Naringin and PDX1 2019

You might also like

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

Stem Cells / Research Article

Cells Tissues Organs Accepted after revision: December 28, 2018


Published online: March 18, 2019
DOI: 10.1159/000496506

Naringin (4′,5,7-Trihydroxyflavanone 7-Rhamnoglucoside)


Attenuates β-Cell Dysfunction in Diabetic Rats through
Upregulation of PDX-1
Manickam Subramanian Balaji Thotakura Swathi Priyadarshini Chandra Sekaran
Ashok kumar Jyothi Indumathi Sundaramurthi
Department of Anatomy, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education,
Chennai, India

Keywords imals. These changes were more pronounced in the 8-week


Naringin · PDX-1 · Diabetes · Insulin naringin-treated diabetic animals. Conclusions: Naringin
was found to be an effective antidiabetic agent which in-
creased Insulin gene expression and insulin secretion by up-
Abstract regulating the PDX-1 gene and protein expression.
Background: Pancreatic duodenal homeobox-1 (PDX-1) is a © 2019 S. Karger AG, Basel
key transcription factor which regulates Insulin gene expres-
sion and insulin secretion in adult β-cells and helps to main-
tain β-cells mass. Naringin, a flavanone, owing to its anti­ Introduction
oxidant property, is reported to have antidiabetic effects.
Objectives: The present study tries to evaluate the role of Diabetes mellitus is a metabolic disease affecting 425
naringin on the β-cell-specific transcription factor PDX-1 in million people globally and around 73 million people in
diabetic rats. Methods: Diabetes was induced in male rats India in particular, placing it second among the countries
using streptozotocin and treated with naringin (100 mg/kg) affected with diabetes. Its prevalence in India is extrapo-
orally for 4 and 8 weeks. Serum insulin level, Pdx-1 and Insu- lated to double by 2045 to reach 134 million [Ogurtsova,
lin gene expression, and PDX-1 protein expression were as- 2017]. It is characterized by hyperglycemia, and the dis-
sessed in the rat pancreas. Histopathological and ultrastruc- ease process includes dysfunction of pancreatic β-cells,
tural changes in the islet and β-cells were observed. Results: reduction in β-cell mass, and impairment in insulin se-
Naringin prevented leukocytic infiltration in the pancreas of cretion irrespective of the type of diabetes [Bell and Po-
diabetic rats and recouped the β-cells with adequate secre- lonsky, 2001; Soleimanpour et al., 2015]. Maintenance of
tory granules. Naringin-treated diabetic rats showed signifi- β-cell mass and insulin secretion are regulated by multi-
cantly increased mRNA expression of Pdx-1 and Insulin ple factors. Pancreatic duodenal homeobox-1 (PDX-1) is
genes, increased expression of transcription factor PDX-1, one of the cardinal transcription factors which binds to
and higher serum insulin levels than the diabetic control an- the promoter region of the insulin gene and regulates its
129.127.145.240 - 3/20/2019 2:12:09 PM

© 2019 S. Karger AG, Basel Manickam Subramanian


Department of Anatomy, Chettinad Hospital and Research Institute
Chettinad Academy of Research and Education, Rajiv Gandhi Salai
E-Mail karger@karger.com
Kelambakkam, Tamil Nadu 603 103 (India)
www.karger.com/cto
Univ.of Adelaide
Downloaded by:

E-Mail gandhiayu @ hotmail.com


expression [Ahlgren et al., 1998]. PDX-1 expression is Materials and Methods
necessary for postnatal rodent β-cells to mature and be- Animals
come glucose-responsive cells [Guo et al., 2013] and for Male adult Wistar albino rats (250–300 g) were procured from
maintaining the adult β-cell mass [Holland et al., 2005]. the Tamil Nadu University of Veterinary and Animal Sciences
PDX-1 regulates the expression of Insulin, Somatostatin, (TANUVAS), Madhavaram (Chennai, India), and maintained at a
Glucokinase, Glucose transporter 2 (GLUT2), and Islet room temperature of 25 ± 1 ° C on a 12-h-light/12-h-dark cycle.
    

