ArticlePDF Available

Peroxiredoxin 6 Attenuates Alloxan-Induced Type 1 Diabetes Mellitus in Mice and Cytokine-Induced Cytotoxicity in RIN-m5F Beta Cells

Wiley
Journal of Diabetes Research
Authors:

Abstract and Figures

Type 1 diabetes is associated with the destruction of pancreatic beta cells, which is mediated via an autoimmune mechanism and consequent inflammatory processes. In this article, we describe a beneficial effect of peroxiredoxin 6 (PRDX6) in a type 1 diabetes mouse model. The main idea of this study was based on the well-known data that oxidative stress plays an important role in pathogenesis of diabetes and its associated complications. We hypothesised that PRDX6, which is well known for its various biological functions, including antioxidant activity, may provide an antidiabetic effect. It was shown that PRDX6 prevented hyperglycemia, lowered the mortality rate, restored the plasma cytokine profile, reversed the splenic cell apoptosis, and reduced the β cell destruction in Langerhans islets in mice with a severe form of alloxan-induced diabetes. In addition, PRDX6 protected rat insulinoma RIN-m5F β cells, cultured with TNF-α and IL-1β, against the cytokine-induced cytotoxicity and reduced the apoptotic cell death and production of ROS. Signal transduction studies showed that PRDX6 prevented the activation of NF-κB and c-Jun N-terminal kinase signaling cascades in RIN-m5F β cells cultured with cytokines. In conclusion, there is a prospect for therapeutic application of PRDX6 to delay or even prevent β cell apoptosis in type 1 diabetes.
This content is subject to copyright. Terms and conditions apply.
Research Article
Peroxiredoxin 6 Attenuates Alloxan-Induced Type 1 Diabetes
Mellitus in Mice and Cytokine-Induced Cytotoxicity in RIN-m5F
Beta Cells
Elena G. Novoselova , Olga V. Glushkova, Sergey M. Lunin, Maxim O. Khrenov,
Svetlana B. Parfenyuk, Tatyana V. Novoselova, Mars G. Sharapov, Vladimir I. Novoselov,
and Evgeny E. Fesenko
Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS, Institutskaya Str. 3, 142290 Pushchino,
Moscow Region, Russia
Correspondence should be addressed to Elena G. Novoselova; elenanov_06@mail.ru
Received 21 January 2020; Revised 1 May 2020; Accepted 17 August 2020; Published 27 August 2020
Academic Editor: Fabrizio Barbetti
Copyright © 2020 Elena G. Novoselova et al. This is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is
properly cited.
Type 1 diabetes is associated with the destruction of pancreatic beta cells, which is mediated via an autoimmune mechanism and
consequent inammatory processes. In this article, we describe a benecial eect of peroxiredoxin 6 (PRDX6) in a type 1 diabetes
mouse model. The main idea of this study was based on the well-known data that oxidative stress plays an important role in
pathogenesis of diabetes and its associated complications. We hypothesised that PRDX6, which is well known for its various
biological functions, including antioxidant activity, may provide an antidiabetic eect. It was shown that PRDX6 prevented
hyperglycemia, lowered the mortality rate, restored the plasma cytokine prole, reversed the splenic cell apoptosis, and reduced the
βcell destruction in Langerhans islets in mice with a severe form of alloxan-induced diabetes. In addition, PRDX6 protected rat
insulinoma RIN-m5F βcells, cultured with TNF-αand IL-1β, against the cytokine-induced cytotoxicity and reduced the apoptotic
cell death and production of ROS. Signal transduction studies showed that PRDX6 prevented the activation of NF-κB and c-Jun
N-terminal kinase signaling cascades in RIN-m5F βcells cultured with cytokines. In conclusion, there is a prospect for therapeutic
application of PRDX6 to delay or even prevent βcell apoptosis in type 1 diabetes.
1. Introduction
Insulin-dependent diabetes mellitus, or type 1 diabetes
(T1D), is a multifactorial disease, in which autoimmune fac-
tors play a key role. Clinical symptoms of T1D manifest
themselves when most of insulin-producing pancreatic beta
cells have already died because of the activation of autoreac-
tive T lymphocytes. The massive death of insulin-producing
βcells, which is caused by cytotoxic T lymphocytes migrating
into the pancreas, leads to the accumulation of glucose in the
blood, and patients with T1D need regular administration of
insulin for the rest of their lives. Despite the administration
of insulin, T1D causes serious inammatory complications
in many systems and organs, including the cardiovascular
system [1], kidneys [2], and eyes [3]. It is known that in dia-
betes, large blood vessels are especially severely damaged;
therefore, mortality from stroke and heart attack is three
times higher among patients with diabetes than in the rest
of the population.
Currently, most of the studies on the pathogenesis of
T1D mellitus focus on pancreatic βcells, which are targets
of the autoimmune attack. Meanwhile, the disease is usually
caused by oxidative stress and imbalances in the immune sys-
tem, which are related to autoaggressive clones of T lympho-
cytes [4, 5]. During autoimmune inammatory reactions,
proinammatory cytokines, including interleukin- (IL-) 1β,
tumor necrosis factor-alpha (TNF-α), and interferon- (IFN-
)γ, are released into the environment of βcells by activated
T cells and macrophages, causing βcell dysfunction and
death [6, 7]. Usually, a proinammatory response protects
Hindawi
Journal of Diabetes Research
Volume 2020, Article ID 7523892, 11 pages
https://doi.org/10.1155/2020/7523892
the mammalian organism from foreign pathogens and main-
tains the integrity of tissues and cellular systems. However, a
defective proinammatory response may cause the opposite
eect, increasing the risk of autoimmune pathologies, which
include T1D [6].
It is known that human T1D is sometimes linked with
altered genes providing susceptibility to diabetes [8]. How-
ever, studies on identical twins with familial diabetes showed
that only approximately half of them develop diabetes [9],
conrming an important role of environmental factors, such
as dietary factors during infancy, vaccination, and others
[10], in the risk of development of T1D [11]. T1D susceptibil-
ity involves a complex interplay between genetic and envi-
ronmental factors and has historically been attributed to
adaptive immunity, although there is now increasing support
for a role of innate inammation [12].
Oxidative stress has been proven to play a key role in the
pathogenesis of diabetes and related complications [13], and
there is evidence that antioxidants, mainly low-molecular-
weight natural and synthetic substances, may be useful for
the treatment of various pathologies associated with diabetes
mellitus [14, 15]. Meanwhile, there are many reasons to
believe that antioxidant enzymes can be more eective in
neutralising reactive oxygen species (ROS) than low-
molecular-weight antioxidants. Previously, we have shown
the therapeutic eects of a recombinant peroxiredoxin 6
(PRDX6) in various pathologies associated with inamma-
tion and oxidative stress, such as intestinal hypoxia/reperfu-
sion [16]. We believe that PRDX6 may be eective as an
agent that suppresses the level of oxidative stress in diabetes
mellitus. Indeed, it was shown that pancreatic βcells contain
lower levels of antioxidant enzymes, such as SOD, catalase,
and GPX, than do other mammalian tissues [17]. Therefore,
these cells are more sensitive to the damaging eects of ROS.
Because of this deciency of endogenous antioxidant
enzymes in βcells, there is an increasing interest in the use
of external proteins with antioxidant activities to protect
pancreatic βcells during diabetes.
Increased superoxide production in the development
and progression of diabetes causes the activation of several
signal pathways involved in the pathogenesis of chronic
complications. Oxidative stress activates cellular signaling
pathways and transcription factors, including protein
kinase C (PKC), c-Jun-N-terminal kinase (JNK), p38
mitogen-activated protein kinase (MAPK), and nuclear
factor kappa-B (NF-κB) [18]. Recently, we have shown
that signal transduction systems of immune cells are
involved in the development of diabetes in animals, with
a special role played by the nuclear factor kappa B (NF-
κB) cascade [19]. Thus, we showed that the use of an
inhibitor of the NF-κB signaling pathway, as well as the
thymic hormone thymulin, and a diet with antioxidants
signicantly reduced the immune imbalance in cells of
mice with alloxan-induced diabetes [20].
In the present study, the ecacy of PRDX6 for reducing
the damaging eects of alloxan-induced diabetes in mice
was studied for the rst time. Furthermore, taking into
account the key role of the pancreatic βcell loss in the devel-
opment of diabetes mellitus, we studied the eects of PRDX6
on the viability and functional activity of the RIN-m5F βcell
line under conditions that simulate diabetes.
2. Materials and Methods
2.1. Animals, Diabetes Model, and Peroxiredoxin 6
Treatment. Six- to eight-week-old male BALB/c mice (22
25 g) were maintained under standard laboratory conditions
(2021
°
C, 1014 h light/dark cycle, and 65% humidity), with
food and water provided ad libitum. Standard food pellets
contained a balanced diet of proteins, vitamins, and minerals
according to the Code of Practice for the Housing and Care
of Animals Used in Scientic Procedures [21]. Experimental
procedures were approved by the Institutional Ethical Com-
mittee (approval #57, 30/12/2011), and the experiments were
performed in accordance with the Guidelines for Ethical
Conduct in the Care and Use of Animals. Mice were sacri-
ced using cervical dislocation and decapitated using a small
animal guillotine with a sharp blade.
Severe diabetes was modelled using a single intraperito-
neal injection of 500 mg/kg alloxan in 0.2mL of physiological
saline, and physiological saline was injected into the control
mice. Experiments were performed 10 days after the injec-
tion, when the blood glucose was consistently greater than
18 mM, indicating that the mice were diabetic. The mortality
rate of the parallel groups of diabetic mice was observed for
32 days. Blood glucose concentration was measured using a
glucometer (Accu-Chek Performa Nano, Germany) and Test
Strips (Accu-Chek Performa Solo, Germany). A drop of
blood was obtained from the tip of the tail of fasting mice.
PRDX6 (20 mg/kg body weight in 0.1 mL saline) was
applied intravenously directly before the onset of diabetes
on the rst day and repeatedly on the eighth day during dia-
betes development. Control mice received intravenously
0.1 mL of physiological saline. Previously, we found that after
intravenous administration, PRDX6 retained the highest
level in the blood for 10 minutes; then, its amount gradually
decreased, but after 6 hours, about 30% of the administered
PRDX6 remained in blood plasma [22]. So, it was proved that
possible PRDX6 eects were caused by its presence in the
animal tissues.
