Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Received: 24 June 2019 | Revised: 23 August 2019 | Accepted: 4 September 2019

DOI: 10.1111/jfbc.13049

FULL ARTICLE

Berberine chloride ameliorated PI3K/Akt‐p/SIRT‐1/PTEN


signaling pathway in insulin resistance syndrome induced in
rats

Marwa El‐Zeftawy1,2 | Doaa Ghareeb2,3 | Eman R. ElBealy4 | Rasha Saad2 |


Salma Mahmoud2 | Nihal Elguindy5 | Attalla F. El‐kott6,7 | Mohamed El‐Sayed5

1
Biochemistry Department, Faculty of
Veterinary Medicine, The New Valley Abstract
University, New Valley, Egypt The liver is the main organ involved in lipid metabolism process and it helps in drug
2
Biological Screening and Preclinical Trial
detoxification. Insulin resistance is considered one of risk reasons which lead to sev‐
Lab, Biochemistry Department, Faculty of
Science, Alexandria University, Alexandria, eral metabolic diseases. Currently, berberine (BER) occupies a huge challenge against
Egypt
multiple diseases with no toxic effect. The present work was aimed to identify, does
3
Pharmaceutical and Fermentation
Industries Development Center, General
BER‐chloride has a poisonous influence on the liver? and investigating the outcome
Authority of City of Scientific Research and of BER‐chloride on PI3K/Akt‐p/SIRT‐1/PTEN pathway during insulin resistance syn‐
Technology Applications, Alexandria, Egypt
4
drome. The insulin resistance model was achieved in experimental female rats via
Biology Department, College of Science
for girls, King Khalid University, Abha, Saudi high‐fat diet (HFD). Glucose, insulin, lipid profiles, and hepatic oxidative stress pa‐
Arabia rameters were assessed. PI3K, AKt‐p, SIRT‐1, and PTEN levels in hepatic tissue were
5
Biochemistry Department, Faculty of
determined at genome and protein levels. Further adiponectin concentration was
Science, Alexandria University, Alexandria,
Egypt performed in serum, hepatic, and white adipose tissues. Molecular study of fold al‐
6
Biology Department, College of teration in insulin, insulin receptor, and retinol binding protein‐4 (RBP4) in liver was
Science, King Khalid University, Abha, Saudi
Arabia done.
Practical applications
7
Zoology Department, College of
Science, Damanhour University, Damanhour,
Egypt Obesity syndrome causes multiple obstacles in modern years. The current results
revealed elevation the body weight of rats, plasma glucose, homeostatic model as‐
Correspondence
Marwa El‐Zeftawy, Biochemistry sessment, glycated hemoglobin, insulin, and lipid profiles concentrations in a group
Department, Faculty of Veterinary Medicine, of rats, which nourished HFD for 8 weeks and this rise, was diminished after 2 weeks
The New Valley University, New Valley,
Egypt. from BER‐chloride administration. Further, BER‐chloride improved transaminases
Email: marwa@vetnv.au.edu.eg, enzymes, pro‐oxidant, and antioxidant defense system, PI3K, AKt‐p, SIRT‐1, and
marwa_3_1983@yahoo.com
PTEN in the liver, with downregulation of hepatic RBP4. Hence, these data provide a
Funding information crucial message that BER‐chloride enhanced both hepatic function and insulin signal‐
deanship of Scientific Research at
King Khalid University, Abha, KSA for ing pathways that might be of therapeutic importance to insulin resistance with no
supported this work under grant number harmful effect on the liver. BER‐chloride is predicted to be a drug of choice for obe‐
(R.G.P.1/117/40), Grant/Award Number:
R.G.P.1/117/40 sity complications cure.

KEYWORDS
berberine chloride, hepatotoxicity, insulin receptor, liver

J Food Biochem. 2019;00:e13049. wileyonlinelibrary.com/journal/jfbc © 2019 Wiley Periodicals, Inc. | 1 of 11


https://doi.org/10.1111/jfbc.13049
2 of 11 | EL‐ZEFTAWY et al.

1 | I NTRO D U C TI O N homolog (PTEN) were purchased from Biovision (USA). ELISA kit of
insulin, PI3K, AKt‐p, SIRT‐1, PTEN, and adiponectin were purchased
Obesity represents one of the severe health trouble worldwide and from DRG (USA), Wuhan Fine Biological Technology Co. (China), Ray
normally linked with hepatic insulin resistance and hyperglycemia Biotech (Georgia), MyBiosource (USA), Abcam (USA), Bioscience
(Kahn, Hull, & Utzschneider, 2006; Ren, Zhou, Huang, Wang, & Li, (USA), respectively. Foline reagent, thiobarbituric acid (TBA), re‐
2019). Multiple metabolic syndromes as hyperlipidemia, type II dia‐ duced glutathione (GSH), 5′5′dithio‐bis‐2‐nitrobenzoic acid (DTNB),
betes mellitus, nonalcoholic fatty liver, and cardiovascular disorders cumene hydroxide, methyl green, polyethylene glycol (PEG), nitro‐
are accompanied to insulin resistance syndrome. All these metabolic cellulose membrane, sodium dodecyl sulfate, acrylamide, and BCIP/
syndromes may lead to bad prognosis and early mortality in some NBT substrate solution, premixed were obtained from Sigma‐Aldrich
diseased individuals (David, Rifkin, Rabbani, & Ceradini, 2017). PI3K/ Chemical Co. (St. Louis, Mo, USA). Organic solvents, namely etha‐
Akt is one of the pathways which involved in metabolic consequence nol 95%, methanol, and formalin solutions were of HPLC‐grade and
of insulin (Gao et al., 2016). Though numerous pharmaceutical drugs brought from Merck (USA). Other reagents were obtained with high
are available for chronic diseases cure, but these drugs show harmful grade. Primary antibodies of β‐actin, Akt‐p, and PTEN were got from
side effects with long term of use due to their poisonous effect on Biotechne (USA), while PI3K and SIRT‐1 were obtained from Abcam
body organs (Koc et al., 2019; Peter, Basha, Giridharan, Lavinya, & (USA). Also mouse and rabbit secondary antibodies were purchased
Sabina, 2017). from Biotechne (USA) and Abcam (USA). Protein marker was pur‐
Berberine (BER) is a natural alkaloid isoquinoline sequestered chased from Biovision (USA).
from different plants like Berberis vulgaris (Jiang, Li, & Li, 2015). BER
is a strong base which is usually unstable when present in free form
2.2 | Experimental animal protocol and sample
so it usually accompanied with chloride ion in the form of BER‐chlo‐
preparation
ride (Bodiwala, Sabde, Mitra, Bhutani, & Singh, 2011; Li, Yuan, Shang,
& Guo, 2017). BER acts as an anticancer (Liu et al., 2015), anti‐in‐ Female albino Sprague‐Dawley rats (Rattus norvegicus), with weight
flammatory (Liu et al., 2015; Vuddanda, Chakraborty, & Singh, 2010), ranged from 130 to 150 g and aged (10–12) weeks old, were taken
anti‐leishmanial (Vuddanda et al., 2010; Zhang et al., 2016), and from the experimental animal house of Medical Research Institute,
anti‐immunodeficiency virus in humans (Dong et al., 2011). BER also Alexandria University, Egypt. Rats were kept in polycarbonate cages
improved some cardiac syndromes and intestinal infections (Zhang (six rats per cage). They were kept under conventional environments
et al., 2016). Recently BER used as a neuroprotective agent against of temperature and humidity with a 12‐hr photoperiod. Diet and
some neurodegenerative disorders as it has the capability of passing water were provided ad libitum. The experimental rats were han‐
the blood–brain barrier (Jiang et al., 2015). To investigate whether dled in agreement with the National Organizations of Health Guide
BER‐chloride has a protective influence on insulin resistance syn‐ for the Care and Use of Laboratory Animals (NIH 1996). This work
drome, the current study sets up in vivo animal model using high‐fat was done in firm accordance with the approvals in the Guide for the
diet (HFD) feeding. The effects of BER‐chloride on the numerous Care and Use of Laboratory Animals. The protocol was approved ac‐
insulin signaling pathway were investigated. cording to the Ethics of Animal House in Medical Technology Center,
Alexandria University, Egypt. All the animals were adapted 1 week
before the experiment begins.
2 | M ATE R I A L S A N D M E TH O DS
After that, 30 rats were divided into 5 groups (six rats per group),
Group 1 (Sham control group) that were healthy and free from any
2.1 | Materials
disease and they fed the standard food for 10 weeks, Group 2
BER‐chloride was got from Sigma‐Aldrich Chemical Co. (USA). Kits (Control vehicle) fed on low‐fat diet (LFD) for 8 weeks then received
of blood glucose level (BGL), protein, lipid profiles (total cholesterol 20% PEG by intragastric tube (10 ml/kg Bwt) for 2 weeks. Group
“TC,” triacylglycerol “TG,” and high density lipoprotein cholesterol 3 (Control BER‐chloride) fed LFD for 8 weeks, then orally given
“HDL‐c”), glycated hemoglobin (HbA1C), as well as creatinine, urea, BER‐chloride dissolved in 20% of PEG (100 mg/kg Bwt) for 2 weeks.
alanine aminotransfersase (ALT), and aspartate aminotransferase Group 4 (Induction group) fed on HFD for 8 weeks, then orally given
(AST) kits were purchased from Spinreact (Spain), Human (Germany), 20% of PEG (10 ml/kg Bwt) for 2 weeks. Group 5 (Induction treated
Biosystem (Egypt), and Biolabo (France), respectively. Ribonucleic group) fed HFD for 8 weeks, then orally given BER‐chloride dis‐
acid (RNA) extraction kit, Maxime reverse transcription (RT) premix solved in 20% of PEG (100 mg/kg Bwt) for 2 weeks. Doses of BER‐
kit, 2x Taq master mix, deoxyribo nucleic acid (DNA) Ladder, RNase‐ chloride and PEG which were used in this protocol referred to other
free water, and the primer sequences of β‐actin, insulin, insulin recep‐ previous studies (El‐Sayed, Ghareeb, Talat, & Sarhan, 2013; Ghareeb
tor (IR), and retinol binding protein‐4 (RBP4) were purchased from et al., 2015; Saleh, Attia, & Ghareeb, 2018).
Qiagen (Germany), Intron Biotechnology (Korea) and Fermentas, Rat's body weights were noticed in the first and last week of ther‐
Thermo fisher scientific (Germany), respectively. Additionally, prim‐ apy. Blood sampling and animal scarification were performed under
ers of phosphatidyl inositol‐3‐kinase (PI3K), phosphorylated protein anesthesia, and all efforts were made to decrease suffering. After the
kinase B (Akt‐p), sirtuin type 1 (SIRT‐1), and phosphatase and tension end of the experimental period, rats were fasted overnight after 8‐hr,
EL‐ZEFTAWY et al. | 3 of 11

