined lesion size. CONCLUSION Anacetrapib dose-dependently reduces atherosclerosis, and adds to the anti-atherogenic effects of atorvastatin, which is mainly ascribed to a reduction in non-HDL-C. In addition, anacetrapib improves lesion stability.
Collapse
Affiliation(s)
- Susan Kühnast
- Gaubius Laboratory, TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK, PO Box 2215, 2301 CE, Leiden, The Netherlands Department of Cardiology, LUMC, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Sam J L van der Tuin
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands Department of Endocrinology and Metabolic Diseases, LUMC, Leiden, The Netherlands
| | - José W A van der Hoorn
- Gaubius Laboratory, TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK, PO Box 2215, 2301 CE, Leiden, The Netherlands Department of Cardiology, LUMC, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Jan B van Klinken
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands Department of Human Genetics, LUMC, Leiden, The Netherlands
| | - Branko Simic
- Center for Molecular Cardiology, Campus Schlieren, University of Zurich, Zurich, Switzerland
| | - Elsbet Pieterman
- Gaubius Laboratory, TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK, PO Box 2215, 2301 CE, Leiden, The Netherlands
| | - Louis M Havekes
- Gaubius Laboratory, TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK, PO Box 2215, 2301 CE, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands Department of Endocrinology and Metabolic Diseases, LUMC, Leiden, The Netherlands
| | - Ulf Landmesser
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Ko Willems van Dijk
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands Department of Endocrinology and Metabolic Diseases, LUMC, Leiden, The Netherlands Department of Human Genetics, LUMC, Leiden, The Netherlands
| | - Patrick C N Rensen
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands Department of Endocrinology and Metabolic Diseases, LUMC, Leiden, The Netherlands
| | | | - Hans M G Princen
- Gaubius Laboratory, TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK, PO Box 2215, 2301 CE, Leiden, The Netherlands
| |
Collapse
|
33
|
Kühnast S, van der Hoorn JWA, Pieterman EJ, van den Hoek AM, Sasiela WJ, Gusarova V, Peyman A, Schäfer HL, Schwahn U, Jukema JW, Princen HMG. Alirocumab inhibits atherosclerosis, improves the plaque morphology, and enhances the effects of a statin. J Lipid Res 2014; 55:2103-12. [PMID: 25139399 PMCID: PMC4174003 DOI: 10.1194/jlr.m051326] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition is a potential novel strategy for treatment of CVD. Alirocumab is a fully human PCSK9 monoclonal antibody in phase 3 clinical development. We evaluated the antiatherogenic potential of alirocumab in APOE*3Leiden.CETP mice. Mice received a Western-type diet and were treated with alirocumab (3 or 10 mg/kg, weekly subcutaneous dosing) alone and in combination with atorvastatin (3.6 mg/kg/d) for 18 weeks. Alirocumab alone dose-dependently decreased total cholesterol (−37%; −46%, P < 0.001) and TGs (−36%; −39%, P < 0.001) and further decreased cholesterol in combination with atorvastatin (−48%; −58%, P < 0.001). Alirocumab increased hepatic LDL receptor protein levels but did not affect hepatic cholesterol and TG content. Fecal output of bile acids and neutral sterols was not changed. Alirocumab dose-dependently decreased atherosclerotic lesion size (−71%; −88%, P < 0.001) and severity and enhanced these effects when added to atorvastatin (−89%; −98%, P < 0.001). Alirocumab reduced monocyte recruitment and improved the lesion composition by increasing the smooth muscle cell and collagen content and decreasing the macrophage and necrotic core content. Alirocumab dose-dependently decreases plasma lipids and, as a result, atherosclerosis development, and it enhances the beneficial effects of atorvastatin in APOE*3Leiden.CETP mice. In addition, alirocumab improves plaque morphology.