The animals were fed with rat/mouse pellet obtained from VRK
amyloid polypeptide (IAPP) genes specific to β-cells [Bell Nutritional Solutions, Maharashtra. The food and water were pro-
and Polonsky, 2001]. Autosomal dominant mutation of vided ad libitum. The study was approved by the Institutional An-
the Pdx-1 gene leads to diabetes in adults at a younger age imal Ethics Committee of the Chettinad Hospital and Research
called maturity onset diabetes of the young 4 (MODY4) Institute (Reg. No. 944/AC/06/CPCSEA, approval No. IAEC1/
Desp. No. 60 and IAEC1/Desp. No. 1 dated 09.06.2014 and
[Kushner et al., 2002].
25.01.2016, respectively).
There is an ever-growing need for newer drugs, which
could combat the disease and probably reduce its compli- Chemicals
cations, in addition to the currently available therapies Streptozotocin (STZ) and naringin (95% purity) were pur-
like insulin and oral antihyperglycemic agents. In this chased from Himedia Laboratories Pvt. Ltd., Mumbai, India (No.
RM6099). Primary rabbit polyclonal anti-PDX-1 antibody (No.
context, pharmacological intervention with natural prod-
ab47267; Abcam, USA) was used in the study. A REAL EnVision
ucts could be a better alternative and might be effective detection system, peroxidase/diaminobenzidine (DAB), rabbit/
and economical as well. mouse kit (Dako, Denmark) was used for primary antibody detec-
Naringin (4′,5,7-trihydroxyflavanone 7-rhamnoglu- tion and labeling. All other chemicals used in this study were of
coside) is a flavanone found in citrus fruits, grapefruits, analytical grade and obtained from Himedia.
beans, cherries, cocoa, oregano, and tomatoes [Bharti et
Induction of Diabetes
al., 2014]. Being a common constituent of dietary prod- Animals were subjected to overnight fasting prior to the induc-
ucts, humans are exposed to naringin in some form or the tion of diabetes mellitus, and their fasting blood glucose levels were
other. Previous in vitro and in vivo studies show naringin recorded. Animals were given a single dose of STZ (50 mg/kg body
has antidiabetic activity, and almost all studies attribute weight [BW] i.p., dissolved in 0.1 M cold citrate buffer, pH 4.5).
The control and nondiabetic group animals were given intraperi-
that effect to its antioxidant property and its ability to toneal injection of citrate buffer. Twenty percent glucose solution
counter oxidative stress [Meier et al., 2008; Pari and was given to STZ-injected animals for 1 day to prevent mortality
Suman, 2010; Liu et al., 2016; Nzuza et al., 2016; Ahmed due to hypoglycemia. Three days after STZ injection, animals were
et al., 2017; Lim et al., 2018]. Most of the studies show that once again tested for their fasting blood glucose level. Animals
naringin has a beneficial effect on diabetes through extra- with a fasting blood glucose level > 250 mg/dL were considered
diabetic and were chosen for the study.
pancreatic mechanisms like reduced glucose absorption
[Reshmi et al., 2017; Taslimi et al., 2017], regulation of Experimental Design
hepatic enzyme dysfunction [Punithavathi et al., 2008; A total of 84 rats were used for the study and were divided into
Sharma et al., 2011; Pu et al., 2012; Ahmed et al., 2017; the following 7 groups (n = 12 animals/group):
• I: normal control animals treated with vehicle alone
Pari and Chandramohan, 2017] and improvement in pe- • II: normal rats + naringin for 4 weeks
ripheral resistance [Priscilla et al., 2015; Ahmed et al., • III: normal rats + naringin for 8 weeks
2017]. Available literature shows that naringin could in- • IV: diabetic control treated with vehicle alone for 4 weeks
• V: diabetic control treated with vehicle alone for 8 weeks
crease insulin levels [Pari and Suman, 2010; Sharma et al., • VI: diabetic rats treated with naringin for 4 weeks
2011; Ahmed et al., 2012; Pu et al., 2012; Priscilla et al., • VII: diabetic rats treated with naringin for 8 weeks
2015; Lim et al., 2018] in diabetic animals, but the mech-
anism proposed for such an activity was rather a general Drug Administration
antioxidative, anti-inflammatory, and antiapoptotic Naringin (100 mg/kg BW) was mixed in distilled water as ve-
hicle (1 mL/kg BW) and was administered once daily orally be-
function. There are few data on the central effect of na­ tween 10 and 11 a.m. using an intragastric tube. Control and un-
ringin on the endocrine pancreas and mechanism by treated animals were given equal volumes of vehicle alone.
which it modulates insulin secretion.
The present study tries to explain the plausible role Sample Collection
At the end of the study, all the animals were sacrificed by an
of naringin on the β-cell-specific transcription factor
overdose of thiopentone sodium (75 mg/kg BW). The ventral ab-
PDX-1 in upregulating insulin gene expression and in dominal wall of the animals was cut open by midline incision, and
turn modulating insulin production in experimentally in- the pancreas was dissected out along with the spleen. Fat sur-
duced diabetes in rats. rounding the pancreas was carefully removed by dissection. Pan-
129.127.145.240 - 3/20/2019 2:12:09 PM

2 Cells Tissues Organs Subramanian/Thotakura/


DOI: 10.1159/000496506 Chandra Sekaran/Jyothi/Sundaramurthi
Univ.of Adelaide
Downloaded by:
Table 1. PCR primer sequences

Genes Forward primer Reverse primer

Gapdh (NM_017008.4) 5′-AGTTCAACGGCACAGTCAAG-3′ 5′-TACTCAGCACCAGCATCACC-3′


Pdx-1 (NM_022852.3) 5′-GGTATAGCCAGCGAGATGCT-3′ 5′-TCAGGTGGGAGCCTGATTCT-3′
Insulin I 5′-ACCTTTGTGGTCCTCACCTG-3′ 5′-AGCTCCAGTTGTGGCACTTG-3′

creatic tissues were stored in neutral buffered formalin for histo- homogenized using Tomy Microsmash tissue homogenizer
pathological and immunohistochemical studies and in glutaralde- (Tomy, Japan) in RIPA buffer. The homogenate was centrifuged
hyde for electron-microscopic studies. For Western blot analysis at 12,000 rpm for 15 min at 4 ° C, and the supernatant was collect-
    

and mRNA extraction, pancreatic tissues were stored at –80 ° C.


     ed and used for protein estimation and Western blot analysis. Pro-
For insulin assays, 2-mL venous blood samples were collected tein concentrations in the pancreatic tissue lysates of the different
by retro-orbital venous puncture after giving ketamine (22 mg/kg groups were determined using the previously published method of
BW i.p.). Blood was centrifuged at 4,000 rpm for 10 min, and se- Lowry et al. [1951] using bovine serum albumin as standard. The
rum separated and stored at –20 ° C.
     cell lysates were subjected to centrifugation at 14,000 rpm for 10
min at 4 ° C, and supernatant was separated. Protein extract (50 µg)
    

