Skip to main content
Erschienen in: Cardiovascular Diabetology 1/2018

Open Access 01.12.2018 | Original investigation

Liraglutide downregulates hepatic LDL receptor and PCSK9 expression in HepG2 cells and db/db mice through a HNF-1a dependent mechanism

verfasst von: Sheng-Hua Yang, Rui-Xia Xu, Chuan-Jue Cui, Yin Wang, Ying Du, Zhi-Guo Chen, Yu-Hong Yao, Chun-Yan Ma, Cheng-Gang Zhu, Yuan-Lin Guo, Na-Qiong Wu, Jing Sun, Bu-Xing Chen, Jian-Jun Li

Erschienen in: Cardiovascular Diabetology | Ausgabe 1/2018

Abstract

Background

Proprotein convertase subtilisin/kexin type 9 (PCSK9), a major regulator of cholesterol homeostasis, is associated with glucose metabolism. Liraglutide, a glucagon-like peptide-1 receptor agonist, can increase insulin secretion in a glucose-dependent manner and lower blood glucose. We aimed to investigate the relationship between liraglutide and PCSK9.

Methods

At the cellular level, the expressions of PCSK9 and hepatocyte nuclear factor 1 alpha (HNF1α) protein in HepG2 cells stimulated by liraglutide was examined using Western blot. Seven-week old db/db mice and wild type (WT) mice were administered either liraglutide (200 μg/kg) or equivoluminal saline subcutaneously, twice daily for 7 weeks. Fasting glucose level, food intake and body weight were measured every week. After the 7-week treatment, the blood was collected for lipid and PCSK9 levels detection and the liver was removed from the mice for oil red O staining, immunohistochemical analysis, immunofluorescence test and Western bolt.

Results

Firstly, liraglutide suppressed both PCSK9 and HNF1α expression in HepG2 cells in a time and concentration dependent manner. Secondly, liraglutide induced weight loss in WT and db/db mice, decreased serum PCSK9, glucose and lipid levels and improved hepatic accumulation in db/db but not WT mice. Thirdly, liraglutide reduced both hepatic PCSK9 and low-density lipoprotein receptor (LDLR) expression with a decrease in HNF1α in db/db mice but not in WT mice.

Conclusions

Liraglutide suppressed PCSK9 expression through HNF1α-dependent mechanism in HepG2 cells and db/db mice, and decreased LDLR possibly via PCSK9-independent pathways in db/db mice.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12933-018-0689-9) contains supplementary material, which is available to authorized users.
Sheng-Hua Yang and Rui-Xia Xu contributed equally to this work
Abkürzungen
GLP-1
glucagon-like peptide-1
HepG2
human hepatoma cell
HNF-1α
hepatocyte nuclear factor 1 alpha
HDL-C
high-density lipoprotein-cholesterol
LDLR
low-density lipoprotein receptor
LDL-C
low-density lipoprotein cholesterol
LIRA
liraglutide
TC
total cholesterol
TG
triglyceride
PCSK9
proprotein convertase subtilisin/kexin type 9
WT
wild type

Background

Proprotein convertase subtilisin/kexin type 9 (PCSK9), mainly secreted by the liver as an important regulator of cholesterol homeostasis, enhances the endosomal and lysosomal degradation of hepatic low-density lipoprotein (LDL) receptors (LDLR), resulting in increased circulating LDL-cholesterol (LDL-C) concentration [1, 2]. Interestingly, recent data suggested a relation of PCSK9 to glucose metabolism [311]. The epidemiological studies previously revealed positive associations between plasma PCSK9 and fasting glucose and insulin [3, 1012]. Several studies have suggested that an increase in PCSK9 levels is associated with higher fasting insulin levels in general populations and patients with diabetes [3, 10, 11]. Conversely, our previous studies and others found no significant difference in PCSK9 levels between the patients with or without diabetes [4, 9]. Awan et al. reported that presence of the loss-of-function PCSK9 p.R46L mutation was associated with insulin resistance in subjects with apolipoprotein E3/E2 genotype [13]. Furthermore, clinical trials showed that PCSK9 inhibitors did not increase the risk of onset-diabetes [14, 15], whereas genetics studies exhibited a link between PCSK9 genetic variants and risk of type 2 diabetes [16, 17]. To date, however, the exact mechanism of PCSK9 involved in glucose metabolism is still undetermined.
Liraglutide, one of glucagon-like peptide-1 (GLP-1) receptor agonists, can stimulate glucose-dependent insulin secretion, suppress glucagon release, and reduce food intake, resulting in glycemic improvement and weight loss in patients with type 2 diabetes [1822]. In addition of anti-diabetic and antiobesity effect, liraglutide can reduce cardiovascular events [23], and thus has been widely used for the treatment of type 2 diabetes. Until now, however, no data concerning the role of liraglutide in PCSK9 levels are available. Therefore, in the present study, the impact of liraglutide on PCSK9 expression was assessed in HepG2 cells and mice.

Materials and methods

Cell culture and treatment

The human hepatoma cell line, HepG2, was obtained from Cell Resource Center, IBMS, CAMS/PUMC (Beijing, China) and cultured in Dulbecco’s Modified Eagle’s medium DMEM (DMEM, Gibco, Grand Island, NY, USA) containing 10% fetal bovine serum (FBS) (Gibco, Grand Island, NY, USA), 1% non-essential amino acids (NEAA) (Life technologies, Carlsbad, CA, USA) and 1% penicillin–streptomycin at 37 °C, 5% (v/v) CO2. HepG2 cells were serum-starved for 18 h and then treated with liraglutide (Novo Nordisk, Bagsværd, Denmark) at various concentrations (0, 10, 50, 100, 500 and 1000 nM) for 24 h or with 500 nM liraglutide for different times (0, 3, 6, 12, 24 h).

Cell viability assay

The cell viability was determined using a Cell Counting Kit-8 (CCK-8; Dojindo Laboratories, Kumamoto, Japan) as previously described [24, 25]. Briefly, HepG2 cells were plated in 96-well plates (5 × 103/well) and serum-starved for 18 h, and then exposed to different concentrations of liraglutide (0, 10, 50, 100, 500 and 1000 nM) for 24 h at 37 °C in a humidified atmosphere containing 5% CO2. After 24 h incubation, the CCK-8 solution was added to each well at a 1:10 dilution and incubated for an additional 3 h at 37 °C. The optical density (OD) value of each well was measured at a wavelength of 450 nm using a Microplate Reader (Thermo, Varioskan Flash). Cell viability was calculated by the following equation: viable cells (%) = (ODtest − ODblank)/(ODcontrol − ODblank). In the equation, ODtest is the optical density of the cells exposed to different concentrations of liraglutide, ODblank is the optical density of the wells without HepG2 cells and ODcontrol is the optical density of the control sample (Phosphate-buffered saline, PBS). Results of cell viability assay are shown as the mean values of three replicate experiments performed in triplicate.

