ArticlePDF Available

Nephroprotective Effects of N-Acetylcysteine Amide against Contrast-Induced Nephropathy through Upregulating Thioredoxin-1, Inhibiting ASK1/p38MAPK Pathway, and Suppressing Oxidative Stress and Apoptosis in Rats

Wiley
Oxidative Medicine and Cellular Longevity
Authors:
  • Shanghai Municipal Hospital of Traditioanl Chinese Medicine

Abstract and Figures

Contrast-induced nephropathy (CIN) is a leading cause of hospital-acquired acute kidney injury (AKI) due to apoptosis induced in renal tubular cells. Our previous study demonstrated the novel N-acetylcysteine amide (NACA); the amide form of N-acetyl cysteine (NAC) prevented renal tubular cells from contrast-induced apoptosis through inhibiting p38 MAPK pathway in vitro. In the present study, we aimed to compare the efficacies of NACA and NAC in preventing CIN in a well-established rat model and investigate whether thioredoxin-1 (Trx1) and apoptosis signal-regulating kinase 1 (ASK1) act as the potential activator for p38 MAPK. NACA significantly attenuated elevations of serum creatinine, blood urea nitrogen, and biomarkers of AKI. At equimolar concentration, NACA was more effective than NAC in reducing histological changes of renal tubular injuries. NACA attenuated activation of p38 MAPK signal, reduced oxidative stress, and diminished apoptosis. Furthermore, we demonstrated that contrast exposure resulted in Trx1 downregulation and increased ASK1/p38 MAPK phosphorylation, which could be reversed by NACA and NAC. To our knowledge, this is the first report that Trx1 and ASK1 are involved in CIN. Our study highlights a renal protective role of NACA against CIN through modulating Trx1 and ASK1/p38 MAPK pathway to result in the inhibition of apoptosis among renal cells.
This content is subject to copyright. Terms and conditions apply.
Research Article
Nephroprotective Effects of N-Acetylcysteine Amide against
Contrast-Induced Nephropathy through Upregulating
Thioredoxin-1, Inhibiting ASK1/p38MAPK Pathway, and
Suppressing Oxidative Stress and Apoptosis in Rats
Xuezhong Gong,1Yiru Duan,1Junli Zheng,1Yiquan Wang,1Guohua Wang,1
Svante Norgren,2and Tom K. Hei3
1Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine,
Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai 200071, China
2Department of Womens and Childrens Health, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
3Center for Radiological Research, Department of Radiation Oncology, College of Physician and Surgeons, Columbia University,
630West168thStreet,NewYork,NY10032,USA
Correspondence should be addressed to Xuezhong Gong; shnanshan@hotmail.com
Received  October ; Accepted  November 
Academic Editor: Mohamed M. Abdel-Daim
Copyright ©  Xuezhong Gong et al. is is an open access article dist ributed under the Creative Commons AttributionL icense,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Contrast-induced nephropathy (CIN) is a leading cause of hospital-acquired acute kidney injury (AKI) due to apoptosis induced
in renal tubular cells. Our previous study demonstrated the novel N-acetylcysteine amide (NACA); the amide form of N-acetyl
cysteine (NAC) prevented renal tubular cells from contrast-induced apoptosis through inhibiting p MAPK pathway in vitro.
In the present study, we aimed to compare the ecacies of NACA and NAC in preventing CIN in a well-established rat model
and investigate whether thioredoxin- (Trx) and apoptosis signal-regulating kinase  (ASK) act as the potential activator for p
MAPK. NACA signicantly attenuated elevations of serum creatinine, blood urea nitrogen, and biomarkers of AKI. At equimolar
concentration, NACA was more eective than NAC in reducing histological changes of renal tubular injuries. NACA attenuated
activation of p MAPK signal, reduced oxidative stress, and diminished apoptosis. Furthermore, we demonstrated that contrast
exposure resulted in Trx downregulation and increased ASK/p MAPK phosphorylation, which could be reversed by NACA
and NAC. To our knowledge, this is the rst report that Trx and ASK are involved in CIN. Our study highlights a renal protective
role of NACA against CIN through modulating Trx and ASK/p MAPK pathway to result in the inhibition of apoptosis among
renal cells.
