You are on page 1of 11

European Journal of Pharmacology 861 (2019) 172594

Contents lists available at ScienceDirect

European Journal of Pharmacology


journal homepage: www.elsevier.com/locate/ejphar

Full length article

Liraglutide improves insulin sensitivity in high fat diet induced diabetic T


mice through multiple pathways
Joseph Yi Zhoua, Anil Poudelb, Ryan Welchkob, Naveen Mekalaa,
⁎⁎ ⁎
Prashanth Chandramani-Shivalingappab, Mariana Georgeta Roscaa, , Lixin Lib,
a
College of Medicine, Central Michigan University, MI, 48859, USA
b
Department of Physician Assistant, College of Health Professions, Central Michigan University MI, 48859, USA

A R T I C LE I N FO A B S T R A C T

Keywords: Glucagon like peptide-1 (GLP-1) promotes postprandial insulin secretion. Liraglutide, a full agonist of the GLP-1
Adipocyte receptor, reduces body weight, improve insulin sensitivity, and alleviate Non Alcoholic Fatty Liver Disease
Metabolism (NAFLD). However, the underlying mechanisms remain unclear. This study aims to explore the underlying
Obesity mechanisms and cell signaling pathways involved in the anti-obesity and anti-inflammatory effects of liraglutide.
Insulin resistance
Mice were fed a high fat high sucrose diet to induce diabetes, diabetic mice were divided into two groups and
Liraglutide
injected with liraglutide or vehicle for 14 days. Liraglutide treatment improved insulin sensitivity, accompanied
with reduced expression of the phosphorylated Acetyl-CoA carboxylase-2 (ACC2) and upregulation of long chain
acyl CoA dehydrogenase (LCAD) in insulin sensitive tissues. Furthermore, liraglutide induced adenosine
monophosphate-activated protein kinase-α (AMPK-α) and Sirtuin-1(Sirt-1) protein expression in liver and
perigonadal fat. Liraglutide induced elevation of fatty acid oxidation in these tissues may be mediated through
the AMPK-Sirt-1 cell signaling pathway. In addition, liraglutide induced brown adipocyte differentiation in
skeletal muscle, including induction of uncoupling protein-1 (UCP-1) and PR-domain-containing-16 (PRDM-16)
protein in association with induction of SIRT-1. Importantly, liraglutide displayed anti-inflammation effect.
Specifically, liraglutide led to a significant reduction in circulating interleukin-1 β (IL-1 β) and interleukin-6 (IL-
6) as well as hepatic IL-1 β and IL-6 content. The expression of inducible nitric oxide synthase (iNOS-1) and
cyclooxygenase-2 (COX-2) in insulin sensitive tissues was also reduced following liraglutide treatment. In
conclusion, liraglutide improves insulin sensitivity through multiple pathways resulting in reduction of in-
flammation, elevation of fatty acid oxidation, and induction of adaptive thermogenesis.

1. Introduction adipocytes and skeletal muscle originate from the same precursor cells,
and the transcription factor PR-domain-containing 16 (PRDM16) con-
Obesity promotes insulin resistance through alteration of metabolic trols the bi-directional cell fate switch between brown fat and skeletal
and endocrine functions in adipose tissue. For instance, obesity is cor- muscle (Harms and Seale, 2013; Harms et al., 2014). BAT activation has
related with elevated levels of pro-inflammatory cytokines such as therefore emerged as an attractive therapeutic target for the treatment
Interleukin-1β (IL-1β), tissue necrosis factor-α (TNF-α), and of obesity.
Interleukin-6 (IL-6) which stimulate lipolysis and lead to hyperlipi- Glucagon like peptide-1 (GLP-1) is released primarily by the L-type
demia (Makki et al., 2013). Furthermore, inflammation and the ensuing cells of the ileal mucosa in response to nutrient ingestion, and increases
metabolic dysfunction is associated with reduced whole body energy postprandial insulin secretion (Drucker, 2002). Beneficial effects of
expenditure in both human and animal studies (Holmes, 2017). GLP-1 in patients with vascular disease, coronary heart disease, and
Brown adipose tissue (BAT) generates heat instead of ATP produc- metabolic syndrome have been reported (Tomas et al., 2011). GLP-1R
tion via uncoupling respiration through the uncoupling protein 1 (UCP- agonists induced moderate weight loss and reduced appetite in type 2
1) (Fuchs et al., 1988). UCP-1 deficient mice gained more body weight diabetic (T2D) patients (Namba et al., 2013; Tang et al., 2013; Zander
than wild-type controls (Feldmann et al., 2009). Importantly, brown et al., 2002). GLP-1 overexpression also promoted insulin induced


Corresponding author. College of Health Professions, Central Michigan University, Mount Pleasant, MI, 48858, USA.
⁎⁎
Corresponding author. College of Medicine, Central Michigan University, Mount Pleasant, MI, USA.
E-mail addresses: rosca1g@cmich.edu (M.G. Rosca), li6l@cmich.edu (L. Li).

https://doi.org/10.1016/j.ejphar.2019.172594
Received 13 June 2019; Received in revised form 1 August 2019; Accepted 7 August 2019
Available online 11 August 2019
0014-2999/ © 2019 Published by Elsevier B.V.
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

hepatic activation of insulin receptor substrate (IRS-1), reduction of (Colbert, GA) and housed in conventional cages at 22 °C on a 12-h light/
hepatic glucose production, and fatty acid synthesis in animal studies dark cycle. Starting at 9–10 weeks of age mice were fed with either
(Lee et al., 2007). GLP-1 analogues have also been reported to be ef- regular chow diet or high fat high sucrose diet (HFHS; 60% calories
fective in treating patients with nonalcoholic fatty liver disease from fat, and 35.7% from carbohydrate; Harlan Teklad, Indianapolis,
(NAFLD) and/or non-alcoholic steatohepatitis (NASH) through redu- IN). Mice fed with HFHS diet developed diabetes after 16 weeks of
cing hepatic fat content (Armstrong et al., 2016a, 2016b; Mells et al., feeding. The diet induced obesity (DIO) mice were further divided into
2012). The use of GLP-1R agonists therefore show promise as a therapy two groups and received daily intraperitoneal injections with either
for treating T2D and obesity-related metabolic disorders. liraglutide (0.2 mg/kg daily) or saline for 2 weeks. Chow diet mice
However, GLP-1 has a short half-life (1–2 min) and is degraded by received 2 weeks of saline injections. At the end of the two week in-
dipeptidyl peptidase-IV (DPP-IV) once released into the plasma jection period mice were fasted for 12 h before they were killed.
(Drucker, 2002). Liraglutide, a full GLP-1 receptor agonist with 97% Harvested tissues were flash frozen in liquid nitrogen and either stored
amino acid sequence similarity with human GLP-1, has long-lasting at −80 °C or fixed in 4% paraformaldehyde (PFA) solution in PBS for
effects and can be injected once daily due to its improved resistance to 48h before being embedded in paraffin. All experimental procedures
DPP-IV (Knudsen, 2010). Liraglutide has been shown to promote he- were approved by the Animal Care Committee at Central Michigan
patic and adipose insulin sensitivity, reduce hepatic steatosis. (Richard University.
and Lingvay, 2011). Liraglutide also enhanced the ability of insulin to
suppress lipolysis in subcutaneous adipose tissue and decreased de novo
2.3. Western blot analysis
lipogenesis in human primary hepatocytes (Armstrong et al., 2016b).
Therefore, liraglutide may be a potential medication for metabolic
Liver, skeletal muscle, and perigonadal fat were homogenized in
syndrome and disease-modifying intervention in NAFLD (Armstrong
radioimmune precipitation assay (RIPA) lysis buffer containing a pro-
et al., 2016b). However, the underlying mechanisms for these benefits
tease inhibitor mixture, EDTA, phenylmethylsulfonyl fluoride, sodium
are not clearly understood and the role of liraglutide in enhancing
fluoride, and sodium orthovanadate (Santa Cruz, Dallas, TX). Protein
thermogenesis has not yet been proven (Heppner et al., 2015). Whether
concentrations were determined using a Bio-Rad protein assay kit.
liraglutide reduces obesity related release of cytokines is also not well
Western blot analyses were performed using standardized laboratory
established. This study was therefore designed to explore the under-
protocol. Signals were detected using Luminata Forte Western HRP
lying mechanisms of the anti-obesity effect and potential anti-in-
substrate (EMD Millipore, MA, USA). Images were taken using LI-COR
flammatory effects of liraglutide.
Odyssey Fc imaging system (LI-COR Biosciences, NE, USA).