2.2. Isolation and Purication of the PRDX6. Genetic con-
structions encoding (human) PRDX6 enzymes were obtained
and expressed earlier in E. coli BL21(DE3) cells [22]. Recom-
binant proteins harbored His-tag, so the enzymes were puri-
ed by anity chromatography on Ni-NTA-agarose
(Thermo Fisher Scientic, USA), according to the manufac-
turers recommendations. The technique of protein isolation
was described earlier [23]. According to electrophoresis in
12% SDS-PAAG, the purity of the obtained enzymes was at
least 98%. PRDX6 in phosphate buer (1.7 mM KH
2
PO
4
,
5.2 mM Na
2
HPO
4
, and 150 mM NaCl, pH 7.4) at a concen-
tration of 10 mg/mL were stored at 20
°
C. No reduction of
enzymatic activity was observed following 2 months of
storage.
The ability of the PRDX6 to reduce hydrogen peroxide
(H
2
O
2
) and tert-butyl-hydroperoxide (t-BOOH) was deter-
mined by Kang et al.s method [24], with minor
2 Journal of Diabetes Research
modications. Peroxidase activity of recombinant PRDX6
was 230 nmol/min/mg of protein (measured with H
2
O
2
)
and 100 nmol/min/mg of protein (measured with t-BOOH).
2.3. Blood Plasma. Plasma was isolated from the blood col-
lected during decapitation of the mice. Blood samples were
kept for 35 h at 4
°
Сand then centrifuged at 200 × g; the
supernatants were collected for cytokine assays. Splenic lym-
phocytes were isolated in Dulbeccos modied Eagles
medium (DMEM; Sigma, USA) containing 10 mM 4-(2-
hydroxyethyl)-1-piperazineethanesulphonic acid solution,
100 μg/mL streptomycin, and 10% fetal bovine serum. Eryth-
rocytes were lysed in Tris-buered ammonium chloride
(0.01 M Tris-HCl, with 0.15 M NaCl, and 0.83% NH
4
Cl at
9 : 1, pH 7.2). After being washed, the samples were stored
at a concentration of 1×10
8cells/mL in RPMI 1640 medium
at 20
°
C.
2.4. Cytokine Measurements. Enzyme-linked immunosor-
bent assays (ELISAs) were used to determine concentrations
of cytokines in blood plasma using ELISA development kits
for mouse TNF-α, IL-5, IL-17, and IFN-γ(PeproTech,
USA). Binding was visualised by adding 100 μL of the 2,2
-azino-bis(3-ethylbenzthiazoline-6-sulphonic acid) green
dye (Sigma), dissolved in 0.05 M citrate buer (pH 4.0) with
0.01% hydrogen peroxide. Absorbance was measured at
405 nm using a Multiscan EX spectro photometer (Thermo
Electron Corporation, Vantaa, Finland).
2.5. Western Blotting Analysis. To prepare specimens, 1×10
8
splenic cells were lysed using a lysis buer containing 50 mM
Tris-HCl (pH 7.4), 150 mM NaCl, 1% Triton X-100 , and
5 mM ethylenediaminetetraacetic acid (Alfa Aesar, UK).
The total protein concentration was determined using Brad-
fords method [25] (Sigma) following protein precipitation
with acetone in an ice bath for 15 min. Next, proteins were
diluted 1 : 1 with a solution containing 65.8 mM Tris-HCl,
pH 6.8, 2.1% sodium dodecyl sulphate, 26.3% (w/v) glycerol,
and 0.01% bromophenol blue, boiled for 5 min, and stored at
4
°
C. Proteins were resolved by 10% polyacrylamide gel elec-
trophoresis using a protein MW marker (Thermo Scientic,
USA) and then transferred onto a nitrocellulose membrane
(GE Healthcare, Amersham, UK) in a transblot chamber.
After being blocked with 5% (w/v) nonfat dry milk in Tris-
buered saline/Tween 20, the membranes were exposed for
2 h to the following antibodies raised against mouse proteins:
a phospho-NF-κB p65 (Ser 536) antibody (#3031, Cell Sig-
naling Technology, Danvers, MA, USA), rabbit phosphoin-
hibitor of NF-κB kinase (IKKα/β) antibody II (Ser 176/180)
(Cell Signaling Technology), rabbit phospho-stress-
activated protein kinase/c-Jun N-terminal kinase
(SAPK/JNK) antibody to synthetic phospho-SAPK/JNK pep-
tide (Cell Signaling Technology), and rabbit caspase-3 mono-
clonal antibody (Cell Signaling Technology). After being
washed, the membranes we re incubated for 1 h with an
anti-rabbit biotinylated antibody (Jackson ImmunoResearch,
West Grove, PA, USA), followed by incubation with
peroxidase-conjugated streptavidin for 1 h. As a loading con-
trol, glyceraldehyde 3-phosphate dehydrogenase (GAPDH)
was used, which was detected with a rabbit monoclonal anti-
body raised against a synthetic peptide corresponding to res-
idues near the C-terminus of human GAPDH (Cell Signaling
Technology). The ECL Plus chemiluminescent cocktail
(Amersham/GE) was used to develop the blots according to
the manufacturers instructions. The developed blots were
photographed using a TFX-35.WL transilluminator (Vilber
Lourmat, France). Protein bands were quantied densitome-
trically using Image Studio Software, version 5.2 (LI-COR,
NE, USA). Two-three independent experiments were per-
formed for each protein using cells from dierent passages
or splenocytes from individual animals. The obtained data
were normalised to the corresponding loading control
(GAPDH) and expressed in relative units.
2.6. Histology and Immunohistochemistry. The pancreases
from dierent groups were xed in 10% formalin, then
embedded in paran, and 3 μm sections were prepared and
stained with haematoxylin and eosin (H&E) for histopatho-
logical examination. For the detect ion of insulin, the 3 μm
sections were dewaxed, rehydrated, and incubated with a
peroxidase-blocking reagent (DAKO Cytomation, Fort Col-
lins, CO, USA) to block endogenous peroxidase. Next, the
slides were incubated with phosphate-buered saline
(PBS)+1% bovine serum albumin to block nonspecic bind-
ing. A rabbit anti-mouse insulin monoclonal antibody (Santa
Cruz Biotechnology, Santa Cruz, CA, USA) was subsequently
applied to the sections, followed by incubation with the
LSABsystem-HRP (DAKO Cytomation). The slides were
stained with diaminobenzidine according to the manufac-
turers instructions (DAKO Cytomation).
2.7. Quantication of βCell Mass. Images of the sections were
analysed using the ImageJ software (National Institutes of
Health, USA, http://rsb.info.nih.gov/ij/) to measure insulin-
positive areas and the total pancreas area. The βcell mass
(mg) per pancreas was calculated by multiplying the relative
insulin-positive area (the percentage of insulin-positive area
over total pancreas area) by the pancreas weight as was
reported earlier [26].
2.8. Culture of RIN-m5F Cells. Rat insulinoma RIN-m5F cells
(Vertebrate Cell Collection, St. Petersburg, Russia) were
grown in culture asks in a medium, with a low glucose con-
tent (8.0 mM), consisting of a mixture of RPMI 1640 med-
ium/DMEM (1 : 1), supplemented with 10% fetal calf serum
(FCS), 100 μg/mL penicillin, 100 μg/mL streptomycin, and
50 μg/mL gentamicin, at 37
°
C and 5% CO
2
. Cells were only
used between passages 3 and 7 and were cultured for 24 h
and washed. To induce apoptosis, 30 ng/mL TNF-α(Recom-
binant Murine TNF-α, PeproTech)+15 ng/mL IL-1β
(Recombinant Murine IL-1β, PeproTech) were added.
Recombinant PRDX6 (150 μg/mL) was added 30 min before
the addition of the cytokines. Cells were cultured for 24 h
and washed before measuring the viability and signal pro-
teins. In each independent experiment performed using sep-
arate passage, the measurements were provided for 912
replicates. The average values from four independent exper-
iments were used to determine the signicance of dierences
3Journal of Diabetes Research
between groups. Cells incubated without the cytokines and
PRDX6 were used as controls.
2.9. Measurement of ROS Using Carboxy-2,7
-Dichlorodihydrouorescein Diacetate (H
2
DCFDA). RIN-
m5F cells were cultured for 24 h in a 96-well plate
(2:5·×10
4cells per well in 100 μL) in DMEM and then
washed with PBS. The cells were then incubated with car-
boxy-H
2
DCFDA (Invitrogen, USA; freshly prepared in ster-
ile DMSO) at a nal concentration of 2.5 μM in the
medium supplemented with 2% depleted FCS in the darkness
for 1 h; then, PRDX6 and the cytokines were added, and the
cells were incubated for another 1 h. Cells incubated in the
absence of PRDX6 and cytokines were used as a control.
The uorescence was measured at an excitation of 485 nm
and an emission of 535 nm using an Innite 200 plate reader
(Tecan, Austria), as described earlier [27].
2.10. Statistical Analysis. Statistical analysis was performed
using the Statistica/Win 6.0 software (Tulsa, OK, USA).
One-way analysis of variance, followed by Tukeys post hoc
test, was performed to determine the signicance of dier-
ences among groups. Values of p0:05 were considered
signicant.
3. Results
3.1. Eects of PRDX6 on Mortality Rate and Plasma Glucose
Levels in Diabetic Mice. Three groups of mice were used
(alloxan-treated, alloxan plus PRDX6-treated, and untreated
age-matched controls), and each group consisted of 10 mice.
It was revealed that the mortality rate following administra-
tion of a high dose of alloxan (500 mg/kg) achieved about
80% on the 32nd day after alloxan treatment (Figure 1(a)).
The mortality rate of diabetic mice pretreated with PRDX6
was markedly lower compared to diabetic mice that did not
receive PRDX6.
Another three mouse groups consisting of 10 mice per
group (alloxan-treated, alloxan plus PRDX6-treated, and
untreated age-matched controls) were observed for blood
glucose. Blood glucose levels were measured for 9 days after
the administration of alloxan (Figure 1(b)). It was demon-
strated that on day 5 after the administration of alloxan, the
average glucose level in the blood of the alloxan-treated mice
exceeded 20 mM, while that in the mice treated with PRDX6
decreased to almost the control level (Figure 1(b)). Thus, the
administration of PRDX6 inhibited the glycemia raise in
mice with alloxan-induced diabetes. So, pathophysiological
manifestations in mice treated with alloxan were signicantly
lowered by PRDX6 application.