blood samples were collected in sodium fluoride tubes for measur‐ CACTGTGTTGGCATA‐3′, RBP4 forward primer 5′‐TTTTCTGTG
ing of fasting BGL. After a full fasting period (12‐hr), blood samples GACGAGAAGGGT‐3′ and reverse primer 5′‐TGGTCATCGTTTCCT
were collected, and then centrifuged at 3,000 rpm for 10 min. The CGCTTG‐3′, IR forward primer 5′‐TGACAATGAGGA ATGTGGGGAC‐3′
obtained serum was kept at −20°C until analyzed. Hepatic tissues and and reverse primer 5′‐GGGCAAACTTTCTGACAATGACTG‐3′, insulin
white adipose tissue from all groups were excised immediately and forward primer 5′‐TTCTACACACCCAAGTCCCGTC‐3′ and reverse
washed with ice‐cold saline. Homogenization was performed in 0.1M primer 5′‐ATCCACAATGCCACGCTTCTGC‐3′, PI3K forward primer
of sodium phosphate‐buffer, pH 7.4 (for hepatic tissue) and in 0.15M 5′‐AACACAGAAGACCAATACTC‐3′, and reverse primer 5′‐TTCGCC
of potassium chloride (for white adipose tissue) (Steinberg, Vaughan, ATCTACCACTAC‐3′, Akt‐p forward primer 5′‐GTGGCAAGATGTGTAT
& Margolis, 1961). The homogenate was centrifuged at 4,000 rpm GAG‐3′, and reverse primer 5′‐CTGGCTGAGTAGGAGAAC‐3′, SIRT‐1
for 15 min at 4°C and supernatant was kept at −80°C until analysis forward primer 5′‐TGAAGCTGTTCGTGGAGATATTTTT‐3′, and reverse
(Pandian, Anuradha, & Viswanathan, 2002). In each group, part of primer 5′‐CATGATGGCAAGTGGCTCAT‐3′, and PTEN forward primer
hepatic tissue was preserved in liquid nitrogen, and stored at −80°C 5′‐CCCAGTTTGTGGTCTGCCAGC‐3′ and reverse primer 5′‐ATGAGC
for total RNA isolation and polymerase chain reaction (PCR) analy‐ TTGTCCTCCCGCCG‐3′.
sis. Another part of liver and white adipose tissue were fixed at 10% PCR program was optimized for the primer pair and the PCR pro‐
neutral buffered formalin solution for histopathological examination. tocol was 40 cycles, each consisted of 30 s denaturation at 95°C, 30 s
annealing at 52.5°C, 51.5°C, 50°C, 52°C, 58°C, 58.8°C, 63.9°C, and
68.9°C for β‐actin, RBP4, IR, insulin, PI3K, Akt‐p, SIRT‐1, and PTEN,
2.3 | Biochemical, molecular, and
respectively, and 60 s extension at 72°C. The primers were run on
histopathological studies
the Mini Cycler (Eppendorf, Labcaire, Germany). The PCR products
were run on 1.5% agarose gel. Gels were stained with ethidium bro‐
2.3.1 | Biochemical and molecular studies
mide, visualized by 30 nm Ultraviolet Radiator (Alpha‐Chem. Imager,
BGL was determined by a glucose assay kit that is dependent on USA), and photographic record were made. The optical density and
glucose oxidase‐peroxidase method. Serum insulin was analyzed the microgram content of bands were calculated by the UVIBAND
through the DRG insulin ELISA kit. The insulin resistance was evalu‐ MAX software program.
ated by calculating the HOMA‐IR as previously described (Han et al., Western blot was performed to determine the protein level ex‐
2018). HbA1C % was assessed by Biosystem kit. Total protein con‐ pression of PI3K, Akt‐p, SIRT‐1, and PTEN in hepatic tissue. After
centration was measured spectrophotometrically using Beirut assay separation of protein (100 μg of each sample was used) by sodium
based kit. Serum lipid profile (TC, TG, and HDL‐c), urea, creatinine, dodecyl sulfate poly acrylamide gel electrophoresis (SDS‐PAGE)
ALT, and AST were performed according to the instructions of their technique, the gel was transferred to nitrocellulose membrane.
commercial kits. LDL‐c and vLDL‐c levels were calculated according The membrane was incubated with labeled antibodies specific for
to previous studied formula by Friedewald, Levy, and Fredrickson the protein of interest and β‐actin was used as an internal control.
(1972). Standardized methods were used to determine the concen‐ Following adding the secondary antibody and incubation time, the
tration of thiobarbituric acid reactive substances (TBARS) (Tappel & unbound antibody was washed off, leaving only antibody which
Zalkin, 1959) and GSH in liver (Ellman, 1959). bounded to the protein of interest so only one band was visible and
In addition, hepatic activities of xanthine oxidase (XO) (Litwack, its thickness was corresponded to the amount of protein which pres‐
Bothwell, Williams, & Elvehjem, 1953), glutathione peroxidase (GPx) ent in a sample and it was detected using BCIP/NBT substrate solu‐
(Chiu, Stults, & Tappel, 1976; Paglia & Valentine, 1967), and adenine tion. The intensities of obtaining bands were estimated using image
triphosphatase (ATPase) (Candeias, Abreu, Pereira, & Cruz‐Morais, analyzing system (UVIBAND MAX software).
2009) were done. PI3K, Akt‐p, SIRT‐1, and PTEN concentrations in
hepatic tissue homogenate and adiponectin level in serum, liver ho‐
2.3.2 | Histopathological study
mogenate, and white adipose tissue were estimated using ELISA kits.
These tests employ the quantitative sandwich enzyme immunoassay Hepatic and white adipose tissues of each rat from each group was
technique. excised and immediately fixed at 10% neutral buffered formalin
Total RNA was isolated from liver using total RNA extraction Kit solution after washing with ice‐cold normal saline. The resultant
and processed according to kit manufacturer's directions. After that fixed tissue samples were used for histopathological examination
the level of total RNA was measured by spectrophotometer at 260 in the Histopathology Laboratory of Medical Technology Center,
and 280 nm. One microgram of the isolated RNA was reverse tran‐ Alexandria University, using the routine procedures developed in
scribed into complementary DNA (cDNA) using reverse transcrip‐ the respective laboratories. The tissue was cut at 3 mm thick, and
tase (Maxime RT Pre‐Mix kit, Fermentas, EU). the blocks were embedded in paraffin. Using a rotary microtome,
Primers used for PCR technique were constructed using the sections of 8 µm thickness were cut. The sections were stained with
known sequences for the respective genes. The gene‐specific hematoxylin and eosin and examined under Olympus microscope
primer sequences were as follows: β‐actin forward primer 5′‐CAT (Olympus, Tokyo, Japan) at (100X) magnification for any histopatho‐
CACTATCGGCAATGAGC‐3′ and reverse primer 5′‐GACAG logical changes.
4 of 11 | EL‐ZEFTAWY et al.

