You are on page 1of 7

Biochemical and Biophysical Research Communications xxx (xxxx) xxx

Contents lists available at ScienceDirect

Biochemical and Biophysical Research Communications


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

Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK


in high fat diet fed mice
Shrirang Inamdar a, Ankita Joshi a, Sajad Malik a, Ramanamurthy Boppana b,
Saroj Ghaskadbi a, *
a
Department of Zoology, Savitribai Phule Pune University, Pune, 411007, India
b
National Centre for Cell Science, Pune, 411007, India

a r t i c l e i n f o a b s t r a c t

Article history: Non-alcoholic fatty liver disease (NAFLD) is a most common liver disorder characterized by accumulation
Received 21 August 2019 of fat in the liver and currently there is no approved treatment for it. Obesity and diabetes being leading
Accepted 25 August 2019 cause of NAFLD, compounds having anti-obesity activity and potential to reduce insulin resistance are
Available online xxx
considered suitable candidate for NAFLD treatment. In this study, we checked effect of vitexin, a naturally
occurring flavonoid, on high fat diet (HFD) induced NAFLD in C57BL/6J mice. In presence of vitexin,
Keywords:
significant reduction in body and liver weight, triglyceride and cholesterol content in serum and liver
Non alcoholic fatty liver disease (NAFLD)
was observed. Serum Alanine aminotransferase (ALT) and Aspartate aminotransferase (AST) levels were
Vitexin
AMP-activated protein kinase (AMPK)
reduced significantly by vitexin which were elevated in HFD group whereas serum lipase activity
Leptin receptor remained unchanged. Vitexin suppressed de novo lipogenesis by downregulating expression of Peroxi-
some proliferator-activated receptor g (PPARg), CCAAT/enhancer-binding protein-a (C/EBP-a), sterol
regulatory element-binding protein-1c (SREBP-1c), Fatty acid synthase (FAS) and Acetyl-CoA Carboxylase
(ACC). Additionally, it also enhanced fatty acid oxidation and lipolysis by upregulating Peroxisome
proliferator-activated receptor a (PPAR-a), carnitine palmitoyltransferase-1a (CPT-1a) and Adipose tri-
glyceride lipase (ATGL). Inhibition of lipogenesis and activation of lipolysis and fatty acid oxidation by
vitexin was found to be mediated by activation of AMP-activated protein kinase (AMPK). Vitexin also
improved insulin signalling by activating insulin receptor substrate-1 (IRS-1) and its downstream target
AKT. AMPK activation of vitexin was possibly through binding of vitexin to leptin receptor (LepR) which
was confirmed by molecular docking studies and by observed enhanced expression of LepR. Thus, we
propose that vitexin alleviates NAFLD by activating AMPK possibly by binding to LepR.
© 2019 Elsevier Inc. All rights reserved.

1. Introduction Since, there is no approved medical treatment for NAFLD,


compounds which control obesity would prove effective in man-
NAFLD is characterized by the accumulation of fat (more than agement of NAFLD [4]. Vitexin, (apigenin-8-C-D-glucopyranoside),
5%) in the liver without a history of alcoholism or known liver is a natural flavonoid found in certain herbs such as hawthorn herb,
pathology. NAFLD has emerged as an important health problem mung bean and fenugreek. It has wide range of pharmacological
worldwide with prevalence of around 25% [1] whereas, in India the uses, including anti-obesity, anti-inflammatory, antioxidant and
prevalence is around 32% [2]. NAFLD includes steatosis and stea- anti-tumor [5]. Vitexin and extract containing vitexin have been
tohepatitis which can progress further to fibrosis, cirrhosis and shown to be effective in reduction of diet induced obesity [6e9]
even to hepatocarcinoma [3]. Obesity and diabetes are a major risk and thus would be a promising candidate for control of NAFLD.
factors leading to NAFLD and their increased prevalence due to High fat diet (HFD) induces NAFLD as consequence of imbalance
modern dietary habits and sedentary lifestyle has resulted in between energy intake and its expenditure. Leptin and LepR
increased NAFLD prevalence. regulate the body weight by balancing food intake and energy
expenditure by activating AMPK [10]. AMPK is a major energy
sensor of the cell and regulates hepatic and adipose lipid meta-
* Corresponding author. bolism by modulating lipogenesis, lipolysis, gluconeogenesis and
E-mail addresses: ssg@unipune.ac.in, sghaskadbi@gmail.com (S. Ghaskadbi). adipogenesis [11]. AMPK is known to inhibit de novo lipogenesis by

https://doi.org/10.1016/j.bbrc.2019.08.139
0006-291X/© 2019 Elsevier Inc. All rights reserved.