Serum Insulin was separated on 12% SDS-polyacrylamide gels and transferred


A rat insulin ELISA kit (RayBiotech, USA) was used to measure onto polyvinylidene difluoride membranes. Then, the membranes
fasting serum insulin levels; 100-µL serum samples were used ac- were incubated with primary antibody to PDX-1 (Abcam) at 1:
cording to the manufacturer’s instruction and read using a micro- 1,000 dilutions in Tris-buffered saline overnight at 4 ° C followed
    

plate reader at 450 nm. by detection using an enzymatically labeled HRP conjugated sec-
ondary antibody (Merck, India) at 1: 5,000 dilutions. Protein ex-
Histopathology pression was visualized by a chemiluminescent method using Che-
Pancreatic tissue fixed in 10% neutral buffered formalin was mi Doc XRS Imaging System (BioRad, USA). Band intensity was
processed and embedded in paraffin, and 4-µm-thick serial sec- quantified by ImageJ software.
tions were cut. The sections were stained with hematoxylin and
eosin (H&E). The sections were examined under an Axio Lab A1 Gene Expression Studies
microscope (Carl Zeiss, Germany), and photomicrographs were Total RNA was extracted from the rat pancreas using TRIzol
taken using an Axiocam ERc5s camera attached to the microscope. reagent (TakaraBio, USA) according to the manufacturer’s in-
structions. The RNA extracted was quantified using a UV spectro-
Immunohistochemistry photometer at 260–280 nm. Then, complementary deoxyribonu-
The 4-µm-thick pancreatic sections were placed on slides coat- cleic acid (cDNA) was obtained with 5 µL total RNA using Prime
ed with poly-L-lysine. Heat-induced antigen retrieval was done by script cDNA master mix (TakaraBio, USA) and using 1 µL of
boiling the sections in a pressure cooker using 10 mM citrate buffer cDNA, amplifications were done in a PCR system (Syngene, UK)
(pH 6) for 20 min. The endogenous enzymes were blocked by in- with different primers (Table 1). PCR products were separated us-
cubation with 3% hydrogen peroxide. The sections were incubated ing agarose gel electrophoresis and visualized using an ultraviolet
with primary antibody against PDX-1 (Abcam) diluted with Ven- gel documentation system. Band intensity was quantified using
tana antibody diluents (Roche Diagnostics, USA) at a ratio of 1:10 ImageJ software. Glyceraldehyde 3-phosphate dehydrogenase
for 45 min. The sections were incubated with secondary antibody (Gapdh) was used as an endogenous control, and the transcription
labeled with horseradish peroxidase (HRP) (Dako Envision, USA) of Pdx-1 and Insulin was compared in relation to Gapdh in the dif-
and with DAB for 30 and 10 min, respectively. Finally, the sections ferent groups. Each value represented the mean value of at least 3
were counterstained with Harris hematoxylin. independent experiments.

Transmission Electron Microscopy Morphometric Analysis


Pancreatic tissues were fixed in 2.5% glutaraldehyde and post- Ten random H&E- and PDX-1-immunostained sections were
fixed in 1% aqueous osmium tetraoxide and embedded in Araldite selected per animal in each group. From the selected sections, 5
6005 resin. Ultrathin sections of 60-nm thickness were cut using an random fields were studied. Totally, 50 fields per animal were ob-
ultramicrotome (Leica Ultracut UCT-GA-D/E-1/100) and stained served. The following parameters were measured:
with saturated aqueous uranyl acetate and counterstained with lead (A) The islet diameter was calculated from H&E-stained sec-
citrate. The sections were viewed under a transmission electron mi- tions at a magnification of ×100 using the following equation:
croscope (Hitachi H-700, Japan), and photographs were taken at
L+B
RUSKA Labs, College of Veterinary Science, P.V. Narsimha Rao Axial ratio (mm) =
Telangana Veterinary University, Hyderabad, India. 2
where L is maximum islet length and B is maximum islet breadth,
PDX-1Western Blot Analysis which is perpendicular to L.
The pancreatic tissues stored at –80 ° C were thawed and washed
    

with cold physiological saline. The tissue (100 µg) was completely
129.127.145.240 - 3/20/2019 2:12:09 PM