Animal model and liraglutide treatment

Twenty-four male db/db mice (BKS.Cg-Dock7m +/+ Leprdb/J, Strain Number: J000642) aged 5 weeks and sixteen non-diabetic littermates (wild-type, WT) were purchased from the Model Animal Research Center of Nanjing University (Nanjing, China). All mice (four mice/cage) were housed under a 12-h light/dark cycle with 50% relative humidity at 22 °C and had free access to regular chow and water. After 2-week habituation, the mice were randomly divided into four treatment groups: db/db + LIRA (liraglutide) (n = 12), db/db + saline (n = 12), WT + LIRA (n = 8), WT + saline (n = 8). Mice were administered either liraglutide (200 μg/kg) or equivoluminal 0.9% saline subcutaneously, twice daily (09:00 and 18:00 h) for 7 weeks. Doses of the liraglutide used in the study were based on previous studies [26, 27]. During this period, body weight and 4-h fasting blood glucose levels from the tail vein were determined weekly. At the end of 7-week treatment period, the mice were euthanized after a 4-h fast and livers were removed after collecting blood for analysis. All experiments were approved by the Ethics Committee for Animal Care and Research at FuWai hospital (Beijing, China).

Serum PCSK9 and serum lipid analysis

Mouse serum PCSK9 levels were detected by enzyme-linked immunosorbent assay (ELISA) using the CircuLex mouse/rat PCSK9 ELISA Kit (MBL, Nagano, Japan) according to the manufacturer’s instructions as previously described [9, 59]. The serum was diluted with 0.9% saline by 1:1. Then the levels of triglyceride (TG), total cholesterol (TC), LDL-C and high-density lipoprotein-cholesterol (HDL-C) were examined by the automatic biochemistry analyser (Beckman CX5, Beckman coulter, Brea, CA, USA).

Hematoxylin and eosin (H&E) staining

Mouse liver slices were processed according to a standard a standard H&E staining technique [28]. Briefly, liver tissues were fixed by 10% neutral formalin, dehydrated in ethanol, and then embedded in paraffin. Liver sections (4 μm) were stained with hematoxylin and eosin (H&E).

Oil red O staining

Oil red O staining was performed as previously described [29]. Briefly, mouse liver tissues were immediately snap-frozen in liquid nitrogen and placed in OCT cryostat embedding compound (Tissue-Tek, Torrance, CA, USA). Frozen liver sections (8 μm) were stained with Oil Red O, washed with 60% isopropanol, and counterstained with hematoxylin. Staining was assessed by bright-field microscopy.

Immunohistochemistry and immunofluorescence

Mouse liver tissues were fixed with formaldehyde, embedded with paraffin and then cut into 4 μm-thick sections. Prior to immunostaining, the sections were dewaxed in xylene and rehydrated through graded alcohol. Antigen retrieval was carried out by boiling in citrate buffer (pH 6.0) for 2 min in a pressure cooker and then the cooker was depressurized and cooled under running water for 20 min [30]. Hydrogen peroxide (3%) was added to the tissue sections and incubated at room temperature for 10 min. Subsequently, the sections were washed with Phosphate buffered saline (PBS) for three times, and then incubated overnight with rabbit polyclonal PCSK9 antibody (1:200, Abcam, ab31762) or rabbit monoclonal LDLR antibody (1:500, Abcam, ab52818) at 4 °C. After that, the slides were incubated with goat anti-rabbit IgG/horseradish peroxidase (HRP) secondary antibody (Beijing Zhongshanjinqiao Biological Technology Co., LTD., China), and then counterstained with hematoxylin.
For immunofluorescence, the slides were blocked with 10% goat serum (Invitrogen, CA, USA) for 1 h and then incubated overnight with rabbit polyclonal PCSK9 antibody (1:200, Abcam, ab31762) or rabbit monoclonal LDLR antibody (1:500, Abcam, ab52818) at 4 °C. Goat anti-rabbit H&G (Alexxa Fluor 488, Abcam, ab150077) antibody was applied as the second antibody. 4′,6-diamidine-2′-phenylindole dihydrochloride (DAPI) (Beijing Zhongshanjinqiao Biological Technology Co., LTD., China) was used as nuclear counterstain.

Western blots

Mouse liver tissue and HepG2 cells samples were homogenized on ice in lysis buffer containing protease and phosphatase inhibitors (Beyotime, Shanghai, China). The homogenate was then centrifuged at 12,000g for 15 min and the supernatant was collected. Protein concentrations were determined using a BCA Protein Assay Kit (Beijing Kangwei Century Biotechnology Co., Ltd, Beijing, China). Subsequently, 30 μg of protein from individual samples was resolved by precast NuPAGE Novex 4–12% (w/v) Bis–Tris gels (Life technologies, Carlsbad, CA, USA), and then transferred onto nitrocellulose membrane using the iBlotTM dry blotting system as described by the manufacturer (Invitrogen, Carlsbad, CA, USA). The membranes were blocked in TBST buffer (20 mM Tris, pH 7.5, 150 mM NaCl, 0.1% tween-20) containing 5% non-fat milk for 2 h at room temperature and then incubated overnight at 4 °C with anti-PCSK9 (1:1000, Abcam, ab31762), anti-HNF1α (Cell Signaling), anti-LDLR (1:5000, Abcam, ab52818) or anti-GAPDH (1:5000, Abcam). Afterwards, the membranes were incubated with the secondary antibodies including goat anti-rabbit IgG/horseradish peroxidase (HRP) and goat anti-mouse IgG/HRP (Abcam) for 2 h at room temperature. Protein expression was detected with chemiluminescence (ECL, Thermo Fisher Scientific, Waltham, MA, USA) on FluorChem M image system.

Statistical analysis

Data are presented as mean ± standard error of the mean unless otherwise stated. Comparisons between two groups were assessed using an unpaired two-tailed Student t test. One-way ANOVA combined with Bonferroni’s post hoc test was used among ≥ 3 groups. Differences were considered statistically significant at P < 0.05. All analyses were performed using SPSS 19.0 (SPSS Inc., Chicago, IL, USA).

Results

Liraglutide down-regulated the protein expression of PCSK9 in HepG2 in a dose- and time-dependent manner

The HepG2 cells were treated with liraglutide (10, 50, 100, 500 and 1000 nM) for 24 h and their viabilities were assessed using the CCK-8 assay. As shown in Fig. 1a, liraglutide showed no cytotoxicity below 1000 nM (1 μM). Subsequently, we determined whether liraglutide could affect the expression of PCSK9 in HepG2 and found that liraglutide down-regulated the protein and mRNA levels of PCSK9 in a dose-dependent manner (Fig. 1b). In parallel, we also found that liraglutide had time-dependent inhibitory effect on the PCSK9 protein and mRNA expression in HepG2 (Fig. 1c). Also, the protein and mRNA expression of HNF1α was found to decrease when HepG2 cells were exposed to liraglutide (500 nM) for 24 h (Fig. 1d). Moreover, the inhibiting effect of liraglutide on PCSK9 was weakened after inhibition of HNF1α by siRNA (Fig. 1d).