Please cite this article as: S. Inamdar et al., Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice,
Biochemical and Biophysical Research Communications, https://doi.org/10.1016/j.bbrc.2019.08.139
2 S. Inamdar et al. / Biochemical and Biophysical Research Communications xxx (xxxx) xxx

down regulating key lipogenic genes such PPARg, C/EBP-a and RNA was assessed by calculating A260/280. cDNA was prepared using
SREBP-1c in HFD fed animals [11,12]. In addition, AMPK is also Mulv-Reverse transciptase (New England Biolabs, USA) and qRT-
known to promote fatty-acid oxidation by suppressing ACC and FAS PCR was performed on Step One Plus (Applied Biosystems, USA)
activity [13]. It also up-regulates expression of CPT-1a, a key using SYBR green and gene specific primers (Supplementary
regulator of fatty-acid oxidation [12]. Vitexin is known to activate Table 1) for PPARg, PPARa, C/EBP-a, SREBP-1c, CPT-1a, PGC-1a,
AMPK signaling during alleviating obesity [8] and endothelial and ATGL. Fold-change in expression was calculated by using 2-DDCT
injury [14]. In addition to alterations in energy homeostasis, HFD is method where b-Actin was used as a house-keeping gene.
also known to induce insulin resistance which in turn leads to
NAFLD [15]. Hence, compounds improving both energy homeo- 2.5. Western blot analysis
stasis and insulin signaling would be advantageous for the
treatment. Liver tissue samples were homogenized in the lysis buffer
This study was undertaken to check effect of vitexin on diet containing 20 mM Tris, pH 7.5, 1 mM EDTA (pH 8.0), 150 mM NaCl,
induced NAFLD in mice and deduce the mechanism behind this. We 1% Triton X-100, 10% glycerol, 1 mM phenylmethylsulfonyl fluoride;
confirmed that vitexin inhibits NAFLD by activating AMPK which in 1 mM sodium orthovanadate, 0.5% sodium deoxycholate, 10 mM
turn inhibits lipogenesis and activates lipolysis and fatty acid sodium fluoride, 1 mM DTT, 12.5 mM b-glycerophosphate and 1X
oxidation. In addition, vitexin also improved insulin signaling in Protease inhibitor cocktail and equal amount of protein was loaded
liver of HFD fed animals. We propose that the vitexin activates on 12% SDS-PAGE. Proteins were electro transferred to PVDF
AMPK by probably binding and activating LepR. membrane (Millipore, USA) at 80 mA constant current. Blots were
then saturated with BSA, reacted with different primary antibodies
2. Materials and methods (PPARg, C/EBP-a, FAS, ACC, p-AMPK, t-AMPK, p-AKT, t-AKT, p-IRS1,
t-IRS1 and b-Actin procured from Elabscience, USA) and then
2.1. Animal studies incubated with secondary anti-rabbit antibody (Sigma, USA) tagged
to horseradish peroxidase. Signals were detected by chem-
C57BL/6J mice used in the study were maintained at Experi- iluminescence using luminol (Advansta, USA) as a substrate and
mental Animal Facility (EAF) at National Centre for Cell Science documented using chemidoc system (Biorad, USA).
(NCCS), Pune, India. Animals were housed at 12 h light/dark cycle
with ad libitum access to water and food. All animal experiments 2.6. Molecular modeling and docking studies
were carried out as per the requirement and guidelines of the
Committee for the Purpose of Control and Supervision of Experi- Sequence of mouse LepR obtained from Uniprot database (www.
ments on Animals (CPCSEA), Government of India, and after uniprot.org) was blasted against protein data bank (PDB) using
obtaining permission of the Institutional Animal Ethics Committee BLASTp. Human LepR crystal structure was used as a template to
(IAEC/2018/B-332). generate homology model of mouse LepR using SWISS-MODEL
Total 30 animals (6e8 weeks old) were used in the study and server (https://swissmodel.expasy.org/). Model was selected
were divided equally into 5 groups namely Control (ND), High fat based on QMEAN score and used further for docking studies. For
diet (HFD), HFD supplemented with 1 mg/kg vitexin (HFDþ 1 mg this, structure of vitexin and structure of mouse LepR constructed
Vit), with 10 mg/kg vitexin (HFDþ 10 mg Vit) and with 20 mg/kg using Swiss model was submitted to AutoDock Vina software and
vitexin (HFDþ 20 mg Vit). ND group animals were fed with normal results obtained were visualized and analyzed using Chimera
diet (5% kcal fat) purchased from Nutrivet Life Sciences Pune, India software [16].
whereas, HFD comprising 60% kcal fat (D12492, Research diet Inc.,
USA) was given to all other groups for eight weeks. Vitexin was 2.7. Statistical analyses
given daily by oral gavage. Body weight of all mice was monitored
weekly. At the end of 8 weeks, blood was collected from orbital Each experiment was performed at least three times and the
plexus and mice were sacrificed by cervical dislocation. Liver and data are shown as mean with standard error (SE). Statistical sig-
adipose tissue were harvested and snap frozen in liquid N2. Part of nificance was calculated by SPSS 19.0 software with one-way
the tissue was fixed in 4% paraformaldehyde and used for histo- ANOVA. P-value<0.05 was considered statistically significant.
logical analysis. Remaining tissue was stored at 80 OC and pro- #,
*p < 0.05, ##,**p < 0.001, ###,***p < 0.0001.
cessed for protein and RNA isolation.
3. Results
2.2. Biochemical analysis
3.1. Vitexin reduces body weight gain and tissue weight in HFD-
Total cholesterol, triglycerides, ALT, AST and lipase activity were induced obese mice
measured using commercially available kits (Accurex Biomedical
PVT LTD, India) following manufacturer's protocol. Significant increase in body and liver weight was observed in
HFD group animals after 8 weeks compared to those of ND group
2.3. Histochemical analysis animals (Fig. 1A). Vitexin treatment significantly reduced the body
weight gain induced by HFD in a dose dependent manner (Fig. 1A)
Liver tissue was fixed in 4% paraformaldehyde and 10 mm sec- concomitant with decreased liver weight (Supplementary Table 2).
tions were collected on poly L-lysine coated slides using cryotome Significantly elevated levels of serum total cholesterol and tri-
(Leika CM 1250, Germany) and stained with oil red O. Images were glycerides were found in HFD group animals compared to ND. This
captured using Nikon eclipse TiS (Nikon, Japan) microscope. increase was significantly reduced by vitexin in a dose dependent
manner (Fig. 1B and C). Both AST and ALT levels found to be
2.4. RNA isolation and quantitative real time PCR (qRT-PCR) increased significantly in HFD fed mice were reduced after vitexin
treatment at 1 and 10 mg/kg body weight and remained unchanged
RNA from liver tissue was isolated using Trizol reagent, quan- at higher concentration (Fig. 1D and E). Intestinal lipase activity was
titated using Nanophotometer (Eppendorf, Germany) and purity of not found to be altered in mice treated with vitexin, indicating that

