137 results on '"Princen, Hans M. G."'
Search Results
2. Effects of repeated weight cycling on non‐alcoholic steatohepatitis in diet‐induced obese mice.
- Author
-
Inia, José A., de Jong, Jelle C. B. C., Keijzer, Nanda, Menke, Aswin L., Princen, Hans M. G., Jukema, J. Wouter, and van den Hoek, Anita M.
- Published
- 2024
- Full Text
- View/download PDF
3. A systems toxicology approach for identification of disruptions in cholesterol homeostasis after aggregated exposure to mixtures of perfluorinated compounds in humans.
- Author
-
Westerhout, Joost, Heijer-Jordaan, Annemiek den, Princen, Hans M G, and Stierum, Rob
- Subjects
PERFLUOROOCTANOIC acid ,FLUOROALKYL compounds ,CHOLESTEROL ,LIPID metabolism ,TOXICOLOGY ,INDUSTRIAL goods ,MIXTURES ,CELL aggregation - Abstract
Per- and polyfluoroalkyl substances (PFAS) are used in various household and industrial products. In humans, positive associations were reported between PFAS, including perfluorsulfonic acid and perfluorooctanoic acid, and cholesterol, a cardiometabolic risk factor. Animal studies show the opposite. Human-centered approaches are needed to better understand the effects of PFAS mixtures on cholesterol. Here, a systems toxicology approach is described, using a gene-centered cholesterol biokinetic model. PFAS exposure-gene expression relations from published data were introduced into the model. An existing PFAS physiologically based kinetic model was augmented with lung and dermal compartments and integrated with the cholesterol model to enable exposure-effect modeling. The final model was populated with data reflecting lifetime mixture exposure from: tolerable weekly intake values; the environment; high occupational exposures (ski waxing, PFAS industry). Results indicate that low level exposures (tolerable weekly intake, environmental) did not change cholesterol. In contrast, occupational exposures clearly resulted in internal PFAS exposure and disruption of cholesterol homeostasis, largely in line with epidemiological observations. Despite model limitations (eg, dynamic range, directionality), changes in cholesterol homeostasis were predicted for ski waxers, hitherto unknown from epidemiological studies. Here, future studies involving lipid metabolism could improve risk assessment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Inhibition of macrophage proliferation dominates plaque regression in response to cholesterol lowering
- Author
-
Härdtner, Carmen, Kornemann, Jan, Krebs, Katja, Ehlert, Carolin A., Jander, Alina, Zou, Jiadai, Starz, Christopher, Rauterberg, Simon, Sharipova, Diana, Dufner, Bianca, Hoppe, Natalie, Dederichs, Tsai-Sang, Willecke, Florian, Stachon, Peter, Heidt, Timo, Wolf, Dennis, von zur Mühlen, Constantin, Madl, Josef, Kohl, Peter, Kaeser, Rafael, Boettler, Tobias, Pieterman, Elsbeth J., Princen, Hans M. G., Ho-Tin-Noé, Benoît, Swirski, Filip K., Robbins, Clinton S., Bode, Christoph, Zirlik, Andreas, and Hilgendorf, Ingo
- Published
- 2020
- Full Text
- View/download PDF
5. Results, meta-analysis and a first evaluation of UNOxR, the urinary nitrate-to-nitrite molar ratio, as a measure of nitrite reabsorption in experimental and clinical settings
- Author
-
Tsikas, Dimitrios, Hanff, Erik, Bollenbach, Alexander, Kruger, Ruan, Pham, Vu Vi, Chobanyan-Jürgens, Kristine, Wedekind, Dirk, Arndt, Tanja, Jörns, Anne, Berbée, Jimmy F. P., Princen, Hans M. G., Lücke, Thomas, Mariotti, François, Huneau, Jean-François, Ückert, Stefan, Frölich, Jürgen C., and Lenzen, Sigurd
- Published
- 2018
- Full Text
- View/download PDF
6. Anti-PCSK9 antibodies inhibit pro-atherogenic mechanisms in APOE*3Leiden.CETP mice
- Author
-
Schuster, Susanne, Rubil, Sandra, Endres, Matthias, Princen, Hans M. G., Boeckel, Jes-Niels, Winter, Karsten, Werner, Christian, and Laufs, Ulrich
- Published
- 2019
- Full Text
- View/download PDF
7. Semaglutide Has Beneficial Effects on Non-Alcoholic Steatohepatitis in Ldlr-/-.Leiden Mice.
- Author
-
Inia, José A., Stokman, Geurt, Morrison, Martine C., Worms, Nicole, Verschuren, Lars, Caspers, Martien P. M., Menke, Aswin L., Petitjean, Louis, Chen, Li, Petitjean, Mathieu, Jukema, J. Wouter, Princen, Hans M. G., and van den Hoek, Anita M.
- Subjects
NON-alcoholic fatty liver disease ,GLUCAGON-like peptide-1 receptor ,SEMAGLUTIDE ,GLUCAGON-like peptide-1 agonists ,FATTY liver ,MICE ,GLUCAGON receptors - Abstract
Semaglutide, a glucagon-like peptide-1 receptor agonist, is an antidiabetic medication that has recently been approved for the treatment of obesity as well. Semaglutide is postulated to be a promising candidate for the treatment of non-alcoholic steatohepatitis (NASH). Here, Ldlr-/-.Leiden mice received a fast-food diet (FFD) for 25 weeks, followed by another 12 weeks on FFD with daily subcutaneous injections of semaglutide or vehicle (control). Plasma parameters were evaluated, livers and hearts were examined, and hepatic transcriptome analysis was performed. In the liver, semaglutide significantly reduced macrovesicular steatosis (−74%, p < 0.001) and inflammation (−73%, p < 0.001) and completely abolished microvesicular steatosis (−100%, p < 0.001). Histological and biochemical assessment of hepatic fibrosis showed no significant effects of semaglutide. However, digital pathology revealed significant improvements in the degree of collagen fiber reticulation (−12%, p < 0.001). Semaglutide did not affect atherosclerosis relative to controls. Additionally, we compared the transcriptome profile of FFD-fed Ldlr-/-.Leiden mice with a human gene set that differentiates human NASH patients with severe fibrosis from those with mild fibrosis. In FFD-fed Ldlr-/-.Leiden control mice, this gene set was upregulated as well, while semaglutide predominantly reversed this gene expression. Using a translational model with advanced NASH, we demonstrated that semaglutide is a promising candidate with particular potential for the treatment of hepatic steatosis and inflammation, while for the reversal of advanced fibrosis, combinations with other NASH agents may be necessary. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Atorvastatin Attenuates Diet-Induced Non-Alcoholic Steatohepatitis in APOE*3-Leiden Mice by Reducing Hepatic Inflammation.
- Author
-
Inia, José A., Stokman, Geurt, Pieterman, Elsbet J., Morrison, Martine C., Menke, Aswin L., Verschuren, Lars, Caspers, Martien P. M., Giera, Martin, Jukema, J. Wouter, van den Hoek, Anita M., and Princen, Hans M. G.
- Subjects
MICE ,NON-alcoholic fatty liver disease ,WESTERN diet ,HEPATIC fibrosis ,ATORVASTATIN ,PREVENTIVE medicine - Abstract
Patients with metabolic syndrome are often prescribed statins to prevent the development of cardiovascular disease. Conversely, data on their effects on non-alcoholic steatohepatitis (NASH) are lacking. We evaluated these effects by feeding APOE*3-Leiden mice a Western-type diet (WTD) with or without atorvastatin to induce NASH and hepatic fibrosis. Besides the well-known plasma cholesterol lowering (−30%) and anti-atherogenic effects (severe lesion size −48%), atorvastatin significantly reduced hepatic steatosis (−22%), the number of aggregated inflammatory cells in the liver (−80%) and hepatic fibrosis (−92%) compared to WTD-fed mice. Furthermore, atorvastatin-treated mice showed less immunohistochemically stained areas of inflammation markers. Atorvastatin prevented accumulation of free cholesterol in the form of cholesterol crystals (−78%). Cholesterol crystals are potent inducers of the NLRP3 inflammasome pathway and atorvastatin prevented its activation, which resulted in reduced expression of the pro-inflammatory cytokines interleukin (IL)-1β (−61%) and IL-18 (−26%). Transcriptome analysis confirmed strong reducing effects of atorvastatin on inflammatory mediators, including NLRP3, NFκB and TLR4. The present study demonstrates that atorvastatin reduces hepatic steatosis, inflammation and fibrosis and prevents cholesterol crystal formation, thereby precluding NLRP3 inflammasome activation. This may render atorvastatin treatment as an attractive approach to reduce NAFLD and prevent progression into NASH in dyslipidemic patients. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. The tyrosine kinase inhibitor nilotinib targets the discoidin domain receptor DDR2 in calcific aortic valve stenosis.