1. Introduction
CIN (contrast-induced nephropathy) has become a leading
cause of hospital-acquired acute kidney injury as a result of
the increasing use of iodine contrast media and the simulta-
neous increase in number of at-risk patients, for example, due
to diabetes or hypertension [–]. e most common clinical
course is a transient nonoliguric and asymptomatic decline in
renal function with serum creatinine levels peaking at days
–, but CIN can also cause long-term adverse events and
need for chronic dialysis [–]. us, it is essential not only
to investigate the pathogenesis of CIN but also to develop
preventive interventions [].
ere is accumulating evidence that CIN is caused by a
combination of a reduction in medullary blood ow resulting
in hypoxia and direct tubular damage, including apoptosis
[–]. Oxidative stress has been identied as an important
driver mechanism in the pathogenesis, and this has trig-
gered trials of antioxidants to prevent CIN [, ]. Although
there is no consensus or standard practice regarding the
most eective intervention to prevent CIN besides adequate
hydration, the international work group Kidney Disease:
Hindawi Publishing Corporation
Oxidative Medicine and Cellular Longevity
Volume 2016, Article ID 8715185, 11 pages
http://dx.doi.org/10.1155/2016/8715185
Oxidative Medicine and Cellular Longevity
Improving Global Outcomes (KDIGO) suggested using oral
administration of the antioxidant N-acetylcysteine (NAC)
along with intravenous uids in patients at increased risk
to develop CIN [, ]. ere is, however, an inconsistency
in guidelines regarding the benet of NAC [, ], which
highlights the need for more investigation to seek new
antioxidants and address the eectiveness of antioxidants in
the prevention of the disease.
NACA,alsotermedAD,astheamideformofNAC,is
a thiol antioxidant with enhanced properties of lipophilicity,
membrane permeability, and antioxidant capacity when com-
pared with NAC []. Recently emerging evidence conrmed
NACA as a protective agent against oxidative stress in vitro
and in vivo [–]. Our previous study also indicated NACA
could protect renal tubular epithelial cell against contrast-
induced apoptosis in vitro []. We thus hypothesized NACA
could be a better renoprotective agent against CIN than NAC
in vivo through its prominent antioxidant activity.
We have previously demonstrated that the low-osmolar,
nonionic contrast agent iohexol, the most widely used radio-
contrast media, induces renal tubular cell apoptosis through
activation of the p MAPK/iNOS signal pathway in vitro
and vivo [, ]. is signal pathway has been conrmed
by others in a human renal tubular cell line (HK) and
cultured renal tubular cells isolated from CIN patients [,
]. We have subsequently identied the Forkhead box O
transcriptional factor (FoxO) as a downstream element of
the p MAPK cascade []. However, little is known of the
upstream modulators of the p MAPK pathway in CIN as
well as in kidney disease [].
One putative candidate for the upstream signal activa-
tor of p MAPK is Apoptosis Signal-regulating Kinase 
(ASK) []. As a serine/threonine kinase belonging to the
mitogen activated protein kinase kinase kinase (MAPK)
family, ASK has been reported to play a critical role in
reactive oxygen species (ROS)-induced apoptosis in vari-
ous cell types and oxidative stress-related diseases such as
D-galactosamine/lipopolysaccharide induced hepatotoxicity
and cardiovascular disease [–]. Meanwhile, as an impor-
tant redox regulator, thioredoxin- (Trx) could bind to the N
terminal non catalytic region of ASK and act as an upstream
inhibitor of ASK [, ].
Inthepresentstudy,weaimedtocomparetheecaciesof
NACA and NAC in preventing CIN and further investigated
whether Trx/ASK signal, as the potential upstream modu-
lators of p MAPK, were involved in CIN pathogenesis.
2. Materials and Methods
2.1. Reagents. All chemicals were purchased from Sigma (St.
Louis, MO, USA) unless otherwise stated. N-Acetylcysteine
amide (NACA) was provided by Dr. Glenn Goldstein (New
York, NY, USA). e contrast media iohexol (Omnipaque)
was purchased from Amersham Health (Princeton, NJ, USA).
2.2. Animals. e study was approved by Medicine Animal
Ethics Committee of Shanghai University of Traditional
Chinese Medicine. A total of  adult –-week-old male
Sprague-Dawley rats weighing – g were purchased
from Shanghai Lab Animal Research Center (certicate num-
ber: -). Rats were housed in an air-conditioned room
at Cwithacycleofh/hlight/dark.Foodandwater
were provided ad libitum except for the day of dehydration.
2.3. Experimental Protocol and Drugs. We us e d a w e l l -
established rat model of CIN [, –]. Rats were randomly
divided into  groups of  rats each: controls (CON), rats
injected with CM (CIN), rats treated with  mg/kg/d NAC
andinjectedwithCM(CIN+NAC),ratstreatedwithmg/
kg/d NACA and injected with CM (CIN+NACA), and
rats treated with  mg/kg/d NACA and injected with CM
(CIN+NACA). NAC and NACA were injected intraperi-
toneally (i.p.) once daily for  consecutive days (days –).