3.2. Eects of PRDX6 on Spleen Cell Apoptosis in Diabetes
Mice. The level of apoptosis in splenocytes was assessed
based on the ratio of activated/nonactivated caspase-3. A
very sharp increase in the level of splenocytic apoptosis was
observed in alloxan-induced diabetes (Figure 2). However,
administration of PRDX6 to the diabetic mice completely
normalised the ratio of activated/nonactivated caspase-3 in
splenocytes, indicating a protective eect of PRDX6 in devel-
oped diabetes.
3.3. Eects of PRDX6 on Cytokine Levels in the Blood of
Diabetic Mice. A study of the cytokine response demon-
strated that in the mice with developed alloxan-induced dia-
betes, the concentrations of all measured cytokines in the
plasma increased on the 10th day after the administration
of alloxan, with the most marked increase observed in
TNF-αand IL-5. The concentrations of IFN-γand IL-17 also
increased, although these changes were relatively small
(Figure 3). The administration of PRDX6 reduced the peaks
of TNF-αand IL-5 in the blood of diabetic mice. It is impor-
tant to emphasise that the use of PRDX6 had no eect on the
level of IL-17 in the blood of diabetic mice.
3.4. Eects of PRDX6 on the Pancreatic Islet Structure, Insulin
Expression, and βCell Mass in Diabetic Mice. To elucidate
PRDX6 eects on the pancreas in diabetic mice, immuno-
staining for insulin was performed. Immunohistochemical
examination of the pancreas revealed a reduction in the islet
density in diabetic mice, and the residual βcells were severely
disorganised (Figure 4). PRDX6 injections substantially
restored the islet density in diabetic mice. Using sections
from the control and diabetic pancreases, we quantitatively
evaluated the βcell mass. As shown in Figure 4, the number
of βcells in the diabetic pancreas was signicantly smaller
than that in the control pancreas. Treatment with PRDX6
markedly increased the βcell numbers, thereby protecting
the islet structure. This nding was consistent with the eects
of PRDX6 on the cytokine prole and cell apoptosis. The data
demonstrated the expected destruction of pancreatic βcells
in advanced diabetes and indicated that PRDX6 markedly
increased the βcell mass, thus supporting a protective func-
tion of the antioxidant enzyme. Moreover, immunostaining
conrmed the protective eect of PRDX6 on the immunity
of mice with alloxan-induced diabetes.
3.5. Eects of Cytokines and PRDX6 on RIN-m5F βCells.
RIN-m5F βcells were cultured under adverse conditions
(in the presence of proinammatory cytokines), and the via-
bility of these cells was studied with or without the PRDX6
addition. To elucidate the molecular mechanisms of the pro-
tective eects of PRDX6, the level of apoptosis and the activ-
ity of the NF-κB and SAPK/JNK signaling cascades were
determined in RIN-m5F βcells. In addition, the PRDX6 anti-
oxidant activity was tested using a carboxy-H
2
DCFDA
probe.
The level of apoptosis in RIN-m5F βcells was assessed by
measuring the ratio of activated to nonactivated form of
caspase-3 (Figure 5). It was demonstrated that the addition
of the mixture of cytokines (TNF-αand IL-1β) to the cell cul-
ture medium led to a signicant increase in the level of apo-
ptosis. At the same time, the presence of PRDX6 in the
medium completely eliminated the toxic eect of the cyto-
kines, reducing the ratio of the activated to nonactivated
form of caspase-3.
The activity of the NF-κB signaling cascade was assessed
based on the level of RelA/p65 protein phosphorylation at the
Ser 536 residue (Figure 5) and IKKα/βactivation. It was
shown that in the cells incubated with the proinammatory
cytokines, the phosphorylation of the RelA/p65 protein at
4 Journal of Diabetes Research
Ser 536 increased more than twofold. It is important to note
that the addition of PRDX6 under these conditions had
almost no eect on the phosphorylation of RelA/p65. On
the other hand, incubation of RIN-m5F cells with the cyto-
kines led to increased phosphorylation of IKKα/β. At the
same time, PRDX6 completely alleviated the cytokine-
induced IKKα/βactivation. Thus, PRDX6 decreased the acti-
vation of the canonical NF-κB pathway in RIN-m5F βcells
under conditions of oxidative stress caused by proinamma-
tory cytokines.
An even more profound protective eect of PRDX6 was
revealed by studying the JNK activity in RIN-m5F cells incu-
bated with the proinammatory cytokines. Indeed, the addi-
tion of the cytokines to the RIN-m5F cell culture medium led
to the activation of the JNK signaling cascade, while the addi-
tion of PRDX6 completely blocked the cytokine-induced
JNK activation in βcells.
Furthermore, the antioxidant eciency of PRDX6 was
also tested in vitro using RIN-m5F cells. As expected, the
addition of the proinammatory cytokine mixture to the cul-
tural medium produced a sharp increase in the ROS content
in these cells. The results also showed that the addition of
PRDX6 signicantly reduced the level of ROS in RIN-m5F
cells cultured in the presence of TNF-αand IL-1β(Figure 6).
Thus, we obtained evidence that the protective eects of
PRDX6 in T1D are related to its antioxidant activity. Inter-
estingly, using another model of oxidative stress in vivo,we
showed that the preliminary PRDX6 treatment reduced the
level of malonic aldehyde in the ischemia-reperfusion kidney
[22].
4. Discussion
Reduced antioxidant activity and increased oxidative stress
are among the intrinsic characteristics of T1D mellitus, both
in patients and in animal diabetes models. T1D is provoked
by the destruction of pancreatic βcells, which is mediated
via an autoimmune mechanism and consequent inamma-
tory processes. Numerous inammatory cytokines and
ROS, which are produced during development of diabetes,
have been proposed to play an important role in βcell
destruction. For example, ROS can penetrate through the cell
membrane and cause damage to βcells of the pancreas [28].
Possible causes of T1D are genetic or involve chemical,
1 4 7 10 14 18 21 25 32
120
100
80
60
40
20
0
Control
PRDX6 + diabetes
Diabetes
Surviving mice (percent to control)
Days aer alloxan treatment
####
(a)
0
5
10
15
20
25
3579
PRDX6 + diabetes
Diabetes
Days aer alloxan treatment
Blood glucose (mM)
#
#
#
Control
(b)
Figure 1: Eects of PRDX6 on the mortality rate of mice and on blood glucose in diabetic animals. (a) Time course of mortality in dierent
groups of mice (mean ± standard deviation ðSDÞof 10 mice). (b) Time course of changes in plasma glucose in dierent groups of mice
(mean ± standard deviation ðSDÞof 10 mice). p<0:05 vs. control;
#
p<0:05 vs. the diabetic group.
0.8
0.7
0.6
0.5
0.4
0.3
0.2
0.1
0
Cleaved caspase-3/caspase-3
ratio
Caspase-3
caspase-3
Cleaved
Control Diabetes PRDX6+
diabetes
GAPDH
Figure 2: Expression of caspase-3 and cleaved caspase-3,
determined using western blot analysis. Representative blots from
three independent experiments are shown. Histograms show the
protein levels normalised to those of GAPDH, used as a loading
control, and to total forms of relative proteins and represent the
average densitometry values for the blots from two experiments
(mean ± SD). p<0:05 vs. control.
5Journal of Diabetes Research
immune, or virus-associated damage to insulin-producing β
cells. Regardless of the cause, a common pathway may exist
that leads to the destruction of βcells.
Animal models play an important role in the develop-
ment of present concepts concerning T1D pathogenesis and
therapeutic approaches. In the present study, we modelled a
severe and rapidly developing form of T1D using a large dose
of alloxan. Indeed, worse blood glucose control in patients
with diabetes is generally associated with a more rapidly pro-
gressing disease and a wider range of complications. Using
0.3
0.25
0.2
0.15
0.1
0.05
0
Concentration (ng/ml)Concentration (ng/ml)
Concentration (ng/ml)Concentration (ng/ml)
IL-5
0.4
0.3
0.2
0.1
0
0.6
0.5
0.4
0.3
0.2
0.1
0
Control Diabetes PRDX6+
diabetes
Control Diabetes PRDX6+
diabetes
IL-17 IFN-𝛾
#
1
0.8
0.6
0.4
0.2
0
TNF-𝛼
#
Figure 3: Eects of PRDX6 on plasma cytokine concentrations in diabetic mice. Each value is the mean ± SD for three mice; six
measurements were performed for each individual mouse. p<0:05 vs. control;
#
p<0:05 vs. the diabetic group.
1.5
H&E
Ins
Control Diabetes PRDX6+
diabetes
Beta cell mass (mg/pancreas)
1
0.5
0
#
100 𝜇m
100 𝜇m
100 𝜇m
100 𝜇m
100 𝜇m
100 𝜇m
Figure 4: Eects of PRDX6 on the pancreas structure and βcell mass in diabetic mice. Representative images show islet histology using H&E
staining and islet immunostaining for insulin in the pancreas of control and diabetic mice. The βcell mass is shown as the mean ± SD for 15
sections/pancreas from three individual mice. p<0:05 vs. control;
#
p<0:05 vs. the diabetic group.
6 Journal of Diabetes Research
this model, we demonstrated the benecial eect of PRDX6
on the T1D pathology in terms of the plasma glucose level,
plasma cytokine prole, and splenic cell apoptosis. Moreover,
using histological and immunohistochemical assays, we stud-
ied the pancreatic islet structure and insulin expression in β
cells after treatment of diabetic mice with PRDX6.
In addition, to elucidate the molecular mechanisms of
PRDX6 protective activity, rat insulinoma RIN-m5F сells
were cultured with cytokines (TNF-αand IL-1β) and used
as an in vitro diabetic model to measure the ROS secretion,
βcell apoptosis, and the activity of the NF-κB and JNK sig-
naling pathways.
Among proinammatory plasma cytokines, TNF-α, IFN-
γ, IL-5, and IL-17 were shown to increase in alloxan-induced
diabetic mice, which may indicate the activation of T helper 1
(Th1) cells, producing TNF-αand IFN-γ; Th2 cells, produc-
ing IL-5; and Th17 cells, secreting IL-17 [29]. The treatment
with PRDX6 reduced the plasma TNF-αand IL-5 levels but
showed no signicant eects on the plasma concentrations
of IFN-γand IL-17 in diabetic mice. Apparently, this can
be explained by the fact that the use of various drugs to
reduce the pathological eects of diabetes can be more or less
eective, depending on the stage and severity of the disease.