2. Materials and methods


2.4. Quantitation of hepatic, adipose tissues, and skeletal muscle mRNA
2.1. Reagents
RNA was extracted from liver, adipose, and skeletal muscle tissues
Liraglutide was purchased from BACHEM (Torrance, CA). Anti-PR using TRIzol (Invitrogen) as described previously (Li et al., 2011a).
domain containing 16 (Prdm16), anti-Inducible Nitric Oxide Synthase RNA was reverse transcribed using the Applied Biosystems High Ca-
(iNOS), anti-cyclooxygenase-2(COX-2), anti-uncoupling protein-1 pacity RNA-to-cDNA kit (Thermo Fisher Scientific) following the man-
(UCP1), anti- Peroxisome proliferator-activated receptor gamma coac- ufacturer's protocol. SYBR Green PCR master mix (Applied Biosystems,
tivator 1-α (PGC1α), anti-Peroxisome proliferator-activated receptor γ Foster City, CA) was used for quantitative PCR performed using the ABI
(PPARγ), anti-Peroxisome proliferator-activated receptor α(PPARα), PRISM machine 7900HT sequence detection system (SDS software
and anti-Long Chain Acyl CoA Dehydrogenase (LCAD) antibodies were version 2.3). GAPDH was used as an internal control. The primers were
purchased from Abcam (Cambridge, MA). Anti-p-acetyl-CoA carbox- synthesized by integrated DNA technologies. Primer pairs used to am-
ylase, anti-(p-ACC2), anti-p-5′ AMP-activated protein kinaseα (p- plify these genes are shown in Table 1. The relative mRNA expression
AMPKα), anti-AMPKα, anti-Sirt-1, anti-mouse IgG, and anti-rabbit Ig was determined using the comparative Ct method by calculating
were from Cell Signaling Technology (Danvers, MA). Anti-β actin was 2−ΔΔCt.
from Sigma Aldrich (St. Louis. MO). Anti-β tubulin and anti-CCAAT-
enhancer-binding proteins-α (C/EBPα) were from Santa Cruz. Mouse 2.5. Intraperitoneal glucose tolerance test
IL1-β and IL-6 Elisa kits were purchased from R & D system, DuoSet
ELISA development system. Mice were fasted overnight then fasting plasma glucose was mea-
sured from tail vein blood using Compact Accu-Check glucometer
2.2. Animals (Roche, Diagnostics, Indianapolis, IN). The mice received in-
traperitoneal injections with 10% glucose at 1 g/kg body weight, and
Male C57BL/6 mice were purchased from Charles River laboratories plasma glucose was measured at 15, 30, 60, and 120 min after injection.

Table 1
Primer sequences.
Primers Forward primer Reverse primer

GAPDH ACCCAGAAGACTGTGGATGG GGATGCAGGGATGATGTTCT


Cidea GCCAGAACTATTCGCTGC TC GCCGCGTCTCTTAAATCACT
PPARϒ ACCACTCGCATT CCT TTG AC AACCATTGGGTCAGCTCT TG
UCP-1 ACTGCCACACCTCCAGTCATT CTTTGCCTCACTCAGGATTGG
PGC-1 α GCAGAGACAAATGTGCTTCG TCTGGGGTCAGAGGAAGAGA
PPAR α CCAATCAGAGATGCCACC TT TCTCCAAGGAGTCTGGCACT
LCAD CACCACACAGAATGGGAGAAAG AATGCCGCCATGTTTCTCTG
IL-1β GCCACCTTTTGACAGTGATG ATGTGCTGCTGCGAGATTTG
IL-6 AGACAAAGCCAGAGTCCTTCAG GAGAGCATTGGAAATTGGGGTAGG
Cox-2 CAGCCAGGCAGCAAATCCTTG CATAGAATCCAGTCCGGGTACAG
Cebp-α AAGCCAAGAAGTCGGTGGAC TCACTGGTCAACTCCAGCAC