TA B L E 1 Effect of berberine (BER) chloride on body weight, blood glucose level (BGL), serum insulin, homeostatic model assessment
insulin resistance (HOMA‐IR), and glycated hemoglobin (HbA1C) after the treatment of diabetic‐induced experimental animals

Serum insulin
Groups Body weight (g) BGL (mg/dl) (µIU/ml) HOMA‐IR (mg/dl) HbA1C (%)

Sham control 141.0 ± 7.4a 76.0 ± 1.4a 10.0 ± 0.2a 1.91 ± 0.061a 4.2 ± 0.4a
a a a a
Control vehicle (LFD + 20% PEG) 142.2 ± 8.04 73.33 ± 2.6 10.1 ± 0.6 1.9 ± 0.13 4.4 ± 0.42a
Control BER‐chloride 144.0 ± 6.07a 75.2 ± 3.31a 10.45 ± 0.33a 1.92 ± 0.12a 4.0 ± 0.24a
b c c c
Induction (HFD + 20% PEG 202.3 ± 5.39 180.1 ± 4.38 20.17 ± 2.93 8.97 ± 1.37 6.1 ± 0.39c
(10 ml/kg Bwt)
Induction treated with 200.5 ± 8.02b 97.7 ± 5.61b 15.67 ± 2.42b 3.76 ± 0.48b 5.1 ± 0.18b
BER‐chloride

Note: Values represent the mean ± SD of six rats. ANOVA (one‐way) followed by Student–Newman–keuls test.
Means with letters (a), (b), and (c) were statistical represented compared to sham control group as follow: a = p < .001, b = p < .01, c = p < .05.

2.4 | Statistical analysis


Data were analyzed using Primer of Biostatistics software program
(Version 5.0) by one‐way ANOVA. Significance of means ± SD was
detected via using the multiple comparisons Student–Newman–
keuls test at p < .05. Adiponectin correlation was analyzed by SPSS
(Version 20.0) software program, using person coefficient (r).

3 | R E S U LT S

3.1 | Body weight, BGL, and insulin resistance


parameters
Feeding of HFD for 8‐weeks leads to body weight elevation more
than control levels. Fasting BGL, homeostatic model assessment in‐
sulin receptor (HOMA‐IR), and HbA1C were also elevated 1.4, 3.7, and
45.2‐folds, in comparison to sham control rats. Moreover, elevation of
insulin concentration was noticed in HFD group 1.0‐fold in serum and
72.3‐fold in liver homogenate. Administration of BER‐chloride leads to
reduction of fasting BGL, serum insulin, HOMA‐IR, and HbA1C to 0.5,
0.2, 0.6, and 0.2‐folds, respectively, compared with HFD rats (Table 1).
Moreover, HFD up regulated expression of insulin gene in liver and
BER‐chloride therapy for 2 weeks wasn't significant (Figure 1).
F I G U R E 1 Effect of berberine (BER) chloride on the fold change
of insulin, insulin receptor (IR) and retinol binding protein‐4 (RBP4)
3.2 | Lipid profiles parameters genes. (A) Agrose gel electrophoresis of gene expression of insulin
(293 bp), IR (129 bp), and RBP4 (392 bp) compared to β‐actin
Lipid profile showed a significant rise in the HFD group than that of
(300 bp). (B) Fold change of gene expression in liver homogenate
sham control where TC increased 1.2‐fold and TG, LDL‐c, and vLDL‐c
after the treatment of diabetic‐induced experimental animals
were 0.9‐fold increase, while HDL‐c was significantly decreased by represented as 6 rats ± SE. ANOVA (one‐way) followed by Student–
43%. Lipid profiles were partially repaired after using BER‐chloride Newman–keuls test. Means with letters (a), (b), (c), and (d) were
as therapy for 2 weeks (Table 2). statistically represented compared to sham control group as a at
p < .001, b at p < .01, c at p < .05, and d at p > .05

3.3 | Oxidative stress markers, serum transaminases


50.9%, 41.9%, and 35.2% in the HFD group compared to sham con‐
activity, and kidney function tests
trol and after administration of BER‐chloride therapy; those previ‐
Experimental HFD rats exhibited an elevation of TBARS and XO (1.5 ous parameters were increased by percentage 42.9, 33.7, and 26.1,
and 0.8‐folds) in comparison to sham control. Both TBARS and XO respectively (Table 3). Also, HFD intake increased liver transami‐
were decreased nearly 0.4‐ and 0.2‐folds after 2 weeks of BER‐chlo‐ nases (ALT and AST) and kidney function tests (urea and creati‐
ride treatment. However, GSH, GPx, and ATPase were decreased by nine) comparing to sham control one. Administration of 100 mg/kg
EL‐ZEFTAWY et al. | 5 of 11

TA B L E 2 Effect of berberine (BER) chloride on total cholesterol (TC), triacylglycerol (TG), high‐density lipoprotein cholesterol (HDL‐c),
low‐density lipoprotein cholesterol (LDL‐c), and very low‐density lipoprotein cholesterol (vLDL‐c) after the treatment of diabetic‐induced
experimental animals

Groups TC (mg/dl) TG (mg/dl) HDL‐c (mg/dl) LDL‐c (mg/dl) vLDL‐c (mg/dl)


a a a a
Sham control 130.5 ± 2.79 110.3 ± 3.6 59.33 ± 3.14 110.33 ± 3.62 22.1 ± 0.7a
a a a a
Control vehicle (LFD + 20% PEG) 127.3 ± 4.41 110.0 ± 4.47 64.3 ± 3.01 41.0 ± 3.74 22.3 ± 1.2a
Control BER‐chloride 129.3 ± 2.25a 111.2 ± 2.40a 63.2 ± 2.56a 43.9 ± 4.24a 21.77 ± 0.5a
c c c c
Induction (HFD + 20% PEG 282.2 ± 3.31 210.5 ± 3.73 33.3 ± 2.88 206.7 ± 2.78 42.9 ± 0.4c
(10 ml/kg Bwt)
Induction treated with 164.5 ± 4.14b 141.5 ± 2.26b 42.8 ± 3.37b 93.4 ± 6.20 b 28.133 ± 0.7b
BER‐chloride

Note: Values represent the mean ± SD of six rats. ANOVA (one‐way) followed by Student–Newman–keuls test.
Means with letters (a), (b), and (c) were statistical represented compared to sham control group as follow: a = p < .001, b = p < .01, c = p < .05.

TA B L E 3 Effect of berberine (BER) chloride on hepatocyte thiobarbituric acid reactive substances (TBARS), reduced glutathione (GSH),
xanthine oxidase (XO), glutathione peroxidase (GPx), and adenine triphosphatase (ATPase) status after the treatment of diabetic‐induced
experimental animals

TBARS (mmol/mg GSH (mg/mg XO (IU/mg GPx (U/mg ATPase (µmol/pi/


Groups protein) protein) protein) protein) min/mg protein)

Sham control 2.1 ± 0.02a 0.285 ± 0.05a 128.0 ± 1.6a 7.06 ± 0.3a 0.71 ± 0.02a
Control vehicle (LFD + 20% PEG) 2.03 ± 0.04a 0.29 ± 0.06a 130.17 ± 4.26a 7.02 ± 0.55a 0.73 ± 0.01a
a a a a
Control BER‐chloride 2.12 ± 0.28 0.29 ± 0.05 129.67 ± 4.46 7.17 ± 0.22 0.71 ± 0.01a
Induction (HFD + 20% PEG 5.25 ± 0.35c 0.14 ± 0.02c 231.5 ± 7.56c 4.1 ± 0.33c 0.46 ± 0.05c
(10 ml/kg Bwt)
Induction treated with 3.15 ± 0.24b 0.20 ± 0.03b 171.3 ± 3.83b 5.48 ± 0.33b 0.58 ± 0.03b
BER‐chloride

Note: Values represent the mean ± SD of six rats. ANOVA (one‐way) followed by Student–Newman–keuls test.
Means with letters (a), (b), and (c) were statistical represented compared to sham control group as follow: a = p < .001, b = p < .01, c = p < .05.