Collapse
Affiliation(s)
- Susan Kühnast
- The Netherlands Organization of Applied Scientific Research (TNO) - Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - José W A van der Hoorn
- The Netherlands Organization of Applied Scientific Research (TNO) - Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Elsbet J Pieterman
- The Netherlands Organization of Applied Scientific Research (TNO) - Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Anita M van den Hoek
- The Netherlands Organization of Applied Scientific Research (TNO) - Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | | | | | - Anusch Peyman
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | | | - Uwe Schwahn
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans M G Princen
- The Netherlands Organization of Applied Scientific Research (TNO) - Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| |
Collapse
|
34
|
A review of paradoxical HDL-C responses to fenofibrate, illustrated by a case report. J Clin Lipidol 2014; 8:455-9. [DOI: 10.1016/j.jacl.2014.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 05/07/2014] [Accepted: 05/18/2014] [Indexed: 11/18/2022]
|
35
|
van den Hoek AM, van der Hoorn JWA, Maas AC, van den Hoogen RM, van Nieuwkoop A, Droog S, Offerman EH, Pieterman EJ, Havekes LM, Princen HMG. APOE*3Leiden.CETP transgenic mice as model for pharmaceutical treatment of the metabolic syndrome. Diabetes Obes Metab 2014; 16:537-44. [PMID: 24373179 DOI: 10.1111/dom.12252] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/08/2013] [Accepted: 12/12/2013] [Indexed: 02/03/2023]
Abstract
AIMS This study aimed to investigate systematically (i) the appropriate dietary conditions to induce the features of the MetS in APOE*3Leiden.humanCholesteryl Ester Transfer Protein (E3L.CETP) mice and (ii) whether the response of this model to different antidiabetic and hypolipidemic drugs is similar as in humans. METHODS Male obese, IR and dyslipidemic E3L.CETP mice were treated with antidiabetic drugs rosiglitazone, liraglutide or an experimental 11β-hydroxysteroid-dehydrogenase-1 (HSD-1) inhibitor, or with hypolipidemic drugs atorvastatin, fenofibrate or niacin for 4-6 weeks. The effects on bw, IR and plasma and liver lipids were assessed. RESULTS Rosiglitazone, liraglutide and HSD-1 inhibitor significantly decreased glucose and insulin levels or IR. Liraglutide and HSD-1 inhibitor also decreased bw. Atorvastatin, fenofibrate and niacin improved the dyslipidemia and fenofibrate and niacin increased high-density lipoprotein (HDL) cholesterol. In addition, hepatic triglycerides were significantly decreased by treatment with rosiglitazone and liraglutide, while hepatic cholesterol esters were significantly decreased by rosiglitazone and atorvastatin. CONCLUSIONS We conclude that the E3L.CETP mouse is a promising novel translational model to investigate the effects of new drugs, alone or in combination, that affect IR, diabetic dyslipidemia and non-alcoholic fatty liver disease (NAFLD).
Collapse
|
36
|
Gierman LM, Kühnast S, Koudijs A, Pieterman EJ, Kloppenburg M, van Osch GJVM, Stojanovic-Susulic V, Huizinga TWJ, Princen HMG, Zuurmond AM. Osteoarthritis development is induced by increased dietary cholesterol and can be inhibited by atorvastatin in APOE*3Leiden.CETP mice--a translational model for atherosclerosis. Ann Rheum Dis 2014; 73:921-7. [PMID: 23625977 DOI: 10.1136/annrheumdis-2013-203248] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Hypercholesterolaemia, a risk factor for atherosclerosis (ATH), has been suggested to have a role in the development of osteoarthritis (OA). To test this hypothesis, the effect of cholesterol and different cholesterol-lowering treatments on OA was investigated in a mouse model resembling human lipoprotein metabolism. METHODS Female ApolipoproteinE*3Leiden.human Cholesteryl Ester Transfer Protein mice received a western-type diet with 0.1% (w/w) cholesterol (LC), 0.3% (w/w) cholesterol alone (HC) or treated with 3 mg/kg/day atorvastatin or 0.3 mg/kg/day ezetimibe. One group remained on chow (control). After 39 weeks, OA grades of the knees and the extent of ATH were determined. Plasma cholesterol levels were measured throughout the study. RESULTS LC and HC groups developed significantly more OA at the medial side than the control group in a dose-dependent manner. Atorvastatin but not ezetimibe treatment significantly suppressed OA development. As expected, features of ATH were significantly increased in the LC and HC groups compared with the control group and suppressed by atorvastatin (48%) and ezetimibe (55%) treatment. There were significant correlations between the development of OA on the medial side of the joint and cholesterol exposure (r=0.4) or ATH features (r=0.3). CONCLUSIONS Dietary cholesterol and accordingly increased plasma levels play a role in the development of OA. The correlation found between OA, cholesterol and ATH demonstrates that these variables are connected, but indicates the contribution of other ongoing processes in the development of OA. The suppressive effect on OA development of atorvastatin but not of ezetimibe, which had similar cholesterol exposure levels, corroborates these findings.