CONandCINratsweregiventhesamevolumeofsaline.
On day , all rats were le without water for h. On day
,  min aer injections of saline, NAC or NACA, CIN,
CIN+NAC, CIN+NACA, and CIN+NACA rats were
injected with a nitric oxide synthase inhibitor (NG-nitro-L-
arginine methyl ester, L-NAME,  mg/kg, i.p.), followed aer
 and  min, respectively, by injection of an inhibitor of
prostaglandin synthesis (indomethacin,  mg/kg, i.p.) and
iohexol (.– g iodine/kg, i.p.). CON rats received injections
of equivalent volume of saline. On day , all rats were allowed
regular chow and tap water in metabolic cages for  h.
Baseline blood samples were collected from the tail vein
under ether anaesthesia for analysis of serum creatine (Scr),
blood urea nitrogen (BUN), and plasma Cystatin-C (CysC).
Urine samples (-h) were collected on day  (baseline)
and on day  for determination of urinary N-acetyl-𝛽-
glucosaminidase (UNAG) and urinary 𝛾-glutamyl transpep-
tidase (UGGT). At the end of day , blood samples were
collected from the abdominal aorta under pentobarbital
( mg/kg) anesthesia for determination of Scr, BUN, and
CysC. Subsequently the rats were killed and kidneys removed
for biochemical and morphological studies.
2.4. Histopathological Examinations
2.4.1. Light Microscopy. Le kidney samples were xed in %
formalin and prepared for examination by light microscopy
by hematoxylin and eosin (HE) staining, TUNEL staining, or
immunohistochemistry (IHC).
For TUNEL staining, sections were stained using In
Situ Cell Death Detection Kit (Roche Applied Science,
Mannheim, Germany). TUNEL-positive tubular cell num-
bers were counted in  nonoverlapping random cortical
elds under a x magnication.
For IHC, sections (mm) were immersed in citrate buer
and autoclaved at C for  min and then immersed in
% aqueous hydrogen peroxide (H2O2). e sections were
incubated with a rabbit polyclonal antibody (phospho-p
MAPK, , Cell Signaling Technology, Danvers, MA, USA,
 : ) for  h at room temperature. Immunodetection was
performed using biotinylated anti-rabbit IgG and peroxidase-
labeled avidin chain working uid (Beijing Zhong Shan
Oxidative Medicine and Cellular Longevity
Golden Bridge Biotechnology Co., China), with diaminoben-
zidine as the substrate. Finally, the slides were lightly counter-
stained with hematoxylin for  sec. e positive signals were
measured using Motic Med . CMIAS Image Analysis Sys-
tem (Motic China Group Co., Ltd., China). e area density
representing the positive staining intensity was calculated as
the ratio between the stained area and the total analyzed eld.
Two blinded examiners independently analyzed all slides.
2.4.2. Transmission Electron Microscopy (TEM). e right
renal cortex samples were cut into pieces ( × mm) on
ice and xed in .% (v/v) glutaraldehyde-polyoxymethylene
solutionforhat
C and subsequently embedded in Epon
. Ultrathin sections (– nm) were stained with uranyl
acetate and alkaline lead citrate and visualized under a JEM
CX transmission electron microscope.
2.4.3. Analysis of Renal Oxidative Stress Indicators. Super-
oxide dismutase (SOD), malondialdehyde (MDA), and glu-
tathione (GSH) levels in renal tissues were determined using
SOD kit, MDA kit, and GSH kit, respectively (Sigma, St.
Louis, MO, USA). Briey, tissue blocks of the appropriate size
were placed in ice-cold normal saline and homogenated in
aratioof:,𝑤(g) : V(ml). Aer  min of centrifugation
at  rpm, the supernatant was removed and used for
determination of SOD, MDA, and GSH using the respective
kitsandanUV-visiblespectrophotometer.
2.4.4. Quantitative Real-Time PCR (QPCR). Renal cortexes
were dissected and total RNA was extracted using Tri-
zol according to the manufacturers instructions (Invitro-
gen, Carlsbad, USA). cDNA was synthesized using random
primer and a High Capacity cDNA Reverse Transcrip-
tion Kit (Applied Biosystems, USA). e following primers
were used: Trx F-GTGGTGTGGACCTTGCAAAA, R-
GGAAGGTCGGCATGCATTTG; beta-actin F-CTGTGT-
GGATTGGTGGCTCT, R-GCAGCTCAGTAACAGTCC-
GC. QPCR was performed on a  Real-Time PCR System
(Applied Biosystems, USA).