Please cite this article as: S. Inamdar et al., Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice,
Biochemical and Biophysical Research Communications, https://doi.org/10.1016/j.bbrc.2019.08.139
S. Inamdar et al. / Biochemical and Biophysical Research Communications xxx (xxxx) xxx 3

Fig. 1. Vitexin attenuates NAFLD by reducing body weight gain and normalizing different serum parameters. Effect of vitexin on body weight (A), total cholesterol (B), total
triglyceride (C) ALT (D) AST (E) and lipase activity (F) after 8 weeks. (G) Liver sections of mice from different groups stained with H & E. (H) Estimation of triglyceride in liver of mice
from different groups. # represents significance level with respect to ND group. * represents significance level with respect to HFD group. #,*p < 0.05, ##,**p < 0.001,
###,
***p < 0.0001.

the effect of vitexin is not mediated by preventing dietary lipid suppressed in liver of HFD-induced obese mice while it was
uptake (Fig. 1F). significantly increased in presence of vitexin in a dose dependent
manner thereby promoting lipolysis (Fig. 2B). PGC-1a is a master
3.2. Vitexin inhibits development of NAFLD regulator of mitochondrial biogenesis and oxidative metabolism
and is known to activate expression of CPT-1a involved in transport
Enhanced accumulation of lipids in liver, a characteristic feature of fatty acids into mitochondria. Expression of both PGC-1a and
of NAFLD, was confirmed by oil red O staining in liver sections of CPT-1a was significantly decreased in liver of HFD fed mice and
HFD fed animals and this was significantly rescued in liver of mice vitexin increased their expression in a dose dependent manner
treated with vitexin in a dose dependent manner (Fig. 1G). Simi- (Fig. 2B).
larly, significant increase in triglyceride accumulation in liver of
HFD group mice was inhibited by treatment with vitexin (Fig. 1H). 3.4. Vitexin alleviates inhibition of insulin signaling