Liraglutide decreased body weight and improved glucose metabolism

The db/db mice had higher levels of fasting blood glucose than those of the WT mice (Fig. 2). Seven-week old male mice (WT or db/db mice) were administered liraglutide (200 μg/kg, twice daily) or vehicle (saline) subcutaneously for 7 weeks. As expected, liraglutide treatment decreased body weight in both WT mice and db/db mice (data not shown) and significantly reduced blood glucose levels in db/db mice but not in WT mice (Fig. 2).

Liraglutide reduced lipid accumulation in the serum and liver in db/db mice

There were no differences in the levels of serum TC, TG, LDL-C and HDL-C among the non-diabetic WT mice with liraglutide or saline treatment (Fig. 3a). In contrast, the levels of TC (3.10 ± 0.10), TG (1.19 ± 0.05) and LDL-C (0.74 ± 0.03) in db/db mice that were treated with liraglutide were significantly decreased compared with those of control db/db mice (TC: 4.59 ± 0.54, P = 0.027; TG: 2.46 ± 0.40, P = 0.016; LDL-C: 1.23 ± 0.21, P = 0.022) (Fig. 3b) Interestingly, liraglutide also reduced the serum HDL-C level in db/db mice (Fig. 3b). Furthermore, oil red O staining of liver sections exhibited no apparent lipid deposition in the livers of non-diabetic WT mice regardless of the treatment of liraglutide (Fig. 4a). Conversely, in db/db mice, marked accumulation of oil red O-stainable lipid droplets was found in liver section and a significantly decrease in the number of the lipid droplets was shown in liraglutide-treated db/db mice compared with the vehicle-treated db/db mice (Fig. 4a).

Liraglutide suppressed PCSK9 levels in db/db mice

To determine whether liraglutide can affects PCSK9 levels, we detected the serum and liver PCSK9 levels in WT and db/db mice. After liraglutide treatment for 7 weeks, there was no difference in serum PCSK9 level in WT mice compared with saline-treated mice (26.57 ± 3.51 vs. 30.82 ± 4.45, P = 0.464), whereas the serum level of PCSK9 in db/db mice exhibited a marked decrease (39.93 ± 4.50 vs. 26.97 ± 2.78, P = 0.021) (Fig. 3i, j). Meanwhile, immunohistochemistry and immunofluorescence staining revealed that liraglutide decreased liver PCSK9 protein level in db/db mice but not in WT mice (Fig. 4b, c), which is consistent with the western blots results (Fig. 5). In parallel, we assessed the liver LDLR level in db/db and WT mice and unexpectedly found that liraglutide reduced the LDLR level in both db/db mice and WT mice (Figs. 4c, 5). Importantly, liraglutide showed an inhibitory effect on the hepatic HNF1α expression in db/db mice not in WT mice (Fig. 5).

Discussion

Based on the data regarding the relation of PCSK9 to cholesterol and glucose metabolism and the beneficial effects of liraglutide on cardiovascular health, we set out to explore whether liraglutide has an effect on PCSK9 expression. Firstly, data showed that liraglutide down-regulated PCSK9 expression in HepG2 cells in a dose- and time-dependent fashion through HNF1α-dependent mechanism. In addition, the results indicated that liraglutide could reduce serum and hepatic PCSK9 levels in db/db mice rather than in WT mice.
Liraglutide, a long-acting GLP-1 receptor agonist, is widely used for treating diabetes by stimulating glucose-dependent insulin secretion and suppressing glucagon secretion with a very low risk for hypoglycemia [19, 31, 32]. In addition, liraglutide can induce body weight loss through reducing food intake, promoting satiety, delaying gastric emptying and inducing autophagy [21, 3335]. Similarly, our data showed that liraglutide lowered serum glucose levels in db/db mice. In fact, previous study suggested that liraglutide could markedly modify circulating lipid profile levels in patients with type 2 diabetes or in diabetic mice [1921, 36], which was also demonstrated by our study. The mechanisms of liraglutide on lipid profile have been speculated: (1) to increase insulin secretion and to reduce eating [37, 38]; (2) to reduce chylomicron production, lipoprotein synthase and ApoB-48 production [39]; (3) to slow gastric emptying and to reduce lipase activity [40]; (4) to decrease VLDL production, lipogenesis and ApoB100 production [41, 42]; (5) to inhibit fat oxidation and to promote thermogenesis [43]; (6) the effect of decreased PCSK9 by liraglutide on cholesteryl ester transfer protein (CETP) inhibition on lipid profiles [44]. Additionally, we observed that liraglutide significantly improved hepatic lipid accumulation in db/db mice, which is consistent with some researches [4548]. Conversely, some other studies showed that liraglutide treatment did not reduce hepatic steatosis [49, 50]. All these inconsistent results may be attributed to the various definitions, evaluation methods, doses or durations of liraglutide treatment.
A novel finding of the present study is that liraglutide can inhibit PCSK9 expression at the cellular and animal levels, which may be an explanation for the beneficial effects of liraglutide on cardiovascular outcomes. Moreover, we found that liraglutide also reduced HNF1α expression. To explore whether HNF1α, a critical regulator of PCSK9 gene transcription, is involved in the regulation of PCSK9 expression, we inhibited HNF1α expression using siRNA in HepG2 cells. As expected, the data showed that the inhibiting effect of liraglutide on PCSK9 was weakened after inhibition of HNF-1α by siRNA, suggesting that liraglutide reduced PCSK9 at least partly via HNF1α-dependent mechanism. Meanwhile, we found that SREBP-2 was not involved in the effect of liraglutide on PCSK9 (data not shown), which is supported by our previous studies [51, 52]. Also, in the present study, liraglutide reduced PCSK9 expression in HepG2 and db/db mice but not in WT mice. This is perhaps due to the fact that pathological states may alter the role of liraglutide in PCSK9 expression, but its real reason is unknown.
Unexpectedly, decreased levels of hepatic PCSK9 protein were not associated with an increase in hepatic LDLR protein in db/db mice, namely, liraglutide also inhibit LDLR. Actually, previous studies reported this phenomenon [5355]. Miao et al. showed that insulin promoted the degradation of LDLR in a PCSK9-dependent manner in HepG2 cells, but in vivo in insulin-deficient states, both PCSK9 and LDLR levels reduced [53]. Similarly, Levenson et al. reported that leptin suppressed both PCSK9 and LDLR in ob/ob mice [54]. Moreover, liraglutide reduced both hepatic PCSK9 and LDLR expression simultaneously, and it is possible that another regulator such as inducible degrader of LDLR (Idol) promotes degradation of LDLR [56]. Moreover, glucagon has been demonstrated to increase LDLR [57]. Liraglutide can suppress glucagon secretion [58] and thus may contribute to LDLR reduction. However, the real season remains unclear. All these data point to the fact that the role of some reagents such insulin, leptin and liraglutide and so on in LDLR regulation is complex, and suggests that in vivo these reagents may act through PCSK9-independent mechanism to affect LDLR expression. Nevertheless, further researches are needed to confirm our finding.
This study has several limitations. Firstly, the db/db model used in the study was characterized by hyperinsulinemia, so we could not find out whether insulin affected LDLR expression. Even so, this model was also used in previous to explore the effect of PCSK9 on glucose and lipid metabolism [59]. Secondly, the mechanism of the effect of liraglutide on LDLR was not explored. Finally, we failed to find out whether liraglutide had a major role in lipid metabolism compared to insulin due to the animal model we used.