Naringin Upregulates PDX-1 in Cells Tissues Organs 3


Diabetic Rats DOI: 10.1159/000496506
Univ.of Adelaide
Downloaded by:
(B) The numerical density of the islets (H&E-stained section Table 2. Serum insulin levels of the different groups
at ×100 magnification) per unit volume of reticule with 121 inter-
sections was calculated using the following equation: Group Insulin, µU/mL
NA
NV (number of cells/mm 3 ) = I Control 18.26±0.33
A (D + T)
II Naringin 4 weeks 17.71±0.41
where NV is the numeric density or number of islets per cubic mil- III Naringin 8 weeks 18.58±0.37
limeter; NA the average number of islets counted within the reti- IV DM 4 weeks 9.81±0.39A
cule; A the area of the reticule in square millimeters; T the thick- V DM 8 weeks 8.56±0.41A
ness of the section in millimeters; and D the mean diameter of islets VI DM + naringin 4 weeks 13.30±0.42A, C
in millimeters. VII DM + naringin 8 weeks 15.21±0.35A, B
(C) The fractional volume of PDX-1-positive β-cells in PDX-
1-immunostained sections (×400 magnification), using a reticule Mean values were significantly different by t test following
with 121 intersections, was calculated using the following equation ANOVA: A p < 0.05 vs. normal controls (group I); B p < 0.05 vs.
(modified Delesse’s method): diabetes mellitus (DM) 8 weeks (group V); C p < 0.05 vs. DM 4
P weeks (group IV).
Vf = int
Ptot
where Vf is the volume fraction of PDX-1-positive cells; Pint the
number of intersections hitting the PDX-1-positive cells; and Ptot
the number of points hitting the whole section. of the islets, the regular contour and absence of other
Statistical Analysis pathological changes showed that the rats were recover-
All the data are expressed as the mean ± standard error of mean ing from the insult caused by STZ (Fig. 1f, 2f).
(SEM) and subjected to statistical analysis using one-way analysis Morphometric analysis showed significant decreases
of variance (ANOVA). The t test was performed to determine sig- in islet diameter and density in group IV and V diabetic
nificant differences between the individual groups using comput- animals compared to group I (Fig. 3a, b). This effect was
er-based software (SPSS, version 21; IBM, USA). p < 0.05 was con-
sidered statistically significant. reversed in the naringin-treated groups VI and VII. This
proves the proliferative effect of naringin on islet cells.
These changes were more conspicuous in group VII when
compared to the corresponding untreated group V ani-
Results mals. In normal rats administered naringin, these param-
eters were similar to the control group.
Fasting Serum Insulin
Groups I, II, and III had similar fasting insulin levels, Transmission Electron Microscopy
while the diabetic untreated groups (groups IV and V) β-Cells of control animals (group I) and naringin con-
presented significantly lower (p < 0.05) insulin levels than trol animals (groups II and III) had large secretory gran-
the control group. In group VI and VII animals, insulin ules in the cytoplasm, which were electron lucent with the
levels were significantly higher (13.30 and 15.21 µU/ characteristic halo around them. Numerous small vesi-
mL, respectively) compared to group IV and V animals cles were seen without secretory content (Fig. 4a).
(Table 2). Group IV showed damaged islets with absence of se-
cretory granules thereby making the β-cell inconspicu-
Histopathology ous. The nuclei of islets were pyknotic and distorted with
H&E-stained pancreatic sections of groups I, II, and III signs of degeneration (Fig. 4b). Group V animals exhib-
showed normal parenchyma and islets (Fig. 1a, b, 2a, b). ited similar pathology with swollen nuclei and indistinct
The diabetic groups IV and V exhibited shrunken and ir- organelles (Fig. 4c).
regularly shaped islets. A lobular pattern of the exocrine β-Cell nuclei of group VI animals had less heterochro-
part was observed due to extensive lymphocytic infiltra- matin and dilated nuclear pores. Cytoplasm exhibits
tion (Fig. 1c, d, 2c, d). agranular vesicles and distorted endoplasmic reticulum
Group VI animals had less pronounced pathological (Fig. 4d). In the 8-week naringin-treated animals (group
changes than the diabetic groups (Fig. 1e, 2e). The dia- VII), β-cells demonstrated increased number of charac-
betic rats treated with naringin for 8 weeks (group VII) teristic small secretory granules suggesting β-cell regen-
showed almost normal parenchyma and islets, and ab- eration (Fig. 4e).
sence of lymphocytic infiltration. Despite the smaller size
129.127.145.240 - 3/20/2019 2:12:09 PM

4 Cells Tissues Organs Subramanian/Thotakura/


DOI: 10.1159/000496506 Chandra Sekaran/Jyothi/Sundaramurthi
Univ.of Adelaide
Downloaded by:
Is Is Ac

Is
Ac Ac

Is
Is
Is
a b Is c

Is
Is

Ac
Is
Ac Ac
Is
d e f

Fig. 1. a–f H&E staining of rat pancreatic tissue. ×100. Scale bar, of lymphocytes damaging the islet. e Group VI exhibits shrunken
1 mm. Group I (a) and group III (b) show normal acinar and islet islets and lymphocytic infiltration but less pronounced than group
architecture. c Group IV shows shrunken islet and lymphocytic V. f Group VII demonstrates near normal islets. Arrows indicate
infiltration. d Group V shows shrunken and extensive infiltration lymphocytic infiltration. Is, Islets; Ac, Acini.

a b c

d e f

Fig. 2. a–f H &E staining of rat pancreatic tissue. ×400. Scale bar, e Group VI shows small islets with few lymphocytes. f Group VII
100 µm. Group I (a) and group III (b) reveal normal islets. c Group demonstrates near normal islets with very few lymphocytes. Ar-
IV shows islets with extensive lymphocytic infiltration. d Group V rowhead indicates lymphocytic infiltration and arrow indicates
demonstrates islets with fewer cells and lymphocytic infiltration. the islets.
129.127.145.240 - 3/20/2019 2:12:09 PM

Naringin Upregulates PDX-1 in Cells Tissues Organs 5


Diabetic Rats DOI: 10.1159/000496506
Univ.of Adelaide
Downloaded by:
14
120 30 $

Fractional volume of PDX1-


Islet density, n of cells/mm3
12 $
$ #
100 25
10
Islet diameter, µm

positive cells, %
* #
80 * 20
8
* *
60 15 6
40 10 4
* *
20 5 2
0 0 0
I II III IV V VI VII I II III IV V VI VII I II III IV V VI VII
a Groups b Groups c Groups

Fig. 3. Morphometric analysis of the rat pancreas. Graphs show the islet diameter (a), islet density (b), and frac-
tional volume of PDX-1-positive β-cells in the islets (c) of the different groups. Mean values were significantly
different by one-way ANOVA. * p < 0.05 vs. control; # p < 0.05 vs. group IV; $ p < 0.05 vs. group V (t test).