TA B L E 4 Effect of berberine (BER) chloride on serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities and
kidney function tests after the treatment of diabetic‐induced experimental animals

Groups ALT (U/ml) AST (U/ml) Urea (mg/dl) Creatinine (mg/dl)


a a a
Sham control 33.5 ± 2.1 46.0 ± 3.2 33.4 ± 2.3 0.71 ± 0.1a
Control vehicle (LFD + 20% PEG) 34.2 ± 4.6a 49.3 ± 1.8a 31.83 ± 4.6a 0.72 ± 0.04a
a a a
Control BER‐chloride 32.2 ± 1.94 49.5 ± 3.78 29.67 ± 2.16 0.71 ± 0.04a
Induction (HFD + 20% PEG (10 ml/kg Bwt) 58.5 ± 5.09c 85.7 ± 5.68c 56.17 ± 2.14c 0.90 ± 0.05b
b b b
Induction treated with BER‐chloride 40.8 ± 3.71 62.5 ± 6.80 39.83 ± 3.49 0.75 ± 0.06a

Note: Values represent the mean ± SD of six rats. ANOVA (one‐way) followed by Student–Newman–keuls test.
Means with letters (a), (b), and (c) were statistical represented compared to sham control group as follow: a = p < .001, b = p < .01, c = p < .05.

Bwt BER‐chloride in HFD rats for 2 weeks leads to decrease liver BER‐chloride to that group of rats leads to increase PI3K, AKt‐p,
transaminases activities and kidney function tests nearly 0.3‐ and and SIRT‐1 to 84.4%, 49%, and 120%, respectively, at ELISA level,
0.2‐folds (Table 4). also they 4.4‐, 0.78‐, 10.5‐ and 0.5‐folds increased at genome
level and also they 8.5‐, 1.7‐ and 0.5‐folds raised at protein level.
However, PTEN was increased in HFD rats by 66.6% in ELISA level
3.4 | Insulin signaling pathway parameters
and 1.25‐ and 21‐folds elevated at genome and protein levels,
As shown in Table 5 and Figures 1‒3 the marked significant de‐ respectively. After BER‐chloride treatment PTEN diminished to
cline of PI3K, AKt‐p and SIRT‐1 by 57.1%, 42.9%, and 61.9%, was 27.5% at ELISA level, 50% at genome level, and 74.2% at protein
noticed in HFD rats compared to sham control at ELISA level, also level. Additionally, RBP4 was reduced from 1.5‐fold to 0.4‐fold
they decreased by 76.2%, 77.5%, and 93.1% at genome level and after BER‐chloride treatment. However, BER‐chloride failed to af‐
by 92.9%, 42.1%, and 76.0% at protein level. Administration of fect the HFD side effect on IR expression.
6 of 11 | EL‐ZEFTAWY et al.

TA B L E 5 Effect of berberine (BER) chloride on phosphatidyl inositol‐3‐kinase (PI3K), phosphorylated protein kinase B (Akt‐p), Sirtuin
type 1 (SIRT‐1), and phosphatase and tension homolog (PTEN) levels in hepatocyte after the treatment of diabetic‐induced experimental
animals

Groups PI3K (pg/g) Akt‐p (pg/g) SIRT‐1 (ng/g) PTEN (pg/g)


c c c
Sham control 493.0 ± 12.3 350.0 ± 23.5 105.0 ± 10.2 1,200 ± 28.9a
c c c
Control vehicle (LFD + 20% PEG) 486.0 ± 12.0 335.0 ± 19.3 110.0 ± 6.9 1,250.0 ± 25.3a
Control BER‐chloride 491.0 ± 23.5c 342.0 ± 18.9c 109.0 ± 9.7c 1,150.0 ± 65.3a
a a a
Induction (HFD + 20% PEG (10 ml/ 211.5 ± 21.5 200.0 ± 12.1 40.0 ± 1.2 2,000.0 ± 28.9c
kg Bwt)
Induction treated with BER‐chloride 390.0 ± 31.2b 298.0 ± 14.2b 86.0 ± 8.5b 1,450.0 ± 23.5b

Note: Values represent the mean ± SD of six rats. ANOVA (one‐way) followed by Student–Newman–keuls test.
Means with letters (a), (b), and (c) were statistical represented compared to sham control group as follow: a = p < .001, b = p < .01, c = p < .05.

F I G U R E 3 Effect of berberine (BER) chloride on the protein


levels of phosphatidyl inositol‐3‐kinase (PI3K), phosphorylated
protein kinase B (Akt‐p), Sirtuin type 1 (SIRT‐1), and phosphatase
and tension homolog (PTEN). (A) Western blot of protein expression
of PI3K (−85 kDa), Akt‐p (−60 kDa), SIRT‐1 (−120 kDa), and PTEN
F I G U R E 2 Effect of berberine (BER) chloride on the fold change (−54 kDa) compared to β‐actin (−45 kDa). (B) Protein levels in liver
of phosphatidyl inositol‐3‐kinase (PI3K), phosphorylated protein homogenate after the treatment of diabetic‐induced experimental
kinase B (Akt‐p), Sirtuin type 1 (SIRT‐1), and phosphatase and animals represented as 6 rats ± SE. ANOVA (one‐way) followed by
tension homolog (PTEN) genes. (A) Agrose gel electrophoresis of Student–Newman–keuls test. Means with letters (a), (b), (c), and (d)
gene expression of PI3K (195 bp), Akt‐p (195 bp), SIRT‐1 (82 bp), and were statistically represented compared to sham control group as a
PTEN (79 bp) compared to β‐actin (300 bp). (B) Fold change of gene at p < .001, b at p < .01, c at p < .05, and d at p > .05
expression in liver homogenate after the treatment of diabetic‐
induced experimental animals represented as 6 rats ± SE. ANOVA
(one‐way) followed by Student–Newman–keuls test. Means with elevated after BER‐chloride administration for 2 weeks (48%, 385%,
letters (a), (b), (c) and (d) were statistically represented compared to
and 65.3%), respectively (Table 6).
sham control group as a at p < .001, b at p < .01, c at p < .05, and d
at p > .05
3.6 | Histopathological results
3.5 | Adiponectin concentration
The biochemical and molecular alterations were confirmed by
Adiponectin percentage in serum, liver, and white adipose tissue of the histopathological study in both liver and white adipose tissue
HFD rats was reduced (52%, 80%, and 45%), and this percentage was (Figures S1a–e and S2a–e).
EL‐ZEFTAWY et al. | 7 of 11

TA B L E 6 Effect of berberine
Adiponectin (ng/ml)
(BER) chloride on adiponectin level in
serum, liver, and white adipose tissue White adi‐
homogenates after the treatment of Groups Serum Hepatic tissue pose tissue
diabetic‐induced experimental animals
Sham control 1.42 ± 0.49a 11.03 ± 0.97a 21.42 ± 1.65a
Control vehicle (LFD + 20% 1.39 ± 0.49a 10.67 ± 0.97a 22.97 ± 1.12a
PEG)
Control BER‐chloride 1.59 ± 0.26a 10.77 ± 1.15a 23.43 ± 1.51a
b b
Induction (HFD + 20% PEG 0.682 ± 0.11 2.0 ± 0.7 11.78 ± 1.89b
(10 ml/kg Bwt)
Induction treated with 1.01 ± 0.18a 9.7 ± 0.9a 19.47 ± 1.67a
BER‐chloride

Note: Values represent the mean ± SD of six rats. SPSS (Version 20.0).
Means with letters (a), (b), and (c) were statistical represented compared to sham control group as
follow: a = p < .001, b = p < .01, c = p < .05.