2
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

2.6. IL-1β and IL-6 measurement (170.24 ± 3.45 pg/ml vs. 65.0 ± 2.34 pg/ml, P < 0.005, n = 5),
respectively. Hepatic IL-1β (Fig. 2B) and IL-6 (Fig. 2E) levels were also
Plasma and tissue IL-1β and IL-6 levels were measured using the significantly reduced by 26% (135.8 ± 44.02, 60.07 ± 1.401,
mouse IL-1β and IL6 Duoset Elisa kit following the manufacturer's P < 0.001, n = 5) and by 78% (201.45 ± 4.56 pg/ml vs
protocol. Briefly, 96 well plates were coated with the capture antibody, 43.04 ± 2.56 pg/ml, P < 0.005, n = 5) respectively by the liraglutide
incubated with samples and standards for 2 h at RT, then followed by treatment. These results are in agreement with the IL-1β and IL-6 gene
exposure to the detection antibody. Plates were then treated with expression in the liver, which were increased by the HFHS diet and
streptavidin-HRP-conjugate and calorimetric changes were measured reduced by the liraglutide treatment (Fig. 2H–I). Both protein and gene
by optical density. expression of IL-1β were reduced in perigonadal fat tissue by liraglutide
(Fig. 2C and H). However, IL-6 expression was not changed in perigo-
2.7. Hematoxylin and eosin (H&E) staining nadal fat after liraglutide treatment (Fig. 2I–F). In contrast to the
findings in the liver, neither IL-1β nor IL-6 levels in skeletal muscle
Mouse tissues were fixed in 4% PFA before being embedded in (data not shown) were altered following liraglutide treatment.
paraffin. Sections were stained with Hematoxylin and eosin (H&E) We investigated the iNOS-COX-2 pro-inflammatory signaling path-
staining using a standard protocol. An Axiocam 506 color camera ways as well in insulin sensitive tissues. Inducible nitric-oxide synthase
captured images with a Carl Zeiss Axio Imager M2 microscope at the (iNOS) specifically binds to S-nitrosylates cyclooxygenase-2 (COX-2)
magnification indicated in the figure legends. leading to enhanced catalytic activity of COX-2 (Kim et al., 2005),
supporting the roles of these two major mediators of inflammation in
2.8. Statistical analysis promoting obesity-associated pathologies. In addition to elevated cy-
tokine levels, HFHS diet induced obesity was correlated with elevated
Data was analyzed by non-parametric, two-sided Student's t-test gene and protein expression of iNOS and COX-2 in our study
when comparing two groups. Data was analyzed by one-way ANOVA (Fig. 3A–G). Liraglutide treatment significantly reduced HFHS diet in-
followed by Bonfferoroni post-hoc tests when 3 groups were compared. duced iNOS protein expression by 37% in liver tissue (P < 0.005,
All data analyses were performed using GraphPad Prism V7.0 software n = 8), 65% in perigonadal fat (P < 0.05, n = 5), and 73%
(GraphPad Software Inc., La Jolla, CA). A P value < 0.05 was con- (P < 0.005, n = 5) in skeletal muscle when compared to diet induced
sidered to be statistically significant. obesity (DIO) animals injected with saline (Fig. 3A–C). Similarly, the
expression of COX-2 was reduced by 28% (P < 0.005, n = 8) in liver
3. Results tissue, 80% in perigonadal fat (P < 0.005, n = 8), and 41%
(P < 0.05, n = 8) in skeletal muscle after two weeks of treatment with
3.1. Liraglutide induced weight loss, reduced food intake, and improved liraglutide compared to saline treated DIO mice. (Fig. 3D–F). Cox-2
glucose tolerance in HFHS diet-fed mice mRNA expression was increased by the HFHS diet, and significantly
reduced after liraglutide treatment in all three insulin sensitive tissues
Mice developed diabetes after 4 months of HFHS diet feeding as well (Fig. 3G).
as impaired glucose tolerance, significant elevation of body weight, and
increased liver and perigondadal fat mass when compared to mice fed a 3.3. Liraglutide activated the AMPK-SIRT1 energy expenditure signaling
regular chow diet (Fig. 1A–F). Two weeks of liraglutide treatment sig- pathway in the liver and adipocytes in HFHS diet mice
nificantly reduced fasting blood glucose (210.5 ± 19.53 mg/dl vs
87.0 ± 6.66 mg/dl, P < 0.05, Fig. 1A) and improved intraperitoneal 5′ AMP-activated protein kinase α (AMPKα) and sirtuin-1 (SIRT-1)
glucose tolerance test (IPGTT) when compared with the HFHS diet play key roles in regulating cellular energy status (Cantó and Auwerx,
group (Fig. 1B–C). A 10% body weight loss (49.5 ± 1.14 g vs 2009; Feige and Auwerx, 2007). HFHS diet led to a significant reduc-
44.17 ± 1.19 g; P < 0.01, n = 5) was observed in the liraglutide tion of both AMPKα phosphorylation (Fig. 4A–B) and SIRT-1 protein
treated mice at the end of the study (Fig. 1D) which was associated with expression (Fig. 4D–E) in the liver and perigonadal fat when compared
a significant reduction of food intake in the first 4 days of injections with mice fed a chow diet. We observed a significant elevation in P-
(Fig. 1H). AMPKα in both liver tissue (67%, P < 0.005, n = 8) and perigonadal
A significant reduction in liver and perigonadal fat mass was also fat (by 40%, P < 0.005, n = 5), but not in skeletal muscle, after lir-
observed after two weeks of treatment with liraglutide (Fig. 1E–F). aglutide treatment (Fig. 4C). In agreement with our AMPKα findings,
Liver mass was reduced by 19% (P < 0.01, n = 11; Fig. 1E) and was liraglutide induced a significant upregulation of SIRT-1 protein ex-
accompanied by reduced hepatocyte ballooning (Fig. 1G) suggesting a pression in the liver (by 22%, P < 0.005, n = 8) and perigonadal fat
decrease in liver fat accumulation. While perigonadal fat mass was (108%, P < 0.005, n = 8) when compared with the HFHS fed mice
reduced by 32% (P < 0.01, n = 11; Fig. 1F), brown fat mass was not (Fig. 4D–E). SIRT-1 protein level was increased by 208% (P < 0.005,
affected after liraglutide treatment (data not shown). Taken together, n = 5) in skeletal muscle with liraglutide treatment (Fig. 4F). Together,
our data indicates that liraglutide inhibits food intake in the first four our results suggest that liraglutide improves the bioenergetics status
days of treatment, reduces fasting plasma glucose, and improves glu- through activation of the AMPKα-SIRT-1 energy-sensing network in the
cose tolerance in association with a reduction in liver weight and liver and perigonadal fat. However, the protective effect of liraglutide
perigonadal fat mass. on skeletal muscle bioenergetics may act through different pathways.

3.2. Liraglutide displayed an anti-inflammatory effect in insulin sensitive 3.4. Liraglutide improved fatty acid oxidation in the liver, perigonadal fat,
tissues of HFHS diet-fed mice and skeletal muscle of HFHS diet-fed mice

To better understand the protective effect of liraglutide against Long-chain acylCoA dehydrogenase (LCAD) is a mitochondrial en-
obesity induced inflammation, IL-6, IL-1β, and other inflammation zyme that participates in fatty acid β-oxidation. We examined the
marker were examined in our study. HFHS diet resulted in increased regulation of ACC2 and LCAD by liraglutide in order to further de-
gene expression of IL-1 β and IL-6 in both liver and adipose tissue lineate the effects of liraglutide on fatty acid oxidation in DIO mice. As
(Fig. 2H–I). Liraglutide treatment significantly reduced plasma IL-1β shown in Fig. 5A–C, a significant increase of LCAD expression was
(Fig. 2A) and IL-6 (Fig. 2D) levels by 27% (456.2 ± 7.689 pg/ml vs. observed in the liver (14%, P < 0.005, n = 5), perigonadal fat (by
332.6 ± 13.81 pg/ml, P < 0.005, n = 5) and 61% 47%, P < 0.005, n = 5) and skeletal muscle (by 49%, P < 0.005,

3
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

Fig. 1. Liraglutide reduces food intake, body weight and improves glucose homeostasis in HFHS diet mice Food intake was measured during the 2 weeks period of
injection. Fasting blood glucose were measured, and IGTT was performed at the end of the study. Liver and tissue weight were measured before snap frozen in liquid
nitrogen. A. Fasting blood glucose; B. Intra-peritoneal glucose tolerance test (IGTT); C. Area under curve (IGTT); D. Body weight; E. Liver weight; F. Perigonada fat
mass F. Liver tissue stained with H&E. H. Food intake. Data is shown as mean ± S.E.M., n = 5–12 experiments. *P < 0.05, **, P < 0.005.