Conclusion

In conclusion, our data indicated that liraglutide suppressed PCSK9 in HepG2 cells and db/db mice through HNF1α-dependent mechanism, and decreased LDLR possibly via PCSK9-independent pathways in db/db mice (Additional files 1, 2, 3, 4).

Authors’ contributions

YSH completed the project, analyzed the data and wrote the manuscript. LJJ established the study, and contributed to reviewed the manuscript. The other co-authors contributed to collected the data and conduct the experiment. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.
Not applicable.
The studies were approved by Fuwai Hospital ethics committees, in accordance with the Helsinki Declaration.

Funding

This work was partially supported by the Capital Health Development Fund (2016-12M-1-011) awarded to J-J Li.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Urban D, Poss J, Bohm M, Laufs U. Targeting the proprotein convertase subtilisin/kexin type 9 for the treatment of dyslipidemia and atherosclerosis. J Am Coll Cardiol. 2013;62:1401–8.CrossRefPubMed Urban D, Poss J, Bohm M, Laufs U. Targeting the proprotein convertase subtilisin/kexin type 9 for the treatment of dyslipidemia and atherosclerosis. J Am Coll Cardiol. 2013;62:1401–8.CrossRefPubMed
2.
Zurück zum Zitat Seidah NG, Awan Z, Chretien M, Mbikay M. PCSK9: a key modulator of cardiovascular health. Circ Res. 2014;114:1022–36.CrossRefPubMed Seidah NG, Awan Z, Chretien M, Mbikay M. PCSK9: a key modulator of cardiovascular health. Circ Res. 2014;114:1022–36.CrossRefPubMed
3.
Zurück zum Zitat Lakoski SG, Lagace TA, Cohen JC, Horton JD, Hobbs HH. Genetic and metabolic determinants of plasma PCSK9 levels. J Clin Endocrinol Metab. 2009;94:2537–43.CrossRefPubMedPubMedCentral Lakoski SG, Lagace TA, Cohen JC, Horton JD, Hobbs HH. Genetic and metabolic determinants of plasma PCSK9 levels. J Clin Endocrinol Metab. 2009;94:2537–43.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Brouwers MCGJ, Troutt JS, van Greevenbroek MMJ, Ferreira I, Feskens EJ, van der Kallen CJH, et al. Plasma proprotein convertase subtilisin kexin type 9 is not altered in subjects with impaired glucose metabolism and type 2 diabetes mellitus, but its relationship with non-HDL cholesterol and apolipoprotein B may be modified by type 2 diabetes mellitus: the CODAM study. Atherosclerosis. 2011;217:263–7.CrossRefPubMed Brouwers MCGJ, Troutt JS, van Greevenbroek MMJ, Ferreira I, Feskens EJ, van der Kallen CJH, et al. Plasma proprotein convertase subtilisin kexin type 9 is not altered in subjects with impaired glucose metabolism and type 2 diabetes mellitus, but its relationship with non-HDL cholesterol and apolipoprotein B may be modified by type 2 diabetes mellitus: the CODAM study. Atherosclerosis. 2011;217:263–7.CrossRefPubMed
5.
Zurück zum Zitat Persson L, Galman C, Angelin B, Rudling M. Importance of proprotein convertase subtilisin/kexin type 9 in the hormonal and dietary regulation of rat liver low-density lipoprotein receptors. Endocrinology. 2009;150:1140–6.CrossRefPubMed Persson L, Galman C, Angelin B, Rudling M. Importance of proprotein convertase subtilisin/kexin type 9 in the hormonal and dietary regulation of rat liver low-density lipoprotein receptors. Endocrinology. 2009;150:1140–6.CrossRefPubMed
6.
Zurück zum Zitat Awan Z, Dubuc G, Faraj M, Dufour R, Seidah NG, Davignon J, et al. The effect of insulin on circulating PCSK9 in postmenopausal obese women. Clin Biochem. 2014;47:1033–9.CrossRefPubMed Awan Z, Dubuc G, Faraj M, Dufour R, Seidah NG, Davignon J, et al. The effect of insulin on circulating PCSK9 in postmenopausal obese women. Clin Biochem. 2014;47:1033–9.CrossRefPubMed
7.
Zurück zum Zitat Mbikay M, Sirois F, Mayne J, Wang GS, Chen A, Dewpura T, et al. PCSK9-deficient mice exhibit impaired glucose tolerance and pancreatic islet abnormalities. FEBS Lett. 2010;584:701–6.CrossRefPubMed Mbikay M, Sirois F, Mayne J, Wang GS, Chen A, Dewpura T, et al. PCSK9-deficient mice exhibit impaired glucose tolerance and pancreatic islet abnormalities. FEBS Lett. 2010;584:701–6.CrossRefPubMed
8.
Zurück zum Zitat Langhi C, Le May C, Gmyr V, Vandewalle B, Kerr-Conte J, Krempf M, et al. PCSK9 is expressed in pancreatic delta-cells and does not alter insulin secretion. Biochem Biophys Res Commun. 2009;390:1288–93.CrossRefPubMed Langhi C, Le May C, Gmyr V, Vandewalle B, Kerr-Conte J, Krempf M, et al. PCSK9 is expressed in pancreatic delta-cells and does not alter insulin secretion. Biochem Biophys Res Commun. 2009;390:1288–93.CrossRefPubMed
9.
Zurück zum Zitat Yang SH, Li S, Zhang Y, Xu RX, Guo YL, Zhu CG, et al. Positive correlation of plasma PCSK9 levels with HbA1c in patients with type 2 diabetes. Diab Metab Res Rev. 2016;32:193–9.CrossRef Yang SH, Li S, Zhang Y, Xu RX, Guo YL, Zhu CG, et al. Positive correlation of plasma PCSK9 levels with HbA1c in patients with type 2 diabetes. Diab Metab Res Rev. 2016;32:193–9.CrossRef
10.
Zurück zum Zitat Baass A, Dubuc G, Tremblay M, Delvin EE, O’Loughlin J, Levy E, et al. Plasma PCSK9 is associated with age, sex, and multiple metabolic markers in a population-based sample of children and adolescents. Clin Chem. 2009;55:1637–45.CrossRefPubMed Baass A, Dubuc G, Tremblay M, Delvin EE, O’Loughlin J, Levy E, et al. Plasma PCSK9 is associated with age, sex, and multiple metabolic markers in a population-based sample of children and adolescents. Clin Chem. 2009;55:1637–45.CrossRefPubMed
11.
Zurück zum Zitat Cui Q, Ju X, Yang T, Zhang M, Tang W, Chen Q, et al. Serum PCSK9 is associated with multiple metabolic factors in a large Han Chinese population. Atherosclerosis. 2010;213:632–6.CrossRefPubMed Cui Q, Ju X, Yang T, Zhang M, Tang W, Chen Q, et al. Serum PCSK9 is associated with multiple metabolic factors in a large Han Chinese population. Atherosclerosis. 2010;213:632–6.CrossRefPubMed
12.