Figure S1a–e discovered liver's histopathological data of all stud‐ elevates gluconeogenesis in the liver, so hyperglycemia is occur‐
ied groups. The liver architecture in control vehicle and control BER‐ ring (Hutchison, Harrison, Stepto, Meyer, & Teede, 2008), hinders
chloride was normal in comparison with a sham control group (Figure the insulin signaling in muscle and decreases the uptake of glucose
S1a–c). On the contrary, pathological studies revealed that the liver by reduction the action of PI3K (Yousefi & TaheriChadorneshin,
of the HFD group contained obvious fat droplets, necrosis, and in‐ 2018). Defects in PI3K reported in other findings (Jiang & Zhang,
flammation (Figure S1d). For HFD treated group with BER‐chloride, 2002). Preceding studies showed the highest concentration of
the necrosis and inflammation sites disappeared, and the steatosis RBP4 are accompanied by increase in body mass index, insulin
decreased from moderate to mild grade (Figure S1e). resistance, and hypertriglyceridemia (Domingos, Queiroz, Lotufo,
Further histopathological results of white adipose tissue were Bensenor, & Titan, 2017). Study of PTEN had a significant impor‐
demonstrated in (Figure S2, a‐e). Control rat's exposed normal tissue tance, PTEN is a lipid phosphatase inhibits insulin signaling by
(Figure S2a). Both PEG and BER‐chloride administrated groups were dephosphorylating phosphatidylinositol (3,4,5) triphosphate (PIP 3)
similar to control rats (Figure S2b and S2c). However, adipose tissue to phosphatidylinositol (4,5) diphosphate (PIP 2) (Cully, You, Levine,
of HFD rats showed multiple fibrosis and degeneration for the archi‐ & Mak, 2006). Hence, PTEN is antagonizing the action of PI3K and
tecture of the adipocytes (Figure S2d). Treatment of HFD rats with inhibits Akt as appear in current results, where HFD rats exhibited
BER‐chloride for 2 weeks lead to the regeneration of the cells and an elevation in PTEN concentration and the decline of PI3K (Yao
reduction of the lipid droplets in it (Figure S2e). & Nyomba, 2008).
It is known that, SIRT1 is a key regulator of lipid mobilization
through its action together with adenosine monophosphate‐acti‐
4 | D I S CU S S I O N vated protein kinase (AMPK) through increasing fatty acid metab‐
olism (Merksamer et al., 2013). So, decrease in SIRT1 concentration
In recent years, insulin resistance has a huge challenge, it repre‐ in HFD rats is linked by hyperlipidemia and insulin resistance due
sented one of the dangerous metabolic conditions, and in many to reduction of phosphorylated and/or activated AMPK is resulting
cases it occurs due to bad dietary habits (Haslam & James, 2005) in lipid synthesis increase (Boulet et al., 2015). The results of the
or due to some diseases like type II diabetes mellitus (Seko et al., present study are in agreement with previous works (Deng, Chen, &
2018). In the current study, HFD feeding for 8 weeks induced hy‐ Li, 2007). Further, SIRT1 has numerous roles in insulin signaling path‐
perinsulinaemia which is attributed to inability of liver to utilize way it regulates insulin secretion from β‐cell of pancreas via reduc‐
the secreted insulin, although the normal role of pancreatic β‐cell tion uncoupling protein‐2 (UCP2) gene expression and improvement
(Arnold et al., 2018). During this time the rats become more obese the depolarization in β‐cell of pancreas (Liang, Kume, & Koya, 2009)
compared to LFD controls due to elevation of insulin concentra‐ and regulates the insulin signaling pathway through deacetylation
tion which inhibit fatty acid oxidation so fats accumulated mainly of insulin receptor substrate‐2 (IRS2) and activation of Akt in cells
in the liver because it is the main organ of oxidation process (Liu et (Yoshizaki et al., 2009). From those mentioned mechanisms of SIRT1
al., 2017). Higher levels of fasting insulin, glucose, and HOMA‐IR and PTEN, the current study exhibited decreased of Akt‐p concen‐
index confirming the state of insulin resistance. Furthermore, in‐ tration in hepatic cells of HFD rats. Moreover adiponectin has a vital
creasing of HbA1c is another indicator of insulin resistance and cor‐ role in the insulin resistance pathway; it has a regulating influence
related with renal function parameters elevation (Fiorentino et al., on insulin sensitivity (Wu, Ni, & Lu, 2016). It was noticed that dis‐
2017). Additionally, the results of this study showed up regulation turbance in lipid metabolism and excessive fat deposition leads to
of RBP4 expression in HFD rats. From previous hypothesis RBP4 abnormal synthesis of adiponectin (Burnstock & Gentile, 2018). HFD
8 of 11 | EL‐ZEFTAWY et al.

rats associated with decline of adiponectin concentration in serum, In recent years with regard to the side effects of artificial med‐
hepatic, and adipose tissues which may be attributed to disruption icines, increasing attention has been paid by researchers to herbal
of both adiponectin receptors‐1 and 2 leading to elevation of glucose medicines. Our in vivo study revealed that treatment with BER‐
concentration and reduce the action of peroxisome proliferator acti‐ chloride has negative influence on insulin resistance through ac‐
vated receptor α‐signaling pathways, respectively, and finally insulin tivating two proteins involved in several physiological processes,
resistance occurs (S. Li, Shin, Ding, & van Dam, 2009). Further, there SIRT‐1 and AMPK (Lin et al., 2018). Those two proteins able to
are some theories suggesting the role of SIRT1 in the regulation of activate each other, AMPK activate SIRT‐1 by elevation the con‐
adiponectin secretion from the adipocytes by deacetylating of fork centration of nicotinamide phosphoribosyltransferase and SIRT‐1
head transcription factor O1 (FOXO‐1) protein and enhance the stimulates AMPK through deacetylation of serine‐threonine
transcription of gene that encodes adiponectin in adipocytes (Qiao & kinase LKB1 (Hardie, 2015; Sun et al., 2014). Also, BER has the
Shao, 2006). Hence, SIRT1 decline effect on adiponectin secretion. capacity to recover insulin resistance syndrome through other
The role of SIRT1 is extended to control the production of reac‐ mechanisms, where it protects β‐cell of islets of Langerhans from
tive oxygen species (ROS) (Colak et al., 2011) as SIRT1 is considered damage, allows glucose uptake by skeletal muscle, improves he‐
one of the important proteins that protect cells from stress damage patic gluconeogenesis and decreases the concentration of lipids
(Deng et al., 2007). Under normal condition the hepatocyte balance in the blood (Pirillo & Catapano, 2015; Yang et al., 2014). As a
the oxidative stress through the action of antioxidant enzymes such result of SIRT‐1 and AMPK pathway activation, adiponectin con‐
as GPx which converts hydrogen peroxide (H2O2) to water (Flores, centration was restored after BER‐chloride therapy in our study
Adhami, & Martins‐Green, 2016). Rats have insulin resistance are which similar to previous results (Wu et al., 2015). Elevation of
showing low GPx activity so H2O2 accumulated and hepatic cells adiponectin concentration is linked by regulation of β‐oxidation of
damaged. These results were confirmed by elevation of liver trans‐ fatty acids and glucose metabolism (Lin & Li, 2012; Yamauchi et al.,
aminases (ALT and AST). H2O2 accumulation also affected on renal 2002). Hence, treated rats with BER‐chloride showed significant
tissue, which confirmed by an increase both urea and creatinine lev‐ decline of lipid profiles and BGL.
els. Another cause of the elevation of ROS in case of insulin resis‐ Also, BER‐chloride ameliorates hyperlipidemia results from in‐
tance is attributed to the dysregulated production of adipocytokines sulin resistance via different mechanisms, BER lowers blood cho‐
where plasma adiponectin concentration is inversely correlated with lesterol levels through inhibiting cholesterol uptake and absorption
systemic oxidative stress (Furukawa et al., 2004). in the intestine (Wang et al., 2014), reduces TC secretion from