Immunohistochemistry PDX-1 Immunoblot


Groups I, II, and III showed PDX-1-immunopositive Western blot analysis of PDX-1 protein showed a
nuclei in the central portion of the islets suggesting the statistically significant difference between the groups
expression and localization of PDX-1 in the nuclei of (Fig. 7). Protein expression was significantly downregu-
β-cells (Fig. 5a, b). In contrast, group IV and V islets ex- lated in group IV and group V in comparison to group I.
hibited very mild reactivity to PDX-1 immunostaining When compared to group IV, group V animals (left un-
showing positivity in very few nuclei (Fig. 5c, d). Group treated) had very low expression of PDX-1 protein. PDX-1
VI islets expressed weak reactivity for PDX-1 in few nu- expression was significantly enhanced in the diabetic
clei (Fig.  5e). Group VII islets were bigger when com- group treated with naringin (group VI) when compared
pared to groups IV and V and displayed more PDX-1-im- to its corresponding untreated diabetic group IV (Fig. 7b).
munopositive cells (Fig. 5f). Moreover, group VII had an almost twofold increase in
The fractional volume of PDX-1-positive β-cells dras- PDX-1 expression when compared to group V, showing
tically reduced in the diabetic untreated groups IV and V. significant increase in the transcription factor which
The diabetic animals treated with naringin (group VI and could influence the upregulatory target genes like Insulin
VII) showed significantly higher PDX-1-positive cells (Fig.  6d). PDX-1 expression was also higher in normal
when compared with the corresponding untreated groups rats treated with naringin for 8 weeks (group III) than the
(groups IV and V) (Fig. 3c). controls.

Gene Expression
Figure 6a, b shows the effect of naringin administra- Discussion
tion on Pdx-1 gene expression. The animals exhibited low
Pdx-1 expression in diabetic groups (groups IV and V), STZ-induced diabetes is a well-accepted animal model
and the same pattern was found in Insulin mRNA expres- for experimental induction of diabetes causing selective
sion (Fig.  6c, d). However, mRNA expression of Pdx-1 destruction of β-cells. It also generates reactive oxygen
and Insulin was increased in the diabetic groups treated species, which accelerates β-cell destruction [Lenzen,
with naringin (groups VI and VII), and they were found 2008]. In the present study, we established the model of
to be significantly higher in group VII than the diabetic experimental diabetes in rats using STZ to investigate
untreated counterparts (group V). Naringin administra- molecular mechanisms through which naringin exerts a
tion to normal rats (groups II and III) did not alter the central effect on the endocrine pancreas.
expression of Pdx-1 and Insulin genes much when com- Untreated diabetic rats show leukocyte infiltration in
pared to control animals. pancreatic parenchyma, perivascular fibrosis, and de-
129.127.145.240 - 3/20/2019 2:12:09 PM

6 Cells Tissues Organs Subramanian/Thotakura/


DOI: 10.1159/000496506 Chandra Sekaran/Jyothi/Sundaramurthi
Univ.of Adelaide
Downloaded by:
ER N

N
N N N

2.5 µm b 25 µm 25 µm
a c

2.5 µm 2.5 µm
d e

Fig. 4. Electron micrographs of pancreatic β-cells of the different empty granules and distorted endoplasmic reticulum. e Group VII
groups (×7,720). a Normal β-cell with characteristic secretory β-cell with numerous small characteristic granules. N, nucleus; ER,
granules. b Group IV islet with indistinguishable cell types and endoplasmic reticulum; *, artifact. Arrows indicate secretory gran-
abnormally shaped nuclei. c Group V islet with indistinguishable ules with characteristic halo around them. Arrowheads indicate
cell types and noncharacteristic granules. d Group VI β-cell with agranular vesicles.

a b c

d e f

Fig. 5. Photomicrographs of pancreatic islets of various groups im- with few PDX-1-positive cells. e, f Group VI and VII islets of
munostained with PDX-1 antibody (×400, scale bar, 100 µm). naringin-treated diabetic rats with immunopositivity for PDX-1.
a, b Group I and III islets with nuclear localization of PDX-1 in Arrowheads show the immunopositive nuclei of the islets.
β-cells present in the center of the islet. c, d Group IV and V islets
129.127.145.240 - 3/20/2019 2:12:09 PM

Naringin Upregulates PDX-1 in Cells Tissues Organs 7


Diabetic Rats DOI: 10.1159/000496506
Univ.of Adelaide
Downloaded by:
Pdx-1
50 $

Relative mRNA expression,


40 *
* *
Groups *

% of GAPDH
30
I IV V VI VII II III

Pdx-1 20

10
Gapdh
0
I IV V VI VII II III
a b Groups

Insulin
70

Relative mRNA expression,


$
60
Groups

% of GAPDH
I IV V VI VII II III
50 *#
Insulin 40
*
30 *
Gapdh
20
I IV V VI VII II III
c d Groups

Fig. 6. Effect of naringin on mRNA expression of Pdx-1 and Insulin genes in rat pancreas. Representative gene
expression (a) and quantification of Pdx-1 gene (b) in the different groups. Representative gene expression (c)
and quantification of Insulin gene (d) in the different groups (n = 6). Mean values were significantly different by
one-way ANOVA: * p < 0.05 vs. control; # p < 0.05 vs. group IV; $ p < 0.05 vs. group V (t test).