3.3. Vitexin alleviates NAFLD state of liver by down regulating NAFLD is associated with impaired insulin signaling [17] due to
lipogenesis and by up regulating lipolysis and fatty acid oxidation enhanced inhibitory phosphorylation at Ser-307 residue of IRS-1.
We indeed, observed significantly increased levels of p-IRS-1(Ser-
PPARg and C/EBP-a, are important transcription factors pro- 307) in liver of HFD fed animals and vitexin reduced this inhibition
moting lipogenesis and their expression was indeed found to be in a dose dependent manner. (Fig. 3A and B). We also checked
significantly increased both at RNA (Fig. 2A) and protein level activation of AKT (phosphorylation levels of Ser-473), a down-
(Fig. 2C and D) in liver of HFD group and vitexin suppressed their stream target of IRS-1 and found it significantly reduced in HFD fed
expression in a dose dependent manner. Expression of SREBP-1c mice. Vitexin reversed this effect in a dose dependent manner
(Fig. 2A) which is activated by PPARg and its downstream targets (Fig. 3A and B).
FAS and ACC, rate limiting enzymes of de novo lipogenesis, was
found to be significantly increased in liver of HFD fed animals and 3.5. Vitexin activates AMPK possibly through LepR
vitexin reduced it in a dose dependent manner (Fig. 2C and D).
Expression of ATGL, gene involved in lipolysis and transcription AMPK is a master regulator of energy homeostasis and is known
factor PPARa was checked by qRT-PCR and was found to be to modulate both lipogenesis by inhibiting SREBP-1c and lipolysis

Please cite this article as: S. Inamdar et al., Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice,
Biochemical and Biophysical Research Communications, https://doi.org/10.1016/j.bbrc.2019.08.139
4 S. Inamdar et al. / Biochemical and Biophysical Research Communications xxx (xxxx) xxx

Fig. 2. Vitexin alleviates NAFLD by inhibiting lipogenesis and by promoting lipolysis and fatty acid oxidation in liver. Relative mRNA expression of genes in lipogenesis (A), and
lipolysis and FFA oxidation (B) in liver of mice from different groups. (C) Expression of protein involved in lipogenesis in liver of mice from different groups and (D) densitometric
analysis of western blot in fig. (C). # represents significance level with respect to ND group. * represents significance level with respect to HFD group. #,*p < 0.05, ##,**p < 0.001,
###,
***p < 0.0001.