- Author
-
Carracedo, Miguel, Pawelzik, Sven‐Christian, Artiach, Gonzalo, Pouwer, Marianne G., Plunde, Oscar, Saliba‐Gustafsson, Peter, Ehrenborg, Ewa, Eriksson, Per, Pieterman, Elsbet, Stenke, Leif, Princen, Hans M. G., Franco‐Cereceda, Anders, Bäck, Magnus, Pawelzik, Sven-Christian, Saliba-Gustafsson, Peter, and Franco-Cereceda, Anders
- Abstract
Background and Purpose: Tyrosine kinase inhibitors (TKI) used to treat chronic myeloid leukaemia (CML) have been associated with cardiovascular side effects, including reports of calcific aortic valve stenosis. The aim of this study was to establish the effects of first and second generation TKIs in aortic valve stenosis and to determine the associated molecular mechanisms.Experimental Approach: Hyperlipidemic APOE*3Leiden.CETP transgenic mice were treated with nilotinib, imatinib or vehicle. Human valvular interstitial cells (VICs) were isolated and studied in vitro. Gene expression analysis was perfromed in aortic valves from 64 patients undergoing aortic valve replacement surgery.Key Results: Nilotinib increased murine aortic valve thickness. Nilotinib, but not imatinib, promoted calcification and osteogenic activation and decreased autophagy in human VICs. Differential tyrosine kinase expression was detected between healthy and calcified valve tissue. Transcriptomic target identification revealed that the discoidin domain receptor DDR2, which is preferentially inhibited by nilotinib, was predominantly expressed in human aortic valves but markedly downregulated in calcified valve tissue. Nilotinib and selective DDR2 targeting in VICs induced a similar osteogenic activation, which was blunted by increasing the DDR2 ligand, collagen.Conclusions and Implications: These findings suggest that inhibition of DDR2 by nilotinib promoted aortic valve thickening and VIC calcification, with possible translational implications for cardiovascular surveillance and possible personalized medicine in CML patients. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
10. Bexarotene Induces Dyslipidemia by Increased Very Low-Density Lipoprotein Production and Cholesteryl Ester Transfer Protein-Mediated Reduction of High-Density Lipoprotein
- Author
-
de Vries-van der Weij, Jitske, de Haan, Willeke, Hu, Lihui, Kuif, Maarten, Oei, H Ling D. W., van der Hoorn, José W. A., Havekes, Louis M., Princen, Hans M. G., Romijn, Johannes A., Smit, Johannes W. A., and Rensen, Patrick C. N.
- Published
- 2009
11. The Cholesterol-Raising Factor from Coffee Beans, Cafestol, as an Agonist Ligand for the Farnesoid and Pregnane X Receptors
- Author
-
Ricketts, Marie-Louise, Boekschoten, Mark V., Kreeft, Arja J., Hooiveld, Guido J. E. J., Moen, Corina J. A., Müller, Michael, Frants, Rune R., Kasanmoentalib, Soemini, Post, Sabine M., Princen, Hans M. G., Porter, J Gordon, Katan, Martijn B., Hofker, Marten H., and Moore, David D.
- Published
- 2007
12. Fish Oil Increases Bile Acid Synthesis in Male Patients with Hypertriglyceridemia1
- Author
-
Jonkers, Iris J. A. M., Smelt, Augustinus H. M., Princen, Hans M. G., Kuipers, Folkert, Romijn, Johannes A., Boverhof, Renze, Masclee, Ad A. M., and Stellaard, Frans
- Published
- 2006
13. Coffee Oil Consumption Increases Plasma Levels of 7α-Hydroxy-4-cholesten-3-one in Humans1
- Author
-
Boekschoten, Mark V., Hofman, Maaike K., Buytenhek, Rien, Schouten, Evert G., Princen, Hans M. G., and Katan, Martijn B.
- Published
- 2005
14. Well-Characterized Garlic-Derived Materials Are Not Hypolipidemic in APOE*3-Leiden Transgenic Mice1
- Author
-
Espirito Santo, Sonia M. S., van Vlijmen, Bart J. M., Buytenhek, Rien, van Duyvenvoorde, Wim, Havekes, Louis M., Arnault, Ingrid, Auger, Jacques, and Princen, Hans M. G.
- Published
- 2004
15. Chronic Oral Administration of Mineral Oil Compared With Corn Oil: Effects on Gut Permeability and Plasma Inflammatory and Lipid Biomarkers.
- Author
-
Pieterman, Elsbet J., Princen, Hans M. G., Jarke, Annica, Nilsson, Ralf, Cavallin, Anders, Bergenholm, Linnéa, Henricsson, Marcus, Gopaul, V. Sashi, Agrawal, Rahul, Nissen, Steven E., and Hurt-Camejo, Eva
- Subjects
BLOOD lipids ,MINERAL oils ,CORN oil ,PERMEABILITY ,ENDOTOXINS ,INTESTINES ,BIOMARKERS - Abstract
We investigated the effects of chronic oral administration of mineral oil, versus corn oil as control, on intestinal permeability, inflammatory markers, and plasma lipids in APOE*3-Leiden.CETP mice. Mice received mineral oil or corn oil 15 or 30 μL/mouse/day for 16 weeks (15 mice/group). Intestinal permeability was increased with mineral versus corn oil 30 µL/day, shown by increased mean plasma FITC-dextran concentrations 2 h post-administration (11 weeks: 1.5 versus 1.1 μg/ml, p = 0.02; 15 weeks: 1.7 versus 1.3 μg/ml, p = 0.08). Mean plasma lipopolysaccharide-binding protein levels were raised with mineral versus corn oil 30 µL/day (12 weeks: 5.8 versus 4.4 μg/ml, p = 0.03; 16 weeks: 5.8 versus 4.5 μg/ml, p = 0.09), indicating increased intestinal bacterial endotoxin absorption and potential pro-inflammatory effects. Plasma cholesterol and triglyceride concentrations were decreased with mineral oil, without affecting liver lipids among treated groups. Fecal neutral sterol measurements indicated increased fecal cholesterol excretion with mineral oil 30 µL/day (+16%; p = 0.04). Chronic oral administration of mineral oil in APOE*3-Leiden.CETP mice increased intestinal permeability, with potential pro-inflammatory effects, and decreased plasma cholesterol and triglyceride levels. Our findings may raise concerns about the use of mineral oil as a placebo in clinical studies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
16. Heterogeneous Expression of Cholesterol 7alpha-Hydroxylase and Sterol 27-Hydroxylase Genes in the Rat Liver Lobulus
- Author
-
Twisk, Jaap, Hoekman, Marco F. M., Mager, Willem H., Moorman, Antoon F. M., de Boer, Piet A. J., Scheja, Ludger, Princen, Hans M. G., and Gebhardt, Rolf.
- Published
- 1995
17. Systemic PFOS and PFOA exposure and disturbed lipid homeostasis in humans: what do we know and what not?
- Author
-
Fragki, Styliani, Dirven, Hubert, Fletcher, Tony, Grasl-Kraupp, Bettina, Bjerve Gützkow, Kristine, Hoogenboom, Ron, Kersten, Sander, Lindeman, Birgitte, Louisse, Jochem, Peijnenburg, Ad, Piersma, Aldert H., Princen, Hans M. G., Uhl, Maria, Westerhout, Joost, Zeilmaker, Marco J., and Luijten, Mirjam
- Subjects
HOMEOSTASIS ,BLOOD lipids ,BLOOD cholesterol ,LIPIDS ,LIPID metabolism ,LIVER cells ,NUCLEAR receptors (Biochemistry) - Abstract
Associations between per- and polyfluoroalkyl substances (PFASs) and increased blood lipids have been repeatedly observed in humans, but a causal relation has been debated. Rodent studies show reverse effects, i.e. decreased blood cholesterol and triglycerides, occurring however at PFAS serum levels at least 100-fold higher than those in humans. This paper aims to present the main issues regarding the modulation of lipid homeostasis by the two most common PFASs, PFOS and PFOA, with emphasis on the underlying mechanisms relevant for humans. Overall, the apparent contrast between human and animal data may be an artifact of dose, with different molecular pathways coming into play upon exposure to PFASs at very low versus high levels. Altogether, the interpretation of existing rodent data on PFOS/PFOA-induced lipid perturbations with respect to the human situation is complex. From a mechanistic perspective, research on human liver cells shows that PFOS/PFOA activate the PPARα pathway, whereas studies on the involvement of other nuclear receptors, like PXR, are less conclusive. Other data indicate that suppression of the nuclear receptor HNF4α signaling pathway, as well as perturbations of bile acid metabolism and transport might be important cellular events that require further investigation. Future studies with human-relevant test systems would help to obtain more insight into the mechanistic pathways pertinent for humans. These studies shall be designed with a careful consideration of appropriate dosing and toxicokinetics, so as to enable biologically plausible quantitative extrapolations. Such research will increase the understanding of possible perturbed lipid homeostasis related to PFOS/ PFOA exposure and the potential implications for human health. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
18. In Vivo Magnetic Resonance Imaging-Based Detection of Heterogeneous Endothelial Response in Thoracic and Abdominal Aorta to Short-Term High-Fat Diet Ascribed to Differences in Perivascular Adipose Tissue in Mice.
- Author
-
Bar, Anna, Kieronska-Rudek, Anna, Proniewski, Bartosz, Suraj-Prażmowska, Joanna, Czamara, Krzysztof, Marczyk, Brygida, Matyjaszczyk-Gwarda, Karolina, Jasztal, Agnieszka, Kuś, Edyta, Majka, Zuzanna, Kaczor, Agnieszka, Kurpińska, Anna, Walczak, Maria, Pieterman, Elsbet J., Princen, Hans M. G., and Chlopicki, Stefan
- Published
- 2020
- Full Text
- View/download PDF
19. Dual targeting of hepatic fibrosis and atherogenesis by icosabutate, an engineered eicosapentaenoic acid derivative.
- Author
-
Stokman, Geurt, Hoek, Anita M., Denker Thorbekk, Ditte, Pieterman, Elsbet J., Skovgård Veidal, Sanne, Basta, Brittany, Iruarrizaga‐Lejarreta, Marta, Hoorn, José W., Verschuren, Lars, Berbée, Jimmy F. P., Rensen, Patrick C. N., Skjæret, Tore, Alonso, Cristina, Feigh, Michael, Kastelein, John J. P., Friedman, Scott L., Princen, Hans M. G., and Fraser, David A.