struction of islets. Naringin treatment reversed these which made them indistinguishable from other islet cell
changes showing markedly reduced leukocyte infiltration types and devoid of secretory granules.
and reappearance of normal islet architecture (Fig. 1, 2), It is established that naringin reduces the β-cell oxida-
and increased islet diameter and density (Fig.  3a, b). tive stress and increases the insulin secretion in a dose-
These changes confirm the regeneration of the diabetic dependent manner in protease inhibitor-treated RIN5F
pancreas and islets in particular from the necrosis caused cell lines [Nzuza et al., 2016]. Similarly, in the present
by STZ. This strongly reaffirms the results of Lim et al. study, administration of naringin increased fasting serum
[2018] that naringin protects the pancreas against leuko- insulin levels in diabetic animals. Moreover, the secretory
cyte infiltration and proinflammatory cytokine produc- granules of the β-cells in diabetic rats increased notice-
tion. ably followed by increasing serum insulin levels with ad-
Ultrastructural changes in the β-cells of naringin- ministration of naringin. These changes confirm that
treated diabetic rats include the presence of characteristic there was a significant improvement in insulin synthesis
secretory granules and increased endoplasmic reticular and secretion in diabetic animals treated with naringin.
network. This indicates the β-cells have recovered from The increased fractional volume of PDX-1-positive
the STZ-created insult, and they are functionally active. cells in naringin-treated animals (Fig. 3) correlates well
This is in accordance with the results of Sharma et al. with the increase in their Pdx-1 transcript level and pro-
[2011], where administration of naringin (100 mg/kg) to tein level indicating that β-cells are regenerating follow-
type II diabetic rats reduced the endoplasmic reticular ing naringin treatment. This accounts for increased ex-
distension and maintained adequate secretory granules. pression of Insulin gene and insulin secretion. Thus, in
In contrast, the diabetic animals had necrosis of β-cells, diabetic rats, the administration of naringin increases the
129.127.145.240 - 3/20/2019 2:12:09 PM

8 Cells Tissues Organs Subramanian/Thotakura/


DOI: 10.1159/000496506 Chandra Sekaran/Jyothi/Sundaramurthi
Univ.of Adelaide
Downloaded by:
4 weeks
#
100
* *

Relative protein expression


compared to GAPDH
Groups 80
I IV VI II 60
PDX-1
40

20
GAPDH
0
I IV VI II
a b Groups

8 weeks
140
*$ *

Relative protein expression


120

compared to GAPDH
Fig. 7. Effect of naringin on PDX-1 protein
expression in rat pancreas. Representative Groups 100
Western blot (a) and quantification of I V VII III 80
PDX-1 (b) in pancreas of groups I, II, IV, *
PDX-1 60
and VI. Representative Western blot (c)
40
and quantification of PDX-1 (d) in pancre-
as of groups I, III, V, and VII (n = 6). Mean GAPDH 20
values were significantly different by one- 0
way ANOVA. * p < 0.05 vs. control; # p < I V VII III
0.05 vs. group IV; $ p < 0.05 vs. group V (t c d Groups
test).

expression of PDX-1, which sequentially influences the mal animals, and naringin-treated diabetic animals. Lo-
expression of the Insulin gene and in turn modulates in- calization of PDX-1 in β-cell nuclei might facilitate its ac-
sulin hormone secretion. tion on Insulin gene expression in diabetic rats.
Moreover, the results show that the protective effect of A previous study suggests that PDX-1 is a signaling
naringin against STZ-induced diabetes was more pro- target of insulin, and insulin causes nuclear localization
nounced in 8-week naringin-treated animals. This sug- of PDX-1 and decreases the apoptosis of islets through
gests the need for prolonged usage of this phytochemical mediation of PDX-1 expression both in vitro and in vivo
to abate the adverse changes involved in the diabetic [Johnson et al., 2006]. This elucidates an additional anti-
pathophysiology. apoptotic role exhibited by PDX-1.
In β-cells, glucose enters the cell through GLUT2 Peroxisome proliferator-activated receptor γ (PPARɣ)
transporters and a signaling pathway involving phos- plays a significant role in cellular proliferation and differ-
phoinositide 3-kinase (PI3K) which phosphorylates entiation, glucose homeostasis, and lipid metabolism. It
PDX-1. The phosphorylated PDX-1 is translocated from regulates the expression of PDX-1 in β-cells and thereby
the cytoplasm to the nucleus and binds the A element lo- improves β-cell function by regulating the downstream
cated in the 5′ flanking sequence of the Insulin gene pro- targets of PDX-1 [Jung et al., 2014]. Naringin upregulates
moter region and activates Insulin gene transcription the PPARɣ receptors in the liver and kidney of rats and in
[Vaulont et al., 2000]. Chronic glucotoxicity shuttles the adipose tissue of db/db mice, and decreases insulin resis-
PDX-1 from the nucleus to the cytoplasm and downregu- tance and proinflammatory cytokines [Jung et al., 2006;
lates Insulin gene transcription and insulin biosynthesis Sharma et al., 2011]. Similar regulation of PPARɣ expres-
[Kitamura et al., 2002]. Figure 5 shows the absence of sion in β-cells by naringin has to be ascertained to under-
PDX-1 immunoreactivity in the nucleus of diabetic islets stand the mechanism resulting in upregulation of PDX-1.
and the increased expression of PDX-1 and its nuclear Chronic hyperglycemia causes oxidative stress and de-
localization in the islets of control, naringin-treated nor- terioration of β-cells via c-Jun N-terminal kinase (JNK)
129.127.145.240 - 3/20/2019 2:12:09 PM