Collapse
|
37
|
Fibrates and fish oil, but not corn oil, up-regulate the expression of the cholesteryl ester transfer protein (CETP) gene. J Nutr Biochem 2014; 25:669-74. [PMID: 24746832 DOI: 10.1016/j.jnutbio.2014.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 02/07/2014] [Accepted: 02/12/2014] [Indexed: 12/28/2022]
Abstract
Cholesteryl ester transfer protein (CETP) is a plasma protein that reduces high density lipoprotein (HDL)-cholesterol (chol) levels and may increase atherosclerosis risk. n-3 and n-6 polyunsaturated fatty acids (PUFAs) are natural ligands, and fibrates are synthetic ligands for peroxisome proliferator activated receptor-alpha (PPARα), a transcription factor that modulates lipid metabolism. In this study, we investigated the effects of PUFA oils and fibrates on CETP expression. Hypertriglyceridemic CETP transgenic mice were treated with gemfibrozil, fenofibrate, bezafibrate or vehicle (control), and normolipidemic CETP transgenic mice were treated with fenofibrate or with fish oil (FO; n-3 PUFA rich), corn oil (CO, n-6 PUFA rich) or saline. Compared with the control treatment, only fenofibrate significantly diminished triglyceridemia (50%), whereas all fibrates decreased the HDL-chol level. Elevation of the CETP liver mRNA levels and plasma activity was observed in the fenofibrate (53%) and gemfibrozil (75%) groups. Compared with saline, FO reduced the plasma levels of nonesterified fatty acid (26%), total chol (15%) and HDL-chol (20%). Neither of the oil treatments affected the plasma triglyceride levels. Compared with saline, FO increased the plasma adiponectin level and reduced plasma leptin levels, whereas CO increased the leptin levels. FO, but not CO, significantly increased the plasma CETP mass (90%) and activity (23%) as well as increased the liver level of CETP mRNA (28%). In conclusion, fibrates and FO, but not CO, up-regulated CETP expression at both the mRNA and protein levels. We propose that these effects are mediated by the activation of PPARα, which acts on a putative PPAR response element in the CETP gene.
Collapse
|
38
|
Aslibekyan S, Straka RJ, Irvin MR, Claas SA, Arnett DK. Pharmacogenomics of high-density lipoprotein-cholesterol-raising therapies. Expert Rev Cardiovasc Ther 2013; 11:355-64. [PMID: 23469915 DOI: 10.1586/erc.12.134] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
High levels of HDL cholesterol (HDL-C) have traditionally been linked to lower incidence of cardiovascular disease, prompting the search for effective and safe HDL-C raising pharmaceutical agents. Although drugs such as niacin and fibrates represent established therapeutic approaches, HDL-C response to such therapies is variable and heritable, suggesting a role for pharmacogenomic determinants. Multiple genetic polymorphisms, located primarily in genes encoding lipoproteins, cholesteryl ester transfer protein, transporters and CYP450 proteins have been shown to associate with HDL-C drug response in vitro and in epidemiologic studies. However, few of the pharmacogenomic findings have been independently validated, precluding the development of clinical tools that can be used to predict HDL-C response and leaving the goal of personalized medicine to future efforts.