2.4.5. Western Blot Analysis. Western blotti n g w a s p e r fo r m e d
as described [, , , ]. Renal cortex samples were lysed in
a lysis buer, separated by electrophoresis in –% poly-
acrylamide gel, and transferred to polyvinylidene uoride
(PVDF) membranes. Protein expression was quantied by
Image J . soware (Wayne Rasband, NIH, Bethesda,
MD, USA) aer scanning the lm. Primary antibodies used
included anti-ASK (abcam); anti-phospho-ASK (abcam,
Ser); anti-Trx (abcam); anti-p MAPK (Cell Signal-
ing Technology); anti-phospho-p MAPK (Cell Signaling
Technology, r/Tyr); anti-cleaved caspase  (Cell
Signaling Technology); and anti-beta-actin (Cell Signaling
Technology). All experiments were performed at least  times
(i.e.,  separate protein preparations) under the same condi-
tions.
2.4.6. Statistical Analysis. Results are expressed as means ±
SD. One-way analysis of variance (ANOVA) with Tukey’s post
hoc multiple-comparison test was used to determine the sig-
nicance of dierences in multiple comparisons. Dierences
were considered signicant if 𝑝 < 0.05 and highly signicant
if 𝑝 < 0.01.
3. Results
3.1. Eects of Induction of CIN (Table 1). At baseline, there was
no dierence in blood levels of markers of renal functional
among the  groups. CIN was eectively induced in CIN
group as evident from drastic deterioration in renal func-
tion (Table ) and morphological changes, including severe
vacuolization of the renal cortex, intratubular cast formation,
and medullary congestion (Figure (b)).
3.2. Eect of NAC and NACA on Renal Function Parameters.
Pretreatment with NAC or NACA preserved renal function as
evident from analysis of renal function parameters (Table ).
At the same dose ( mg/kg/d), NACA was uniformly more
eective than NAC as demonstrated by lower Scr, BUN,
UNAG, and UGGT (Table , 𝑝 < 0.05 versus CIN+NAC).
In fact,  mg/kg/d NACA had similar eects to  mg/kg/d
of NAC.
3.3. Eects of NAC and NACA on Histopathology and Ultra-
structures (Figure 1). Histopathological examinations of renal
samples from CIN rats revealed normal glomerulus structure
but severe renal tubular interstitial injury (Figure (b)).
Pretreatment with NAC or NACA markedly attenuated the
development of these lesions (Figures (c), (d), and (e)).
Renal tubular epithelial cell apoptosis in CIN rats was
evident by TEM. Cells undergoing apoptosis were character-
ized by injuries to ultrastructures (Figure (g)), for example,
condensation of the nuclear chromatin, wrinkling of nuclear
membranes, swelling of mitochondria, fracture of cristae, and
shedding of microvilli of cell cavity. In comparison, both
the apoptotic cell number and injuries to ultrastructures
were obviously reduced in CIN+NAC, CIN+NACA, and
CIN+NACA rats compared to CIN rats (Figures (h),
(i), and (j)). Consistent with the ndings under light
microscopy, no obvious glomerular lesions were observed by
TEM in CIN rats (Figures (k) and (l)).
3.4. Eects of NAC and NACA on Renal Tubular Apoptosis as
Assessed by TUNEL Staining and Analysis of Cleaved Caspase
3 (Figure 2). In addition to TEM, apoptosis was assessed
with two independent methods: TUNEL staining in kidney
sections and analysis of cleaved caspase  by western blot-
ting. Compared to CON rats, CIN rats exhibited markedly
increased numbers of TUNEL-positive tubular cells (Figures
(a), (b), and (f), 𝑝 < 0.01 versus CON) and also
increased cleavage of caspase  (Figures (g) and (h), 𝑝<
0.01 versus CON). Pretreatment with NAC or NACA signif-
icantly decreased both apoptotic cell numbers (Figures (c)–
(f), 𝑝 < 0.01 versus CIN) and caspase  cleavage (Figures
(g) and (h), 𝑝 < 0.01 versus CIN). At the same dose
( mg/kg/d), NACA exhibited better protection than NAC
Oxidative Medicine and Cellular Longevity
T : Eects of NACA on renal function parameters. e baseline levels of Scr, BUN, CysC, UNAG, and UGGT did not dier among the
groups. In CIN rats, BUN, Scr, CysC, UNAG, and UGGT were markedly increased on day  as compared to CON rats (𝑝 < 0.01). Pretreatment
with NAC or NACA attenuated the eect of CM, and the increases in Scr, BUN, CysC, UGGT, and UNAG were signicantly lower than in
CIN group (𝑝 < 0.05 or .). At the same dose ( mg/kg/d), NACA was signicantly more eective than NAC (CIN+NACA versus
CIN+NAC).