F I G U R E 4 Schematic diagram for the effect of berberine (BER) chloride on insulin signaling in high‐fat diet (HFD) insulin resistance
induced rats
EL‐ZEFTAWY et al. | 9 of 11

enterocytes to the circulation by down regulation acetyl CoA trans‐ REFERENCES


ferase II enzyme (Wang et al., 2017) and increases the regulation of
Abidi, P., Zhou, Y., Jiang, J. D., & Liu, J. (2005). Extracellular signal‐regu‐
LDL‐receptor and hence, BER decreases the concentration of LDL‐c lated kinase‐dependent stabilization of hepatic low‐density lipopro‐
(Abidi, Zhou, Jiang, & Liu, 2005). Moreover, BER chloride is able to tein receptor mRNA by herbal medicine berberine. Arteriosclerosis,
improve glucose control by stimulation the glycolysis in peripheral Thrombosis, and Vascular Biology, 25(10), 2170–2176. https​://doi.
tissue, inhibition of FOXO‐1 and hepatic nuclear factor 4, lead to org/10.1161/01.ATV.00001​81761.16341.2b
Arnold, S. E., Arvanitakis, Z., Macauley‐Rambach, S. L., Koenig, A. M.,
suppression of glucose‐6‐phosphatase and phosphoenolpyruvate
Wang, H.‐Y., Ahima, R. S., … Nathan, D. M. (2018). Brain insulin re‐
carboxykinase enzymes which responsible for liver gluconeogenesis sistance in type 2 diabetes and Alzheimer disease: Concepts and
(Kim et al., 2009; Xia et al., 2011) and activation of glucose trans‐ conundrums. Nature Reviews Neurology, 14(3), 168–181. https​://doi.
port‐1 (GLUT1) (Cok et al., 2011). org/10.1038/nrneu​rol.2017.185
Bodiwala, H. S., Sabde, S., Mitra, D., Bhutani, K. K., & Singh, I. P. (2011).
The current study revealed BER‐chloride has the ability to
Synthesis of 9‐substituted derivatives of berberine as anti‐HIV
raise the concentration of antioxidant enzymes (GSH, GPx, and agents. European Journal of Medicinal Chemistry, 46(4), 1045–1049.
XO) by reducing the high level of lipid peroxidation (Lao‐ong, https​://doi.org/10.1016/j.ejmech.2011.01.016
Chatuphonprasert, Nemoto, & Jarukamjorn, 2012). Also BER has Boulet, M. M., Chevrier, G., Grenier‐Larouche, T., Pelletier, M., Nadeau,
M., Scarpa, J., … Tchernof, A. (2015). Alterations of plasma me‐
the ability to prevent nicotinamide adenine dinucleotide phosphate
tabolite profiles related to adipose tissue distribution and car‐
(NADPH) oxidase, which is the main source of ROS production diometabolic risk. American Journal of Physiology‐Endocrinology and
(Cheng et al., 2013). These results are in agreement with previous Metabolism, 309(8), E736–E746. https​://doi.org/10.1152/ajpen​
theories which proved that BER is a strong antioxidant molecule due do.00231.2015
Burnstock, G., & Gentile, D. (2018). The involvement of purinergic
to its ability to scavenge free radicals (Kumar et al., 2015). Moreover,
signalling in obesity. Purinergic Signal, 14(2), 97–108. https​://doi.
BER‐chloride exerts protective effect against ROS through SIRT‐1 org/10.1007/s11302-018-9605-8
activation where SIRT‐1 is capable to modulate NOX4/NADPH ox‐ Candeias, M. F., Abreu, P., Pereira, A., & Cruz‐Morais, J. (2009). Effects of
idative subunit (Karbasforooshan & Karimi, 2018). Fall of ROS pro‐ strictosamide on mouse brain and kidney Na+, K+‐ATPase and Mg2+‐
ATPase activities. Journal of Ethnopharmacology, 121(1), 117–122.
duction lead to decrease the level of liver transaminases activity in
https​://doi.org/10.1016/j.jep.2008.08.032
rats, group administrated BER‐chloride. In the present study, BER‐ Cheng, F., Wang, Y., Li, J., Su, C., Wu, F., Xia, W.‐H., … Tao, J. (2013).
chloride down regulates RBP4 which acts as an effective insulin sen‐ Berberine improves endothelial function by reducing endothelial
sitizing function (Zhang, Xu, Guo, Meng, & Li, 2008). From previous microparticles‐mediated oxidative stress in humans. International
studies, it was noticed that reduction of RBP4 concentration is re‐ Journal of Cardiology, 167(3), 936–942. https​://doi.org/10.1016/j.
ijcard.2012.03.090
lated to elevation of HDL‐c and decrease TG levels in some patients.
Chiu, D. T., Stults, F. H., & Tappel, A. L. (1976). Purification and properties of
Also, as a result of the decline of SIRT‐1 in HFD rats, elevation of rat lung soluble glutathione peroxidase. Biochimica et Biophysica Acta,
TBARS and XO, and decrease in GPx and GSH was reported and this 445(3), 558–566. https​://doi.org/10.1016/0005-2744(76)90110-8
was improved after BER‐chloride administration. From this study, we Cok, A., Plaisier, C., Salie, M. J., Oram, D. S., Chenge, J., & Louters, L.
L. (2011). Berberine acutely activates the glucose transport activity
conclude that BER‐chloride can be considered one of therapeutics
of GLUT1. Biochimie, 93(7), 1187–1192. https​://doi.org/10.1016/j.
used to help the treatment of insulin resistance syndrome through biochi.2011.04.013
its influence on multiple insulin signaling pathways. A schematic rep‐ Colak, Y., Ozturk, O., Senates, E., Tuncer, I., Yorulmaz, E., Adali, G., … Enc,
resentation was designed (Figure 4) to summarize the modification F. Y. (2011). SIRT1 as a potential therapeutic target for treatment
of nonalcoholic fatty liver disease. Medical Science Monitor, 17(5),
of insulin resistance by BER‐chloride.
HY5‐9. https​://doi.org/10.12659/​MSM.881749
Cully, M., You, H., Levine, A. J., & Mak, T. W. (2006). Beyond PTEN mu‐
AC K N OW L E D G M E N T S tations: The PI3K pathway as an integrator of multiple inputs during
tumorigenesis. Nature Reviews Cancer, 6(3), 184–192. https​://doi.
The authors extend their appreciation to the deanship of Scientific org/10.1038/nrc1819
Research at King Khalid University, Abha, KSA for supported this David, J. A., Rifkin, W. J., Rabbani, P. S., & Ceradini, D. J. (2017). The Nrf2/
Keap1/ARE Pathway and Oxidative Stress as a Therapeutic Target in
work under grant number (R.G.P.1/117/40)
Type II Diabetes Mellitus. Journal of Diabetes Research, 2017, 1–15.
https​://doi.org/10.1155/2017/4826724
C O N FL I C T O F I N T E R E S T Deng, X. Q., Chen, L. L., & Li, N. X. (2007). The expression of
SIRT1 in nonalcoholic fatty liver disease induced by high‐fat
The authors declared that they have no conflict of interest. diet in rats. Liver International, 27(5), 708–715. https​://doi.
org/10.1111/j.1478-3231.2007.01497.x
Domingos, M. A. M., Queiroz, M., Lotufo, P. A., Bensenor, I. J., & Titan,
ORCID S. M. O. (2017). Serum RBP4 and CKD: Association with insulin re‐
sistance and lipids. Journal of Diabetes and Its Complications, 31(7),
Marwa El‐Zeftawy https://orcid.org/0000-0002-2884-8148 1132–1138. https​://doi.org/10.1016/j.jdiac​omp.2017.04.013
Dong, S. F., Hong, Y., Liu, M., Hao, Y. Z., Yu, H. S., Liu, Y., & Sun, J. N.
Doaa Ghareeb https://orcid.org/0000-0003-3327-4377
(2011). Berberine attenuates cardiac dysfunction in hyperglycemic
Nihal Elguindy https://orcid.org/0000-0002-5705-1349 and hypercholesterolemic rats. European Journal of Pharmacology,
660(2–3), 368–374. https​://doi.org/10.1016/j.ejphar.2011.03.024
Attalla F. El‐kott https://orcid.org/0000-0001-5060-0790
10 of 11 | EL‐ZEFTAWY et al.