Collapse
Affiliation(s)
- Stella Aslibekyan
- Department of Epidemiology, Ryals School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | | | | | | |
Collapse
|
39
|
Yakala GK, Wielinga PY, Suarez M, Bunschoten A, van Golde JM, Arola L, Keijer J, Kleemann R, Kooistra T, Heeringa P. Effects of chocolate supplementation on metabolic and cardiovascular parameters in ApoE3L mice fed a high-cholesterol atherogenic diet. Mol Nutr Food Res 2013; 57:2039-48. [PMID: 23946229 DOI: 10.1002/mnfr.201200858] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 04/05/2013] [Accepted: 05/17/2013] [Indexed: 11/11/2022]
Abstract
SCOPE Dietary intake of cocoa and/or chocolate has been suggested to exhibit protective cardiovascular effects although this is still controversial. The aim of this study was to investigate the effects of chocolate supplementation on metabolic and cardiovascular parameters. METHODS AND RESULTS Four groups of ApoE*3Leiden mice were exposed to the following diet regimens. Group 1: cholesterol-free control diet (CO). Group 2: high-dose (1.0% w/w) control cholesterol (CC). Group 3: CC supplemented chocolate A (CCA) and Group 4: CC supplemented chocolate B (CCB). Both chocolates differed in polyphenol and fiber content, CCA had a relatively high-polyphenol and low-fiber content compared to CCB. Mice fed a high-cholesterol diet showed increased plasma-cholesterol and developed atherosclerosis. Both chocolate treatments, particularly CCA, further increased plasma-cholesterol and increased atherosclerotic plaque formation. Moreover, compared to mice fed a high-cholesterol diet, both chocolate-treated groups displayed increased liver injury. Mice on high-cholesterol diet had elevated plasma levels of sVCAM-1, sE-selectin and SAA, which was further increased in the CCB group. Similar effects were observed for renal inflammation markers. CONCLUSION The two chocolate preparations showed unfavorable, but different effects on cardiometabolic health in E3L mice, which dissimilarities may be related to differences in chocolate composition. We conclude that discrepancies reported on the effects of chocolate on cardiometabolic health may at least partly be due to differences in chocolate composition.
Collapse
Affiliation(s)
- Gopala K Yakala
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Top Institute Food and Nutrition, Wageningen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Auvinen HE, Wang Y, Princen H, Romijn JA, Havekes LM, Smit JWA, Meijer OC, Biermasz NR, Rensen PCN, Pereira AM. Both transient and continuous corticosterone excess inhibit atherosclerotic plaque formation in APOE*3-leiden.CETP mice. PLoS One 2013; 8:e63882. [PMID: 23717502 PMCID: PMC3661690 DOI: 10.1371/journal.pone.0063882] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 04/09/2013] [Indexed: 01/28/2023] Open
Abstract
Introduction The role of glucocorticoids in atherosclerosis development is not clearly established. Human studies show a clear association between glucocorticoid excess and cardiovascular disease, whereas most animal models indicate an inhibitory effect of glucocorticoids on atherosclerosis development. These animal models, however, neither reflect long-term glucocorticoid overexposure nor display human-like lipoprotein metabolism. Aim To investigate the effects of transient and continuous glucocorticoid excess on atherosclerosis development in a mouse model with human-like lipoprotein metabolism upon feeding a Western-type diet. Methods Pair-housed female APOE*3-Leiden.CETP (E3L.CETP) mice fed a Western-type containing 0.1% cholesterol for 20 weeks were given corticosterone (50 µg/ml) for either 5 (transient group) or 17 weeks (continuous group), or vehicle (control group) in the drinking water. At the end of the study, atherosclerosis severity, lesion area in the aortic root, the number of monocytes adhering to the endothelial wall and macrophage content of the plaque were measured. Results Corticosterone treatment increased body weight and food intake for the duration of the treatment and increased gonadal and subcutaneous white adipose tissue weight in transient group by +35% and +31%, and in the continuous group by +140% and 110%. Strikingly, both transient and continuous corticosterone treatment decreased total atherosclerotic lesion area by −39% without lowering plasma cholesterol levels. In addition, there was a decrease of −56% in macrophage content of the plaque with continuous corticosterone treatment, and a similar trend was present with the transient treatment. Conclusion Increased corticosterone exposure in mice with human-like lipoprotein metabolism has beneficial, long-lasting effects on atherosclerosis, but negatively affects body fat distribution by promoting fat accumulation in the long-term. This indicates that the increased atherosclerosis observed in humans in states of glucocorticoid excess may not be related to cortisol per se, but might be the result of complex indirect effects of cortisol.