n = 5) during HFHS diet and was further increased with liraglutide tissue (45% reduction, P < 0.05, n = 5), perigonadal fat (74% reduc-
treatment. These results indicate that in the presence of an excess of tion, P < 0.05, n = 5), and skeletal muscle (37% reduction n = 5,
energetic substrates, these insulin sensitive tissues experience an in- P < 0.005) following liraglutide treatment when compared to mice fed
crease in the enzymatic ability to oxidize long-chain fatty acids, and a HFHS diet (Fig. 5D–F), Hence, liraglutide increased LCAD expression
that liraglutide facilitates this ability. and decreased ACC phosphorylation in all three peripheral tissues ex-
Additionally, ACC2 phosphorylation decreased significantly in liver amined. Taken together, our results clearly indicate that liraglutide

4
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

Fig. 2. Liraglutide reduces systemic and hepatic IL-1 β and IL-6 level in HFHS diet mice. Blood samples were collected from tail vein at the end of the study. Plasma,
liver, fat IL1- β and IL-6 content were measured using mouse douset Elisa kit, and gene expression was measured using real-time PCR. A. Plasma IL-1 β, B. Liver IL-1 β
content, C. Perigonadal fat IL-1 β content. D. Plasma IL-6 content, E. Hepatic IL-6 content, F. Perigonadal fat IL-6 content, H. IL-1 β mRNA expression in liver and
perigonadal fat tissue. I. IL-6 mRNA expression in liver and fat tissue. Data is shown as mean ± S.E.M., n = 5–6. *P < 0.05, **P < 0.005.

improved fatty acid oxidation in all insulin sensitive tissues studied. 3.7. Liraglutide promoted brown fat differentiation in skeletal muscles of
HFHS diet fed mice

3.5. Liraglutide reduced adipogenesis in perigonadal fat and skeletal muscle To further determine the effect of liraglutide on energy expenditure,
tissue in mice fed a HFHS diet we investigated brown fat differentiation in skeletal muscle and peri-
gonadal fat. We observed that liraglutide induced a 229% increase
High fat high sucrose diet or obesity suppressed the activity of (P < 0.005) in UCP-1 expression and a 175% increase (P < 0.005) in
AMPK while activating the expression of lipogenetic transcription fac- PRDM-16 in the skeletal muscle of DIO mice when compared to control
tors such as PPARγ and C/EBPα, two major regulators of adipocyte mice (Fig. 8A–B). Liraglutide therefore failed to induce beige fat in
differentiation during adipogenesis, in our study. Both gene and protein HFHS diet mice. Furthermore, the expression of PPARα, another brown
expression of PPARγ were reduced in all three insulin sensitive per- fat marker and inducer of mitochondrial biosynthesis, was significantly
ipheral tissues after liraglutide treatment (Fig. 6E–J). As shown in enhanced in skeletal muscle of DIO mice treated with liraglutide (109%
Fig. 6E–G, there was a 13% decrease in PPARγ protein expression in increase, P < 0.05, n = 5, Fig. 8C). In concordance with the protein
liver tissue (P < 0.005, n = 5), a 27% reduction in perigonadal fat expression, gene expression markers of BAT were also elevated
(P < 0.05, n = 5), and a 96% decrease in skeletal muscle (P < 0.005, (Fig. 8D). In contrast, liraglutide failed to induce expression of UCP-1 or
n = 5). Liraglutide also significantly decreased C/EBPα protein ex- PRDM-16 in perigonadal fat in DIO mice (data now shown). Overall,
pression in liver tissue (41%, P < 0.005, n = 5), perigonadal fat (by our data suggests that liraglutide induced brown fat differentiation in
43%, P < 0.005, n = 5) and skeletal muscle (by 44%, P < 0.005, skeletal muscle.
n = 5) of DIO mice (Fig. 6A–C). Similar findings were observed in C/
EBPα mRNA levels (Fig. 6D). In summary, liraglutide inhibited adipo- 4. Discussion
genesis in insulin sensitive tissues in DIO mice.
In humans, and experimental animal models, HFHS diet often in-
itiates a series of molecular events that lead to obesity, insulin re-
3.6. Liraglutide induced PPARα in the liver and perigonadal fat of HFHS sistance, or metabolic syndrome. Obesity is associated with an in-
diet-fed mice flammatory state that contributes to the development of insulin
resistance in peripheral tissues. In the present study, we demonstrated
HFHS diet induced a reduction of PPARα gene expression in the that liraglutide improves insulin sensitivity, increases fatty acid oxi-
liver and perigonadal fat (Fig. 7C–D). Liraglutide induced 46% increase dation, and reduces systemic and tissue inflammation induced by
in the level of PPARα in liver and 112% increase (P < 0.05, n = 5) in obesity in the DIO mouse model. Crucially, we found that liraglutide
perigonadal fat in comparison to saline treated DIO mice (Fig. 7A–B). induces brown adipocyte differentiation in skeletal muscle in diet

5
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

Fig. 3. Liraglutide reduces iNOS and COX-2 expression in liver, perigonadal fat and skeletal muscle tissue in HFHS diet mice Mice tissues were collected, protein and
RNA were extracted. Protein expression of iNOS and COX-2 were assessed by Western blot, mRNA expression was assessed by real-time PCR. Representaive image of
western blotting and protien expression level of A. iNOS expression in liver tissue; B. Perigonadal fat iNOS expression in perigonadal fat; C. iNOS from skeletal
muscle; D. COX-2 expression in liver; E. COX-2 expression in perigonadoal fat. F. COX-2 expression in skeletal muscle. G. Cox-2 mRNA expression in liver, peri-
gonadal fat and skeletal muscle are shown. Data is shown as mean ± S.E.M., n = 5–12. β-actin and β-tubulin were used as loading control *, P < 0.05, **,
P < 0.005.

Fig. 4. Liraglutide activates AMPKα-SIRT-1 cell signaling in liver and perigonadal fat in HFHS diet mice. Mice tissues were collected, and protein were extraced.
Expression of AMPKα and SIRT-1 were measured by western blotting. A. AMPKα expression in liver tissue; B. AMPKα expression in paragonadal fat; C. AMPKα
expression in skeletal tissue, D. SIRT-1 expression in liver, E. SIRT-1 expresion in perigonadal fat, F. SIRT-1 expression in skeletal muscle. Data is shown as
mean ± S.E.M., n = 5–12. β-actin and β-tubulin were used as loading control *, P < 0.05, **, P < 0.005.