Indeed, it has earlier been demonstrated that antidiabetic
approaches are more eective in prediabetic mice than in
mice with advanced diabetes [20]. In addition, IFN-γis
known to inhibit Th17 cells, and the role of Th17 cells in
T1D remains largely unknown [30].
More clear evidence of the protective activity of PRDX6
was obtained by the measurement of the blood glucose level,
as well as by the assessment of the level of apoptosis of sple-
nocytes. Indeed, administration of a large dose of alloxan
caused a sharp increase in the blood glucose level in diabetic
mice within 5 days after the administration, and glucose
remained at a high level throughout the experiment. In the
mice treated with PRDX6, a slight increase in plasma glucose
was observed on day 5, but by the end of the experiment, the
plasma glucose level was fully normalised. These observa-
tions are in agreement with the data of our recent study
showing the protective eects of PRDX6 on RIN-m5F сells
cultured with high glucose concentrations [31]. It should be
emphasised that according to our previous study, PRDX6
itself did not cause changes in either the activity of the NF-
κB and JNK signaling cascades or the level of apoptosis of β
cells, estimated by the ratio of activated to nonactivated form
of caspase-3 [31]. Thus, preliminary results indicate the non-
toxicity of the PRDX6 recombinant protein.
It is generally known that the pathogenic eect of hyper-
glycemia is mediated, to a signicant extent, via increased
production of ROS. Therefore, it seems quite possible that
PRDX6 may actually reduce the oxidative stress in diabetic
mice, as was shown in the present in vitro study using RIN-
5mF cells. In addition, alloxan-induced diabetes is a rapidly
developing severe form of diabetes, which may be more
dependent on oxidative stress than other forms. Consistent
with our results, another study demonstrated that acute ele-
vation of glucose resulted in a more specic triggering eect
on oxidative stress than did chronic sustained hyperglycemia
[32].
0
200
400
600
800
1000
1200
1400
Relative units
0
400
200
600
800
1000
1200
1400
1600
Relative units
0
200
100
300
400
500
600
800
700
900
Relative units
0
0.2
0.1
0.3
0.4
0.5
0.6
0.8
Control Cytokines PRDX6+
cytokines
0.7
Caspase/cleaved caspase
ratio
GAPDH
Caspase-3
Cleaved
caspase-3
phRelA
phIKK
phJNK
Figure 5: Eects of PRDX6 on the activation of caspase-3, NF-κB,
IKK, and JNK pathways in RIN-m5F βcells cultured with
proinammatory cytokines. Blot images are shown from a single
representative experiment. Histograms show the protein levels (in
relative units) normalised to those of GAPDH, used as a loading
control, and to total forms of relative proteins, representing the
average densitometry values for blots from two-three experiments
with cells from dierent passages (mean ± SD). p<0:05 vs. control.
7Journal of Diabetes Research
It is commonly known that βcell death in T1D involves
necrosis and apoptosis [33]. One of the immunocytochemi-
cal markers for apoptosis is cleaved caspase-3. The caspase-
3 protein is a member of the cysteine-aspartic acid protease
(caspase) family and plays a central role in the execution
phase of cell apoptosis [34]. The ubiquitously distributed
caspase-3 is the main eector of the apoptotic cascade within
cells and is activated through cleavage [35].
In the present study, we demonstrated that both βcells
in vitro and splenic cells in vivo underwent apoptosis in situ-
ations modelling T1D, and the treatment with PRDX6 sub-
stantially reduced the diabetogenic apoptosis, thus
indicating a protective eect of PRDX6. In addition, the loss
of the βcell mass, observed in mice with alloxan-induced dia-
betes and mediated by the activation of proapoptotic signal-
ing events, is increasingly recognised as the causal and
committed stage in the development of T1D mellitus. Thus,
our data suggest that this stage may also be alleviated by
PRDX6 administration.
The recombinant PRDX6 can aect the level of ROS in
animals, inhibiting the development of oxidative stress and
normalising the redox status of cells. However, the question
arises: how does recombinant PRDX6 located in the extracel-
lular space neutralise ROS in cells into which it does not pen-
etrate? It is known that hydroperoxides, in addition to
passive diusion through the cell membrane, are actively
transported to the intercellular space using aquaporins [36].
Probably, being in the extracellular space, recombinant
PRDX6 can participate in the elimination of peroxides
formed in the intercellular space and released from the cells
by the aquaporins.
Earlier, it was shown that PRDX6 can aect the level of
NF-κB via the TLR4/NF-κB signaling pathway [37]. The
authors showed that during ischemic damage of the brain,
PRDX6 released during the destruction of cells can act as
an endogenous ligand for the TLR4 receptor. The interaction
of PRDX6 with TLR4 triggers a cascade of processes in which
NF-κB plays a major role, resulting in an emergency cell
repair and suppression of apoptosis [38]. It is possible that
intravenous application of the recombinant PRDX6 before
exposure to alloxan can lead to a preconditioning eect,
inducing the mechanisms of repair and antioxidant response
through stimulation of the TLR4/NF-κB signaling pathway
in the cells. Therefore, exposure to alloxan does not lead to
a synergistic increase in NF-κB expression.
In addition to the peroxidase activity of PRDX6, a Ca
2+
-
dependent phospholipase A2 activity (aiPLA2) of PRDX6
was also shown, which normally manifests itself only under
acidic conditions and plays an important role in the metabo-
lism of phospholipids and the transmission of intracellular/-
intercellular signals [39]. Interestingly, with an excess of
ROS, peroxidase cysteine center of PRDX6 is oxidized, which
leads to a signicant increase in the Ca
2+
-dependent phos-
pholipase A2 activity (aiPLA2) [39]. In addition, regardless
of the oxidation of the peroxidase center, an increase in the
phospholipase activity (more than 10 times) of PRDX6 is
observed after specic phosphorylation of the Thr177 residue
by mitogen-activated protein kinases (MAPKs) (ERK2, p38γ,
and p38δ) [40]. Accordingly, with the induction of the
aiPLA2-activity of PRDX6, there is an increase in the level
of lysophospholipids and fatty acids, which serve as second-
ary messengers both in normal and in pathologies. It has
been shown that the phospholipase activity of PRDX6 stimu-
lates signaling pathways (p38, PI3K/Akt) and also promotes
the formation of arachidonic acid, which, in turn, aects
the activity of Src (SFK) kinases, stimulating cell growth
and division [41].
We have shown earlier that in slowly developing diabetes,
induced by a small dose of alloxan, the most signicant acti-
vation of several signaling cascades, including the interferon
regulatory factor 3, Toll-like receptor 4, and NF-κB path-
ways, as well as an increase in the expression of heat shock
protein 70 in splenic cells, was observed only at the prediabe-
tes stage but not in advanced diabetes [20]. However, the JNK
pathway was an exception in this regard, being activated
more signicantly at the stage of advanced diabetes. In the
present study, we demonstrated that the JNK pathway was
profoundly activated in βcells by proinammatory cytokines
and oxidative stress. This does not contradict the ndings
that demonstrated the role of both NF-κB and JNK signaling
220
200
160
180
120
140
80
100
0 20 40 60 80 100 120 140
Fluorescence intensity (percent to control)
Time (min)
Cytokines
Control
PRDX6
+cytokines
⁎#
⁎⁎
#####
#
Figure 6: Eect of PRDX6 on the ROS levels in RIN-m5F cells in the presence of proinammatory cytokines. Cells were incubated for 1 h in
the presence of the uorescent dye carboxy-H
2
DCFDA, with or without cytokines (TNF-α+IL-1β) and PRDX6 (150 μg/mL). Each value
presents the mean green uorescence intensity from 912 repeats (as a percentage of control). p<0:01 vs. control;
#
p<0:05 vs. the
cytokine treatment group.
8 Journal of Diabetes Research
in rat βcell death that is induced by proinammatory cyto-
kines [42]. Interestingly, PRDX6 downregulated the JNK
activity in βcells exposed to TNF-αand IL-1β, suggesting
that oxidative stress and subsequent activation of the JNK
pathway could be involved in the pathogenesis of T1D. This
study investigated whether the NF-κB and JNK pathways are
involved in the protective eect of exogenous PRX6 against β
cell destruction induced by alloxan treatment in mice or by
proinammatory cytokines in RIN-m5F cells. We demon-
strated that the decrease in the ROS levels in RIN-m5F cells
was accompanied by improvement of activity of caspase-3,
IKK, and JNK pathways in RIN-m5F βcells cultured with
proinammatory cytokines. In addition, PRDX6 adminis-
trated to diabetic mice partially improved the plasma cyto-
kine prole and tended to restore the pancreas structure
and βcell mass in diabetic mice.
The importance of the NF-κB pathway was demonstrated
in both T1D and T2D, due to its role in inammatory
responses [43]. We have previously shown involvement of
NF-κB, namely, RelA/p65, in the protection of pancreatic β
cells in alloxan-induced diabetic mice, using an inhibitor of
this cascade (IKK inhibitor XII) [20]. It was demonstrated
that knockdown of PRDX6 increased susceptibility of RIN-
m5F cells to the deleterious eects of proinammatory cyto-
kines and to oxidative stress [44]. These results show that
among the PRDXs signicantly expressed in RIN-m5F cells,
only PRDX6 is modulated by the proinammatory cytokines,
and the PRDX6 downregulation depends on the calpain, pro-
teasome systems, and JNK signaling [44]. Moreover, the link
between PRX6 and NF-κB, which is one of the most promi-
nent redox-regulated proinammatory regulators, has been
previously observed in hypoxic mouse hippocampal cells
[45]. Similarly, PRDX6 expression is inversely correlated
with NF-κB during Clonorchis sinensis infection [46]. Inter-
estingly, NF-κB activation is crucial for maturation and acti-
vation of immune cells, but in βcells, it has mostly a
deleterious eect [47]. So, understanding the innate inam-
mation and mechanisms by which βcell susceptibility to pro-
inammatory cytokines is potentiated or mitigated oers
important insight into T1D progression and avenues for
therapeutic intervention [12].
In conclusion, PRDX6 treatment reduced cell apoptosis
in both in vitro and in vivo models of T1D. Thus, PRDX6
protected RIN-5mF cells against cytokine-induced cytotoxic-
ity in vitro, and it also prevented T1D, normalised blood glu-
cose, lowered mortality rate, and restored the pancreatic islet
structure in vivo. Moreover, PRDX6 normalised the activity
of the NF-κB and JNK pathways in βcells cultured with pro-
inammatory cytokines and protected these cells from ROS.
Therefore, there is a prospect for therapeutic application of
PRDX6 to alleviate or even prevent βcell apoptosis in T1D.