Naringin Upregulates PDX-1 in Cells Tissues Organs 9


Diabetic Rats DOI: 10.1159/000496506
Univ.of Adelaide
Downloaded by:
pathway activation. Translocation of PDX-1 to the cyto- Acknowledgments
plasm and its reduced binding to the insulin gene are
The authors thank the following individuals for their contribu-
linked to the activation of JNK. Further, oxidative stress tions to this article: S. Govindaraju for statistical support and R.
downregulates Akt expression which phosphorylates an- Vijayashree for pathological interpretation.
other important negative regulator of PDX-1, namely
Forkhead box protein O1 (FoxO1), leading to nucleocy-
toplasmic translocation of PDX-1 [Tabatabaie and Yaz- Statement of Ethics
danparast, 2017]. Naringin, a potent antioxidant flava-
none, may counteract the hyperglycemia-induced β-cell Animal experiments were conducted according to the guide-
oxidative stress. We hypothesize that naringin by virtue lines of the Committee for the Purpose of Control and Supervision
of Experiments on Animals (CPCSEA). The experimental proto-
of its antioxidant property might probably maintain the col was approved by the Institutional Animal Ethics Committee of
normal localization and increased expression of PDX-1 the Chettinad Hospital and Research Institute (Reg. No. 944/
through JNK kinase pathway suppression and downregu- AC/06/CPCSEA, approval No. IAEC1/Desp. No. 60 and IAEC1/
lation of FoxO1. Desp. No. 1 dated 09.06.2014 and 25.01.2016, respectively).
The results of the present study confirm that increased
expression of PDX-1 in diabetic rats treated with narin-
gin upregulates the insulin gene expression and insulin Disclosure Statement
secretion. This in turn can eventually result in a reduction
The authors have no conflict of interest to declare.
in hyperglycemia. Research on the effect of naringin on
the Pdx-1 gene and protein, the key controllers of insulin
secretion, has not been reported and hence the need for
Funding Sources
the present study.
In summary, the present study demonstrates naringin The study was supported by the Chettinad Academy of Re-
as an effective antidiabetic agent and reveals the mecha- search and Education.
nism by which naringin increases insulin secretion in an
experimental model of diabetes. Increased expression of
PDX-1 by naringin positively modulates the Insulin gene Author Contributions
and its expression resulting in increased insulin biosyn-
Study conception and design: Manickam Subramanian, Balaji
thesis and secretion. Further research is required to inves- Thotakura, and Indumathi Sundaramurthi; acquisition, analysis,
tigate the up- and downregulatory mechanisms by which and interpretation of data: Manickam Subramanian, Swathi Priya-
naringin regulates the transcription factor PDX-1 to es- darshini Chandra Sekaran, and Ashok Kumar Jyothi; drafting of
tablish its clinical value. the manuscript: Manickam Subramanian; critical revision: Balaji
Thotakura and Indumathi Sundaramurthi, and final approval of
the version to be published: Manickam Subramanian, Balaji
Thotakura, Indumathi Sundaramurthi, Swathi Priyadarshini
Chandra Sekaran, and Ashok Kumar Jyothi.

References
Ahlgren U, Jonsson J, Jonsson L, Simu K, Edlund Bell GI, Polonsky KS. Diabetes mellitus and ge- Holland AM, Góñez LJ, Naselli G, Macdonald
H. Beta-cell-specific inactivation of the mouse netically programmed defects in β-cell func- RJ, Harrison LC. Conditional expression
Ipf1/Pdx1 gene results in loss of the beta-cell tion. Nature. 2001 Dec;414(6865):788–91. demonstrates the role of the homeodo-
phenotype and maturity onset diabetes. Bharti S, Rani N, Krishnamurthy B, Arya DS. Pre- main transcription factor Pdx1 in mainte-
Genes Dev. 1998 Jun;12(12):1763–8. clinical evidence for the pharmacological ac- nance and regeneration of β-cells in the
Ahmed OM, Hassan MA, Abdel-Twab SM, Abdel tions of naringin: a review. Planta Med. 2014 adult pancreas. Diabetes. 2005 Sep; 54(9):
Azeem MN. Navel orange peel hydroethano- Apr;80(6):437–51. 2586–95.
lic extract, naringin and naringenin have anti- Guo L, Inada A, Aguayo-Mazzucato C, Hollister- Johnson JD, Bernal-Mizrachi E, Alejandro EU,
diabetic potentials in type 2 diabetic rats. Lock J, Fujitani Y, Weir GC, et al. PDX1 in Han Z, Kalynyak TB, Li H, et al. Insulin pro-
Biomed Pharmacother. 2017 Oct;94:197–205. ducts is not required for postnatal formation tects islets from apoptosis via Pdx1 and spe-
Ahmed OM, Mahmoud AM, Abdel-Moneim A, of β-cells but is necessary for their subsequent cific changes in the human islet proteome.
Ashour MB. Antidiabetic effects of hesperidin maturation. Diabetes. 2013 Oct;62(10):3459– Proc Natl Acad Sci USA. 2006 Dec; 103(51):
and naringin in type 2 diabetic rats. Diabetol 68. 19575–80.
Croat. 2012;41(2):53–67.
129.127.145.240 - 3/20/2019 2:12:09 PM