6
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

Fig. 5. Liraglutide increases fatty acid oxdation in liver, perigondal and skeletal muscle in HFHS diet mice Mice tissues were collected, and protein and RNA were
extraced. Expression of LCAD and ACC were measured by western blotting and real-time PCR. A. LCAD expression in liver tissue; B. LCAD expression in perigonadal
fat; C. LCAD expression in skeletal tissue; D. ACC expression in liver, E. ACC expression in perigonadal fat, F. ACC expresison in skeletal muscle. G. LCAD mRNA
expression in liver, perigonadal fat and skeletal muscle tissue. Data is shown as mean ± S.E.M., n = 5–12. β-actin and β-tubulin were used as loading control *,
P < 0.05, **, P < 0.005.

induced obese diabetic mice. human liver and improve insulin sensitivity, the mechanisms are not
IL-6 and IL-1β are systemic inflammatory cytokines involved in well understood (Armstrong et al., 2016b). Impaired fatty acid oxida-
obesity-linked pathological states including hepatic steatosis and in- tion and the ensuing decrease in energy expenditure contribute to the
sulin resistance (Bastard et al., 2002; Kern et al., 2001; Parthsarathy development of insulin resistance and type 2 diabetes (Montgomery and
and Hölscher, 2013; Vozarova et al., 2001). In adipose tissue, in- Turner, 2015). HFHS diet has been shown to alter β-oxidation in var-
filtrating macrophages are a major source of pro-inflammatory cyto- ious tissues and contribute to liver steatosis (Kakimoto and
kines and chemokines, and contribute to up to 35% of circulating IL-6 Kowaltowski, 2016). ACC and LCAD are key enzymes in regulating fatty
levels (Kim et al., 2009). Activated macrophages further propagate in- acid synthesis and oxidation, respectively (Abu-Elheiga, L et al., 2003).
flammation and induce insulin resistance in liver, white adipose tissue, Two ACC isoforms, ACC1 and ACC2, are expressed in mammals. The
and skeletal muscle. These insulin sensitive tissues are important sites ACC2 isoform regulates mitochondrial malonyl-CoA that controls fatty
of inflammation in obesity (Olefsky and Glass, 2010; Senn et al., 2002; acids oxidation. A significant elevation of fatty acid oxidation rates
Sheedfar et al., 2013). Liraglutide has been shown to reduce pro-in- have been observed in Acc2−/− mutant mice when compared to wild
flammatory cytokines including IL-6 and IL-1β in the brain type mice (Wakil and Abu-Elheiga, 2009). Additionally, LCAD defi-
(Parthsarathy and Hölscher, 2013) and endothelial cells (Krasner et al., ciency has been linked to mitochondrial dysfunction, fatty liver, and
2014). However, its anti-inflammatory effect in systemic and insulin- hepatic insulin resistance in mice (Zhang et al., 2007). Importantly,
sensitive tissues has not been well studied. Our results revealed for the ACC is the downstream effector of AMPK signaling, which is impaired
first time that liraglutide significantly reduces the increased circulating by high-fat diet as shown in our study. Our study indicates an important
IL-6 and IL-1β levels induced by a HFHS diet. Furthermore, we also role of liraglutide in the upregulation of mitochondrial fatty acid oxi-
demonstrated that liraglutide reduces IL-6 and IL-1β content in the liver dation. We observed that liraglutide modulated LCAD and ACC2, two
and fat tissues in HFHS diet induced diabetic mice. To our knowledge, key enzymes in fatty acid metabolism. Specifically, a reduction of ACC2
this is the first report on the anti-inflammatory effects of liraglutide on phosphorylation in association with an upregulation of LCAD after lir-
both systemic and hepatic IL-1 β and IL-6 content. COX-2 enzyme ac- aglutide treatment was observed in liver, adipose tissue and skeletal
tivates inflammation in part through induction of pro-inflammatory muscle. Hence, an increase of mitochondrial fatty oxidation in insulin
transcription factors, such as NF-kB, and cytokines such as IL-6 and sensitive tissues during the diabetic state after liraglutide treatment
TNF-α (Leclercq et al., 2004; Yu et al., 2006). Our results showed that may account for the reduced body weight and improved insulin sensi-
liraglutide significantly reduces iNOS and COX-2 in liver, skeletal tivity observed.
muscle, and adipose tissue. In summary, our observations clearly in- We further investigated cellular signaling pathways that participate
dicate that liraglutide improves insulin sensitivity through reduction of in liraglutide induced fatty acid β-oxidation. AMPK is an enzyme which
both systemic and liver pro-inflammation cytokines and inhibition of plays an important role in regulating metabolic and energy homeostasis
iNOS and Cox-2 expression in insulin sensitive tissues. (Dyck et al., 1996). AMPK phosphorylates ACC2 at Ser 79 and inhibits
Although liraglutide is known to decrease de novo lipogenesis in the its enzymatic activity (Fullerton et al., 2013; Marcinko and Steinberg,

7
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

Fig. 6. Liraglutide elevates C/EBP α and reduces PPAR-γ expresion in liver, perigonadal fat and skeletal muscle in HFHS diet mice Mice tissues were collected, and
protein and RNA were extraced. Expression of C/EBPα and PPARα was measured by both western blotting and real-time PCR. A. C/EBPα expression in liver tissue; B.
C/EBPα in perigonadal fat; C. C/EBPα expression in skeletal tissue, D. PPARα expression in liver, E. PPARα expression in perigonadal fat. F. PPAR-α expression in
skeletal muscle. G. Hepatic PPARγ mRNA expression. H. Perigonadal fat PPAR-γ mRNA expresson. I. Skeletal muscle PPARγ mRNA. Data is shown as
mean ± S.E.M., n = 5–12. β-actin and β-tubulin were used as loading control *, P < 0.05, **, P < 0.005.

2014). Our results indicate that the inhibition of ACC2 phosphorylation through down regulation of PPARγ induced by activation of the AMPK-
and elevation of LCAD after liraglutide treatment is associated with SIRT-1 pathway (Inoue et al., 2015). In agreement with the findings in
activation of AMPK-α in both liver and fat tissues. However, liraglutide cardiac tissue, our study indicates that liraglutide increases SIRT-1 ex-
fails to activate AMPK phosphorylation in skeletal muscle, suggesting pression and elevates fatty acid oxidation in insulin sensitive tissues,
the modulatory role of liraglutide on fatty acid oxidation in skeletal including liver, adipose, and skeletal muscle, suggesting that the AMPK-
muscle may not occur through the AMPK-α pathway. SIRT-1 cell signaling pathway is a key mediator of liraglutide induced
SIRT-1 controls hepatic gluconeogenesis by regulating transcription fatty acid oxidation in liver and fat tissues of DIO mice.
factors and co-regulators such as PPARγ and PGC-1α (Cantó and The anti-adipogenic role of liraglutide is also supported by its in-
Auwerx, 2009), and has been shown to ameliorate hepatic steatosis in hibitory effect on lipogenic signaling pathways. A high fat diet or
T2D patients with NAFLD complications (Xu et al., 2014). The AMPK- obesity suppresses AMPK activity while activating the expression of
SIRT-1 pathway therefore plays a major role in regulating metabolism. adipogenic transcription factors such as PPARγ and C/EBPα in the liver
Liraglutide is reported to inhibit cardiac steatosis in diabetic mice (Armstrong et al., 2016b). Our study confirmed the activation of this