5. Conclusion
The study demonstrates that administration of recombinant,
exogenous PRDX6 reduces impact of the alloxan toxicity on
pancreatic βcells in mice, improving animal mortality and
lowering the serum glucose level. The benecial eects of
recombinant PRDX6 treatment are associated with reduced
levels of inammatory cytokines in vivo. Meanwhile, in the
rat insulinoma cell line, the recombinant PRDX6 attenuated
eect of exogenous proinammatory cytokines on the ROS
production and activation of NF-κB and JNK signaling path-
ways. In summary, the manuscript provides novel observa-
tions, which have clear implications in understanding the
cytotoxic mechanism underlying death of βcells in DM1
and in forming potential therapeutic approaches.
Data Availability
The data used to support the ndings of this study are
included within the article.
Disclosure
The funding sponsors had no role in the design of the study;
in the collection, analyses, or interpretation of data; in the
writing of the manuscript; and in the decision to publish
the results.
Conflicts of Interest
The authors report no conict of interest.
Acknowledgments
The work was supported by the Russian Foundation for Basic
Research, project Nos. 18-04-00091 and 20-015-00216, and
by the Program of Russian Academy of Sciences 1.18 Molec-
ular and Cellular Biology and Post-Genomic Technologies.
The authors thank Editage for help with language editing.
References
[1] D. V. Cokkinos and C. Pantos, Le diabète de type 1 diminue la
réponse compensatrice après un infarctus du myocarde. Rôle
de lhypothyroïdie tissulaire et eets de ladministration dhor-
mones thyroïdiennes,Bulletin de l'Académie Nationale de
Médecine, vol. 195, no. 1, pp. 151165, 2011.
[2] A. S. Perlman, J. M. Chevalier, P. Wilkinson et al., Serum
inammatory and immune mediators are elevated in early
stage diabetic nephropathy,Annals of Clinical and Laboratory
Science, vol. 45, no. 3, pp. 256263, 2015.
[3] S. Wang, S. Du, Q. Wu, J. Hu, and T. Li, Decorin prevents ret-
inal pigment epithelial barrier breakdown under diabetic con-
ditions by suppressing p38 MAPK activation,Investigative
Ophthalmology & Visual Science, vol. 56, no. 5, pp. 2971
2979, 2015.
[4] C. P. Domingueti, L. M. Dusse, M. D. Carvalho, L. P. de Sousa,
K. B. Gomes, and A. P. Fernandes, Diabetes mellitus: the link-
age between oxidative stress, inammation, hypercoagulability
and vascular complications,Journal of Diabetes and its Com-
plications, vol. 30, no. 4, pp. 738745, 2016.
[5] K. Stadler, Oxidative stress in diabetes,Advances in Experi-
mental Medicine and Biology, vol. 771, pp. 272287, 2013.
[6] D. L. Eizirik, M. L. Colli, and F. Ortis, The role of inamma-
tion in insulitis and beta-cell loss in type 1 diabetes,Nature
Reviews. Endocrinology, vol. 5, no. 4, pp. 219226, 2009.
9Journal of Diabetes Research
[7] J. W. Yoon and H. S. Jun, Autoimmune destruction of pan-
creatic beta cells,American journal of therapeutics, vol. 12,
no. 6, pp. 580591, 2005.
[8] R. Bergholdt, J. Nerup, and F. Pociot, Fine mapping of a
region on chromosome 21q21.11-q22.3 showing linkage to
type 1 diabetes,Journal of medical genetics, vol. 42, no. 1,
pp. 1725, 2005.
[9] D. D. Adams, J. G. Knight, P. White et al., A solution to the
genetic and environmental puzzles of insulin-dependent dia-
betes mellitus,Lancet, vol. 323, no. 8374, pp. 420424, 1984.
[10] K. S. Rønningen, Environmental trigger(s) of type 1 diabetes:
why so dicult to identify?,BioMed research international,
vol. 2015, Article ID 321656, 10 pages, 2015.
[11] M. Rewers and J. Ludvigsson, Environmental risk factors for
type 1 diabetes,Lancet, vol. 387, no. 10035, pp. 23402348,
2016.
[12] S. M. Cabrera, A. M. Henschel, and M. J. Hessner, Innate
inammation in type 1 diabetes,Translational Research,
vol. 167, no. 1, pp. 214227, 2016.
[13] P. Manna and P. C. Sil, Arjunolic acid: benecial role in type 1
diabetes and its associated organ pathophysiology,Free Rad-
ical Research, vol. 46, no. 7, pp. 815830, 2012.
[14] W. Wojnar, M. Zych, and I. Kaczmarczyk-Sedlak, Antioxida-
tive eect of avonoid naringenin in the lenses of type 1 dia-
betic rats,Biomedicine & Pharmacotherapy, vol. 108,
pp. 974984, 2018.
[15] M. E. Czerwińska, E. Gąsińska, A. Leśniak et al., Inhibitory
eect of Ligustrum vulgare leaf extract on the development of
neuropathic pain in a streptozotocin-induced rat model of dia-
betes,Phytomedicine, vol. 49, pp. 7582, 2018.
[16] A. E. Gordeeva, A. A. Temnov, A. A. Charnagalov, M. G. Shar-
apov, E. E. Fesenko, and V. I. Novoselov, Protective eect of
peroxiredoxin 6 in ischemia/reperfusion-induced damage of
small intestine,Digestive Diseases and Sciences, vol. 60,
no. 12, pp. 36103619, 2015.
[17] H. Kaneto, Y. Kajimoto, J. Miyagawa et al., Benecial eects
of antioxidants in diabetes - possible protection of pancreatic
beta-cells against glucose toxicity,Diabetes, vol. 48, no. 12,
pp. 23982406, 1999.
[18] B. Feng, M. A. Ruiz, and S. Chakrabarti, Oxidative-stress-
induced epigenetic changes in chronic diabetic complica-
tions,Canadian Journal of Physiology and Pharmacology,
vol. 91, no. 3, pp. 213220, 2013.
[19] E. G. Novoselova, M. O. Khrenov, S. B. Parfenyuk, T. V. Novo-
selova, S. M. Lunin, and E. E. Fesenko, The NF-κB, IRF3, and
SAPK/JNK signaling cascades of animal immune cells and
their role in the progress of type 1 diabetes mellitus,Doklady
Biological Sciences, vol. 457, no. 1, pp. 255257, 2014.
[20] E. G. Novoselova, O. V. Glushkova, S. M. Lunin et al., Signal-
ing, stress response and apoptosis in pre-diabetes and diabetes:
restoring immune balance in mice with alloxan-induced type 1
diabetes mellitus,International Immunopharmacology,
vol. 31, pp. 2431, 2016.
[21] Home oce, Animals (scientic procedures) . Act 1986. Code
of practice for the housing and care of animals used in scien-
tic procedures.
[22] R. G. Goncharov, K. A. Rogov, A. A. Temnov, V. I. Novoselov,
and M. G. Sharapov, Protective role of exogenous recombi-
nant peroxiredoxin 6 under ischemia-reperfusion injury of
kidney,Cell and Tissue Research, vol. 378, no. 2, pp. 319
332, 2019.
[23] M. G. Sharapov, V. I. Novoselov, E. E. Fesenko, V. I. Bruskov,
and S. V. Gudkov, The role of peroxiredoxin 6 in neutraliza-
tion of X-ray mediated oxidative stress: eects on gene expres-
sion, preservation of radiosensitive tissues and postradiation
survival of animals,Free Radical Research, vol. 51, no. 2,
pp. 148166, 2017.
[24] S. W. Kang, I. C. Baines, and S. G. Rhee, Characterization of a
mammalian peroxiredoxin that contains one conserved cyste-
ine,Journal of Biological Chemistry, vol. 273, no. 11,
pp. 63036311, 1998.
[25] M. M. Bradford, A rapid and sensitive method for the quan-
titation of microgram quantities of protein utilizing the princi-
ple of protein-dye binding,Analytical Biochemistry, vol. 72,
no. 1-2, pp. 248254, 1976.
[26] M. Chintinne, G. Stangé, B. Denys, Z. Ling, P. in t Veld, and
D. Pipeleers, Beta cell count instead of beta cell mass to assess
and localize growth in beta cell population following pancre-
atic duct ligation in mice,PLoS One, vol. 7, no. 8, article
e43959, 2012.
[27] D. Wu and P. Yotnda, Production and detection of reactive
oxygen species (ROS) in cancers,Journal of Visualized Exper-
iments, no. 57, Article ID e3357, 2011.
[28] K. Maiese, Programming apoptosis and autophagy with novel
approaches for diabetes mellitus,Current Neurovascular
Research, vol. 12, no. 2, pp. 173188, 2015.
[29] M. Pietropaolo, J. M. Surhigh, P. W. Nelson, and G. S. Eisen-
barth, Primer: immunity and autoimmunity,Diabetes,
vol. 57, no. 11, pp. 28722882, 2008.
[30] R. Jain, D. M. Tartar, R. K. Gregg et al., Innocuous IFN-
gamma induced by adjuvant-free antigen restores normogly-
cemia in NOD mice through inhibition of IL-17
production,Journal of Experimental Medicine, vol. 205,
no. 1, pp. 207218, 2008.
[31] E. G. Novoselova, O. V. Glushkova, S. B. Parfenuyk et al., Pro-
tective eect of peroxiredoxin 6 against toxic eects of glucose
and cytokines in pancreatic RIN-m5F β-cells,Biochemistry
(Moscow), vol. 84, no. 6, pp. 637643, 2019.
[32] L. Monnier, E. Mas, C. Ginet et al., Activation of oxidative
stress by acute glucose uctuations compared with sustained
chronic hyperglycemia in patients with type 2 diabetes,
JAMA,vol. 295, no. 14, pp. 16811687, 2006.
[33] N. S. Wilcox, J. Rui, M. Hebrok, and K. C. Herold, Life and
death of βcells in type 1 diabetes: a comprehensive review,
Journal of Autoimmunity, vol. 71, pp. 5158, 2016.
[34] T. Tomita, Apoptosis of pancreatic β-cells in type 1 diabetes,
Bosnian journal of basic medical sciences, vol. 17, no. 3,
pp. 183193, 2017.
[35] N. Katunuma, A. Matsui, Q. T. le, K. Utsumi, G. Salvesen, and
A. Ohashi, Novel procaspase-3 activating cascade mediated
by lysoapoptases and its biological signicances in apoptosis,
Advances in Enzyme Regulation, vol. 41, no. 1, pp. 237250,
2001.