- Subjects
HEPATIC fibrosis ,EICOSAPENTAENOIC acid ,ACID derivatives ,ARACHIDONIC acid ,BLOOD lipids ,CARDIOVASCULAR diseases ,ATHEROSCLEROSIS - Abstract
Background & Aims: While fibrosis stage predicts liver‐associated mortality, cardiovascular disease (CVD) is still the major overall cause of mortality in patients with NASH. Novel NASH drugs should thus ideally reduce both liver fibrosis and CVD. Icosabutate is a semi‐synthetic, liver‐targeted eicosapentaenoic acid (EPA) derivative in clinical development for NASH. The primary aims of the current studies were to establish both the anti‐fibrotic and anti‐atherogenic efficacy of icosabutate in conjunction with changes in lipotoxic and atherogenic lipids in liver and plasma respectively. Methods: The effects of icosabutate on fibrosis progression and lipotoxicity were investigated in amylin liver NASH (AMLN) diet (high fat, cholesterol and fructose) fed ob/ob mice with biopsy‐confirmed steatohepatitis and fibrosis and compared with the activity of obeticholic acid. APOE*3Leiden.CETP mice, a translational model for hyperlipidaemia and atherosclerosis, were used to evaluate the mechanisms underlying the lipid‐lowering effect of icosabutate and its effect on atherosclerosis. Results: In AMLN ob/ob mice, icosabutate significantly reduced hepatic fibrosis and myofibroblast content in association with downregulation of the arachidonic acid cascade and a reduction in both hepatic oxidised phospholipids and apoptosis. In APOE*3Leiden.CETP mice, icosabutate reduced plasma cholesterol and TAG levels via increased hepatic uptake, upregulated hepatic lipid metabolism and downregulated inflammation pathways, and effectively decreased atherosclerosis development. Conclusions: Icosabutate, a structurally engineered EPA derivative, effectively attenuates both hepatic fibrosis and atherogenesis and offers an attractive therapeutic approach to both liver‐ and CV‐related morbidity and mortality in NASH patients. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
20. Salsalate attenuates diet induced non-alcoholic steatohepatitis in mice by decreasing lipogenic and inflammatory processes
- Author
-
Liang, Wen, Verschuren, Lars, Mulder, Petra, van der Hoorn, José W. A., Verheij, Joanne, van Dam, Andrea D., Boon, Mariette R., Princen, Hans M. G., Havekes, Louis M., Kleemann, Robert, van den Hoek, Anita M., and Pathology
- Abstract
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. 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. 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. 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
- Published
- 2015
21. Oncostatin M reduces atherosclerosis development in APOE*3Leiden.CETP mice and is associated with increased survival probability in humans.
- Author
-
Keulen, Danielle van, Pouwer, Marianne G., Emilsson, Valur, Matic, Ljubica Perisic, Pieterman, Elsbet J., Hedin, Ulf, Gudnason, Vilmundur, Jennings, Lori L., Holmstrøm, Kim, Nielsen, Boye Schnack, Pasterkamp, Gerard, Lindeman, Jan H. N., van Gool, Alain J., Sollewijn Gelpke, Maarten D., Princen, Hans M. G., and Tempel, Dennie
- Subjects
LEUKEMIA inhibitory factor ,ONCOSTATIN M ,IN situ hybridization ,ATHEROSCLEROSIS ,ATHEROSCLEROTIC plaque ,CAROTID artery ,MICE - Abstract
Objective: Previous studies indicate a role for Oncostatin M (OSM) in atherosclerosis and other chronic inflammatory diseases for which inhibitory antibodies are in development. However, to date no intervention studies with OSM have been performed, and its relation to coronary heart disease (CHD) has not been studied. Approach and results: Gene expression analysis on human normal arteries (n = 10) and late stage/advanced carotid atherosclerotic arteries (n = 127) and in situ hybridization on early human plaques (n = 9) showed that OSM, and its receptors, OSM receptor (OSMR) and Leukemia Inhibitory Factor Receptor (LIFR) are expressed in normal arteries and atherosclerotic plaques. Chronic OSM administration in APOE*3Leiden.CETP mice (n = 15/group) increased plasma E-selectin levels and monocyte adhesion to the activated endothelium independently of cholesterol but reduced the amount of inflammatory Ly-6C
High monocytes and atherosclerotic lesion size and severity. Using aptamer-based proteomics profiling assays high circulating OSM levels were shown to correlate with post incident CHD survival probability in the AGES-Reykjavik study (n = 5457). Conclusions: Chronic OSM administration in APOE*3Leiden.CETP mice reduced atherosclerosis development. In line, higher serum OSM levels were correlated with improved post incident CHD survival probability in patients, suggesting a protective cardiovascular effect. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
22. Dose Effects of Ammonium Perfluorooctanoate on Lipoprotein Metabolism in APOE*3-Leiden.CETP Mice.
- Author
-
Pouwer, Marianne G, Pieterman, Elsbet J, Chang, Shu-Ching, Olsen, Geary W, Caspers, Martien P M, Verschuren, Lars, Jukema, J Wouter, and Princen, Hans M G
- Subjects
CHOLESTERYL ester transfer protein ,BLOOD lipids ,PEROXISOME proliferator-activated receptors ,CHOLESTEROL metabolism ,LIPOPROTEIN lipase ,PERFLUOROOCTANOIC acid - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
23. The APOE∗3-Leiden Heterozygous Glucokinase Knockout Mouse as Novel Translational Disease Model for Type 2 Diabetes, Dyslipidemia, and Diabetic Atherosclerosis.
- Author
-
Pouwer, Marianne G., Heinonen, Suvi E., Behrendt, Margareta, Andréasson, Anne-Christine, van Koppen, Arianne, Menke, Aswin L., Pieterman, Elsbet J., van den Hoek, Anita M., Jukema, J. Wouter, Leighton, Brendan, Jönsson-Rylander, Ann-Cathrine, and Princen, Hans M. G.
- Subjects
DYSLIPIDEMIA ,TYPE 2 diabetes ,MEDICAL model ,GLUCOKINASE ,KNOCKOUT mice ,ATHEROSCLEROSIS - Abstract
Background. There is a lack of predictive preclinical animal models combining atherosclerosis and type 2 diabetes. APOE∗3-Leiden (E3L) mice are a well-established model for diet-induced hyperlipidemia and atherosclerosis, and glucokinase
+/− (GK+/− ) mice are a translatable disease model for glucose control in type 2 diabetes. The respective mice respond similarly to lipid-lowering and antidiabetic drugs as humans. The objective of this study was to evaluate/characterize the APOE∗3-Leiden.glucokinase+/− (E3L.GK+/− ) mouse as a novel disease model to study the metabolic syndrome and diabetic complications. Methods. Female E3L.GK+/− , E3L, and GK+/− mice were fed fat- and cholesterol-containing diets for 37 weeks, and plasma parameters were measured throughout. Development of diabetic macro- and microvascular complications was evaluated. Results. Cholesterol and triglyceride levels were significantly elevated in E3L and E3L.GK+/− mice compared to GK+/− mice, whereas fasting glucose was significantly increased in E3L.GK+/− and GK+/− mice compared to E3L. Atherosclerotic lesion size was increased 2.2-fold in E3L.GK+/− mice as compared to E3L (p=0.037), which was predicted by glucose exposure (R2 =0.636, p=0.001). E3L and E3L.GK+/− mice developed NASH with severe inflammation and fibrosis which, however, was not altered by introduction of the defective GK phenotype, whereas mild kidney pathology with tubular vacuolization was present in all three phenotypes. Conclusions. We conclude that the E3L.GK+/− mouse is a promising novel diet-inducible disease model for investigation of the etiology and evaluation of drug treatment on diabetic atherosclerosis. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
24. Inhibition and induction of bile acid synthesis by ketoconazole effects on bile formation in the rat
- Author
-
Kuipers, Folkert, Havinga, Rick, Huijsmans, Christine M. G., Vonk, Roel J., and Princen, Hans M. G.
- Published
- 1989
- Full Text
- View/download PDF
25. Free cytoplasmic messenger ribonucleoprotein complexes from rabbit reticulocytes
- Author
-
Princen, Hans M. G., van Eekelen, Chris A. G., Asselbergs, Fred A. M., and van Venrooij, Walther J.
- Published
- 1979
- Full Text
- View/download PDF
26. Inflammatory cytokine oncostatin M induces endothelial activation in macro- and microvascular endothelial cells and in APOE*3Leiden.CETP mice.
- Author
-
van Keulen, Danielle, Pouwer, Marianne G., Pasterkamp, Gerard, van Gool, Alain J., Sollewijn Gelpke, Maarten D., Princen, Hans M. G., and Tempel, Dennie
- Subjects
CYTOKINES ,ONCOSTATIN M ,ENDOTHELIAL cells ,APOLIPOPROTEIN E ,ATHEROSCLEROSIS ,PHOSPHORYLATION ,LABORATORY mice - Abstract
Aims: Endothelial activation is involved in many chronic inflammatory diseases, such as atherosclerosis, and is often initiated by cytokines. Oncostatin M (OSM) is a relatively unknown cytokine that has been suggested to play a role in both endothelial activation and atherosclerosis. We comprehensively investigated the effect of OSM on endothelial cell activation from different vascular beds and in APOE*3Leiden.CETP mice. Methods and results: Human umbilical vein endothelial cells, human aortic endothelial cells and human microvascular endothelial cells cultured in the presence of OSM express elevated MCP-1, IL-6 and ICAM-1 mRNA levels. Human umbilical vein endothelial cells and human aortic endothelial cells additionally expressed increased VCAM-1 and E-selectin mRNA levels. Moreover, ICAM-1 membrane expression is increased as well as MCP-1, IL-6 and E-selectin protein release. A marked increase was observed in STAT1 and STAT3 phosphorylation indicating that the JAK/STAT pathway is involved in OSM signaling. OSM signals through the LIF receptor alfa (LIFR) and the OSM receptor (OSMR). siRNA knockdown of the LIFR and the OSMR revealed that simultaneous knockdown is necessary to significantly reduce MCP-1 and IL-6 secretion, VCAM-1 and E-selectin shedding and STAT1 and STAT3 phosphorylation after OSM stimulation. Moreover, OSM administration to APOE*3Leiden.CETP mice enhances plasma E-selectin levels and increases ICAM-1 expression and monocyte adhesion in the aortic root area. Furthermore, Il-6 mRNA expression was elevated in the aorta of OSM treated mice. Conclusion: OSM induces endothelial activation in vitro in endothelial cells from different vascular beds through activation of the JAK/STAT cascade and in vivo in APOE*3Leiden.CETP mice. Since endothelial activation is an initial step in atherosclerosis development, OSM may play a role in the initiation of atherosclerotic lesion formation. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
27. Results, meta-analysis and a first evaluation of UNOxR, the urinary nitrate-to-nitrite molar ratio, as a measure of nitrite reabsorption in experimental and clinical settings.