8
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

Fig. 7. Liralutide induces PPARα in liver and perigonadal fat in HFHS diet mice. Mice tissues were collected, and protein and RNA were extraced A. PPARα protein
expression in liver. B. PPARα protein expression in perigonadal fat. C. Liver PPARα mRNA expression. D. Perigonadal fat PPARα mNA expression Data is shown as
mean ± S.E.M., n = 5. β-actin were used as loading control. *, P < 0.05, **, P < 0.005.

signaling pathway in insulin sensitive tissues. Furthermore, we detected recent evidence also suggests it plays a role in modulating acute or
an inhibitory effect; liraglutide downregulated PPAR-γ and C/EBPα in chronic inflammation (Bougarne et al., 2018; Gervois and Mansouri,
the liver, adipose tissue, and skeletal muscle, indicating an anti-lipo- 2012). We have shown that liraglutide treatment induces PPARα up-
genic effect of liraglutide in these tissues. This can be explained by the regulation in association with reduced inflammation markers in insulin
cross-talk between PPARγ and C/EBPα in regulating adipogenesis sensitive tissues, a finding that supports the anti-inflammatory effect of
(Rosen et al., 2002; Wu et al., 1999), with PPARγ being the proximal liraglutide.
effector of adipogensis (Walton and Maung Tun, 1978), lipogenesis, and One possible mechanism by which liraglutide promotes energy ex-
lipid accumulation in steatotic hepatocytes (Schadinger et al., 2005). penditure is through the activation and/or expansion of brown adipose
The reductions in adiposity as a result of liraglutide treatment, speci- tissue and mitochondrial uncoupling of respiration in brown adipo-
fically visceral adiposity, suggests a modulatory role of liraglutide in de cytes, which may be present in skeletal muscles, as reported in our
novo lipogenic pathways in liver tissue and an anti-lipogenic effect of previous studies (Li et al., 2011b). PRDM-16 is a transcriptional reg-
liraglutide in fat and skeletal muscle. ulator of a bidirectional switch in cell fate between skeletal muscle and
Benefits of liraglutide on NAFLD have not been well established. brown fat (Seale et al., 2008) via regulation of PGC1-α and mi-
Forty-eight weeks of liraglutide treatment led to the histological re- tochondrial UCP-1 (Seale et al., 2008). Following treatment with lir-
solution of NASH in overweight subjects (Armstrong et al., 2016a). aglutide we observed substantial increases in the expression of PRDM16
However, twelve-week liraglutide treatment did not reduce hepatic and brown fat markers such as UCP-1 and PPARα in skeletal muscle
steatosis or fibrosis in T2D patients in a randomized, placebo-controlled tissue. These findings indicate that liraglutide induces brown fat dif-
trial (Smits et al., 2016). In contrast, eight-weeks of liraglutide treat- ferentiation in skeletal muscle in an insulin resistant state. Despite
ment improved steatosis scores in DIO-NASH in rodents (Ipsen et al., upregulating brown fat markers, two weeks of liraglutide treatment
2018; Tølbøl et al., 2018), and this beneficial role may be mediated failed to induce beige fat in our study, possibly due to the insufficient
through the elevation of adiponectin and the inactivation of JNK-1 (Gao duration of the treatment. Twelve-weeks of liraglutide treatment has
et al., 2015). Our study provides new evidence that liraglutide reduces been reported to induce beige fat in high fat diet-fed KK/Upj-Ay/J
hepatic inflammation and hepatocyte ballooning, which may be (KKAy) mice (Zhu et al., 2016). Hence, a prolonged treatment period
mediated through the AMPK-Sirt-1 cell signaling pathway. However, with liraglutide may be required to induce beige fat differentiation in
long term treatment with liraglutide and extensive study of chronic HFHS-fed mice.
HFHS diet induced hepatic steatosis are warranted. In conclusion, our study indicates that liraglutide improves insulin
PPARα plays a major role in regulating energy metabolism and is sensitivity through multiple pathways including reduction of in-
the most important PPAR in regulating liver fatty acid mitochondrial flammation, elevation of fatty acid oxidation, and inducing skeletal
and peroxisomal β-oxidation (Bjørndal et al., 2011). Importantly, muscle adaptive thermogenesis. Our data that derived from animal

9
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

Fig. 8. Liralutide induces brown adipocyte diferentiation in skeletal muscle in HFHS diet mice. Mice tissues were collected, and protein and RNA were extraced.
Expression of UCP-1, PRDM-16 and PPARα was measured by both western blotting and real-time PCR. A Expression of UCP-1 in skeletal muscle, B. PRDM-16
expression in skeletal muscle, C. PPARα expression in skeletal muscle. D. mRNA expression of brown fat markers in skeletal muscle. Data is shown as
mean ± S.E.M., n = 5. β-actin and β-tubulin were used as loading control *, P < 0.05, **, P < 0.005.