[36] G. P. Bienert, A. L. Møller, K. A. Kristiansen et al., Specic
aquaporins facilitate the diusion of hydrogen peroxide across
membranes,The Journal of Biological Chemistry, vol. 282,
no. 2, pp. 11831192, 2007.
[37] X. Kuang, L.-F. Wang, L. Yu et al., Ligustilide ameliorates
neuroinammation and brain injury in focal cerebral ische-
mia/reperfusion rats: involvement of inhibition of TLR4/per-
oxiredoxin 6 signaling,Free Radical Biology & Medicine,
vol. 71, pp. 165175, 2014.
10 Journal of Diabetes Research
[38] C. E. Hellweg, The nuclear factor κB pathway: a link to the
immune system in the radiation response,Cancer Letters,
vol. 368, no. 2, pp. 275289, 2015.
[39] S. Y. Kim, H.-Y. Jo, M. H. Kim et al., H
2
O
2
-dependent hyper-
oxidation of peroxiredoxin 6 (Prdx6) plays a role in cellular
toxicity via up-regulation of iPLA2 activity,The Journal of
Biological Chemistry, vol. 283, no. 48, pp. 3356333568, 2008.
[40] Y. Wu, S. I. Feinstein, Y. Manevich et al., Mitogen-activated
protein kinase-mediated phosphorylation of peroxiredoxin 6
regulates its phospholipase A
2
activity,Biochemical Journal,
vol. 419, no. 3, pp. 669679, 2009.
[41] A. Schmitt, W. Schmitz, A. Hufnagel, M. Schartl, and
S. Meierjohann, Peroxiredoxin 6 triggers melanoma cell
growth by increasing arachidonic acid-dependent lipid signal-
ling,Biochemical Journal, vol. 471, no. 2, pp. 267279, 2015.
[42] T. R. Nardelli, E. C. Vanzela, K. C. Benedicto et al., Prolactin
protects against cytokine-induced beta-cell death by NFκB and
JNK inhibition,Journal of Molecular Endocrinology, vol. 61,
no. 1, pp. 2536, 2018.
[43] Q. Zhang, M. J. Lenardo, and D. Baltimore, 30 years of NF-
κB: a blossoming of relevance to human pathobiology,Cell,
vol. 168, no. 1-2, pp. 3757, 2017.
[44] F. M. Paula, S. M. Ferreira, A. C. Boschero, and K. L. Souza,
Modulation of the peroxiredoxin system by cytokines in
insulin-producing RINm5F cells: down-regulation of PRDX6
increases susceptibility of beta cells to oxidative stress,Molec-
ular and Cellular Endocrinology, vol. 374, no. 1-2, pp. 5664,
2013.
[45] B. Chhunchha, N. Fatma, E. Kubo, P. Rai, S. P. Singh, and D. P.
Singh, Curcumin abates hypoxia-induced oxidative stress
based-ER stress-mediated cell death in mouse hippocampal
cells (HT22) by controlling Prdx6 and NF-κB regulation,
American Journal of Physiology. Cell Physiology, vol. 304,
no. 7, pp. C636C655, 2013.
[46] J. H. Pak, W. C. Son, S. B. Seo et al., Peroxiredoxin 6 expres-
sion is inversely correlated with nuclear factor-κB activation
during Clonorchis sinensis infestation,Free Radical Biology
& Medicine, vol. 99, pp. 273285, 2016.
[47] K. Meyerovich, F. Ortis, F. Allagnat, and A. K. Cardozo,
Endoplasmic reticulum stress and the unfolded protein
response in pancreatic islet inammation,Journal of molecu-
lar endocrinology, vol. 57, no. 1, pp. R1R17, 2016.
11Journal of Diabetes Research
... Furthermore, Prdx6 has an essential stimulating effect on insulin secretion in β cells (Novoselova et al. 2019). Prdx6 reduces the effect of exogenous proinflammatory cytokines that cause reactive oxygen species (ROS) production in β cells and can inhibit β-cell apoptosis (Novoselova et al. 2020b). Knockdown of Prdx6 in β cells activates intrinsic and extrinsic apoptotic pathways. ...
... Prdx6 protects β cells against oxidative stress (Paula et al. 2013) and significantly reduces reactive oxygen species levels and reduces hyperglycemia-induced cell death in β cells (Novoselova et al. 2019). Additionally, Prdx6 reduces the effect of exogenous proinflammatory cytokines that cause oxidative stress in β cells and inhibits apoptosis of these cells (Novoselova et al. 2020b). Suppression of Prdx6 in β cells causes the activation of intrinsic and extrinsic apoptotic pathways (Pacifici et al. 2022). ...
Article
Full-text available
Diabetes mellitus is a widespread endocrine disease worldwide, accompanying chronic hyperglycemia. In this study, we investigated the effect of hydroxytyrosol, which exerts an antioxidant effect, on the expressions of insulin and peroxiredoxin-6 (Prdx6), which protect cells against oxidative injury in diabetic rat pancreas. This experimental study had four groups with ten animals in each group: control (nondiabetic) group, hydroxytyrosol group [10 mg/kg/day intraperitoneal injection (ip) hydroxytyrosol for 30 days], streptozotocin group (single ip injection of 55 mg/kg streptozotocin), and streptozotocin + hydroxytyrosol group (single ip injection of streptozotocin and ip injection of 10 mg/kg/day hydroxytyrosol for 30 days). During the experiment, blood glucose levels were measured at regular intervals. Insulin expression was determined by immunohistochemistry and Prdx6 expression was determined by immunohistochemistry and western blot. Immunohistochemistry and western blot results were analyzed by one-way ANOVA with applied Holm–Sidak multiple comparison test, and blood glucose results were analyzed by two-way repeated measures ANOVA with applied Tukey’s multiple comparison test. Blood glucose levels on days 21 and 28 were significantly lower in the streptozotocin + hydroxytyrosol group compared with the streptozotocin group (day 21, p = 0.049 and day 28, p = 0.003). Expression of both insulin and Prdx6 were lower in the streptozotocin and the streptozotocin + hydroxytyrosol groups compared with the control and hydroxytyrosol groups (p < 0.001). Insulin and Prdx6 expression in the streptozotocin + hydroxytyrosol group were higher compared with the streptozotocin group (p < 0.001). The immunohistochemical findings of Prdx6 and western blot were the same. In conclusion, hydroxytyrosol, which is an antioxidant compound, increased Prdx6 and insulin expression in diabetic rats. Insulin increased by hydroxytyrosol may have been effective in reducing blood glucose levels. Furthermore, hydroxytyrosol may exert its effect on insulin by increasing Prdx6 expression. Thus, hydroxytyrosol may decrease or prevent several hyperglycemia-dependent complications by increasing the expression of these proteins.
... The roles of peroxiredoxins and their partners, thioredoxin and thioredoxin reductase, in β-cell antioxidant defense are well known [10,11]. We have previously demonstrated the protective role of exogenous peroxiredoxin 6 under T1D conditions in a diabetic mouse model and in a rat insulinoma RIN-m5F β-cell model [12][13][14]. In addition, we have recently discussed the role of senescent β-cells in the development of type 1 diabetes, in which we concluded that peroxiredoxin 6 can be qualified as a new protective and senolytic agent in diabetes mellitus [15]. ...
Article
Full-text available
Diabetes mellitus type 1 (T1D) and type 2 (T2D) develop due to dysfunction of the Langerhans islet β-cells in the pancreas, and this dysfunction is mediated by oxidative, endoplasmic reticulum (ER), and mitochondrial stresses. Although the two types of diabetes are significantly different, β-cell failure and death play a key role in the pathogenesis of both diseases, resulting in hyperglycemia due to a reduced ability to produce insulin. In T1D, β-cell apoptosis is the main event leading to hyperglycemia, while in T2D, insulin resistance results in an inability to meet insulin requirements. It has been suggested that autophagy promotes β-cell survival by delaying apoptosis and providing adaptive responses to mitigate the detrimental effects of ER stress and DNA damage, which is directly related to oxidative stress. As people with diabetes are now living longer, they are more susceptible to a different set of complications. There has been a diversification in causes of death, whereby a larger proportion of deaths among individuals with diabetes is attributable to nonvascular conditions; on the other hand, the proportion of cancer-related deaths has remained stable or even increased in some countries. Due to the increasing cases of both T1D and T2D, these diseases become even more socially significant. Hence, we believe that search for any opportunities for control of this disease is an overwhelmingly important target for the modern science. We focus on two differences that are characteristic of the development of diabetes’s last periods. One of them shows that all-cause death rates have declined in several diabetes populations, driven in part by large declines in vascular disease mortality but large increases in oncological diseases. Another hypothesis is that some T2D medications could be repurposed to control glycemia in patients with T1D.
... Regarding the relative weights of the organs, we notice nephromgaly, hepatic, pancreatic and ovarian hypertrophy. This increase in relative organ weight may be due to alloxan-induced necrosis and apoptosis [21]. The results obtained show an improvement in the weight of the members of the herbal treatment group, and this is due to the presence of some active ingredients in the herb, which led to the reduction of alloxan toxicity and the reduction of the secondary effects of diabetes [22]. ...
Article
Full-text available
Objective: To investigate the effect of polyherb (formulated by some plants) on physiological, biochemical alterations and oxidative stress induced by experimental diabetes in rats. Materials and methods: Twenty female albino Wistar rats were randomly divided into 4 groups (n=5); Control, Polyherb, diabetic rats and Diabetic rats treated with polyherb. All types of treatments were given to rats orally for 21 days. Results: In vitro phytochemical analysis results revealed that polyherb contains phenols, flavonoids, alkaloids, terpenoids, tannins and steroids. In vivo results, in diabetic group showed an alteration in lipid profile and biochemical parameters. While, results showed an increase in MDA level and a decrease in GSH and SOD levels compared to control. Moreover, the hematological parameters revealed that Diabetes induce a significant diminution (P<0.01) of RBC, MCV and HCT levels. In the other hand, histopathological analysis noted an alteration in pancreas tissues of diabetic rats group compared to control. However, treatment of Diabetic rats by Polyherb ensured a partial amelioration and correction of the previous parameters. Conclusion: Use of Polyherb appears to be the dominant limited of development and complications of diabetes which can have a benefic effect against others diseases which oxidative stress plays an important role.