Zurück zum Zitat Dong B, Singh AB, Azhar S, Seidah NG, Liu J. High-fructose feeding promotes accelerated degradation of hepatic LDL receptor and hypercholesterolemia in hamsters via elevated circulating PCSK9 levels. Atherosclerosis. 2015;239:364–74.CrossRefPubMedPubMedCentral Dong B, Singh AB, Azhar S, Seidah NG, Liu J. High-fructose feeding promotes accelerated degradation of hepatic LDL receptor and hypercholesterolemia in hamsters via elevated circulating PCSK9 levels. Atherosclerosis. 2015;239:364–74.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Awan Z, Delvin EE, Levy E, Genest J, Davignon J, Seidah NG, et al. Regional distribution and metabolic effect of PCSK9 insLEU and R46L gene mutations and apoE genotype. Can J Cardiol. 2013;29:927–33.CrossRefPubMed Awan Z, Delvin EE, Levy E, Genest J, Davignon J, Seidah NG, et al. Regional distribution and metabolic effect of PCSK9 insLEU and R46L gene mutations and apoE genotype. Can J Cardiol. 2013;29:927–33.CrossRefPubMed
14.
Zurück zum Zitat Colhoun HM, Ginsberg HN, Robinson JG, Leiter LA, Muller-Wieland D, Henry RR, et al. No effect of PCSK9 inhibitor alirocumab on the incidence of diabetes in a pooled analysis from 10 ODYSSEY Phase 3 studies. Eur Heart J. 2016;37:2981–9.CrossRefPubMedPubMedCentral Colhoun HM, Ginsberg HN, Robinson JG, Leiter LA, Muller-Wieland D, Henry RR, et al. No effect of PCSK9 inhibitor alirocumab on the incidence of diabetes in a pooled analysis from 10 ODYSSEY Phase 3 studies. Eur Heart J. 2016;37:2981–9.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Sabatine MS, Leiter LA, Wiviott SD, Giugliano RP, Deedwania P, De Ferrari GM, et al. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without diabetes and the effect of evolocumab on glycaemia and risk of new-onset diabetes: a prespecified analysis of the FOURIER randomised controlled trial. Lancet Diab Endocrinol. 2017;5:941–50.CrossRef Sabatine MS, Leiter LA, Wiviott SD, Giugliano RP, Deedwania P, De Ferrari GM, et al. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without diabetes and the effect of evolocumab on glycaemia and risk of new-onset diabetes: a prespecified analysis of the FOURIER randomised controlled trial. Lancet Diab Endocrinol. 2017;5:941–50.CrossRef
16.
Zurück zum Zitat Lotta LA, Sharp SJ, Burgess S, Perry JRB, Stewart ID, Willems SM, et al. Association between low-density lipoprotein cholesterol-lowering genetic variants and risk of type 2 diabetes: a meta-analysis. JAMA. 2016;316:1383–91.CrossRefPubMedPubMedCentral Lotta LA, Sharp SJ, Burgess S, Perry JRB, Stewart ID, Willems SM, et al. Association between low-density lipoprotein cholesterol-lowering genetic variants and risk of type 2 diabetes: a meta-analysis. JAMA. 2016;316:1383–91.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Ference BA, Robinson JG, Brook RD, Catapano AL, Chapman MJ, Neff DR, et al. Variation in PCSK9 and HMGCR and risk of cardiovascular disease and diabetes. N Engl J Med. 2016;375:2144–53.CrossRefPubMed Ference BA, Robinson JG, Brook RD, Catapano AL, Chapman MJ, Neff DR, et al. Variation in PCSK9 and HMGCR and risk of cardiovascular disease and diabetes. N Engl J Med. 2016;375:2144–53.CrossRefPubMed
18.
Zurück zum Zitat Zinman B, Gerich J, Buse JB, Lewin A, Schwartz S, Raskin P, et al. Efficacy and safety of the human glucagon-like peptide-1 analog liraglutide in combination with metformin and thiazolidinedione in patients with type 2 diabetes (LEAD-4 Met + TZD). Diabetes Care. 2009;32:1224–30.CrossRefPubMedPubMedCentral Zinman B, Gerich J, Buse JB, Lewin A, Schwartz S, Raskin P, et al. Efficacy and safety of the human glucagon-like peptide-1 analog liraglutide in combination with metformin and thiazolidinedione in patients with type 2 diabetes (LEAD-4 Met + TZD). Diabetes Care. 2009;32:1224–30.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Marre M, Shaw J, Brandle M, Bebakar WM, Kamaruddin NA, Strand J, et al. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabet Med. 2009;26:268–78.CrossRefPubMedPubMedCentral Marre M, Shaw J, Brandle M, Bebakar WM, Kamaruddin NA, Strand J, et al. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabet Med. 2009;26:268–78.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Yang W, Chen L, Ji Q, Liu X, Ma J, Tandon N, et al. Liraglutide provides similar glycaemic control as glimepiride (both in combination with metformin) and reduces body weight and systolic blood pressure in Asian population with type 2 diabetes from China, South Korea and India: a 16-week, randomized, double-blind, active control trial. Diab Obes Metab. 2011;13:81–8.CrossRef Yang W, Chen L, Ji Q, Liu X, Ma J, Tandon N, et al. Liraglutide provides similar glycaemic control as glimepiride (both in combination with metformin) and reduces body weight and systolic blood pressure in Asian population with type 2 diabetes from China, South Korea and India: a 16-week, randomized, double-blind, active control trial. Diab Obes Metab. 2011;13:81–8.CrossRef
21.
Zurück zum Zitat Davies MJ, Bergenstal R, Bode B, Kushner RF, Lewin A, Skjoth TV, et al. Efficacy of liraglutide for weight loss among patients with type 2 diabetes: the SCALE diabetes randomized clinical trial. JAMA. 2015;314:687–99.CrossRefPubMed Davies MJ, Bergenstal R, Bode B, Kushner RF, Lewin A, Skjoth TV, et al. Efficacy of liraglutide for weight loss among patients with type 2 diabetes: the SCALE diabetes randomized clinical trial. JAMA. 2015;314:687–99.CrossRefPubMed
22.
Zurück zum Zitat Astrup A, Rossner S, Van Gaal L, Rissanen A, Niskanen L, Al Hakim M, et al. Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet. 2009;374:1606–16.CrossRefPubMed Astrup A, Rossner S, Van Gaal L, Rissanen A, Niskanen L, Al Hakim M, et al. Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet. 2009;374:1606–16.CrossRefPubMed
23.