models provides more evidence that liraglutide may be an effective 2018. Molecular actions of PPARα in lipid metabolism and inflammation. Endocr.
treatment for obesity and related metabolic disorders. However, further Rev. 39, 760–802.
Cantó, C., Auwerx, J., 2009. PGC-1 alpha, SIRT1 and AMPK, an energy sensing network
human studies regarding the potential therapeutic role of liraglutide on that controls energy expenditure. Curr. Opin. Lipidol. 20, 98–105.
metabolic disorder and NAFLD are needed. Drucker, D.J., 2002. Biological actions and therapeutic potential of the glucagon-like
peptides. Gastroenterology 122, 531–544.
Dyck, J.R., Gao, G., Widmer, J., Stapleton, D., Fernandez, C.S., Kemp, B.E., Witters, L.A.,
Conflicts of interest 1996. Regulation of 5'-AMP-activated protein kinase activity by the noncatalytic beta
and gamma subunits. J. Biol. Chem. 271, 17798–17803.
The authors have no conflict of interests to declare. Feige, J.N., Auwerx, J., 2007. Transcriptional coregulators in the control of energy
homeostasis. Trends Cell Biol. 17, 292–301.
Feldmann, H.M., Golozoubova, V., Cannon, B., Nedergaard, J., 2009. UCP1 ablation in-
Acknowledgement duces obesity and abolishes diet-induced thermogenesis in mice exempt from thermal
stress by living at thermoneutrality. Cell Metabol. 9, 203–209.
Fuchs, O., Borová, J., Hradilek, A., Neuwirt, J., 1988. Non-transferrin donors of iron for
L.L. is supported by faculty start-up fund from College of Health
heme synthesis in immature erythroid cells. Biochim. Biophys. Acta 969, 158–165.
Professions, Central Michigan University. Rosca M is supported by Fullerton, M.D., Galic, S., Marcinko, K., Sikkema, S., Pulinilkunnil, T., Chen, Z.P., O'Neill,
American Heart Association (grant # 18AIREA33990023). The authors H.M., Ford, R.J., Palanivel, R., O'Brien, M., Hardie, D.G., Macaulay, S.L., Schertzer,
thank Jamsheed Bahaee and Melisa Andrews for technical support and J.D., Dyck, J.R., van Denderen, B.J., Kemp, B.E., Steinberg, G.R., 2013. Single
phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-
Darren Story for editing English grammars for the manuscript. sensitizing effects of metformin. Nat. Med. 19, 1649–1654.
Gao, H., Zeng, Z., Zhang, H., Zhou, X., Guan, L., Deng, W., Xu, L., 2015. The glucagon-like
References peptide-1 analogue liraglutide inhibits oxidative stress and inflammatory response in
the liver of rats with diet-induced non-alcoholic fatty liver disease. Biol. Pharm. Bull.
38, 694–702.
Abu-Elheiga, L, Oh, W, Kordari, P, Wakil, S.J, 2003. Acetyl-CoA carboxylase 2 mutant Gervois, P., Mansouri, R.M., 2012. PPARα as a therapeutic target in inflammation-asso-
mice are protected against obesity and diabetes induced by high-fat/high-carbohy- ciated diseases. Expert Opin. Ther. Targets 16, 1113–1125.
drate diets. Proc Natl Acad Sci U S A 100, 10207–10212.. Harms, M., Seale, P., 2013. Brown and beige fat: development, function and therapeutic
Armstrong, M.J., Gaunt, P., Aithal, G.P., Barton, D., Hull, D., Parker, R., Hazlehurst, J.M., potential. Nat. Med. 19, 1252–1263.
Guo, K., Abouda, G., Aldersley, M.A., Stocken, D., Gough, S.C., Tomlinson, J.W., Harms, M.J., Ishibashi, J., Wang, W., Lim, H.W., Goyama, S., Sato, T., Kurokawa, M.,
Brown, R.M., Hübscher, S.G., Newsome, P.N., team, L.t., 2016a. Liraglutide safety Won, K.J., Seale, P., 2014. Prdm16 is required for the maintenance of brown adi-
and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, pocyte identity and function in adult mice. Cell Metabol. 19, 593–604.
double-blind, randomised, placebo-controlled phase 2 study. Lancet 387, 679–690. Heppner, K.M., Marks, S., Holland, J., Ottaway, N., Smiley, D., Dimarchi, R., Perez-Tilve,
Armstrong, M.J., Hull, D., Guo, K., Barton, D., Hazlehurst, J.M., Gathercole, L.L., Nasiri, D., 2015. Contribution of brown adipose tissue activity to the control of energy
M., Yu, J., Gough, S.C., Newsome, P.N., Tomlinson, J.W., 2016b. Glucagon-like balance by GLP-1 receptor signalling in mice. Diabetologia 58, 2124–2132.
peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis. J. Hepatol. 64, Holmes, D., 2017. Pharmacotherapy: a smarter way to treat obesity. Nat. Rev. Endocrinol.
399–408. 13, 626.
Bastard, J.P., Maachi, M., Van Nhieu, J.T., Jardel, C., Bruckert, E., Grimaldi, A., Robert, Inoue, T., Inoguchi, T., Sonoda, N., Hendarto, H., Makimura, H., Sasaki, S., Yokomizo, H.,
J.J., Capeau, J., Hainque, B., 2002. Adipose tissue IL-6 content correlates with re- Fujimura, Y., Miura, D., Takayanagi, R., 2015. GLP-1 analog liraglutide protects
sistance to insulin activation of glucose uptake both in vivo and in vitro. J. Clin. against cardiac steatosis, oxidative stress and apoptosis in streptozotocin-induced
Endocrinol. Metab. 87, 2084–2089. diabetic rats. Atherosclerosis 240, 250–259.
Bjørndal, B., Burri, L., Staalesen, V., Skorve, J., Berge, R.K., 2011. Different adipose de- Ipsen, D.H., Rolin, B., Rakipovski, G., Skovsted, G.F., Madsen, A., Kolstrup, S., Schou-
pots: their role in the development of metabolic syndrome and mitochondrial re- Pedersen, A.M., Skat-Rørdam, J., Lykkesfeldt, J., Tveden-Nyborg, P., 2018.
sponse to hypolipidemic agents. J Obes 2011, 490650. Liraglutide decreases hepatic inflammation and injury in advanced lean non-alco-
Bougarne, N., Weyers, B., Desmet, S.J., Deckers, J., Ray, D.W., Staels, B., De Bosscher, K., holic steatohepatitis. Basic Clin. Pharmacol. Toxicol. 123 (6), 704–713 2018.