... Interestingly, the insulin-stimulating activity of Prx6 in vitro was detected both when RIN-m5F β-cells were cultivated under normal conditions and under stressful conditions, inducing cell death [99]. In an alloxan-or streptozotocin-induced mouse model of diabetes, intravenous administration of recombinant Prdx6 prevented hyperglycemia, reduced mortality, restored the plasma cytokine profile, and reduced β-cell destruction in the islets of Langerhans in the mouse pancreas [100][101][102]. ...
Article
Full-text available
This minireview discusses the very important biomedical problem of treating type 2 diabetes mellitus (T2D). T2D accounts for more than 90% of the total number of diagnosed cases of diabetes mellitus and can result from aging, inflammation, obesity and β-cell senescence. The main symptom of both T2D and type 1 diabetes (T1D) is an increase in blood glucose concentration. While T1D is insulin-dependent and is associated with the destruction of pancreatic β-cells, T2D does not require lifelong insulin administration. In this case, pancreatic β-cells are not destroyed, but their functional activity is deregulated. In T2D, metabolic stress increases the number of senescent β-cells while impairing glucose tolerance. The potential paracrine effects of senescent β-cells highlight the importance of the β-cell senescenceassociated secretory phenotype (SASP) in driving metabolic dysfunction. We believe that the main reason for the deregulation of the functional activity of pancreatic β-cells in T2D is associated with their "aging" or senescence, which may be induced by various stressors. We propose the use of peroxiredoxin 6 as a new senolytic drug, and the role of β-cell senescence in the development of T2D is discussed in this review.
... PRDX6 has also been shown to be involved in cerebral ischemia/reperfusion inflammatory injury (16). Additionally, research has shown that PRDX6 plays a role in the pathogenesis of diabetes (17) and protects against type 1 diabetes (18). Notably, PRDX6 elevation suppresses oxidative stress and ferroptosis to ease podocyte injury in DN. ...
Article
Full-text available
Background: This research sought to elucidate the effects of peroxiredoxin 6 (PRDX6) on the biological processes in diabetic nephropathy (DN) and to identify the underlying regulatory mechanism related to toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling. Methods: To induce an in vitro DN cellular model, human kidney 2 (HK-2) cells were treated with high glucose (HG). The mitochondrial membrane potential, adenosine triphosphate level, reactive oxygen species generation, and oxidative stress of the cells were then evaluated. After the PRDX6 level had been determined, the effects of its overexpression on the mitochondrial membrane potential, adenosine triphosphate level, reactive oxygen species generation, and oxidative stress of the cells were assessed. Next, cytochrome c expression, cell viability, cell apoptosis, the inflammatory level, and the TLR4/NF-κB signaling-related factors were assessed. After the addition of the TLR4 activator CRX-527 or the NF-κB activator phorbol 12-myristate 13-acetate (PMA), cell viability, cell apoptosis and the inflammatory level were evaluated again. Results: The results revealed that HG exposure triggered mitochondrial dysfunction and oxidative stress and decreased PRDX6 expression in the HK-2 cells. PRDX6 elevation exacerbated cell viability while alleviating mitochondrial membrane potential loss, oxidative stress, apoptosis, and inflammation in the HG-treated HK-2 cells. Further, PRDX6 inhibited HG-induced TLR4/NF-κB activation. The administration of CRX-527 or PMA reversed the effects of PRDX6 on the cell viability, apoptosis, and inflammation of the HG-exposed HK-2 cells. Conclusions: To conclude, PRDX6 appears to protect HG-exposed HK-2 cells against inflammation and apoptosis by inhibiting TLR4/NF-κB signaling.
... Investigations by Nano et al. have documented protective effects of islet neogenesis associated protein (INGAP) against proinflammatory cytokineinduced metabolic defects, including accelerated NFκB signaling S536 phosphorylation and nuclear accumulation of p65 [70]. Novoselova and coworkers studied potential protective effects of peroxiredoxin 6 (Prx6) against metabolic dysfunction in rat insulinoma RIN-m5F cells following exposure to high glucose or proinflammatory cytokines [71]. Based on data accrued from a series of complementary studies, these investigators surmised that NF-κB signaling module, specifically S536 phosphorylation of p65, as a target for Prx6 mediated protective effects. ...
Article
Full-text available
Background/aims: We recently reported increased phosphorylation (at S536) of the p65 subunit of NFκB (Rel A) in pancreatic beta (INS-1 832/13) cells following exposure to hyperglycemic (HG) conditions. We also demonstrated that HG-induced S536 phosphorylation of p65 is downstream to the regulatory effects of CARD9 since deletion of CARD9 expression significantly attenuated HG-induced S536 phosphorylation of p65 in beta cells. The overall objective of the current investigation is to identify putative mechanisms underlying HG-induced phosphorylation of p65 in islet beta cells following exposure to HG conditions. Methods: INS-1 832/13 cells were incubated in low glucose (LG; 2.5 mM) or high glucose (HG; 20 mM) containing media for 24 hours in the absence or presence of small molecule inhibitors of G protein prenylation and activation. Non-nuclear and nuclear fractions were isolated from INS-1 832/13 cells using a commercially available (NE-PER) kit. Degree of S536 phosphorylation of the p65 subunit was quantified by western blotting and densitometry. Results: HG-induced p65 phosphorylation was significantly attenuated by inhibitors of protein prenylation (e.g., simvastatin and L-788,123). Pharmacological inhibition of Tiam1-Rac1 (e.g., NSC23766) and Vav2-Rac1 (e.g., Ehop-016) signaling pathways exerted minimal effects on HG-induced p65 phosphorylation. However, EHT-1864, a small molecule compound, which binds to Rac1 thereby preventing GDP/GTP exchange, markedly suppressed HG-induced p65 phosphorylation, suggesting that Rac1 activation is requisite for HG-mediated p65 phosphorylation. Lastly, EHT-1864 significantly inhibited nuclear association of STAT3, but not total p65, in INS-1 832/13 cells exposed to HG conditions. Conclusion: Activation of Rac1, a step downstream to HG-induced activation of CARD9, might represent a requisite signaling step in the cascade of events leading to HG-induced S536 phosphorylation of p65 and nuclear association of STAT3 in pancreatic beta cells. Data from these investigations further affirm the role(s) of Rac1 as a mediator of metabolic stress- induced dysfunction of the islet beta cell.
... Regarding the relative weights of the organs, we notice kidney, hepatic, and pancreatic hypertrophy. This increase in relative organ weight may be due to Alloxan-induced necrosis and apoptosis (Novoselova et al., 2020). For the therapeutic effect of zinc oxide nanoparticles. ...
Article
This study aimed to assess the effects of the immunomodulator thymulin, a thymic peptide with anti-inflammatory effects, and peroxiredoxin 6 (Prdx6), an antioxidant enzyme with dual peroxidase and phospholipase A2 activities, on the blood‒brain barrier (BBB) condition and general health status of animals with relapsing-remitting experimental autoimmune encephalomyelitis (EAE), which is a model of multiple sclerosis in humans. Both thymulin and Prdx6 significantly improved the condition of the BBB, which was impaired by EAE induction, as measured by Evans blue dye accumulation, tight-junction protein loss in brain tissue, and lymphocyte infiltration through the BBB. The effect was associated with significant amelioration of EAE symptoms. Thymulin treatment was accompanied by a decrease in immune cell activation as judged by interleukin-6, -17, and interferon-gamma cytokine levels in serum and NF-kappaB cascade activation in splenocytes of mice with EAE. Prdx6 did not induce significant immunomodulatory effects but abruptly decreased EAE-induced NOX1 and NOX4 gene expression in brain tissue, which may be one of the possible mechanisms of its beneficial effects on BBB conditions and health status. The simultaneous administration of thymulin and Prdx6 resulted in complete symptomatic restoration of mice with EAE. The results demonstrate prospective strategies for multiple sclerosis treatment.
Article
Significance: Oxidative stress is a common feature of autoimmune diseases. Low levels of reactive oxygen species (ROS) generation are important for various biological processes. Redox homeostasis can be disrupted when there is an imbalance between the production of ROS and the detoxification effect of antioxidants. Peroxiredoxins (PRDXs) are essential regulators of cellular redox signaling. Recent advances: PRDXs are widely expressed antioxidant enzymes, and their physiological role is mainly to remove excess ROS in cells and reduce oxidative stress. Recent studies have shown that almost all PRDX subtypes are involved in the development of autoimmune diseases. Critical issues: The pathogenesis of autoimmune diseases is complex, and effective treatments are lacking. Therefore, there is an urgent need to find new therapeutic targets. In this review, we discuss the functions of PRDXs and their pathophysiological roles in several autoimmune diseases. PRDXs may serve as potential targets for the treatment of autoimmune diseases. Future directions: PRDXs are important in oxidative stress-mediated pathological situations. Future in-depth exploration of the mechanisms involved in regulating PRDXs in autoimmune diseases is needed to develop strategies the targeting PRDXs for the treatment of autoimmune diseases.
Article
The reduction of insulin secretion due to pancreatic β cells injury caused by autoimmune reaction is the pathological basis of Type 1 diabetes mellitus (T1DM). Therefore, seeking new molecular targets for alleviating pancreatic β cells injury will provide experimental basis for the prevention and treatment of T1DM. SRY-box 9 (Sox9) is not only an important molecule regulating the development of various organs, but also its high expression can aggravate the pathological process of various diseases. In addition, Sox9⁺ cells are also pancreatic progenitor cells, participating in pancreatic repair reaction induced by injury. In our study, elevated blood glucose and lack of pancreatic β cells almost returned to normal over time after streptozotocin (STZ)-induced pancreatic β cells damage, implying that pancreatic β cells were regenerated after STZ-induced injury. In particular, the expression of Sox9 was significantly elevated during pancreatic β cells regeneration. On this basis, we conducted in vitro experiments to verify whether overexpression of Sox9 could inhibit the damage of pancreatic β cells by inflammatory factors. Our results showed that overexpression of Sox9 alleviated the damage of pancreatic β cells by inflammatory factors and improved the inhibitory effect of inflammatory factors on insulin secretion of pancreatic β cells. Unsurprising, blood glucose levels, insulin content and pancreatic β cells number failed to return to near-normal levels timely after pancreatic β cells specific knockout Sox9 mice were treated with STZ, further confirming the importance of Sox9 in facilitating pancreatic β cells repair or regeneration. Our study indicate that enhanced Sox9 activity might protect pancreatic β cells from autoimmune induced damage and thus improve the pathological process of T1DM.