by activating PGC-1a and PPARa. We found decreased p-AMPK in 4. Discussion


liver of HFD fed animals while in case of vitexin treated mice this
decrease was inhibited in a dose dependent manner (Fig. 3A and B). In the current study, we demonstrate the potential of vitexin to
AMPK is known to be activated by LepR [10]. Therefore, in order inhibit liver steatosis observed in NAFLD in mice fed with HFD for
to determine whether vitexin would also activate AMPK by binding eight weeks. Obesity induced NAFLD is associated with increased
to LepR, molecular docking studies were performed. Since crystal body and liver weight, enhanced serum triglyceride and cholesterol
structure of mouse LepR is not available, a blast was performed level along with accumulation of fat in liver. This was confirmed by
using mouse LepR sequence and PDB database wherein mouse staining liver tissue sections by lipid specific stain, oil red O and by
LepR showed maximum similarity (86.87%) with human LepR (PDB estimating the triglyceride and cholesterol levels in liver and
id 3V6O) (Fig. 3C). Therefore, crystal structure of human LepR was serum. HT048, a combination of Crataegus pinnatifida leaf and Cit-
used as a template for generation of homology model of mouse rus unshiu peel extracts containing vitexin has been shown to
LepR using Swiss model server. The model with the best QMEAN reduce body and liver weight and serum levels of triglyceride and
score was selected (QMEAN score ¼ 1.5), validated by Ramchan- cholesterol in HFD fed rat [6]. Similarly, ethanol extract of mung
dran plot and was used for further study (Supplementary Fig. 1). bean containing vitexin, significantly reduced the body weight and
Superimposing human and homology model generated mouse serum lipid levels in KK-Ay mice when challenged with HFD [7].
LepR structure showed good match with RMSD value of 0.096 Ao More recently Peng et al. [8] and Seyedan et al. [9] have reported
(Fig. 3D). Vitexin showed docking at the leptin binding site pocket that vitexin or extract of Cynometra cauliflora containing vitexin
of the LepR with vena score of 5.9 indicating strong affinity. reduced body and liver weight and serum lipid levels in HFD fed
Vitexin formed hydrogen bonds with the LYS-484 and ASN-485 mice similar to our observations.
amino acids from the active site region which lie in binding re- Serum lipase hydrolyzes dietary fats into triglycerides and free
gion of leptin with its receptor (Fig. 3E and F). We also performed fatty acids which are then absorbed in the intestine. Commercially
docking studies with LepR inhibitor curcumin and observed that available anti-obesity drugs such as orlistat and resveratrol are
the inhibitor binds strongly to LepR at a site similar to that of known to exert their effect by inhibiting pancreatic lipase activity.
vitexin with vena score of 6.5. Similar amino acids are involved in We did not find alteration in the serum lipase activity at all con-
the formation of H bonds as that of vitexin (Fig. 3G). To further centrations of vitexin used indicating that effect of vitexin is not
confirm we checked expression of LepR by qRT-PCR in presence of mediated by inhibiting digestion of fat and its intestinal absorption.
vitexin and indeed found it to be activated (Fig. 3H). Based on this Increased triglyceride accumulation in liver can be either due to
and in silico analysis, vitexin seems to activate LepR which further increased FFA uptake and de novo lipogenesis in liver and/or
activates AMPK. decreased oxidation of stored fatty acids. FAS and ACC are the

Please cite this article as: S. Inamdar et al., Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice,
Biochemical and Biophysical Research Communications, https://doi.org/10.1016/j.bbrc.2019.08.139
S. Inamdar et al. / Biochemical and Biophysical Research Communications xxx (xxxx) xxx 5

Fig. 3. Vitexin alleviates inhibition of insulin signaling and activates AMPK possibly through LepR. (A) Protein expression of p-IRS-1, t-IRS1 p-AKT t-AKT and p-AMPK and t-
AMPK in liver of mice from different groups (B) Densitometric analysis of western blot in (a) (C) Pairwise sequence alignment of human and mouse LepR sequence. (D) Super-
imposed crystal structures of human and mouse LepR. Binding of vitexin (shown in green in E and blue in F) to LepR in hydrophobic surface (E) and ribbon (F) representation.
Hydrogen bonds are represented in red. (G) Binding of antagonist curcumin (shown in blue) to LepR. Hydrogen bonds are represented in red. # represents significance level with
respect to ND group. * represents significance level with respect to HFD group. #,*p < 0.05, ##,**p < 0.001, ###,***p < 0.0001. (For interpretation of the references to colour in this
figure legend, the reader is referred to the Web version of this article.)