Collapse
Affiliation(s)
- Hanna E Auvinen
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Karalis I, Rensen PCN, Jukema JW. Journey through cholesteryl ester transfer protein inhibition: from bench to bedside. Circ Cardiovasc Qual Outcomes 2013; 6:360-6. [PMID: 23674310 DOI: 10.1161/circoutcomes.111.000014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Ioannis Karalis
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
42
|
Hulsmans M, Geeraert B, Arnould T, Tsatsanis C, Holvoet P. PPAR agonist-induced reduction of Mcp1 in atherosclerotic plaques of obese, insulin-resistant mice depends on adiponectin-induced Irak3 expression. PLoS One 2013; 8:e62253. [PMID: 23620818 PMCID: PMC3631170 DOI: 10.1371/journal.pone.0062253] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 03/19/2013] [Indexed: 11/18/2022] Open
Abstract
Synthetic peroxisome proliferator-activated receptor (PPAR) agonists are used to treat dyslipidemia and insulin resistance. In this study, we examined molecular mechanisms that explain differential effects of a PPARα agonist (fenofibrate) and a PPARγ agonist (rosiglitazone) on macrophages during obesity-induced atherogenesis. Twelve-week-old mice with combined leptin and LDL-receptor deficiency (DKO) were treated with fenofibrate, rosiglitazone or placebo for 12 weeks. Only rosiglitazone improved adipocyte function, restored insulin sensitivity, and inhibited atherosclerosis by decreasing lipid-loaded macrophages. In addition, it increased interleukin-1 receptor-associated kinase-3 (Irak3) and decreased monocyte chemoattractant protein-1 (Mcp1) expressions, indicative of a switch from M1 to M2 macrophages. The differences between fenofibrate and rosiglitazone were independent of Pparγ expression. In bone marrow-derived macrophages (BMDM), we identified the rosiglitazone-associated increase in adiponectin as cause of the increase in Irak3. Interestingly, the deletion of Irak3 in BMDM (IRAK3−/− BMDM) resulted in activation of the canonical NFκB signaling pathway and increased Mcp1 protein secretion. Rosiglitazone could not decrease the elevated Mcp1 secretion in IRAK3−/− BMDM directly and fenofibrate even increased the secretion, possibly due to increased mitochondrial reactive oxygen species production. Furthermore, aortic extracts of high-fat insulin-resistant LDL-receptor deficient mice, with lower adiponectin and Irak3 and higher Mcp1, showed accelerated atherosclerosis. In aggregate, our results emphasize an interaction between PPAR agonist-mediated increase in adiponectin and macrophage-associated Irak3 in the protection against atherosclerosis by PPAR agonists.
Collapse
Affiliation(s)
- Maarten Hulsmans
- Atherosclerosis and Metabolism Unit, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
43
|
Zhao H, Zhu C, Qin C, Tao T, Li J, Cheng G, Li P, Cao Q, Meng X, Ju X, Shao P, Hua L, Gu M, Yin C. Fenofibrate down-regulates the expressions of androgen receptor (AR) and AR target genes and induces oxidative stress in the prostate cancer cell line LNCaP. Biochem Biophys Res Commun 2013; 432:320-5. [PMID: 23399562 DOI: 10.1016/j.bbrc.2013.01.105] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 01/24/2013] [Indexed: 01/22/2023]
Abstract
Fenofibrate, a peroxisome proliferator-androgen receptor-alpha agonist, is widely used in treating different forms of hyperlipidemia and hypercholesterolemia. Recent reports have indicated that fenofibrate exerts anti-proliferative and pro-apoptotic properties. This study aims to investigate the effects of fenofibrate on the prostate cancer (PCa) cell line LNCaP. The effects of fenofibrate on LNCaP cells were evaluated by flow cytometry, reverse transcription-polymerase chain reaction, enzyme-linked immunosorbent assays, Western blot analysis, and dual-luciferase reporter assay. Fenofibrate induces cell cycle arrest in G1 phase and apoptosis in LNCaP cells, reduces the expressions of androgen receptor (AR) and AR target genes (prostate-specific antigen and TMPRSS2), and inhibits Akt phosphorylation. Fenofibrate can induce the accumulation of intracellular reactive oxygen species and malondialdehyde, and decrease the activities of total anti-oxidant and superoxide dismutase in LNCaP cells. Fenofibrate exerts an anti-proliferative property by inhibiting the expression of AR and induces apoptosis by causing oxidative stress. Therefore, our data suggest fenofibrate may have beneficial effects in fenofibrate users by preventing prostate cancer growth through inhibition of androgen activation and expression.