Ellman, G. L. (1959). Tissue sulfhydryl groups. Archives of Physiology‐Endocrinology and Metabolism, 296(4), E812–819. https​://
Biochemistry and Biophysics, 82(1), 70–77. https​://doi. doi.org/10.1152/ajpen​do.90710.2008
org/10.1016/0003-9861(59)90090-6 Koc, K., Cerig, S., Ozek, N. S., Aysin, F., Yildirim, S., Cakmak, O., …
El‐Sayed, M. M., Ghareeb, D. A., Talat, H. A., & Sarhan, E. M. (2013). High Geyikoglu, F. (2019). The efficacy of oleuropein against non‐ste‐
fat diet induced insulin resistance and elevated retinol binding pro‐ roidal anti‐inflammatory drug induced toxicity in rat kidney.
tein 4 in female rats; treatment and protection with Berberis vulgaris Environmental Toxicology, 34(1), 67–72. https​://doi.org/10.1002/
extract and vitamin A. Pakistan Journal of Pharmaceutical Sciences, tox.22658​
26(6), 1189–1195. Kumar, A., Ekavali, Chopra, K., Mukherjee, M., Pottabathini, R., & Dhull,
Fiorentino, T. V., Marini, M. A., Succurro, E., Sciacqua, A., Andreozzi, F., D. K. (2015). Current knowledge and pharmacological profile of ber‐
Perticone, F., & Sesti, G. (2017). Elevated hemoglobin glycation index berine: An update. European Journal of Pharmacology, 761, 288–297.
identify non‐diabetic individuals at increased risk of kidney dysfunc‐ https​://doi.org/10.1016/j.ejphar.2015.05.068
tion. Oncotarget., 8(45), 79576–79586. https​://doi.org/10.18632/​ Lao‐ong, T., Chatuphonprasert, W., Nemoto, N., & Jarukamjorn, K.
oncot​arget.18572​ (2012). Alteration of hepatic glutathione peroxidase and superox‐
Flores, C., Adhami, N., & Martins‐Green, M. (2016). THS tox‐ ide dismutase expression in streptozotocin‐induced diabetic mice
ins induce hepatic steatosis by altering oxidative stress and by berberine. Pharmaceutical Biology, 50(8), 1007–1012. https​://doi.
SIRT1 levels. Journal of Clinical Toxicology, 6, 318. https​ ://doi. org/10.3109/13880​209.2012.655377
org/10.10.4172/2161-0495.1000318 Li, J.‐W., Yuan, K., Shang, S.‐C., & Guo, Y. (2017). A safer hypoglyce‐
Friedewald, W. T., Levy, R. I., & Fredrickson, D. S. (1972). Estimation of mic agent for type 2 diabetes—Berberine organic acid salt. Journal
the concentration of low density lipoprotein cholesterol in plasma of Functional Foods, 38, 399–408. https​ ://doi.org/10.1016/j.
without use of the preparative ultracentrifuge. Clinical Chemistry, jff.2017.09.031
18(6), 499–502. Li, S., Shin, H. J., Ding, E. L., & van Dam, R. M. (2009). Adiponectin levels
Furukawa, S., Fujita, T., Shimabukuro, M., Iwaki, M., Yamada, Y., Nakajima, and risk of type 2 diabetes: A systematic review and meta‐analysis.
Y., … Shimomura, I. (2004). Increased oxidative stress in obesity and JAMA, 302(2), 179–188. https​://doi.org/10.1001/jama.2009.976
its impact on metabolic syndrome. Journal of Clinical Investigation, Liang, F., Kume, S., & Koya, D. (2009). SIRT1 and insulin resistance. Nature
114(12), 1752–1761. https​://doi.org/10.1172/jci21625 Reviews Endocrinology, 5(7), 367–373. https​://doi.org/10.1038/
Gao, Y. F., Zhang, M. N., Wang, T. X., Wu, T. C., Ai, R. D., & Zhang, Z. nrendo.2009.101
S. (2016). Hypoglycemic effect of D‐chiro‐inositol in type 2 diabe‐ Lin, H., & Li, Z. (2012). Adiponectin self‐regulates its expression and
tes mellitus rats through the PI3K/Akt signaling pathway. Molecular multimerization in adipose tissue: An autocrine/paracrine mecha‐
and Cellular Endocrinology, 433, 26–34. https​ ://doi.org/10.1016/j. nism? Medical Hypotheses, 78(1), 75–78. https​://doi.org/10.1016/j.
mce.2016.05.013 mehy.2011.07.063
Ghareeb, D. A., Khalil, S., Hafez, H. S., Bajorath, J., Ahmed, H. E. A., Lin, Y., Sheng, M., Ding, Y., Zhang, N., Song, Y., Du, H., … Yu, W. (2018).
Sarhan, E., … El‐Demellawy, M. A. (2015). Berberine reduces neu‐ Berberine protects renal tubular cells against hypoxia/reoxygenation
rotoxicity related to nonalcoholic steatohepatitis in rats. Evidence‐ injury via the Sirt1/p53 pathway. Journal of Natural Medicines, 72(3),
Based Complementary and Alternative Medicine, 2015, 1–13. https​:// 715–723. https​://doi.org/10.1007/s11418-018-1210-1
doi.org/10.1155/2015/361847 Litwack, G., Bothwell, J. W., Williams, J. N. J., & Elvehjem, C. A. (1953).
Han, S. J., Boyko, E. J., Kim, S. K., Fujimoto, W. Y., Kahn, S. E., & A colorimetric assay for xanthine oxidase in rat liver homogenates.
Leonetti, D. L. (2018). Association of Thigh Muscle Mass with Insulin Journal of Biological Chemistry, 200(1), 303–310.
Resistance and Incident Type 2 Diabetes Mellitus in Japanese Liu, Q., Xu, X., Zhao, M., Wei, Z., Li, X., Zhang, X., … Shao, C. (2015).
Americans. Diabetes & Metabolism Journal, 42(6), 488–495. https​:// Berberine induces senescence of human glioblastoma cells by down‐
doi.org/10.4093/dmj.2018.0022 regulating the EGFR‐MEK‐ERK signaling pathway. Molecular Cancer
Hardie, D. G. (2015). AMPK: Positive and negative regulation, and its role Therapeutics, 14(2), 355–363. https​://doi.org/10.1158/1535-7163.
in whole‐body energy homeostasis. Current Opinion in Cell Biology, MCT-14-0634
33, 1–7. https​://doi.org/10.1016/j.ceb.2014.09.004 Liu, R., Li, H., Fan, W., Jin, Q., Chao, T., Wu, Y., & Yang, X. (2017). Leucine
Haslam, D. W., & James, W. P. (2005). Obesity. Lancet, 366(9492), 1197– supplementation differently modulates branched‐chain amino acid
1209. https​://doi.org/10.1016/S0140-6736(05)67483-1 catabolism, mitochondrial function and metabolic profiles at the dif‐
Hutchison, S. K., Harrison, C., Stepto, N., Meyer, C., & Teede, H. J. (2008). ferent stage of insulin resistance in rats on high‐fat diet. Nutrients,
Retinol‐binding protein 4 and insulin resistance in polycystic ovary 9(6), https​://doi.org/10.3390/nu906​0565
syndrome. Diabetes Care, 31(7), 1427–1432. https​://doi.org/10.2337/ Merksamer, P. I., Liu, Y., He, W., Hirschey, M. D., Chen, D., & Verdin, E.
dc07-2265 (2013). The sirtuins, oxidative stress and aging: An emerging link.
Jiang, G., & Zhang, B. B. (2002). Pi 3‐kinase and its up‐ and down‐stream Aging, 5(3), 144–150. https​://doi.org/10.18632/​aging.100544
modulators as potential targets for the treatment of type II diabetes. Paglia, D. E., & Valentine, W. N. (1967). Studies on the quantitative and
Frontiers in Bioscience, 7, d903–907. https​://doi.org/10.2741/jiang​ qualitative characterization of erythrocyte glutathione peroxidase.
Jiang, W., Li, S., & Li, X. (2015). Therapeutic potential of berberine against Journal of Laboratory and Clinical Medicine, 70(1), 158–169.
neurodegenerative diseases. Science China Life Sciences, 58(6), 564– Pandian, R. S., Anuradha, C. V., & Viswanathan, P. (2002).
569. https​://doi.org/10.1007/s11427-015-4829-0 Gastroprotective effect of fenugreek seeds (Trigonella foe‐
Kahn, S. E., Hull, R. L., & Utzschneider, K. M. (2006). Mechanisms linking num graecum) on experimental gastric ulcer in rats. Journal
obesity to insulin resistance and type 2 diabetes. Nature, 444(7121), of Ethnopharmacology, 81, 393–397. https​ ://doi.org/10.1016/
840–846. https​://doi.org/10.1038/natur​e 05482 S0378-8741(02)00117-4
Karbasforooshan, H., & Karimi, G. (2018). The role of SIRT1 in diabetic Peter, S. J., Basha, S. K., Giridharan, R., Lavinya, B. U., & Sabina, E. P.
retinopathy. Biomedicine & Pharmacotherapy, 97, 190–194. https​:// (2017). Suppressive effect of Spirulina fusiformis on diclofenac‐in‐
doi.org/10.1016/j.biopha.2017.10.075 duced hepato‐renal injury and gastrointestinal ulcer in Wistar al‐
Kim, W. S., Lee, Y. S., Cha, S. H., Jeong, H. W., Choe, S. S., Lee, M.‐R., … bino rats: A biochemical and histological approach. Biomedicine
Kim, J. B. (2009). Berberine improves lipid dysregulation in obesity by & Pharmacotherapy, 88, 11–18. https​
://doi.org/10.1016/j.
controlling central and peripheral AMPK activity. American Journal of biopha.2017.01.032
EL‐ZEFTAWY et al. | 11 of 11