important enzymes in de novo lipogenesis which are regulated by inhibits lipogenesis by suppressing the expression of C/EBP-a,
transcription factor, SREBP-1c, which in turn is activated by PPARg SREBP-1c, FAS as well as ACC and enhances lipolysis and b-oxida-
and C/EBP-a [18]. We found that vitexin inhibited expression of all tion by activating the expression of PGC-1a, ATGL and CPT-1a [21].
these genes in a dose dependent manner. Herbal formulation We found that in presence of vitexin AMPK was activated signifi-
HT048 containing vitexin has also been shown to inhibit lipogen- cantly in a dose dependent manner suggesting the effect of vitexin
esis in 3T3-L1 pre-adipocytes and HFD fed rats by inhibiting is mediated by activation of AMPK signaling.
expression of PPARg, C/EBP-a and FAS [6]. Similarly, mung bean In addition to altered lipogenesis and lipolysis, impaired insulin
ethanol extract containing vitexin suppressed the lipogenic genes signaling due to development of hepatic insulin resistance is very
PPARg, C/EBP-a and ACC in muscle of KK-Ay diabetic mice [7]. closely linked to NAFLD [17,22]. Different plant extracts with po-
Recently, Peng et al. [8] have shown the potential of vitexin in tential to improve insulin sensitivity and activation of downstream
reducing the expression of C/EBP-a and ACC in adipose tissue in AKT have shown promising therapeutic effects in NAFLD [17,23,24].
HFD fed mice. We then checked the expression of lipolysis and fatty In this study, we also observed that vitexin improved insulin
acid oxidation related genes in liver. PPARa and PGC-1a govern the signaling by inhibiting Ser-307 phosphorylation of IRS-1 and acti-
fatty acid oxidation by enhancing the expression of ATGL, a rate vating AKT; a master regulator of Insulin signaling.
limiting enzyme in lipolysis, and CPT-1a, which is essential for LepR is known to activate AMPK by activating Calcium/
transport of long-chain fatty acids into mitochondria [19]. Results calmodulin-dependent protein kinase kinase 2 (CaMKK2) and
revealed that vitexin indeed increased expression of these genes in IRS-1 via JAK/STAT3 pathway [10]. We therefore, speculated that
a dose dependent manner in liver suggesting that vitexin mediates vitexin may govern its action by activating LepR. To confirm this,
its effect also by activating lipolysis and subsequent fatty acid initially we performed docking studies. Since, the crystal structure
oxidation. of mouse LepR is not known, we generated and validated homology
AMPK is a master regulator of energy homeostasis and known to model using human LepR crystal structure as a template [25]. The
play role in pathogenesis of hepatic steatosis [20]. AMPK activation LepR (LR), encoded by db gene contains two cytokine receptor

Please cite this article as: S. Inamdar et al., Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice,
Biochemical and Biophysical Research Communications, https://doi.org/10.1016/j.bbrc.2019.08.139
6 S. Inamdar et al. / Biochemical and Biophysical Research Communications xxx (xxxx) xxx

Fig. 4. Schematic diagram showing mechanism of action of protective effects of vitexin against NAFLD. Vitexin binds to LepR leading to activation of AMPK. Phosphorylated
AMPK activates PGC-1a, PPARa and ATGL leading to increased lipolysis and fatty acid oxidation. And inhibits PPARg, SREBP-1c, ACC and FAS leading to inhibition of de novo
lipogenesis. Vitexin also improves insulin signaling by activating IRS-1 leading to activation of AKT.

homology (CRH) modules. CRH2 domain which is composed of two Acknowledgements


sub-domains with fibronectin type III folds (residues 428e535 and
536e635, respectively in human) was identified as the main high- Authors acknowledge financial support from Department of
affinity binding site for leptin on the LepR [26,27]. Blast studies and Science and Technology - Promotion of University Research and
earlier reports confirm that this region is highly conserved in Scientific Excellence (DST-PURSE, GOI-A-670) and University Grant
mouse [28]. Our docking studies revealed that vitexin binds Commission-Career Advancement Scheme (UGCCAS, F-5-2/
strongly at CRH2 domain of mouse LepR i.e. the leptin binding re- 2005(SAP-II) program of the Department of Zoology, Savitribai
gion. In addition, we found that binding site of LepR inhibitor Phule Pune University, India and SERB project (GOI-A-752) to SG.
curcumin overlaps with that of vitexin. We also observed increased Fellowship provided by Dr. D. S. Kothari fellowship from UGC to SI
expression of LepR in presence of vitexin by q-RT PCR. Thus, and DST-PURSE fellowship to SM is duly acknowledged. Authors
docking studies and enhanced LepR expression suggest that the thank Dr. Abhijit Kulkarni, Department of Bioinformatics, SPPU and
effect of vitexin is probably governed by activation of LepR. How- Dr. Susmit Sambhare,ACTREC, Mumbai for their help in bioinfor-
ever, this needs to be confirmed by further experimentation. Leptin matics related work.
bound at CRH2 domain also interacts with Ig like domain of the
second LepR via binding site-III thereby inducing 2:4 leptin:LepR Appendix A. Supplementary data
complex and receptor activation [10]. It would be interesting to see
whether vitexin acts in a similar manner. Supplementary data to this article can be found online at
In summary, present study provides evidence to support the https://doi.org/10.1016/j.bbrc.2019.08.139.
potential therapeutic effect of vitexin on reducing liver steatosis in
HFD fed mice. This observed effect is attributed to enhanced fatty
acid oxidation and lipolysis in addition to anti-lipogenesis activity. References
Vitexin was also found to enhance insulin signaling in the liver. [1] P.S. Dulai, S. Singh, J. Patel, M. Soni, L.J. Prokop, Z. Younossi, G. Sebastiani,
These effects are achieved by activating the AMPK signaling M. Ekstedt, H. Hagstrom, P. Nasr, P. Stal, Increased risk of mortality by fibrosis
possibly by binding and activating LepR (Fig. 4). stage in nonalcoholic fatty liver disease: systematic review and meta-analysis,
Hepatology 65 (2017) 1557e1565.
[2] V. Jain, M. Jana, B. Upadhyay, N. Ahmad, O. Jain, A.D. Upadhyay,
L. Ramakrishnan, N.K. Vikram, Prevalence, clinical & biochemical correlates of
Conflicts of interest non-alcoholic fatty liver disease in overweight adolescents, Indian J. Med. Res.
148 (2018) 291e301.
[3] E.L. Anderson, L.D. Howe, H.E. Jones, J.P. Higgins, D.A. Lawlor, A. Fraser, The
Authors declare that there is no conflict of interest. prevalence of non-alcoholic fatty liver disease in children and adolescents: a