Collapse
Affiliation(s)
- Hu Zhao
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Aslibekyan S, Goodarzi MO, Frazier-Wood AC, Yan X, Irvin MR, Kim E, Tiwari HK, Guo X, Straka RJ, Taylor KD, Tsai MY, Hopkins PN, Korenman SG, Borecki IB, Chen YDI, Ordovas JM, Rotter JI, Arnett DK. Variants identified in a GWAS meta-analysis for blood lipids are associated with the lipid response to fenofibrate. PLoS One 2012; 7:e48663. [PMID: 23119086 PMCID: PMC3485381 DOI: 10.1371/journal.pone.0048663] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 09/28/2012] [Indexed: 12/23/2022] Open
Abstract
A recent large-scale meta-analysis of genome-wide studies has identified 95 loci, 59 of them novel, as statistically significant predictors of blood lipid traits; we tested whether the same loci explain the observed heterogeneity in response to lipid-lowering therapy with fenofibrate. Using data from the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN, n = 861) we fit linear mixed models with the genetic markers as predictors and high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, total cholesterol, and triglyceride concentrations as outcomes. For all four traits, we analyzed both baseline levels and changes in response to treatment with fenofibrate. For the markers that were significantly associated with fenofibrate response, we fit additional models evaluating potential epistatic interactions. All models were adjusted for age, sex, and study center as fixed effects, and pedigree as a random effect. Statistically significant associations were observed between the rs964184 polymorphism near APOA1 (P-value≤0.0001) and fenofibrate response for HDL and triglycerides. The association was replicated in the Pharmacogenetics of Hypertriglyceridemia in Hispanics study (HyperTG, n = 267). Suggestive associations with fenofibrate response were observed for markers in or near PDE3A, MOSC1, FLJ36070, CETP, the APOE-APOC1-APOC4-APOC2, and CILP2. Finally, we present strong evidence for epistasis (P-value for interaction = 0.0006 in GOLDN, 0.05 in HyperTG) between rs10401969 near CILP2 and rs4420638 in the APOE-APOC1-APOC4-APOC2 cluster with total cholesterol response to fenofibrate. In conclusion, we present evidence linking several novel and biologically relevant genetic polymorphisms to lipid lowering drug response, as well as suggesting novel gene-gene interactions in fenofibrate pharmacogenetics.
Collapse
Affiliation(s)
- Stella Aslibekyan
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Torsney E, Pirianov G, Charolidi N, Shoreim A, Gaze D, Petrova S, Laing K, Meisinger T, Xiong W, Baxter BT, Cockerill GW. Elevation of plasma high-density lipoproteins inhibits development of experimental abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol 2012; 32:2678-86. [PMID: 23023368 DOI: 10.1161/atvbaha.112.00009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Patients with abdominal aortic aneurysms have lower concentrations of high-density lipoproteins (HDLs), leading us to investigate whether increasing plasma HDLs could influence aneurysm formation. METHODS AND RESULTS Using the angiotensin II-induced hypercholesterolemic and the CaCl(2)-induced normocholesterolemic mouse model of AAA, we investigated the hypothesis that elevation of HDLs inhibits AAA. HDLs elevated before or at the time of AAA induction reduced AAA formation in both models but had no effect on early ruptures. Analysis of protein lysates from specific aortic segments demonstrated site-specific effects of HDLs on early signal transduction and cellular attrition. We found that HDLs reduced extracellular signal related kinases 1/2 activation in the suprarenal segment, while having no effect on p38 mitogen-associated protein kinase activation in any aortic segment and inhibiting c-Jun N-terminal kinase activation in all aortic segments. In addition, HDL elevation inhibited angiotensin II-induced apoptosis while inducing autophagy in the suprarenal segment of the aorta. Using Illumina gene array profiling we investigated the ability of HDL to modulate basal suprarenal aortic gene expression. CONCLUSIONS Increasing plasma HDLs inhibit experimental AAA formation, independent of hypercholesterolemia via reduced extracellular signal related kinases 1/2 activation and alteration of the balance of cellular attrition. HDLs modulate genes involved in matrix remodelling, cell migration, and proliferation.