- Author
-
Tsikas, Dimitrios, Hanff, Erik, Bollenbach, Alexander, Kruger, Ruan, Pham, Vu Vi, Chobanyan-Jürgens, Kristine, Wedekind, Dirk, Arndt, Tanja, Jörns, Anne, Berbée, Jimmy F. P., Princen, Hans M. G., Lücke, Thomas, Mariotti, François, Huneau, Jean-François, Ückert, Stefan, Frölich, Jürgen C., and Lenzen, Sigurd
- Subjects
DIABETES ,DRUG therapy ,MASS spectrometry ,NITRIC oxide ,CARBONIC anhydrase - Abstract
We recently found that renal carbonic anhydrase (CA) is involved in the reabsorption of inorganic nitrite (NO
2 − ), 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 N2 O3 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., UNOx R, 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 UNOx R. We determined UNOx R 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 UNOx R values in healthy young men who ingested isosorbide dinitrate (ISDN), pentaerythrityl tetranitrate (PETN), or inorganic nitrate. In addition, we tested the utility of UNOx R 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 UNOx R 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 UNOx R were of the order of 50% in young and elderly healthy subjects. UNOx R 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 UNOx R 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 UNOx R 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), UNOx R was higher than in those who ingested the same dose of NaCl (1709 vs. 369). In LEW.1AR1-iddm rats, mean UNOx R values were lower than in healthy rats (198 vs. 308) and comparable to those in APOE*3-Leiden.CETP mice (151). [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
28. The AT04A vaccine against proprotein convertase subtilisin/kexin type 9 reduces total cholesterol, vascular inflammation, and atherosclerosis in APOE*3Leiden.CETP mice.
- Author
-
Landlinger, Christine, Pouwer, Marianne G., Juno, Claudia, van der Hoorn, José W. A., Pieterman, Elsbet J., Jukema, J. Wouter, Staffler, Guenther, Princen, Hans M. G., and Galabova, Gergana
- Abstract
Aims: Proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as a promising therapeutic target for the treatment of hypercholesterolaemia and atherosclerosis. PCSK9 binds to the low density lipoprotein receptor and enhances its degradation, which leads to the reduced clearance of low density lipoprotein cholesterol (LDLc) and a higher risk of atherosclerosis. In this study, the AT04A anti-PCSK9 vaccine was evaluated for its therapeutic potential in ameliorating or even preventing coronary heart disease in the atherogenic APOE*3Leiden.CETP mouse model. Methods and results: Control and AT04A vaccine-treated mice were fed western-type diet for 18weeks. Antibody titres, plasma lipids, and inflammatory markers were monitored by ELISA, FPLC, and multiplexed immunoassay, respectively. The progression of atherosclerosis was evaluated by histological analysis of serial cross-sections from the aortic sinus. The AT04A vaccine induced high and persistent antibody levels against PCSK9, causing a significant reduction in plasma total cholesterol (-53%, P< 0.001) and LDLc compared with controls. Plasma inflammatory markers such as serum amyloid A (SAA), macrophage inflammatory protein-1β (MIP-1β/CCL4), macrophage-derived chemokine (MDC/CCL22), cytokine stem cell factor (SCF), and vascular endothelial growth factor A (VEGF-A) were significantly diminished in AT04A-treated mice. As a consequence, treatment with the AT04A vaccine resulted in a decrease in atherosclerotic lesion area (-64%, P= 0.004) and aortic inflammation as well as in more lesion-free aortic segments (+119%, P= 0.026), compared with control. Conclusions: AT04A vaccine induces an effective immune response against PCSK9 in APOE*3Leiden.CETP mice, leading to a significant reduction of plasma lipids, systemic and vascular inflammation, and atherosclerotic lesions in the aorta. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
29. Metformin Lowers Plasma Triglycerides by Promoting VLDL-Triglyceride Clearance by Brown Adipose Tissue in Mice.
- Author
-
Geerling, Janine J., Boon, Mariëtte R., van der Zon, Gerard C., den Berg, Sjoerd A. A. van, den Hoek, Anita M. van, Lombès, Marc, Princen, Hans M. G., Havekes, Louis M., Rensen, Patrick C. N., and Guigas, Bruno
- Subjects
TYPE 2 diabetes treatment ,BROWN adipose tissue ,TRIGLYCERIDES ,LIPOPROTEINS ,LABORATORY mice - Abstract
Metformin is the first-line drug for the treatment of type 2 diabetes. Besides its well-characterized antihyperglycemic properties, metformin also lowers plasma VLDL triglyceride (TG). In this study, we investigated the underlying mechanisms in APOE*3-Leiden.CETP mice, a well-established model for human-like lipoprotein metabolism. We found that metformin markedly lowered plasma total cholesterol and TG levels, an effect mostly due to a decrease in VLDL-TG, whereas HDL was slightly increased. Strikingly, metformin did not affect hepatic VLDL-TG production, VLDL particle composition, and hepatic lipid composition but selectively enhanced clearance of glycerol tri[³H]oleate-labeled VLDL-like emulsion particles into brown adipose tissue (BAT). BAT mass and lipid droplet content were reduced in metformin-treated mice, pointing to increased BAT activation. In addition, both AMP-activated protein kinase α1 (AMPKα1) expression and activity and HSL and mitochondrial content were increased in BAT. Furthermore, therapeutic concentrations of metformin increased AMPK and HSL activities and promoted lipolysis in T37i differentiated brown adipocytes. Collectively, our results identify BAT as an important player in the TG-lowering effect of metformin by enhancing VLDL-TG uptake, intracellular TG lipolysis, and subsequent mitochondrial fatty acid oxidation. Targeting BAT might therefore be considered as a future therapeutic strategy for the treatment of dyslipidemia. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
30. Niacin Reduces Atherosclerosis Development in APOE*3Leiden.CETP Mice Mainly by Reducing NonHDL-Cholesterol.
- Author
-
Kühnast, Susan, Louwe, Mieke C., Heemskerk, Mattijs M., Pieterman, Elsbet J., van Klinken, Jan B., van den Berg, Sjoerd A. A., Smit, Johannes W. A., Havekes, Louis M., Rensen, Patrick C. N., van der Hoorn, José W. A., Princen, Hans M. G., and Jukema, J. Wouter
- Subjects
NIACIN ,ATHEROSCLEROSIS ,APOLIPOPROTEIN E ,LABORATORY mice ,CHOLESTEROL ,TRIGLYCERIDES ,HIGH density lipoproteins - Abstract
Objective: Niacin potently lowers triglycerides, mildly decreases LDL-cholesterol, and largely increases HDL-cholesterol. Despite evidence for an atheroprotective effect of niacin from previous small clinical studies, the large outcome trials, AIM-HIGH and HPS2-THRIVE did not reveal additional beneficial effects of niacin (alone or in combination with laropiprant) on top of statin treatment. We aimed to address this apparent discrepancy by investigating the effects of niacin without and with simvastatin on atherosclerosis development and determine the underlying mechanisms, in APOE*3Leiden.CETP mice, a model for familial dysbetalipoproteinemia (FD). Approach and Results: Mice were fed a western-type diet containing cholesterol without or with niacin (120 mg/kg/day), simvastatin (36 mg/kg/day) or their combination for 18 weeks. Similarly as in FD patients, niacin reduced total cholesterol by -39% and triglycerides by −50%, (both P<0.001). Simvastatin and the combination reduced total cholesterol (−30%; −55%, P<0.001) where the combination revealed a greater reduction compared to simvastatin (−36%, P<0.001). Niacin decreased total cholesterol and triglycerides primarily by increasing VLDL clearance. Niacin increased HDL-cholesterol (+28%, P<0.01) and mildly increased reverse cholesterol transport. All treatments reduced monocyte adhesion to the endothelium (−46%; −47%, P<0.01; −53%, P<0.001), atherosclerotic lesion area (−78%; −49%, P<0.01; −87%, P<0.001) and severity. Compared to simvastatin, the combination increased plaque stability index [(SMC+collagen)/macrophages] (3-fold, P<0.01). Niacin and the combination reduced T cells in the aortic root (−71%, P<0.01; −81%, P<0.001). Lesion area was strongly predicted by nonHDL-cholesterol (R
2 = 0.69, P<0.001) and to a much lesser extent by HDL-cholesterol (R2 = 0.20, P<0.001). Conclusion: Niacin decreases atherosclerosis development mainly by reducing nonHDL-cholesterol with modest HDL-cholesterol-raising and additional anti-inflammatory effects. The additive effect of niacin on top of simvastatin is mostly dependent on its nonHDL-cholesterol-lowering capacities. These data suggest that clinical beneficial effects of niacin are largely dependent on its ability to lower LDL-cholesterol on top of concomitant lipid-lowering therapy. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
31. CYP7A1 A-278C polymorphism affects the response of plasma lipids after dietary cholesterol or cafestol interventions in humans.