Please cite this article as: S. Inamdar et al., Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice,
Biochemical and Biophysical Research Communications, https://doi.org/10.1016/j.bbrc.2019.08.139
S. Inamdar et al. / Biochemical and Biophysical Research Communications xxx (xxxx) xxx 7

systematic review and meta-analysis, PLoS One 10 (2015), e0140908. research and analysis, J. Comput. Chem. 25 (2004) 1605e1612.
[4] S. Kenneally, J.H. Sier, J.B. Moore, Efficacy of dietary and physical activity [17] K.A. Hwang, Y.J. Hwang, G.R. Kim, J.S. Choe, Extracts from Aralia elata (Miq)
intervention in non-alcoholic fatty liver disease: a systematic review, BMJ Seem alleviate hepatosteatosis via improving hepatic insulin sensitivity, BMC
Open Gastroenterol. 4 (2017), e000139. Complement Altern. Med. 15 (2015) 347.
[5] M. He, J.W. Min, W.L. Kong, X.H. He, J.X. Li, B.W. Peng, A review on the [18] J.D. Horton, J.L. Goldstein, M.S. Brown, SREBPs: activators of the complete
pharmacological effects of vitexin and isovitexin, Fitoterapia 115 (2016) program of cholesterol and fatty acid synthesis in the liver, J. Clin. Investig.
74e85. 109 (2002) 1125e1131.
[6] Y. Lee, Y.S. Kim, M. Song, M. Lee, J. Park, H. Kim, A herbal formula HT048, Citrus [19] M. Schweiger, R. Schreiber, G. Haemmerle, A. Lass, C. Fledelius, P. Jacobsen,
unshiu and Crataegus pinnatifida, prevents obesity by inhibiting adipogenesis H. Tornqvist, R. Zechner, R. Zimmermann, Adipose triglyceride lipase and
and lipogenesis in 3T3-L1 preadipocytes and HFD-induced obese rats, Mole- hormone-sensitive lipase are the major enzymes in adipose tissue tri-
cules 20 (2015) 9656e9670. acylglycerol catabolism, J. Biol. Chem. 281 (2006) 40236e40241.
[7] I. Kang, S. Choi, T.J. Ha, M. Choi, H.R. Wi, B.W. Lee, M. Lee, Effects of mung bean [20] E.A. Day, R.J. Ford, G.R. Steinberg, AMPK as a therapeutic target for treating
(Vigna radiata L.) ethanol extracts decrease proinflammatory cytokine- metabolic diseases, Trends Endocrinol. Metab. 28 (2017) 545e560.
induced lipogenesis in the KK-Ay diabese mouse model, J. Med. Food 18 [21] C. Canto  , J. Auwerx, AMP-activated protein kinase and its downstream tran-
(2015) 841e849. scriptional pathways, Cell. Mol. Life Sci. 67 (2010) 3407e3423.
[8] Y. Peng, Q. Sun, W. Xu, Y. He, W. Jin, L. Yuan, R. Gao, Vitexin ameliorates high [22] T. Galbo, G.I. Shulman, Lipid-induced hepatic insulin resistance, Aging 5
fat diet-induced obesity in male C57BL/6J mice via the AMPKa-mediated (2013) 582e583.
pathway, Food Funct. 10 (2019) 1940e1947. [23] L.C. Araujo, K.B. Feitosa, G.M. Murata, I.C. Furigo, S.A. Teixeira, C.F. Lucena,
[9] A. Seyedan, Z. Mohamed, M.A. Alshagga, S. Koosha, M.A. Alshawsh, Cynometra L.M. Ribeiro, M.N. Muscar a, S.K. Costa, J. Donato, S. Bordin, Uncaria tomentosa
cauliflora Linn. Attenuates metabolic abnormalities in high-fat diet-induced improves insulin sensitivity and inflammation in experimental NAFLD, Sci.