Collapse
Affiliation(s)
- Evelyn Torsney
- Division of Clinical Sciences, St George's University of London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Giordano A, Macaluso M. Fenofibrate triggers apoptosis of glioblastoma cells in vitro: new insights for therapy. Cell Cycle 2012; 11:3154. [PMID: 22895169 PMCID: PMC3466510 DOI: 10.4161/cc.21719] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
47
|
Zema MJ. Colesevelam hydrochloride: evidence for its use in the treatment of hypercholesterolemia and type 2 diabetes mellitus with insights into mechanism of action. CORE EVIDENCE 2012; 7:61-75. [PMID: 22936894 PMCID: PMC3426253 DOI: 10.2147/ce.s26725] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Colesevelam hydrochloride is a molecularly engineered, second-generation bile acid sequestrant demonstrating enhanced specificity for bile acids which has been approved for use as adjunctive therapy to diet and exercise as monotherapy or in combination with a β-hydroxymethylglutaryl-coenzyme A reductase inhibitor for the reduction of elevated low-density lipoprotein cholesterol in patients with primary hypercholesterolemia. It is also the only lipid-lowering agent currently available in the United States which has been approved for use as adjunctive therapy in patients with type 2 diabetes mellitus whose glycemia remains inadequately controlled on therapy with metformin, sulfonylurea, or insulin. With the recent emphasis upon drug safety by the Food and Drug Administration and various consumer agencies, it is fitting that the role of nonsystemic lipid-lowering therapies such as bile acid sequestrants – with nearly 90 years of in-class, clinically safe experience – should be reexamined. This paper presents information on the major pharmacologic effects of colesevelam, including a discussion of recent data derived from both in vitro and in vivo rodent and human studies, which shed light on the putative mechanisms involved.
Collapse
|
48
|
Chen WL, Chen YL, Chiang YM, Wang SG, Lee HM. Fenofibrate lowers lipid accumulation in myotubes by modulating the PPARα/AMPK/FoxO1/ATGL pathway. Biochem Pharmacol 2012; 84:522-31. [PMID: 22687626 DOI: 10.1016/j.bcp.2012.05.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 05/28/2012] [Accepted: 05/31/2012] [Indexed: 12/14/2022]
Abstract
Fenofibrate, a fibric acid derivative, is known to possess lipid-lowering effects. Although fenofibrate may activate peroxisome proliferator-activated receptor (PPAR)α and regulate the transcription of several genes, the underlying mechanisms are poorly understood. In this study, we demonstrated that incubation of C2C12 myotubes with fenofibrate increased adipose triglyceride lipase (ATGL) expression and suppressed fatty acid synthase (FAS) level, thereby decreasing intracellular triglyceride accumulation when cells were incubated at high-glucose condition. Fenofibrate increased the phosphorylation of AMP-activated protein kinase (AMPK), which subsequently increased fatty acid β-oxidation. AMPK phosphorylation was reduced by pretreatment with GW9662 (a PPARα inhibitor), suggesting that AMPK may be a downstream effector of PPARα. Pretreatment with compound C (an AMPK inhibitor) or GW9662 blocked fenofibrate-induced ATGL expression and the lipid-lowering effect. Our results suggest that AMPK is as an upstream regulator of ATGL. With further exploration, we demonstrated that fenofibrate stimulated FoxO1 translocation from the cytosol to nuclei by immunefluorescence assay, chromatin immuneprecipitation assay, and reporter assay. Furthermore, oral administration of fenofibrate ameliorated the body weight, visceral fat and serum biochemical indexes in db/db mice. Taken together, our results suggest that the lipid-lowering effect of fenofibrate was achieved by activating PPARα and AMPK signaling pathway that resulted in increasing ATGL expression, lipolysis, and fatty acid β-oxidation.
Collapse
Affiliation(s)
- Wei-Lu Chen
- Graduate Institute of Medical Sciences, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Aliskiren inhibits atherosclerosis development and improves plaque stability in APOE*3Leiden.CETP transgenic mice with or without treatment with atorvastatin. J Hypertens 2012; 30:107-16. [DOI: 10.1097/hjh.0b013e32834ddd8e] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|