Pirillo, A., & Catapano, A. L. (2015). Berberine, a plant alkaloid with lipid‐ Xia, X., Yan, J., Shen, Y., Tang, K., Yin, J., Zhang, Y., … Weng, J. (2011).
and glucose‐lowering properties: From in vitro evidence to clinical Berberine improves glucose metabolism in diabetic rats by inhibi‐
studies. Atherosclerosis, 243(2), 449–461. https​://doi.org/10.1016/j. tion of hepatic gluconeogenesis. PLoS ONE, 6(2), e16556. https​://doi.
ather​oscle​rosis.2015.09.032 org/10.1371/journ​al.pone.0016556
Qiao, L., & Shao, J. (2006). SIRT1 regulates adiponectin gene expression Yamauchi, T., Kamon, J., Minokoshi, Y., Ito, Y., Waki, H., Uchida, S., …
through Foxo1‐C/enhancer‐binding protein alpha transcriptional Kadowaki, T. (2002). Adiponectin stimulates glucose utilization and
complex. Journal of Biological Chemistry, 281(52), 39915–39924. fatty‐acid oxidation by activating AMP‐activated protein kinase.
https​://doi.org/10.1074/jbc.M6072​15200​ Nature Medicine, 8(11), 1288–1295. https​://doi.org/10.1038/nm788​
Ren, L., Zhou, X., Huang, X., Wang, C., & Li, Y. (2019). The IRS/PI3K/ Yang, T. C., Chao, H. F., Shi, L. S., Chang, T. C., Lin, H. C., & Chang, W.
Akt signaling pathway mediates olanzapine‐induced hepatic insu‐ L. (2014). Alkaloids from Coptis chinensis root promote glucose
lin resistance in male rats. Life Sciences, 217, 229–236. https​://doi. uptake in C2C12 myotubes. Fitoterapia, 93, 239–244. https​://doi.
org/10.1016/j.lfs.2018.12.015 org/10.1016/j.fitote.2014.01.008
Saleh, S. R., Attia, R., & Ghareeb, D. A. (2018). The ameliorating effect of Yao, X. H., & Nyomba, B. (2008). Hepatic insulin resistance induced
berberine‐rich fraction against gossypol‐induced testicular inflam‐ by prenatal alcohol exposure is associated with reduced PTEN
mation and oxidative stress. Oxid Med Cell Longev, 2018, 1056173. and TRB3 acetylation in adult rat offspring. American Journal of
https​://doi.org/10.1155/2018/1056173 Physiology‐Regulatory, Integrative and Comparative Physiology, 294(6),
Seko, Y., Sumida, Y., Tanaka, S., Mori, K., Taketani, H., Ishiba, H., … Itoh, Y. R1797–R1806. https​://doi.org/10.1152/ajpre​gu.00804.2007
(2018). Insulin resistance increases the risk of incident type 2 diabetes Yoshizaki, T., Milne, J. C., Imamura, T., Schenk, S., Sonoda, N., Babendure,
mellitus in patients with non‐alcoholic fatty liver disease. Hepatology J. L., … Olefsky, J. M. (2009). SIRT1 exerts anti‐inflammatory effects
Research, 48(3), E42–E51. https​://doi.org/10.1111/hepr.12925​ and improves insulin sensitivity in adipocytes. Molecular and Cellular
Steinberg, D., Vaughan, M., & Margolis, S. (1961). Studies of triglyceride Biology, 29(5), 1363–1374. https​://doi.org/10.1128/MCB.00705-08
biosynthesis in homogenates of adipose tissue. Journal of Biological Yousefi, M. R., & TaheriChadorneshin, H. (2018). The effect of moder‐
Chemistry, 236(6), 1631–1637. ate endurance training on gastrocnemius retinol‐binding protein
Sun, Y., Li, J., Xiao, N. A., Wang, M., Kou, J., Qi, L., … Liu, K. (2014). 4 and insulin resistance in streptozotocin‐induced diabetic rats.
Pharmacological activation of AMPK ameliorates perivascular ad‐ Interventional Medicine and Applied Science, 10(1), 59–63. https​://doi.
ipose/endothelial dysfunction in a manner interdependent on org/10.1556/1646.9.2017.34
AMPK and SIRT1. Pharmacological Research, 89, 19–28. https​://doi. Zhang, S., Wang, X., Yin, W., Liu, Z., Zhou, M. I., Xiao, D., … Peng, D. (2016).
org/10.1016/j.phrs.2014.07.006 Synthesis and hypoglycemic activity of 9‐O‐(lipophilic group substi‐
Tappel, A. L., & Zalkin, H. (1959). Inhibition of lipid peroxidation in mi‐ tuted) berberine derivatives. Bioorganic & Medicinal Chemistry Letters,
tochondria by vitamin E. Archives of Biochemistry and Biophysics, 80, 26(19), 4799–4803. https​://doi.org/10.1016/j.bmcl.2016.08.027
333–336. https​://doi.org/10.1016/0003-9861(59)90259-0 Zhang, W., Xu, Y. C., Guo, F. J., Meng, Y., & Li, M. L. (2008). Anti‐dia‐
Vuddanda, P. R., Chakraborty, S., & Singh, S. (2010). Berberine: A po‐ betic effects of cinnamaldehyde and berberine and their impacts
tential phytochemical with multispectrum therapeutic activities. on retinol‐binding protein 4 expression in rats with type 2 diabetes
Expert Opinion on Investigational Drugs, 19(10), 1297–1307. https​:// mellitus. Chinese Medical Journal, 121(21), 2124–2128. https​://doi.
doi.org/10.1517/13543​784.2010.517745 org/10.1097/00029​330-20081​1010-00003​
Wang, H., Zhu, C., Ying, Y., Luo, L., Huang, D., & Luo, Z. (2017). Metformin
and berberine, two versatile drugs in treatment of common metabolic
diseases. Oncotarget, 9(11), 10135–10146. https​://doi.org/10.18632/​
oncot​arget.20807​ S U P P O R T I N G I N FO R M AT I O N
Wang, Y., Yi, X., Ghanam, K., Zhang, S., Zhao, T., & Zhu, X. (2014). Berberine
decreases cholesterol levels in rats through multiple mechanisms, Additional supporting information may be found online in the
including inhibition of cholesterol absorption. Metabolism ‐ Clinical Supporting Information section at the end of the article.
and Experimental, 63(9), 1167–1177. https​://doi.org/10.1016/j.metab​
ol.2014.05.013
Wu, Q. M., Ni, H. X., & Lu, X. (2016). Changes of adipocytokine expres‐
sion after diabetic rats received sitagliptin and the molecular mecha‐ How to cite this article: El‐Zeftawy M, Ghareeb D, ElBealy
nism. Asian Pacific Journal of Tropical Medicine, 9(9), 893–897. https​://
ER, et al. Berberine chloride ameliorated PI3K/Akt‐p/SIRT‐1/
doi.org/10.1016/j.apjtm.2016.07.014
Wu, U., Cha, Y., Huang, X., Liu, J., Chen, Z., Wang, F., & Ding, H. (2015).
PTEN signaling pathway in insulin resistance syndrome
Protective effects of berberine on high fat‐induced kidney damage induced in rats. J Food Biochem. 2019;00:e13049. https​://doi.
by increasing serum adiponectin and promoting insulin sensitivity. org/10.1111/jfbc.13049​
International Journal of Clinical and Experimental Pathology, 8(11),
4486–4492.

You might also like