Zurück zum Zitat Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311–22.CrossRefPubMedPubMedCentral Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311–22.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Chang Y, Yang ST, Liu JH, Dong E, Wang Y, Cao A, et al. In vitro toxicity evaluation of graphene oxide on A549 cells. Toxicol Lett. 2011;200:201–10.CrossRefPubMed Chang Y, Yang ST, Liu JH, Dong E, Wang Y, Cao A, et al. In vitro toxicity evaluation of graphene oxide on A549 cells. Toxicol Lett. 2011;200:201–10.CrossRefPubMed
25.
Zurück zum Zitat Deng X, Luan Q, Chen W, Wang Y, Wu M, Zhang H, et al. Nanosized zinc oxide particles induce neural stem cell apoptosis. Nanotechnology. 2009;20:115101.CrossRefPubMed Deng X, Luan Q, Chen W, Wang Y, Wu M, Zhang H, et al. Nanosized zinc oxide particles induce neural stem cell apoptosis. Nanotechnology. 2009;20:115101.CrossRefPubMed
26.
Zurück zum Zitat Shimoda M, Kanda Y, Hamamoto S, Tawaramoto K, Hashiramoto M, Matsuki M, et al. The human glucagon-like peptide-1 analogue liraglutide preserves pancreatic beta cells via regulation of cell kinetics and suppression of oxidative and endoplasmic reticulum stress in a mouse model of diabetes. Diabetologia. 2011;54:1098–108.CrossRefPubMedPubMedCentral Shimoda M, Kanda Y, Hamamoto S, Tawaramoto K, Hashiramoto M, Matsuki M, et al. The human glucagon-like peptide-1 analogue liraglutide preserves pancreatic beta cells via regulation of cell kinetics and suppression of oxidative and endoplasmic reticulum stress in a mouse model of diabetes. Diabetologia. 2011;54:1098–108.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Kimura T, Kaneto H, Shimoda M, Hirukawa H, Okauchi S, Kohara K, et al. Protective effects of pioglitazone and/or liraglutide on pancreatic beta-cells in db/db mice: comparison of their effects between in an early and advanced stage of diabetes. Mol Cell Endocrinol. 2015;400:78–89.CrossRefPubMed Kimura T, Kaneto H, Shimoda M, Hirukawa H, Okauchi S, Kohara K, et al. Protective effects of pioglitazone and/or liraglutide on pancreatic beta-cells in db/db mice: comparison of their effects between in an early and advanced stage of diabetes. Mol Cell Endocrinol. 2015;400:78–89.CrossRefPubMed
28.
Zurück zum Zitat Chong BF, Murphy JE, Kupper TS, Fuhlbrigge RC. E-selectin, thymus- and activation-regulated chemokine/CCL17, and intercellular adhesion molecule-1 are constitutively coexpressed in dermal microvessels: a foundation for a cutaneous immunosurveillance system. J Immunol. 2004;172:1575–81.CrossRefPubMed Chong BF, Murphy JE, Kupper TS, Fuhlbrigge RC. E-selectin, thymus- and activation-regulated chemokine/CCL17, and intercellular adhesion molecule-1 are constitutively coexpressed in dermal microvessels: a foundation for a cutaneous immunosurveillance system. J Immunol. 2004;172:1575–81.CrossRefPubMed
29.
Zurück zum Zitat Lerat H, Honda M, Beard MR, Loesch K, Sun J, Yang Y, et al. Steatosis and liver cancer in transgenic mice expressing the structural and nonstructural proteins of hepatitis C virus. Gastroenterology. 2002;122:352–65.CrossRefPubMed Lerat H, Honda M, Beard MR, Loesch K, Sun J, Yang Y, et al. Steatosis and liver cancer in transgenic mice expressing the structural and nonstructural proteins of hepatitis C virus. Gastroenterology. 2002;122:352–65.CrossRefPubMed
30.
Zurück zum Zitat Norton AJ, Jordan S, Yeomans P. Brief, high-temperature heat denaturation (pressure cooking): a simple and effective method of antigen retrieval for routinely processed tissues. J Pathol. 1994;173:371–9.CrossRefPubMed Norton AJ, Jordan S, Yeomans P. Brief, high-temperature heat denaturation (pressure cooking): a simple and effective method of antigen retrieval for routinely processed tissues. J Pathol. 1994;173:371–9.CrossRefPubMed
31.
Zurück zum Zitat Drucker DJ, Buse JB, Taylor K, Kendall DM, Trautmann M, Zhuang D, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet. 2008;372:1240–50.CrossRefPubMed Drucker DJ, Buse JB, Taylor K, Kendall DM, Trautmann M, Zhuang D, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet. 2008;372:1240–50.CrossRefPubMed
32.
Zurück zum Zitat Trujillo JM, Nuffer W. GLP-1 receptor agonists for type 2 diabetes mellitus: recent developments and emerging agents. Pharmacotherapy. 2014;34:1174–86.CrossRefPubMed Trujillo JM, Nuffer W. GLP-1 receptor agonists for type 2 diabetes mellitus: recent developments and emerging agents. Pharmacotherapy. 2014;34:1174–86.CrossRefPubMed
33.
Zurück zum Zitat Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373:11–22.CrossRefPubMed Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373:11–22.CrossRefPubMed
34.
Zurück zum Zitat Fujishima Y, Maeda N, Inoue K, Kashine S, Nishizawa H, Hirata A, et al. Efficacy of liraglutide, a glucagon-like peptide-1 (GLP-1) analogue, on body weight, eating behavior, and glycemic control, in Japanese obese type 2 diabetes. Cardiovasc Diabetol. 2012;11:107.CrossRefPubMedPubMedCentral Fujishima Y, Maeda N, Inoue K, Kashine S, Nishizawa H, Hirata A, et al. Efficacy of liraglutide, a glucagon-like peptide-1 (GLP-1) analogue, on body weight, eating behavior, and glycemic control, in Japanese obese type 2 diabetes. Cardiovasc Diabetol. 2012;11:107.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Krause GC, Lima KG, Dias HB, da Silva EFG, Haute GV, Basso BS, et al. Liraglutide, a glucagon-like peptide-1 analog, induce autophagy and senescence in HepG2 cells. Eur J Pharmacol. 2017;809:32–41.CrossRefPubMed Krause GC, Lima KG, Dias HB, da Silva EFG, Haute GV, Basso BS, et al. Liraglutide, a glucagon-like peptide-1 analog, induce autophagy and senescence in HepG2 cells. Eur J Pharmacol. 2017;809:32–41.CrossRefPubMed
36.
Zurück zum Zitat Panjwani N, Mulvihill EE, Longuet C, Yusta B, Campbell JE, Brown TJ, et al. GLP-1 receptor activation indirectly reduces hepatic lipid accumulation but does not attenuate development of atherosclerosis in diabetic male ApoE(−/−) mice. Endocrinology. 2013;154:127–39.CrossRefPubMed Panjwani N, Mulvihill EE, Longuet C, Yusta B, Campbell JE, Brown TJ, et al. GLP-1 receptor activation indirectly reduces hepatic lipid accumulation but does not attenuate development of atherosclerosis in diabetic male ApoE(−/−) mice. Endocrinology. 2013;154:127–39.CrossRefPubMed