- Author
-
Hofman, Maaike K., Weggemans, Rianne M., Zock, Peter L., Schouten, Evert G., Katan, Martijn B., and Princen, Hans M. G.
- Subjects
BLOOD lipids ,BLOOD cholesterol ,GENETIC polymorphisms ,NUTRITION ,HIGH density lipoproteins ,BLOOD lipoproteins ,ALLELES ,CHOLESTEROL ,CLINICAL trials ,COMPARATIVE studies ,DOSE-effect relationship in pharmacology ,CHOLESTEROL content of food ,GENES ,HETEROCYCLIC compounds ,HYDROCARBONS ,LIPIDS ,RESEARCH methodology ,MEDICAL cooperation ,META-analysis ,OXIDOREDUCTASES ,PURINES ,RESEARCH ,EVALUATION research ,GENOTYPES - Abstract
The response of plasma lipids to dietary cholesterol and fat varies among individuals. Variations in genes involved in cholesterol metabolism can be important in these interindividual differences. The rate-limiting enzyme in the conversion of cholesterol into bile acids is cholesterol 7alpha-hydroxylase (CYP7A1). We investigated the effect of the A278-C promoter polymorphism in the CYP7A1 gene on responses of plasma lipids to an increased intake in dietary cholesterol (742 +/- 114 mg/d), cafestol (57 +/- 6 mg/d), saturated fat [change of 8-9 energy percent/d (en%/d)] and trans fat (change of 10-11 en%/d) in 496 normolipidemic subjects. These responses were measured in 26 previously published dietary trials. After adjustment for the apolipoprotein E genotype effect, AA-subjects consuming a cholesterol-rich diet had a smaller increase in plasma HDL cholesterol than CC-subjects (0.00 +/- 0.02 vs. 0.17 +/- 0.04 mmol/L; P < 0.001). Upon intake of cafestol, AA-subjects had a smaller increase in plasma total cholesterol than CC-subjects (0.69 +/- 0.10 vs. 1.01 +/- 0.10 mmol/L; P = 0.028). No effects of the polymorphism were found in the saturated and trans fat interventions. In conclusion, the CYP7A1 polymorphism has a small but significant effect on the increase in plasma HDL cholesterol and plasma total cholesterol after an increased intake of dietary cholesterol and cafestol, respectively. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
32. Acyl-coenzyme A: Cholesterol acyltransferase inhibitor, avasimibe, stimulates bile acid synthesis and cholesterol 7α-hydroxylase in cultured rat hepatocytes and in vivo in the rat.
- Author
-
Post, Sabine M., Zoeteweij, J. Paul, Bos, Mettine H., de Wit, Elly C., Havinga, Rick, Kuipers, Folkert, and Princen, Hans M. G.
- Published
- 1999
- Full Text
- View/download PDF
33. Bile acids exert negative feedback control on bile acid synthesis in cultured pig hepatocytes by suppression of cholesterol 7α-hydroxylase activity.
- Author
-
Kwekkeboom, Jaap, Princen, Hans M. G., van Voorthuizen, Eline M., and Kempen, Herman Jan M.
- Published
- 1990
- Full Text
- View/download PDF
34. Regulation of low density lipoprotein receptor activity in primary cultures of human hepatocytes by serum lipoproteins.
- Author
-
Havekes, Louis M., Verboom, Hans, de Wit, Elly, Yap, Sing Hiem, and Princen, Hans M. G.
- Published
- 1986
- Full Text
- View/download PDF
35. Insulin suppresses bile acid synthesis in cultured rat hepatocytes by down-regulation of cholesterol 7α-hydroxylase and sterol 27-hydroxylase gene transcription.
- Author
-
Twisk, Jaap, Hoekman, Marco F. M., Lehmann, Eline M., Meijer, Piet, Mager, Willem H., and Princen, Hans M. G.
- Published
- 1995
- Full Text
- View/download PDF
36. Fenofibrate Increases Very Low Density Lipoprotein Triglyceride Production Despite Reducing Plasma Triglyceride Levels in APOE*3-Leiden.CETP Mice.
- Author
-
Bijland, Silvia, Pieterman, Elsbet J., Maas, Annemarie C. E., Van der Hoorn, José W. A., Van Erk, Marjan J., Van Klinken, Jan B., Havekes, Louis M., Van Dijk, Ko Willems, Princen, Hans M. G., and Rensen, Patrick C. N.
- Subjects
- *
PEROXISOMES , *LOW density lipoproteins , *LABORATORY mice , *FENOFIBRATE , *TRIGLYCERIDES - Abstract
The peroxisome proliferator-activated receptor alpha (PPARα) activator fenofibrate efficiently decreases plasma triglycerides (TG), which is generally attributed to enhanced very low density lipoprotein (VLDL)-TG clearance and decreased VLDL-TG production. However, because data on the effect of fenofibrate on VLDL production are controversial, we aimed to investigate in (more) detail the mechanism underlying the TG-lowering effect by studying VLDL-TG production and clearance using APOE*3-Leiden.CETP mice, a unique mouse model for human-like lipoprotein metabolism. Male mice were fed a Western-type diet for 4 weeks, followed by the same diet without or with fenofibrate (30 mg/kg bodyweight/day) for 4 weeks. Fenofibrate strongly lowered plasma cholesterol (-38%) and TG (-60%) caused by reduction of VLDL. Fenofibrate markedly accelerated VLDL-TG clearance, as judged from a reduced plasma half-life of glycerol tri[³H]oleate-labeled VLDL-like emulsion particles (-68%). This was associated with an increased post-heparin lipoprotein lipase (LPL) activity (+110%) and an increased uptake of VLDL-derived fatty acids by skeletal muscle, white adipose tissue, and liver. Concomitantly, fenofibrate markedly increased the VLDL-TG production rate (+73%) but not the VLDL-apolipoprotein B (apoB) production rate. Kinetic studies using [³H] palmitic acid showed that fenofibrate increased VLDL-TG production by equally increasing incorporation of re-esterified plasma fatty acids and liver TG into VLDL, which was supported by hepatic gene expression profiling data. We conclude that fenofibrate decreases plasma TG by enhancing LPL-mediated VLDL-TG clearance, which results in a compensatory increase in VLDL-TG production by the liver. ] [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
37. Anacetrapib reduces progression of atherosclerosis, mainly by reducing non-HDL-cholesterol, improves lesion stability and adds to the beneficial effects of atorvastatin
- Author
-
Elsbet J. Pieterman, Ko Willems van Dijk, Sam van der Tuin, Susan Kühnast, Jan B. van Klinken, Thomas F. Lüscher, Louis M. Havekes, Patrick C.N. Rensen, J. Wouter Jukema, Branko Simic, Ulf Landmesser, José W.A. van der Hoorn, Hans M.G. Princen, University of Zurich, and Princen, Hans M G
- Subjects
Mouse ,Atorvastatin ,Apolipoprotein E3 ,Biomedical Innovation ,Pharmacology ,Efficacy ,In vivo study ,chemistry.chemical_compound ,Basic Science ,Life ,Anacetrapib ,Enzyme activity ,High density lipoprotein cholesterol ,Atherosclerotic plaque ,biology ,Anticholesteremic Agents ,Cholesteryl ester transfer protein ,HDL-cholesterol ,Drug Combinations ,Cholesterol blood level ,Disease Progression ,10209 Clinic for Cardiology ,Aorta atherosclerosis ,Female ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Animal cell ,MHR - Metabolic Health Research ,Cardiology and Cardiovascular Medicine ,Healthy Living ,medicine.drug ,HDL function ,Dose ,Drug potentiation ,Mice, Transgenic ,610 Medicine & health ,Non-HDL-cholesterol ,2705 Cardiology and Cardiovascular Medicine ,Treatment duration ,Lesion ,Cholesterylester transfer protein ,medicine ,Animals ,Pyrroles ,Animal model ,Biology ,Disease severity ,CETP inhibitor ,Oxazolidinones ,Serum Amyloid A Protein ,Cholesterol ,business.industry ,Cholesterol, HDL ,nutritional and metabolic diseases ,Nonhuman ,Atherosclerosis ,Cholesterol Ester Transfer Proteins ,Diet ,Drug efficacy ,chemistry ,Heptanoic Acids ,Analysis of covariance ,Risk reduction ,biology.protein ,ELSS - Earth, Life and Social Sciences ,business ,Controlled study ,Aorta root - Abstract
The present study is the first intervention study in a well-established, translational mouse model for hyperlipidaemia and atherosclerosis showing that anacetrapib dose-dependently reduces atherosclerosis development and adds to the anti-atherogenic effects of atorvastatin. This effect is mainly ascribed to the reduction in non-HDL-C despite a remarkable increase in HDL-C and without affecting HDL functionality. In addition, anacetrapib improves lesion stability., 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.