10 Cells Tissues Organs Subramanian/Thotakura/


DOI: 10.1159/000496506 Chandra Sekaran/Jyothi/Sundaramurthi
Univ.of Adelaide
Downloaded by:
Jung KY, Kim KM, Lim S. Therapeutic Approach- Meier JJ, Butler AE, Saisho Y, Monchamp T, Ga- Punithavathi VR, Anuthama R, Prince PS. Com-
es for Preserving or Restoring Pancreatic lasso R, Bhushan A, et al. Beta-cell replication bined treatment with naringin and vitamin C
β-Cell Function and Mass. Diabetes Metab J. is the primary mechanism subserving the ameliorates streptozotocin-induced diabetes
2014 Dec;38(6):426–36. postnatal expansion of beta-cell mass in hu- in male Wistar rats. J Appl Toxicol. 2008 Aug;
Jung UJ, Lee MK, Park YB, Kang MA, Choi MS. mans. Diabetes. 2008 Jun;57(6):1584–94. 28(6):806–13.
Effect of citrus flavonoids on lipid metabo- Nzuza S, Ndwandwe DE, Owira PM. Naringin Reshmi SK, Sudha ML, Shashirekha MN. Starch
lism and glucose-regulating enzyme mRNA protects against HIV-1 protease inhibitors- digestibility and predicted glycemic index in
levels in type-2 diabetic mice. Int J Biochem induced pancreatic β-cell dysfunction and the bread fortified with pomelo (Citrus max-
Cell Biol. 2006;38(7):1134–45. apoptosis. Mol Cell Endocrinol. 2016 Dec 5; ima) fruit segments. Food Chem. 2017 Dec;
Kitamura T, Nakae J, Kitamura Y, Kido Y, Biggs 437:1–10. 237:957–65.
WH 3rd, Wright CV, et al. The forkhead tran- Ogurtsova K, da Rocha Fernandes JD, Huang Y, Sharma AK, Bharti S, Ojha S, Bhatia J, Kumar N,
scription factor Foxo1 links insulin signaling Linnenkamp U, Guariguata L, et al. IDF Dia- Ray R, et al. Up-regulation of PPARγ, heat
to Pdx1 regulation of pancreatic β cell growth. betes Atlas. Brussels: International Diabetes shock protein-27 and -72 by naringin attenu-
J Clin Invest. 2002 Dec;110(12):1839–47. Federation (IDF); 2017. ates insulin resistance, β-cell dysfunction, he-
Kushner JA, Ye J, Schubert M, Burks DJ, Dow Pari L, Chandramohan R. Modulatory effects of patic steatosis and kidney damage in a rat
MA, Flint CL, et al. Pdx1 restores β cell func- naringin on hepatic key enzymes of carbohy- model of type 2 diabetes. Br J Nutr. 2011 Dec;
tion in Irs2 knockout mice. J Clin Invest. 2002 drate metabolism in high-fat diet/low-dose 106(11):1713–23.
May;109(9):1193–201. streptozotocin-induced diabetes in rats. Gen Soleimanpour SA, Ferrari AM, Raum JC, Groff
Lenzen S. The mechanisms of alloxan- and strep- Physiol Biophys. 2017 Jul;36(3):343–52. DN, Yang J, Kaufman BA, et al. Diabetes sus-
tozotocin-induced diabetes. Diabetologia. Pari L, Suman S. Antihyperglycemic and antilip- ceptibility genes Pdx1 and Clec16a function
2008 Feb;51(2):216–26. idperoxidative effects of flavanoid naringin in in a pathway regulating mitophagy in β-cells.
Lim YJ, Kim JH, Pan JH, Kim JK, Park TS, Kim streptozotocin-nicotinamide induced diabet- Diabetes. 2015 Oct;64(10):3475–84.
YJ, et al. Naringin Protects Pancreatic β-Cells ic rats. Int J Biol Med Res. 2010;1(4):206–10. Tabatabaie PS, Yazdanparast R. Teucrium polium
against Oxidative Stress-Induced Apoptosis Priscilla DH, Jayakumar M, Thirumurugan K. extract reverses symptoms of streptozotocin-
by Inhibiting Both Intrinsic and Extrinsic Flavanone naringenin: an effective antihyper- induced diabetes in rats via rebalancing the
Pathways in Insulin-Deficient Diabetic Mice. glycemic and antihyperlipidemic nutraceuti- Pdx1 and FoxO1 expressions. Biomed Phar-
Mol Nutr Food Res. 2018 Mar;62(5):1700810. cal agent on high fat diet fed streptozotocin macother. 2017 Sep;93:1033–39.
Liu X, Liu M, Mo Y, Peng H, Gong J, Li Z, et al. induced type 2 diabetic rats. J Funct Foods. Taslimi P, Caglayan C, Gulcin İ. The impact of
Naringin ameliorates cognitive deficits in 2015;14:363–73. some natural phenolic compounds on car-
streptozotocin-induced diabetic rats. Iran J Pu P, Gao DM, Mohamed S, Chen J, Zhang J, bonic anhydrase, acetylcholinesterase, butyr-
Basic Med Sci. 2016 Apr;19(4):417–22. Zhou XY, et al. Naringin ameliorates meta- ylcholinesterase, and α-glycosidase enzymes:
Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. bolic syndrome by activating AMP-activated an antidiabetic, anticholinergic, and anti­ -
Protein measurement with the Folin phenol protein kinase in mice fed a high-fat diet. epileptic study. J Biochem Mol Toxicol. 2017
reagent. J Biol Chem. 1951 Nov; 193(1): 265– Arch Biochem Biophys. 2012 Feb; 518(1):61– Dec;31(12):e21995.
75. 70. Vaulont S, Vasseur-Cognet M, Kahn A. Glucose
regulation of gene transcription. J Biol Chem.
2000 Oct;275(41):31555–8.

129.127.145.240 - 3/20/2019 2:12:09 PM

Naringin Upregulates PDX-1 in Cells Tissues Organs 11


Diabetic Rats DOI: 10.1159/000496506
Univ.of Adelaide
Downloaded by:

You might also like