37.
Zurück zum Zitat Ruttimann EB, Arnold M, Hillebrand JJ, Geary N, Langhans W. Intrameal hepatic portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous meal size in the rat via different mechanisms. Endocrinology. 2009;150:1174–81.CrossRefPubMed Ruttimann EB, Arnold M, Hillebrand JJ, Geary N, Langhans W. Intrameal hepatic portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous meal size in the rat via different mechanisms. Endocrinology. 2009;150:1174–81.CrossRefPubMed
38.
Zurück zum Zitat Tang-Christensen M, Vrang N, Larsen PJ. Glucagon-like peptide 1 (7-36) amide’s central inhibition of feeding and peripheral inhibition of drinking are abolished by neonatal monosodium glutamate treatment. Diabetes. 1998;47:530–7.CrossRefPubMed Tang-Christensen M, Vrang N, Larsen PJ. Glucagon-like peptide 1 (7-36) amide’s central inhibition of feeding and peripheral inhibition of drinking are abolished by neonatal monosodium glutamate treatment. Diabetes. 1998;47:530–7.CrossRefPubMed
39.
Zurück zum Zitat Xiao C, Dash S, Morgantini C, Adeli K, Lewis GF. Gut peptides are novel regulators of intestinal lipoprotein secretion: experimental and pharmacological manipulation of lipoprotein metabolism. Diabetes. 2015;64:2310–8.CrossRefPubMed Xiao C, Dash S, Morgantini C, Adeli K, Lewis GF. Gut peptides are novel regulators of intestinal lipoprotein secretion: experimental and pharmacological manipulation of lipoprotein metabolism. Diabetes. 2015;64:2310–8.CrossRefPubMed
40.
Zurück zum Zitat Farr S, Baker C, Naples M, Taher J, Iqbal J, Hussain M, et al. Central nervous system regulation of intestinal lipoprotein metabolism by glucagon-like peptide-1 via a brain–gut axis. Arterioscler Thromb Vasc Biol. 2015;35:1092–100.CrossRefPubMed Farr S, Baker C, Naples M, Taher J, Iqbal J, Hussain M, et al. Central nervous system regulation of intestinal lipoprotein metabolism by glucagon-like peptide-1 via a brain–gut axis. Arterioscler Thromb Vasc Biol. 2015;35:1092–100.CrossRefPubMed
41.
Zurück zum Zitat Taher J, Baker CL, Cuizon C, Masoudpour H, Zhang R, Farr S, et al. GLP-1 receptor agonism ameliorates hepatic VLDL overproduction and de novo lipogenesis in insulin resistance. Mol Metab. 2014;3:823–33.CrossRefPubMedPubMedCentral Taher J, Baker CL, Cuizon C, Masoudpour H, Zhang R, Farr S, et al. GLP-1 receptor agonism ameliorates hepatic VLDL overproduction and de novo lipogenesis in insulin resistance. Mol Metab. 2014;3:823–33.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Parlevliet ET, Wang Y, Geerling JJ, Schroder-Van der Elst JP, Picha K, O’Neil K, et al. GLP-1 receptor activation inhibits VLDL production and reverses hepatic steatosis by decreasing hepatic lipogenesis in high-fat-fed APOE*3-Leiden mice. PLoS ONE. 2012;7:e49152.CrossRefPubMedPubMedCentral Parlevliet ET, Wang Y, Geerling JJ, Schroder-Van der Elst JP, Picha K, O’Neil K, et al. GLP-1 receptor activation inhibits VLDL production and reverses hepatic steatosis by decreasing hepatic lipogenesis in high-fat-fed APOE*3-Leiden mice. PLoS ONE. 2012;7:e49152.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Kooijman S, Wang Y, Parlevliet ET, Boon MR, Edelschaap D, Snaterse G, et al. Central GLP-1 receptor signalling accelerates plasma clearance of triacylglycerol and glucose by activating brown adipose tissue in mice. Diabetologia. 2015;58:2637–46.CrossRefPubMedPubMedCentral Kooijman S, Wang Y, Parlevliet ET, Boon MR, Edelschaap D, Snaterse G, et al. Central GLP-1 receptor signalling accelerates plasma clearance of triacylglycerol and glucose by activating brown adipose tissue in mice. Diabetologia. 2015;58:2637–46.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Girona J, Ibarretxe D, Plana N, Guaita-Esteruelas S, Amigo N, Heras M, et al. Circulating PCSK9 levels and CETP plasma activity are independently associated in patients with metabolic diseases. Cardiovasc Diabetol. 2016;15:107.CrossRefPubMedPubMedCentral Girona J, Ibarretxe D, Plana N, Guaita-Esteruelas S, Amigo N, Heras M, et al. Circulating PCSK9 levels and CETP plasma activity are independently associated in patients with metabolic diseases. Cardiovasc Diabetol. 2016;15:107.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R, et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet. 2016;387:679–90.CrossRefPubMed Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R, et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet. 2016;387:679–90.CrossRefPubMed
46.
Zurück zum Zitat Petit JM, Cercueil JP, Loffroy R, Denimal D, Bouillet B, Fourmont C, et al. Effect of liraglutide therapy on liver fat content in patients with inadequately controlled type 2 diabetes: the Lira-NAFLD study. J Clin Endocrinol Metab. 2017;102:407–15.PubMed Petit JM, Cercueil JP, Loffroy R, Denimal D, Bouillet B, Fourmont C, et al. Effect of liraglutide therapy on liver fat content in patients with inadequately controlled type 2 diabetes: the Lira-NAFLD study. J Clin Endocrinol Metab. 2017;102:407–15.PubMed
47.
Zurück zum Zitat Baumeier C, Schluter L, Saussenthaler S, Laeger T, Rodiger M, Alaze SA, et al. Elevated hepatic DPP4 activity promotes insulin resistance and non-alcoholic fatty liver disease. Mol Metab. 2017;6:1254–63.CrossRefPubMedPubMedCentral Baumeier C, Schluter L, Saussenthaler S, Laeger T, Rodiger M, Alaze SA, et al. Elevated hepatic DPP4 activity promotes insulin resistance and non-alcoholic fatty liver disease. Mol Metab. 2017;6:1254–63.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Ao N, Yang J, Wang X, Du J. Glucagon-like peptide-1 preserves non-alcoholic fatty liver disease through inhibition of the endoplasmic reticulum stress-associated pathway. Hepatol Res. 2016;46:343–53.CrossRefPubMed Ao N, Yang J, Wang X, Du J. Glucagon-like peptide-1 preserves non-alcoholic fatty liver disease through inhibition of the endoplasmic reticulum stress-associated pathway. Hepatol Res. 2016;46:343–53.CrossRefPubMed