Article
Full-text available
Peroxiredoxin 6 (Prx6) is an important antioxidant enzyme with various functions in the cell. Prx6 reduces a wide range of peroxide substrates, playing a leading role in maintaining the redox homeostasis of mammalian cells. In addition to the peroxidase activity, a phospholipase A2-like activity was demonstrated for Prx6, which plays an important role in the metabolism of membrane phospholipids. Besides that, due to its peroxidase and phospholipase activities, Prx6 participates in intracellular and intercellular signal transduction, thus triggering regenerative processes in the cell, suppressing apoptosis caused by various factors, including ischemia-reperfusion injuries. A nephroprotective effect of exogenous recombinant Prx6 administered before ischemia-reperfusion injury was demonstrated on an animal model. Exogenous Prx6 effectively alleviates the severeness of renal ischemia-reperfusion injuries and facilitates normalization of their structural and functional conditions. Infusion of exogenous Prx6 increases the survival rate of experimental animals by almost 3 times. Application of exogenous Prx6 can be an effective approach in the prevention and treatment of renal ischemia-reperfusion kidney lesions and in preserving isolated kidneys during transplantation.
Article
Full-text available
Oxidative stress arising during diabetes may lead to cataract formation. Thus, in order to prevent oxidative stress development, antioxidants could be considered helpful agents. Naringenin, a flavonoid with a well-documented antioxidative activity, can be found in many plant-derived products, especially citrus fruits. The aim of the study was to examine the effect of naringenin on oxidative stress markers in the lenses of type 1 diabetic rats. The study was conducted on 3-month-old male Wistar rats with streptozotocin-induced type 1 diabetes. The rats were treated orally with naringenin at the doses of 50 and 100 mg/kg for 4 weeks. In the lenses obtained from the animals, enzymatic and non-enzymatic parameters connected with oxidative stress were measured. The enzymatic parameters included superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase and glucose-6-phosphate dehydrogenase activity. For non-enzymatic parameters, the total thiol groups, reduced and oxidized glutathione, protein carbonyl groups, advanced oxidation protein products, malondialdehyde and vitamin C level were assayed. Oral administration of naringenin counteracted most of the unfavorable changes induced by diabetes, including reduction of elevated antioxidative enzymes activity and amelioration of oxidative damage in proteins and lipids. Naringenin administered orally reduces oxidative stress markers in the lenses of type 1 diabetic rats.
Article
Taking into account a special role of pancreatic β-cells in the development of diabetes mellitus, the effects of peroxiredoxin 6 (Prx6) on the viability and functional activity of rat insulinoma RIN-m5F β-cells were studied under diabetes-simulating conditions. For this purpose, the cells were cultured at elevated glucose concentrations or in the presence of proinflammatory cytokines (TNF-α and IL-1) known for their special role in the cytotoxic autoimmune response in diabetes. It was found that the increased glucose concentration of 23-43 mM caused death of 20-60% β-cells. Prx6 added to cells significantly reduced the level of reactive oxygen species and protected the RIN-m5F ß-cells from hyperglycemia, reducing the death of these cells by several fold. A measurement of insulin secretion by the RIN-m5F ß-cells showed a significant stimulatory effect of Prx6 on the insulin-producing activity of pancreatic β-cells. It should be noted that the stimulatory activity of Prx6 was detected during culturing the cells under both normal and unfavorable conditions. The regulation of the NF-ϰB signaling cascade could be one of the mechanisms of Prx6 action on β-cells, in particular, through activation of RelA/p65 phosphorylation at Ser536.
Article
Background: Chronic hyperalgesia and allodynia associated with progressive damage of peripheral neurons are the most prevalent complications of diabetes mellitus. Plants belonging to the family of Oleaceae were traditionally used in folk medicine for the management of diabetes. Hypothesis/purpose: The aim of this study was to investigate whether an aqueous extract from the leaves of Ligustrum vulgare (common privet) could be useful to target neuropathic pain in a rat streptozotocin (STZ) model of diabetes. Methods: The chemical composition of the aqueous extract from privet leaf was characterized with the UHPLC-DAD-MS method and the analytical quantification of its constituents was performed with HPLC-DAD. Mechanical hyperalgesia and allodynia were evaluated with the Randall-Selitto and von Frey tests. Results: Our investigation revealed the presence of secoiridoids: oleacein (23.48 ± 0.87 mg/g), oleocanthal (8.44 ± 0.08 mg/g), oleuropein (1.50 ± 0.01 mg/g), as well as phenylpropanoids: echinacoside (6.46 ± 0.07 mg/g), verbascoside (4.03 ± 0.04 mg/g) and p-coumaroyl glucarates in the dried aqueous extract of privet leaves. Behavioral data indicated that chronic intraperitoneal administration of the extract (50-200 mg/kg) for 21 days resulted in a decrease in diabetes-induced hyperalgesia and allodynia. Blood glucose levels remained unaltered, while body weight and water intake decreased significantly. Conclusion: The aqueous privet leaf extract could serve useful in facilitating treatment of painful diabetic neuropathy. Additionally, the study showed that the antihyperalgesic activity of Ligustrum vulgare leaf extract is not likely related to its antihyperglycemic properties.
Article
Type 1 diabetes is caused by an autoimmune assault that induces progressive beta cell dysfunction and dead. Pro-inflammatory cytokines, such as interleukin 1 beta (IL1b), tumor necrosis factor (TNF) and interferon gamma (IFNg) contribute for beta cell death, which involves activation of the nuclear factor kappa B (NFkB) and jun N-terminal kinase (JNK). Prolactin (PRL), a physiological mediator for beta cell proliferation, was shown to protect beta cells against cytokines pro-apoptotic effects. We presently investigated the mechanisms involved in the protective effects of prolactin against cytokine-induced beta cell death. The findings obtained indicate that STAT3 activation is involved in the anti-apoptotic role of PRL in rat beta cells. PRL prevents the activation of JNK via AKT and promotes a shift from expression of pro- to anti-apoptotic proteins downstream of the JNK cascade. Furthermore, PRL partially prevents the activation of NFkB and the transcription of its target genes IkBa, FAS, MCP1, A20 and CXCL10, and also decreases NO production. On the other hand, the pro-survival effects of PRL do not involve modulation of cytokine-induced endoplasmic reticulum stress. These results suggest that the beneficial effects of PRL in beta cells involve augmentation of anti-apoptotic mechanisms and, at the same time, reduction of pro-apoptotic effectors, rendering beta cells better prepared to deal with inflammatory insults. The better understanding of the pro-survival mechanisms modulated by PRL in beta cells can provide tools to prevent cell demise during an autoimmune attack or following islet transplantation.
Article
Type 1 diabetes mellitus (T1DM) results from autoimmune destruction of pancreatic ?-cells after an asymptomatic period over years. Insulitis activates antigen presenting cells, which trigger activating CD4+ helper-T cells, releasing chemokines/cytokines. Cytokines activate CD8+ cytotoxic -T cells, which lead to ?-cell destruction. Apoptosis pathway consists of extrinsic (receptor-mediated) and intrinsic (mitochondria-driven) pathway. Extrinsic pathway includes Fas pathway to CD4+-CD8+ interaction, whereas intrinsic pathway includes mitochondria-driven pathway at a balance between pro-apoptotic B-cell lymphoma (Bcl) 2 and Bcl-xL and anti-apoptotic Bad, Bid, and Bik proteins. Activated cleaved caspse-3 is the converging point between extrinsic and intrinsic pathway. Apoptosis takes place only when pro-apoptotic proteins exceed anti-apoptotic proteins. Since the concordance rate of T1DM in identical twins is about 50%, environmental factors are involved in the development of T1DM, opening a door to find means to detect and prevent further development of autoimmune ?-cell destruction for a therapeutic application.
Article
Peroxiredoxins are redox-sensing multifunctional enzymes, аmong them peroxiredoxin 6 (Prx6) can neutralize the most broadest range of hydroperoxides and playing an important role in maintaining the redox homeostasis of the cell. In the present study, radioprotective and signaling regulatory effects of Prx6 were demonstrated and characterized. Intravenously administered exogenous Prx6 protects the organism of mice from the destructive action of ionizing radiation in the lethal dose range of 5-10 Gy. Dose reduction factor of 1.4. Prx6 injection reduces the severity of radiation-induced leuko- and thrombopenia in irradiated animals, also preventing the destruction of epithelial cells in the small intestine. Injecting exogenous Prx6 also as its mutated form of Prx6-C47S lacking peroxidase activity affects the expression of genes involved in antioxidant response, DNA reparation, apoptosis and inflammatory processes, in bone marrow cells both in intact animals and in those subjected to ionizing radiation. The radioprotective properties of Prx6 are based, on the one hand, on the capability for ROS neutralization, and on the other hand - on the potentiality for activation of reparation processes of the cell under oxidative stress conditions. Prx6 can be considered as a potentially perspective radioprotective agent for the reduction of risks from the damaging action of ionizing radiation on the mammalian organism.
Article
Figure 1. The NF-κB Paradigm of Timely and Flexible Biochemical Control of Cell Behavior
Article
Clonorchis sinensis is a carcinogenic human liver fluke. Its infection promotes persistent oxidative stress and chronic inflammation environments in the bile duct and surrounding liver tissues owing to direct contact with worms and their excretory-secretory products (ESPs), provoking epithelial hyperplasia, periductal fibrosis, and cholangiocarcinogenesis. We examined the reciprocal regulation of two ESP-induced redox-active proteins, NF-κB and peroxiredoxin 6 (Prdx6), during C. sinensis infection. Prdx6 overexpression suppressed intracellular free-radical generation by inhibiting NADPH oxidase2 and inducible nitric oxide synthase activation in the ESP-treated cholangiocarcinoma cells, substantially attenuating NF-κB-mediated inflammation. NF-κB overexpression decreased Prdx6 transcription levels by binding to two κB sites within the promoter. This transcriptional repression was compensated for by other ESP-induced redox-active transcription factors, including erythroid 2-related factor 2 (Nrf2), hypoxia inducible factor 1α (HIF1α), and CCAAT/enhancer-binding protein β (C/EBPβ). Distribution of immunoreactive Prdx6 and NF-κB was distinct in the early stages of infection in mouse livers but shared concomitant localization in the later stages. The intensity and extent of their immunoreactive staining in infected mouse livers are proportional to lesion severity and infection duration. The constitutive elevations of Prdx6 and NF-κB during C. sinensis infection may be associated with more severe persistent hepatobiliary abnormalities mediated by clonorchiasis.