- Published
- 2015
38. Therapeutic effects of FGF21 mimetic bFKB1 on MASH and atherosclerosis in Ldlr-/-.Leiden mice.
- Author
-
Inia JA, Attema J, de Ruiter C, Menke AL, Caspers MPM, Verschuren L, Wilson M, Arlantico A, Brightbill HD, Jukema JW, van den Hoek AM, Princen HMG, Chen MZ, and Morrison MC
- Subjects
- Animals, Mice, Male, Diet, High-Fat adverse effects, Liver metabolism, Liver pathology, Liver drug effects, Atherosclerosis metabolism, Atherosclerosis drug therapy, Atherosclerosis genetics, Atherosclerosis pathology, Fibroblast Growth Factors metabolism, Fibroblast Growth Factors genetics, Fatty Liver drug therapy, Fatty Liver metabolism, Fatty Liver pathology, Mice, Knockout, Receptors, LDL genetics, Receptors, LDL metabolism
- Abstract
Fibroblast growth factor 21 (FGF21) is a promising target for treatment of obesity-associated diseases including metabolic dysfunction-associated steatohepatitis (MASH) and atherosclerosis. We evaluated the effects of the bispecific anti-FGF21-β klotho (KLB) agonist antibody bFKB1 in a preclinical model of MASH and atherosclerosis. Low-density lipoprotein receptor knockout (Ldlr-/-).Leiden mice received a high-fat diet for 20 weeks, followed by treatment with an isotype control antibody or bFKB1 for 12 weeks. Effects on plasma risk markers and (histo)pathology of liver, adipose tissue, and heart were evaluated alongside hepatic transcriptomics analysis. bFKB1 lowered body weight (-21%) and adipose tissue mass (-22%) without reducing food intake. The treatment also improved plasma insulin (-80%), cholesterol (-48%), triglycerides (-76%), alanine transaminase (ALT: -79%), and liver weight (-43%). Hepatic steatosis and inflammation were strongly reduced (macrovesicular steatosis -34%; microvesicular steatosis -100%; inflammation -74%) and while the total amount of fibrosis was not affected, bFKB1 did decrease new collagen formation (-49%). Correspondingly, hepatic transcriptomics and pathway analysis revealed the mechanistic background underlying these histological improvements, demonstrating broad inactivation of inflammatory and profibrotic transcriptional programs by bFKB1. In epididymal white adipose tissue, bFKB1 reduced adipocyte size (-16%) and inflammation (-52%) and induced browning, signified by increased uncoupling protein-1 (UCP1) protein expression (8.5-fold increase). In the vasculature, bFKB1 had anti-atherogenic effects, lowering total atherosclerotic lesion area (-38%). bFKB1 has strong beneficial metabolic effects associated with a reduction in hepatic steatosis, inflammation, and atherosclerosis. Analysis of new collagen formation and profibrotic transcriptional programs indicate that bFKB1 treatment may have antifibrotic potential in a longer treatment duration as well., (© 2024 The Author(s). The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)
- Published
- 2024
- Full Text
- View/download PDF
39. A Novel, Orally Bioavailable, Small-Molecule Inhibitor of PCSK9 With Significant Cholesterol-Lowering Properties In Vivo.
- Author
-
Suchowerska AK, Stokman G, Palmer JT, Coghlan PA, Pieterman EJ, Keijzer N, Lambert G, Chemello K, Jaafar AK, Parmar J, Yan L, Tong Y, Mu L, Princen HMG, Bonnar J, and Evison BJ
- Subjects
- Animals, Humans, Mice, Apolipoproteins E, Cholesterol, Cholesterol, LDL, Receptors, LDL genetics, Receptors, LDL metabolism, PCSK9 Inhibitors pharmacology, Hyperlipidemias drug therapy, Anticholesteremic Agents pharmacology
- Abstract
Proprotein convertase subtilisin kexin type 9 (PCSK9) inhibits the clearance of low-density lipoprotein (LDL) cholesterol (LDL-C) from plasma by directly binding with the LDL receptor (LDLR) and sending the receptor for lysosomal degradation. As the interaction promotes elevated plasma LDL-C levels, and therefore a predisposition to cardiovascular disease, PCSK9 has attracted intense interest as a therapeutic target. Despite this interest, an orally bioavailable small-molecule inhibitor of PCSK9 with extensive lipid-lowering activity is yet to enter the clinic. We report herein the discovery of NYX-PCSK9i, an orally bioavailable small-molecule inhibitor of PCSK9 with significant cholesterol-lowering activity in hyperlipidemic APOE∗3-Leiden.CETP mice. NYX-PCSK9i emerged from a medicinal chemistry campaign demonstrating potent disruption of the PCSK9-LDLR interaction in vitro and functional protection of the LDLR of human lymphocytes from PCSK9-directed degradation ex vivo. APOE∗3-Leiden.CETP mice orally treated with NYX-PCSK9i demonstrated a dose-dependent decrease in plasma total cholesterol of up to 57%, while its combination with atorvastatin additively suppressed plasma total cholesterol levels. Importantly, the majority of cholesterol lowering by NYX-PCSK9i was in non-HDL fractions. A concomitant increase in total plasma PCSK9 levels and significant increase in hepatic LDLR protein expression strongly indicated on-target function by NYX-PCSK9i. Determinations of hepatic lipid and fecal cholesterol content demonstrated depletion of liver cholesteryl esters and promotion of fecal cholesterol elimination with NYX-PCSK9i treatment. All measured in vivo biomarkers of health indicate that NYX-PCSK9i has a good safety profile. NYX-PCSK9i is a potential new therapy for hypercholesterolemia with the capacity to further enhance the lipid-lowering activities of statins., Competing Interests: Conflict of Interest A. K. S., J. P., J. B., and B. J. E. are employees of Nyrada Inc. J. T. P. and G. L. are members of the Scientific Advisory Board of Nyrada Inc. J. T. P., G. L., J. B., and B. J. E. have share/stock options in Nyrada Inc. A. K. S., P. A. C., J. T. P., J. B., and B. J. E. are listed as inventors on the patent that discloses NYX-PCSK9i and related compounds. G. S., P. A. C., E. J. P., N. K., G. L., K. C., A. K. J., and H. M. G. P. are employees or members of their respective companies or laboratories who were contracted and/or consulted by Nyrada Inc. to perform specific research activities., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
40. Effects of mineral oil administration on the pharmacokinetics, metabolism and pharmacodynamics of atorvastatin and pravastatin in mice and dogs.
- Author
-
Gopaul VS, Pieterman EJ, Princen HMG, Bergenholm L, Lundborg E, Cavallin A, Johansson MJ, Hawthorne G, Björkbom A, Hammarberg M, Li X, Jarke A, Bright J, Svensson L, Jansson-Löfmark R, Abrahamsson B, Agrawal R, and Hurt-Camejo E
- Subjects
- Animals, Atorvastatin, Dogs, Mice, Mice, Inbred C57BL, Mineral Oil, Pravastatin, Pyrroles, Heptanoic Acids, Hydroxymethylglutaryl-CoA Reductase Inhibitors
- Abstract
We investigated the effects of mineral oil on statin pharmacokinetics and inflammatory markers in animal models. A new synthesis strategy produced regioisomers that facilitated the characterization of the main metabolite (M1) of atorvastatin, a lipophilic statin, in C57BL/6NCrl mice. The chemical structure of M1 in mice was confirmed as ortho-hydroxy β-oxidized atorvastatin. Atorvastatin and M1 pharmacokinetics and inflammatory markers were assessed in C57BL6/J mice given atorvastatin 5 mg/kg/day or 10 mg/kg/day, as a single dose or for 21 days, with or without 10 µL or 30 µL mineral oil. No consistent differences in plasma exposure of atorvastatin or M1 were observed in mice after single or repeat dosing of atorvastatin with or without mineral oil. However, mice administered atorvastatin 10 mg/kg with 30 µL mineral oil for 21 days had significantly increased plasma levels of serum amyloid A (mean 9.6 µg/mL vs 7.9 µg/mL without mineral oil; p < 0.01) and significantly increased proportions of C62L
high B cells (mean 18% vs 12% without mineral oil; p = 0.04). There were no statistically significant differences for other inflammatory markers assessed. In dogs, pharmacokinetics of atorvastatin, its two hydroxy metabolites and pravastatin (a hydrophilic statin) were evaluated after single administration of atorvastatin 10 mg plus pravastatin 40 mg with or without 2 g mineral oil. Pharmacokinetics of atorvastatin, hydroxylated atorvastatin metabolites or pravastatin were not significantly different after single dosing with or without mineral oil in dogs. Collectively, the results in mice and dogs indicate that mineral oil does not affect atorvastatin or pravastatin pharmacokinetics, but could cause low-grade inflammation with chronic oral administration, which warrants further investigation., (Copyright © 2021. Published by Elsevier B.V.)- Published
- 2021
- Full Text
- View/download PDF
41. Common Variants Associated With OSMR Expression Contribute to Carotid Plaque Vulnerability, but Not to Cardiovascular Disease in Humans.