49.
Zurück zum Zitat Smits MM, Tonneijck L, Muskiet MH, Kramer MH, Pouwels PJ, Pieters-van den Bos IC, et al. Twelve week liraglutide or sitagliptin does not affect hepatic fat in type 2 diabetes: a randomised placebo-controlled trial. Diabetologia. 2016;59:2588–93.CrossRefPubMed Smits MM, Tonneijck L, Muskiet MH, Kramer MH, Pouwels PJ, Pieters-van den Bos IC, et al. Twelve week liraglutide or sitagliptin does not affect hepatic fat in type 2 diabetes: a randomised placebo-controlled trial. Diabetologia. 2016;59:2588–93.CrossRefPubMed
50.
Zurück zum Zitat Tang A, Rabasa-Lhoret R, Castel H, Wartelle-Bladou C, Gilbert G, Massicotte-Tisluck K, et al. Effects of insulin glargine and liraglutide therapy on liver fat as measured by magnetic resonance in patients with type 2 diabetes: a randomized trial. Diabetes Care. 2015;38:1339–46.CrossRefPubMed Tang A, Rabasa-Lhoret R, Castel H, Wartelle-Bladou C, Gilbert G, Massicotte-Tisluck K, et al. Effects of insulin glargine and liraglutide therapy on liver fat as measured by magnetic resonance in patients with type 2 diabetes: a randomized trial. Diabetes Care. 2015;38:1339–46.CrossRefPubMed
51.
Zurück zum Zitat Du Y, Li S, Cui CJ, Zhang Y, Yang SH, Li JJ. Leptin decreases the expression of low-density lipoprotein receptor via PCSK9 pathway: linking dyslipidemia with obesity. J Transl Med. 2016;14:276.CrossRefPubMedPubMedCentral Du Y, Li S, Cui CJ, Zhang Y, Yang SH, Li JJ. Leptin decreases the expression of low-density lipoprotein receptor via PCSK9 pathway: linking dyslipidemia with obesity. J Transl Med. 2016;14:276.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Cui CJ, Li S, Zhu CG, Sun J, Du Y, Zhang Y, et al. Enhanced pro-protein convertase subtilisin/kexin type 9 expression by C-reactive protein through p38MAPK-HNF1alpha pathway in HepG2 cells. J Cell Mol Med. 2016;20:2374–83.CrossRefPubMedPubMedCentral Cui CJ, Li S, Zhu CG, Sun J, Du Y, Zhang Y, et al. Enhanced pro-protein convertase subtilisin/kexin type 9 expression by C-reactive protein through p38MAPK-HNF1alpha pathway in HepG2 cells. J Cell Mol Med. 2016;20:2374–83.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Miao J, Manthena PV, Haas ME, Ling AV, Shin DJ, Graham MJ, et al. Role of insulin in the regulation of proprotein convertase subtilisin/kexin type 9. Arterioscler Thromb Vasc Biol. 2015;35:1589–96.CrossRefPubMedPubMedCentral Miao J, Manthena PV, Haas ME, Ling AV, Shin DJ, Graham MJ, et al. Role of insulin in the regulation of proprotein convertase subtilisin/kexin type 9. Arterioscler Thromb Vasc Biol. 2015;35:1589–96.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Levenson AE, Haas ME, Miao J, Brown RJ, de Ferranti SD, Muniyappa R, et al. Effect of leptin replacement on PCSK9 in ob/ob mice and female lipodystrophic patients. Endocrinology. 2016;157:1421–9.CrossRefPubMedPubMedCentral Levenson AE, Haas ME, Miao J, Brown RJ, de Ferranti SD, Muniyappa R, et al. Effect of leptin replacement on PCSK9 in ob/ob mice and female lipodystrophic patients. Endocrinology. 2016;157:1421–9.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Dong B, Singh AB, Fung C, Kan K, Liu J. CETP inhibitors downregulate hepatic LDL receptor and PCSK9 expression in vitro and in vivo through a SREBP2 dependent mechanism. Atherosclerosis. 2014;235:449–62.CrossRefPubMedPubMedCentral Dong B, Singh AB, Fung C, Kan K, Liu J. CETP inhibitors downregulate hepatic LDL receptor and PCSK9 expression in vitro and in vivo through a SREBP2 dependent mechanism. Atherosclerosis. 2014;235:449–62.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Zelcer N, Hong C, Boyadjian R, Tontonoz P. LXR regulates cholesterol uptake through Idol-dependent ubiquitination of the LDL receptor. Science. 2009;325:100–4.CrossRefPubMedPubMedCentral Zelcer N, Hong C, Boyadjian R, Tontonoz P. LXR regulates cholesterol uptake through Idol-dependent ubiquitination of the LDL receptor. Science. 2009;325:100–4.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Rudling M, Angelin B. Stimulation of rat hepatic low density lipoprotein receptors by glucagon. Evidence of a novel regulatory mechanism in vivo. J Clin Investig. 1993;91:2796–805.CrossRefPubMedPubMedCentral Rudling M, Angelin B. Stimulation of rat hepatic low density lipoprotein receptors by glucagon. Evidence of a novel regulatory mechanism in vivo. J Clin Investig. 1993;91:2796–805.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368:1696–705.CrossRefPubMed Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368:1696–705.CrossRefPubMed
59.
Zurück zum Zitat Wang Y, Ye J, Li J, Chen C, Huang J, Liu P, et al. Polydatin ameliorates lipid and glucose metabolism in type 2 diabetes mellitus by downregulating proprotein convertase subtilisin/kexin type 9 (PCSK9). Cardiovasc Diabetol. 2016;15:19.CrossRefPubMedPubMedCentral Wang Y, Ye J, Li J, Chen C, Huang J, Liu P, et al. Polydatin ameliorates lipid and glucose metabolism in type 2 diabetes mellitus by downregulating proprotein convertase subtilisin/kexin type 9 (PCSK9). Cardiovasc Diabetol. 2016;15:19.CrossRefPubMedPubMedCentral
Metadaten
Titel
Liraglutide downregulates hepatic LDL receptor and PCSK9 expression in HepG2 cells and db/db mice through a HNF-1a dependent mechanism
verfasst von
Sheng-Hua Yang
Rui-Xia Xu
Chuan-Jue Cui
Yin Wang
Ying Du
Zhi-Guo Chen
Yu-Hong Yao
Chun-Yan Ma
Cheng-Gang Zhu
Yuan-Lin Guo
Na-Qiong Wu
Jing Sun
Bu-Xing Chen
Jian-Jun Li
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Cardiovascular Diabetology / Ausgabe 1/2018
Elektronische ISSN: 1475-2840
DOI
https://doi.org/10.1186/s12933-018-0689-9

Weitere Artikel der Ausgabe 1/2018

Cardiovascular Diabetology 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.