obese mice, J. Ethnopharmacol. 236 (2019) 173e182. Rep. 8 (2018) 11013.
[10] J. Wauman, L. Zabeau, J. Tavernier, The leptin receptor complex: heavier than [24] T. Jian, C. Yu, X. Ding, J. Chen, J. Li, Y. Zuo, B. Ren, H. Lv, W. Li, Hepatoprotective
expected? Front. Endocrinol. 8 (2017) 30. effect of seed coat of Euryale ferox extract in non-alcoholic fatty liver disease
[11] B.B. Zhang, G. Zhou, C. Li, AMPK: an emerging drug target for diabetes and the induced by high-fat diet in mice by increasing IRs-1 and inhibiting CYP2E1,
metabolic syndrome, Cell Metabol. 9 (2009) 407e416. J. Oleo Sci. 68 (2019) 581e589.
[12] N.A. Shirwany, M.H. Zou, AMPK: a cellular metabolic and redox sensor. A [25] B. Carpenter, G.R. Hemsworth, Z. Wu, M. Maamra, C.J. Strasburger, R.J. Ross,
minireview, Front. Biosci. 19 (2014) 447. P.J. Artymiuk, Structure of the human obesity receptor leptin-binding domain
[13] Y. Li, S. Xu, M.M. Mihaylova, B. Zheng, X. Hou, B. Jiang, O. Park, Z. Luo, E. Lefai, reveals the mechanism of leptin antagonism by a monoclonal antibody,
J.Y. Shyy, B. Gao, AMPK phosphorylates and inhibits SREBP activity to atten- Structure 20 (2012) 487e497.
uate hepatic steatosis and atherosclerosis in diet-induced insulin-resistant [26] T.M. Fong, R.R. Huang, M.R. Tota, C. Mao, T. Smith, J. Varnerin, V.V. Karpitskiy,
mice, Cell Metabol. 13 (2011) 376e388. J.E. Krause, L.H. Van der Ploeg, Localization of leptin binding domain in the
[14] S. Zhang, C. Guo, Z. Chen, P. Zhang, J. Li, Y. Li, Vitexin alleviates ox-LDL- leptin receptor, Mol. Pharmacol. 53 (1998) 234e240.
mediated endothelial injury by inducing autophagy via AMPK signaling [27] Y. Sandowski, N. Raver, E.E. Gussakovsky, S. Shochat, O. Dym, O. Livnah,
activation, Mol. Immunol. 85 (2017) 214e221. M. Rubinstein, R. Krishna, A. Gertler, Subcloning, expression, purification, and
[15] D.W. Lim, H. Kim, S.J. Lee, G.R. Yu, J.E. Kim, W.H. Park, Jwa Kum whan at- characterization of recombinant human leptin-binding domain, J. Biol. Chem.
tenuates nonalcoholic fatty liver disease by modulating glucose metabolism 277 (2002) 46304e46309.
and the insulin signaling pathway, evid. Based complement, Altern. Med. [28] H. Iserentant, F. Peelman, D. Defeau, J. Vandekerckhove, L. Zabeau, J. Tavernier,
2019 (2019), 4589810. https://doi.org/10.1155/2019/4589810, 10 pages. Mapping of the interface between leptin and the leptin receptor CRH2
[16] E.F. Pettersen, T.D. Goddard, C.C. Huang, G.S. Couch, D.M. Greenblatt, domain, J. Cell Sci. 118 (2005) 2519e2527.
E.C. Meng, T.E. Ferrin, UCSF Chimerada visualization system for exploratory

Please cite this article as: S. Inamdar et al., Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice,
Biochemical and Biophysical Research Communications, https://doi.org/10.1016/j.bbrc.2019.08.139

You might also like