- Author
-
van Keulen D, van Koeverden ID, Boltjes A, Princen HMG, van Gool AJ, de Borst GJ, Asselbergs FW, Tempel D, Pasterkamp G, and van der Laan SW
- Abstract
Background and Aims: Oncostatin M (OSM) signaling is implicated in atherosclerosis, however the mechanism remains unclear. We investigated the impact of common genetic variants in OSM and its receptors, OSMR and LIFR , on overall plaque vulnerability, plaque phenotype, intraplaque OSMR and LIFR expression, coronary artery calcification burden and cardiovascular disease susceptibility. Methods and Results: We queried Genotype-Tissue Expression data and found that rs13168867 (C allele) was associated with decreased OSMR expression and that rs10491509 (A allele) was associated with increased LIFR expression in arterial tissues. No variant was significantly associated with OSM expression. We associated these two variants with plaque characteristics from 1,443 genotyped carotid endarterectomy patients in the Athero-Express Biobank Study. After correction for multiple testing, rs13168867 was significantly associated with an increased overall plaque vulnerability (β = 0.118 ± s.e. = 0.040, p = 3.00 × 10
-3 , C allele). Looking at individual plaque characteristics, rs13168867 showed strongest associations with intraplaque fat (β = 0.248 ± s.e. = 0.088, p = 4.66 × 10-3 , C allele) and collagen content (β = -0.259 ± s.e. = 0.095, p = 6.22 × 10-3 , C allele), but these associations were not significant after correction for multiple testing. rs13168867 was not associated with intraplaque OSMR expression. Neither was intraplaque OSMR expression associated with plaque vulnerability and no known OSMR eQTLs were associated with coronary artery calcification burden, or cardiovascular disease susceptibility. No associations were found for rs10491509 in the LIFR locus. Conclusions: Our study suggests that rs1316887 in the OSMR locus is associated with increased plaque vulnerability, but not with coronary calcification or cardiovascular disease risk. It remains unclear through which precise biological mechanisms OSM signaling exerts its effects on plaque morphology. However, the OSM-OSMR/LIFR pathway is unlikely to be causally involved in lifetime cardiovascular disease susceptibility., Competing Interests: DvK is employed by Quorics B.V., and DT is employed by SkylineDx B.V and Quorics B.V. Quorics B.V. and SkylineDx B.V. had no part whatsoever in the conception, design, or execution of this study, nor the preparation and contents of this manuscript. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 van Keulen, van Koeverden, Boltjes, Princen, van Gool, de Borst, Asselbergs, Tempel, Pasterkamp and van der Laan.)- Published
- 2021
- Full Text
- View/download PDF
42. No effects of PCSK9-inhibitor treatment on spatial learning, locomotor activity, and novel object recognition in mice.
- Author
-
Schlunk F, Fischer P, Princen HMG, Rex A, Prinz V, Foddis M, Lütjohann D, Laufs U, and Endres M
- Subjects
- Animals, Antibodies, Mice, Proprotein Convertase 9 immunology, Behavior, Animal drug effects, Locomotion drug effects, PCSK9 Inhibitors, Protease Inhibitors pharmacology, Recognition, Psychology drug effects, Spatial Learning drug effects
- Abstract
Monoclonal anti-proprotein convertase subtilisin/kexin type 9 (PSCK9) neutralizing antibodies effectively lower plasma cholesterol levels and decrease cardiovascular events but also raised some concern that cognitive function could worsen as a side effect. Here, we performed experiments in mice to characterize the effect of anti-PCSK9 antibodies on behavior and cognitive function in detail. APOE*3Leiden.CETP mice and B6129SF1/J wildtype mice were fed a Western type diet and treated with the fully human anti-PCSK9 antibody CmAb1 (PL-45134; 10mg*kg
-1 s.c.) or vehicle for 6 weeks. Locomotor activity, anxiety levels, recognition memory, and spatial learning were investigated using the open field, novel object recognition test, and Morris water maze, respectively. Serum cholesterol levels in APOE*3Leiden.CETP mice after treatment with anti-PCSK9 antibody were significantly lower compared to controls whereas cholesterol levels in B6129SF1/J wildtype mice remained unchanged at low levels. No apparent differences were found regarding locomotor activity, anxiety, recognition memory, and spatial learning between animals treated with anti-PCSK9 antibody or vehicle in APOE*3Leiden.CETP and B6129SF1/J wildtype mice. In this study, we found no evidence that treatment with anti-PCSK9 antibodies lead to differences in behavior or changes of cognition in mice., (Copyright © 2020 Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
43. Effects of Inhibition or Deletion of PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) on Intracerebral Hemorrhage Volumes in Mice.
- Author
-
Schlunk F, Fischer P, Princen HMG, Rex A, Prinz V, Foddis M, Lütjohann D, Laufs U, and Endres M
- Subjects
- Animals, Apolipoprotein E3 genetics, Cerebral Hemorrhage pathology, Cholesterol Ester Transfer Proteins genetics, Diet, Western, Mice, Mice, Knockout, PCSK9 Inhibitors, Cerebral Hemorrhage genetics, Hypercholesterolemia genetics, Proprotein Convertase 9 genetics
- Published
- 2020
- Full Text
- View/download PDF
44. Alirocumab, evinacumab, and atorvastatin triple therapy regresses plaque lesions and improves lesion composition in mice.
- Author
-
Pouwer MG, Pieterman EJ, Worms N, Keijzer N, Jukema JW, Gromada J, Gusarova V, and Princen HMG
- Subjects
- Administration, Oral, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized administration & dosage, Anticholesteremic Agents administration & dosage, Atorvastatin administration & dosage, Drug Therapy, Combination, Female, Mice, Mice, Inbred C57BL, Mice, Transgenic, Plaque, Atherosclerotic chemically induced, Plaque, Atherosclerotic pathology, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized therapeutic use, Anticholesteremic Agents therapeutic use, Atorvastatin therapeutic use, Plaque, Atherosclerotic drug therapy
- Abstract
Atherosclerosis-related CVD causes nearly 20 million deaths annually. Most patients are treated after plaques develop, so therapies must regress existing lesions. Current therapies reduce plaque volume, but targeting all apoB-containing lipoproteins with intensive combinations that include alirocumab or evinacumab, monoclonal antibodies against cholesterol-regulating proprotein convertase subtilisin/kexin type 9 and angiopoietin-like protein 3, may provide more benefit. We investigated the effect of such lipid-lowering interventions on atherosclerosis in APOE*3-Leiden.CETP mice, a well-established model for hyperlipidemia. Mice were fed a Western-type diet for 13 weeks and thereafter matched into a baseline group (euthanized at 13 weeks) and five groups that received diet alone (control) or with treatment [atorvastatin; atorvastatin and alirocumab; atorvastatin and evinacumab; or atorvastatin, alirocumab, and evinacumab (triple therapy)] for 25 weeks. We measured effects on cholesterol levels, plaque composition and morphology, monocyte adherence, and macrophage proliferation. All interventions reduced plasma total cholesterol (37% with atorvastatin to 80% with triple treatment; all P < 0.001). Triple treatment decreased non-HDL-C to 1.0 mmol/l (91% difference from control; P < 0.001). Atorvastatin reduced atherosclerosis progression by 28% versus control ( P < 0.001); double treatment completely blocked progression and diminished lesion severity. Triple treatment regressed lesion size versus baseline in the thoracic aorta by 50% and the aortic root by 36% (both P < 0.05 vs. baseline), decreased macrophage accumulation through reduced proliferation, and abated lesion severity. Thus, high-intensive cholesterol-lowering triple treatment targeting all apoB-containing lipoproteins regresses atherosclerotic lesion area and improves lesion composition in mice, making it a promising potential approach for treating atherosclerosis., (Copyright © 2020 Pouwer et al.)
- Published
- 2020
- Full Text
- View/download PDF
45. Icosabutate Exerts Beneficial Effects Upon Insulin Sensitivity, Hepatic Inflammation, Lipotoxicity, and Fibrosis in Mice.
- Author
-
van den Hoek AM, Pieterman EJ, van der Hoorn JW, Iruarrizaga-Lejarreta M, Alonso C, Verschuren L, Skjæret T, Princen HMG, and Fraser DA
- Abstract
Icosabutate is a structurally engineered eicosapentaenoic acid derivative under development for nonalcoholic steatohepatitis (NASH). In this study, we investigated the absorption and distribution properties of icosabutate in relation to liver targeting and used rodents to evaluate the effects of icosabutate on glucose metabolism, insulin resistance, as well as hepatic steatosis, inflammation, lipotoxicity, and fibrosis. The absorption, tissue distribution, and excretion of icosabutate was investigated in rats along with its effects in mouse models of insulin resistance ( ob/ob ) and metabolic inflammation/NASH (high-fat/cholesterol-fed APOE*3Leiden.CETP mice) and efficacy was compared with synthetic peroxisome proliferator-activated receptor α (PPAR-α) (fenofibrate) and/or PPAR-γ/(α) (pioglitazone and rosiglitazone) agonists. Icosabutate was absorbed almost entirely through the portal vein, resulting in rapid hepatic accumulation. Icosabutate demonstrated potent insulin-sensitizing effects in ob/ob mice, and unlike fenofibrate or pioglitazone, it significantly reduced plasma alanine aminotransferase. In high-fat/cholesterol-fed APOE*3Leiden.CETP mice, icosabutate, but not rosiglitazone, reduced microvesicular steatosis and hepatocellular hypertrophy. Although both rosiglitazone and icosabutate reduced hepatic inflammation, only icosabutate elicited antifibrotic effects in association with decreased hepatic concentrations of multiple lipotoxic lipid species and an oxidative stress marker. Hepatic gene-expression analysis confirmed the changes in lipid metabolism, inflammatory and fibrogenic response, and energy metabolism, and revealed the involved upstream regulators. In conclusion, icosabutate selectively targets the liver through the portal vein and demonstrates broad beneficial effects following insulin sensitivity, hepatic microvesicular steatosis, inflammation, lipotoxicity, oxidative stress, and fibrosis. Icosabutate therefore offers a promising approach to the treatment of both dysregulated glucose/lipid metabolism and inflammatory disorders of the liver, including NASH., (© 2019 The Authors. Hepatology Communications published by Wiley Periodicals, Inc., on behalf of the American Association for the Study of Liver Diseases.)
- Published
- 2019
- Full Text
- View/download PDF
46. The APOE ∗ 3-Leiden Heterozygous Glucokinase Knockout Mouse as Novel Translational Disease Model for Type 2 Diabetes, Dyslipidemia, and Diabetic Atherosclerosis.