10
J.Y. Zhou, et al. European Journal of Pharmacology 861 (2019) 172594

Kakimoto, P.A., Kowaltowski, A.J., 2016. Effects of high fat diets on rodent liver bioe- Schadinger, S.E., Bucher, N.L., Schreiber, B.M., Farmer, S.R., 2005. PPARgamma2 reg-
nergetics and oxidative imbalance. Redox Biol. 216–225. ulates lipogenesis and lipid accumulation in steatotic hepatocytes. Am. J. Physiol.
Kern, P.A., Ranganathan, S., Li, C., Wood, L., Ranganathan, G., 2001. Adipose tissue Endocrinol. Metab. 288, E1195–E1205.
tumor necrosis factor and interleukin-6 expression in human obesity and insulin re- Seale, P., Bjork, B., Yang, W., Kajimura, S., Chin, S., Kuang, S., Scimè, A., Devarakonda,
sistance. Am. J. Physiol. Endocrinol. Metab. 280, E745–E751. S., Conroe, H.M., Erdjument-Bromage, H., Tempst, P., Rudnicki, M.A., Beier, D.R.,
Kim, J.H., Bachmann, R.A., Chen, J., 2009. Interleukin-6 and insulin resistance. Vitam. Spiegelman, B.M., 2008. PRDM16 controls a brown fat/skeletal muscle switch.
Horm. 80, 613–633. Nature 454, 961–967.
Kim, S.F., Huri, D.A., Snyder, S.H., 2005. Inducible nitric oxide synthase binds, S-ni- Senn, J.J., Klover, P.J., Nowak, I.A., Mooney, R.A., 2002. Interleukin-6 induces cellular
trosylates, and activates cyclooxygenase-2. Science 310, 1966–1970. insulin resistance in hepatocytes. Diabetes 51, 3391–3399.
Knudsen, L.B., 2010. Liraglutide: the therapeutic promise from animal models. Int. J. Clin. Sheedfar, F., Di Biase, S., Koonen, D., Vinciguerra, M., 2013. Liver diseases and aging:
Pract. Suppl, 4–11. friends or foes? Aging Cell 12, 950–954.
Krasner, N.M., Ido, Y., Ruderman, N.B., Cacicedo, J.M., 2014. Glucagon-like peptide-1 Smits, M.M., Tonneijck, L., Muskiet, M.H., Kramer, M.H., Pouwels, P.J., Pieters-van den
(GLP-1) analog liraglutide inhibits endothelial cell inflammation through a calcium Bos, I.C., Hoekstra, T., Diamant, M., van Raalte, D.H., Cahen, D.L., 2016. Twelve
and AMPK dependent mechanism. PLoS One 9, e97554. week liraglutide or sitagliptin does not affect hepatic fat in type 2 diabetes: a ran-
Leclercq, I.A., Farrell, G.C., Sempoux, C., dela Peña, A., Horsmans, Y., 2004. Curcumin domised placebo-controlled trial. Diabetologia 59, 2588–2593.
inhibits NF-kappaB activation and reduces the severity of experimental steatohepa- Tang, S.C., Hendrikx, J.J., Beijnen, J.H., Schinkel, A.H., 2013. Genetically modified
titis in mice. J. Hepatol. 41, 926–934. mouse models for oral drug absorption and disposition. Curr. Opin. Pharmacol. 13,
Lee, Y.S., Shin, S., Shigihara, T., Hahm, E., Liu, M.J., Han, J., Yoon, J.W., Jun, H.S., 2007. 853–858.
Glucagon-like peptide-1 gene therapy in obese diabetic mice results in long-term cure Tomas, E., Wood, J.A., Stanojevic, V., Habener, J.F., 2011. Glucagon-like peptide-1(9-36)
of diabetes by improving insulin sensitivity and reducing hepatic gluconeogenesis. amide metabolite inhibits weight gain and attenuates diabetes and hepatic steatosis
Diabetes 56, 1671–1679. in diet-induced obese mice. Diabetes Obes. Metab. 13, 26–33.
Li, L., Hossain, M.A., Sadat, S., Hager, L., Liu, L., Tam, L., Schroer, S., Huogen, L., Fantus, Tølbøl, K.S., Kristiansen, M.N., Hansen, H.H., Veidal, S.S., Rigbolt, K.T., Gillum, M.P.,
I.G., Connelly, P.W., Woo, M., Ng, D.S., 2011a. Lecithin cholesterol acyltransferase Jelsing, J., Vrang, N., Feigh, M., 2018. Metabolic and hepatic effects of liraglutide,
null mice are protected from diet-induced obesity and insulin resistance in a gender- obeticholic acid and elafibranor in diet-induced obese mouse models of biopsy-con-
specific manner through multiple pathways. J. Biol. Chem. 286, 17809–17820. firmed nonalcoholic steatohepatitis. World J. Gastroenterol. 24, 179–194.
Li, L., Hossain, M.A., Sadat, S., Hager, L., Liu, L., Tam, L., Schroer, S., Huogen, L., Fantus, Vozarova, B., Weyer, C., Hanson, K., Tataranni, P.A., Bogardus, C., Pratley, R.E., 2001.
I.G., Connelly, P.W., Woo, M., Ng, D.S., 2011b. Lecithin cholesterol acyltransferase Circulating interleukin-6 in relation to adiposity, insulin action, and insulin secretion.
null mice are protected from diet-induced obesity and insulin resistance in a gender- Obes. Res. 9, 414–417.
specific manner through multiple pathways. J. Biol. Chem. 286, 17809–17820. Wakil, S.J., Abu-Elheiga, L.A., 2009. Fatty acid metabolism: target for metabolic syn-
Makki, K., Froguel, P., Wolowczuk, I., 2013. Adipose tissue in obesity-related in- drome. J. Lipid Res. 50 (Suppl. l), S138–S143.
flammation and insulin resistance: cells, cytokines, and chemokines. ISRN Inflamm Walton, D.W., Maung Tun, U.M., 1978. Fleas of small mammals from Rangoon, Burma.
2013, 139239. Southeast Asian J. Trop. Med. Public Health 9, 369–377.
Marcinko, K., Steinberg, G.R., 2014. The role of AMPK in controlling metabolism and Wu, Z., Rosen, E.D., Brun, R., Hauser, S., Adelmant, G., Troy, A.E., McKeon, C.,
mitochondrial biogenesis during exercise. Exp. Physiol. 99, 1581–1585. Darlington, G.J., Spiegelman, B.M., 1999. Cross-regulation of C/EBP alpha and PPAR
Mells, J.E., Fu, P.P., Sharma, S., Olson, D., Cheng, L., Handy, J.A., Saxena, N.K., Sorescu, gamma controls the transcriptional pathway of adipogenesis and insulin sensitivity.
D., Anania, F.A., 2012. Glp-1 analog, liraglutide, ameliorates hepatic steatosis and Mol. Cell 3, 151–158.
cardiac hypertrophy in C57BL/6J mice fed a Western diet. Am. J. Physiol. Xu, F., Li, Z., Zheng, X., Liu, H., Liang, H., Xu, H., Chen, Z., Zeng, K., Weng, J., 2014.
Gastrointest. Liver Physiol. 302, G225–G235. SIRT1 mediates the effect of GLP-1 receptor agonist exenatide on ameliorating he-
Montgomery, M.K., Turner, N., 2015. Mitochondrial dysfunction and insulin resistance: patic steatosis. Diabetes 63, 3637–3646.
an update. Endocr Connect 4, R1–R15. Yu, J., Ip, E., Dela Peña, A., Hou, J.Y., Sesha, J., Pera, N., Hall, P., Kirsch, R., Leclercq, I.,
Namba, M., Katsuno, T., Kusunoki, Y., Matsuo, T., Miuchi, M., Miyagawa, J., 2013. New Farrell, G.C., 2006. COX-2 induction in mice with experimental nutritional steato-
strategy for the treatment of type 2 diabetes mellitus with incretin-based therapy. hepatitis: role as pro-inflammatory mediator. Hepatology 43, 826–836.
Clin. Exp. Nephrol. 17, 10–15. Zander, M., Madsbad, S., Madsen, J.L., Holst, J.J., 2002. Effect of 6-week course of glu-
Olefsky, J.M., Glass, C.K., 2010. Macrophages, inflammation, and insulin resistance. cagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function
Annu. Rev. Physiol. 72, 219–246. in type 2 diabetes: a parallel-group study. Lancet 359, 824–830.
Parthsarathy, V., Hölscher, C., 2013. The type 2 diabetes drug liraglutide reduces chronic Zhang, D., Liu, Z.X., Choi, C.S., Tian, L., Kibbey, R., Dong, J., Cline, G.W., Wood, P.A.,
inflammation induced by irradiation in the mouse brain. Eur. J. Pharmacol. 700, Shulman, G., 2007. Mitochondrial dysfunction due to long-chain Acyl-CoA dehy-
42–50. drogenase deficiency causes hepatic steatosis and hepatic insulin resistance. Proc Natl
Richard, J., Lingvay, I., 2011. Hepatic steatosis and Type 2 diabetes: current and future Acad Sci U S A 104, 17075–17080.
treatment considerations. Expert Rev. Cardiovasc Ther. 9, 321–328. Zhu, E., Yang, Y., Zhang, J., Li, Y., Li, C., Chen, L., Sun, B., 2016. Liraglutide suppresses
Rosen, E.D., Hsu, C.H., Wang, X., Sakai, S., Freeman, M.W., Gonzalez, F.J., Spiegelman, obesity and induces brown fat-like phenotype via Soluble Guanylyl Cyclase mediated
B.M., 2002. C/EBPalpha induces adipogenesis through PPARgamma: a unified pathway in vivo and in vitro. Oncotarget 7, 81077–81089.
pathway. Genes Dev. 16, 22–26.

11

You might also like