- Author
-
Pouwer MG, Heinonen SE, Behrendt M, Andréasson AC, van Koppen A, Menke AL, Pieterman EJ, van den Hoek AM, Jukema JW, Leighton B, Jönsson-Rylander AC, and Princen HMG
- Subjects
- Animals, Atherosclerosis blood, Blood Glucose metabolism, Cholesterol blood, Diabetes Complications blood, Diabetes Mellitus, Type 2 blood, Dyslipidemias blood, Female, Heterozygote, Inflammation, Lipids blood, Mice, Mice, Knockout, Phenotype, Risk, Translational Research, Biomedical, Triglycerides metabolism, Apolipoprotein E3 genetics, Atherosclerosis genetics, Diabetes Complications genetics, Diabetes Mellitus, Type 2 genetics, Disease Models, Animal, Dyslipidemias genetics
- Abstract
Background: There is a lack of predictive preclinical animal models combining atherosclerosis and type 2 diabetes. APOE∗3-Leiden (E3L) mice are a well-established model for diet-induced hyperlipidemia and atherosclerosis, and glucokinase
+/- (GK+/- ) mice are a translatable disease model for glucose control in type 2 diabetes. The respective mice respond similarly to lipid-lowering and antidiabetic drugs as humans. The objective of this study was to evaluate/characterize the APOE∗ 3-Leiden.glucokinase+/- (E3L.GK+/- ) mouse as a novel disease model to study the metabolic syndrome and diabetic complications., Methods: Female E3L.GK+/- , E3L, and GK+/- mice were fed fat- and cholesterol-containing diets for 37 weeks, and plasma parameters were measured throughout. Development of diabetic macro- and microvascular complications was evaluated., Results: Cholesterol and triglyceride levels were significantly elevated in E3L and E3L.GK+/- mice compared to GK+/- mice, whereas fasting glucose was significantly increased in E3L.GK+/- and GK+/- mice compared to E3L. Atherosclerotic lesion size was increased 2.2-fold in E3L.GK+/- mice as compared to E3L ( p = 0.037), which was predicted by glucose exposure ( R2 = 0.636, p = 0.001). E3L and E3L.GK+/- mice developed NASH with severe inflammation and fibrosis which, however, was not altered by introduction of the defective GK phenotype, whereas mild kidney pathology with tubular vacuolization was present in all three phenotypes., Conclusions: We conclude that the E3L.GK+/- mouse is a promising novel diet-inducible disease model for investigation of the etiology and evaluation of drug treatment on diabetic atherosclerosis.- Published
- 2019
- Full Text
- View/download PDF
47. The BCR-ABL1 Inhibitors Imatinib and Ponatinib Decrease Plasma Cholesterol and Atherosclerosis, and Nilotinib and Ponatinib Activate Coagulation in a Translational Mouse Model.
- Author
-
Pouwer MG, Pieterman EJ, Verschuren L, Caspers MPM, Kluft C, Garcia RA, Aman J, Jukema JW, and Princen HMG
- 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.
- Published
- 2018
- Full Text
- View/download PDF
48. Atorvastatin accelerates clearance of lipoprotein remnants generated by activated brown fat to further reduce hypercholesterolemia and atherosclerosis.
- Author
-
Hoeke G, Wang Y, van Dam AD, Mol IM, Gart E, Klop HG, van den Berg SM, Pieterman EH, Princen HMG, Groen AK, Rensen PCN, Berbée JFP, and Boon MR
- Subjects
- Adipose Tissue metabolism, Animals, Atherosclerosis metabolism, Calorimetry, Indirect, Cholesterol Ester Transfer Proteins blood, Cholesterol Ester Transfer Proteins genetics, Female, Gene Expression Profiling, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Hypercholesterolemia metabolism, Hyperlipidemias metabolism, Lipids blood, Liver metabolism, Mice, Mice, Knockout, ApoE, Proprotein Convertase 9 blood, Proprotein Convertase 9 genetics, Receptors, Adrenergic, beta-3 metabolism, Triglycerides metabolism, Adipose Tissue, Brown metabolism, Atherosclerosis drug therapy, Atorvastatin pharmacology, Hypercholesterolemia drug therapy, Lipoproteins metabolism
- Abstract
Background and Aims: Activation of brown adipose tissue (BAT) reduces both hyperlipidemia and atherosclerosis by increasing the uptake of triglyceride-derived fatty acids by BAT, accompanied by formation and clearance of lipoprotein remnants. We tested the hypothesis that the hepatic uptake of lipoprotein remnants generated by BAT activation would be accelerated by concomitant statin treatment, thereby further reducing hypercholesterolemia and atherosclerosis., Methods: APOE*3-Leiden.CETP mice were fed a Western-type diet and treated without or with the selective β3-adrenergic receptor (AR) agonist CL316,243 that activates BAT, atorvastatin (statin) or both., Results: β3-AR agonism increased energy expenditure as a result of an increased fat oxidation by activated BAT, which was not further enhanced by statin addition. Accordingly, statin treatment neither influenced the increased uptake of triglyceride-derived fatty acids from triglyceride-rich lipoprotein-like particles by BAT nor further lowered plasma triglyceride levels induced by β3-AR agonism. Statin treatment increased the hepatic uptake of the formed cholesterol-enriched remnants generated by β3-AR agonism. Consequently, statin treatment further lowered plasma cholesterol levels. Importantly, statin, in addition to β3-AR agonism, also further reduced the atherosclerotic lesion size as compared to β3-AR agonism alone, without altering lesion severity and composition., Conclusions: Statin treatment accelerates the hepatic uptake of remnants generated by BAT activation, thereby increasing the lipid-lowering and anti-atherogenic effects of BAT activation in an additive fashion. We postulate that, in clinical practice, combining statin treatment with BAT activation is a promising new avenue to combat hyperlipidemia and cardiovascular disease., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
49. Genetic and Pharmacologic Inactivation of ANGPTL3 and Cardiovascular Disease.
- Author
-
Dewey FE, Gusarova V, Dunbar RL, O'Dushlaine C, Schurmann C, Gottesman O, McCarthy S, Van Hout CV, Bruse S, Dansky HM, Leader JB, Murray MF, Ritchie MD, Kirchner HL, Habegger L, Lopez A, Penn J, Zhao A, Shao W, Stahl N, Murphy AJ, Hamon S, Bouzelmat A, Zhang R, Shumel B, Pordy R, Gipe D, Herman GA, Sheu WHH, Lee IT, Liang KW, Guo X, Rotter JI, Chen YI, Kraus WE, Shah SH, Damrauer S, Small A, Rader DJ, Wulff AB, Nordestgaard BG, Tybjærg-Hansen A, van den Hoek AM, Princen HMG, Ledbetter DH, Carey DJ, Overton JD, Reid JG, Sasiela WJ, Banerjee P, Shuldiner AR, Borecki IB, Teslovich TM, Yancopoulos GD, Mellis SJ, Gromada J, and Baras A
- Subjects
- Aged, Angiopoietin-Like Protein 3, Angiopoietin-like Proteins, Angiopoietins genetics, Animals, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacology, Atherosclerosis metabolism, Cardiovascular Diseases prevention & control, Coronary Artery Disease metabolism, Disease Models, Animal, Dose-Response Relationship, Drug, Double-Blind Method, Dyslipidemias blood, Female, Humans, Lipid Metabolism drug effects, Male, Mice, Mice, Inbred Strains, Middle Aged, Angiopoietins antagonists & inhibitors, Antibodies, Monoclonal administration & dosage, Atherosclerosis drug therapy, Coronary Artery Disease genetics, Dyslipidemias drug therapy, Lipids blood, Mutation
- Abstract
Background: Loss-of-function variants in the angiopoietin-like 3 gene (ANGPTL3) have been associated with decreased plasma levels of triglycerides, low-density lipoprotein (LDL) cholesterol, and high-density lipoprotein (HDL) cholesterol. It is not known whether such variants or therapeutic antagonism of ANGPTL3 are associated with a reduced risk of atherosclerotic cardiovascular disease., Methods: We sequenced the exons of ANGPTL3 in 58,335 participants in the DiscovEHR human genetics study. We performed tests of association for loss-of-function variants in ANGPTL3 with lipid levels and with coronary artery disease in 13,102 case patients and 40,430 controls from the DiscovEHR study, with follow-up studies involving 23,317 case patients and 107,166 controls from four population studies. We also tested the effects of a human monoclonal antibody, evinacumab, against Angptl3 in dyslipidemic mice and against ANGPTL3 in healthy human volunteers with elevated levels of triglycerides or LDL cholesterol., Results: In the DiscovEHR study, participants with heterozygous loss-of-function variants in ANGPTL3 had significantly lower serum levels of triglycerides, HDL cholesterol, and LDL cholesterol than participants without these variants. Loss-of-function variants were found in 0.33% of case patients with coronary artery disease and in 0.45% of controls (adjusted odds ratio, 0.59; 95% confidence interval, 0.41 to 0.85; P=0.004). These results were confirmed in the follow-up studies. In dyslipidemic mice, inhibition of Angptl3 with evinacumab resulted in a greater decrease in atherosclerotic lesion area and necrotic content than a control antibody. In humans, evinacumab caused a dose-dependent placebo-adjusted reduction in fasting triglyceride levels of up to 76% and LDL cholesterol levels of up to 23%., Conclusions: Genetic and therapeutic antagonism of ANGPTL3 in humans and of Angptl3 in mice was associated with decreased levels of all three major lipid fractions and decreased odds of atherosclerotic cardiovascular disease. (Funded by Regeneron Pharmaceuticals and others; ClinicalTrials.gov number, NCT01749878 .).
- Published
- 2017
- Full Text
- View/download PDF
50. 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.
- Author
-
Princen HMG, Pouwer MG, and Pieterman EJ
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.