CON (𝑛=8)CIN(𝑛=8)CIN+NAC(𝑛=8)CIN+NACA(𝑛=8)CIN+NACA(𝑛=8)
Scr (𝜇mol/l)
Baseline . ±. . ±. . ±. . ±. . ±.
Day  . ±. . ±.∗∗ . ±.∗∗#. ±.   ∗∗#. ±.##&
Serum BUN (mmol/l)
Baseline . ±. . ±. . ±. . ±. . ±.
Day  . ±. . ±.∗∗ . ±.∗∗## . ±.∗∗## . ±.##&
Serum Cystatin-C (U/l)
Baseline . ±. . ±. . ±. . ±. . ±.
Day  . ±. . ±.∗∗ . ±.∗∗#. ±.∗∗#. ±.∗∗##
UNAG (U/l)
Baseline . ±. . ±. . ±. . ±. . ±.
Day  . ±. . ±.∗∗ . ±.   ∗∗#. ±.∗∗#. ±. ##&
UGGT (IU/l)
Baseline . ±. . ±. . ±. . ±. . ±.
Day  . ±. . ±.∗∗ . ±.∗∗## . ±.∗∗## . ±.∗∗##&
Data are means ±SD. 𝑝<0.05versus CON, ∗∗𝑝<0.01versus CON, #𝑝<0.05versus CIN, ##𝑝<0.01versus CIN, and &𝑝<0.05versus CIN+NAC.
(a) (b) (c) (d) (e)
(f) (g) (h) (i) (j)
(k) (l)
F : NACA attenuated CM-induced morphological changes. HE staining of kidney sections (magnication ×) from CON rats
(a), CIN rats (b), NAC+CIN rats (c), NACA+CIN rats (d), and NACA+CIN rats (e). Representative changes of ultrastructure by TEM
(magnications ×) from CON rats (f), CIN rats (g), NAC+CIN rats (h), NACA+CIN rats (i), NACA+CIN group (j), normal glomerular
basement membrane and podocyte in CON rats (k), and CIN rats (l). Note the condensation of the nuclear chromatin (red arrow) and
cytoplasm vacuoles (orange arrow) in CIN rats. Blue arrows refer to severe vacuolization of the renal cortex. Figures are representative of 
to  rats from each group.
Oxidative Medicine and Cellular Longevity
(a) (b) (c) (d) (e)
Con CIN CIN+CIN+CIN+
NAC NACA1NACA2
∗∗##
∗∗## ∗∗##&
∗∗
10
0
20
30
40
% of apoptotic cells
(TUNEL-positive)
(f)
Con CIN CIN+
NAC
CIN+
NACA1
CIN+
NACA2
Cleaved
caspase 3
Beta-actin
19 kd
17 kd
(g)
Con CIN CIN+CIN+CIN+
NAC NACA1NACA2
## ##
##&
∗∗
0.0
0.2
0.4
0.6
0.8
1.0
Relative density (cleaved
caspase 3/actin)
(h)
F : NACA inhibited CM-induced renal tubular cell apoptosis as detected by TUNEL staining and western blot analyses of cleaved
caspase.CMincreasedthenumberofTUNEL-positiverenaltubularcells(bluearrows),andpretreatmentwithNACorNACAblocked
this eect. TUNEL-stained kidney sections (magnications ×) from CON rats (a), CIN rats (b), NAC+CIN rats (c), NACA+CIN rats
(d), and NACA+CIN rats (e). TUNEL-positive cells are marked by blue arrows. (f) Quantitative analysis of TUNEL-positive cell number.
(g) Western blot analyses of cleaved capsase-. (h) Relative densitometry analysis of the ratio of cleaved caspase  to beta-actin. Figures are
representative of  to  rats from each group. e values are means ±SD (𝑛=5). 𝑝< 0.05 versus CON, ∗∗ 𝑝< 0.01 versus CON, ##𝑝< 0.01
versus CIN, and &𝑝< 0.05 versus CIN+NAC.
(Figure (f), 𝑝 < 0.05 versus CIN+NAC, and Figures (g) and
(h), 𝑝 < 0.05 versus CIN+NAC).
3.5. Eects of NAC and NACA on Indicators of Oxidative Stress
in Renal Tissue. Induction of CIN attenuated renal SOD
(Figure (a)) and GSH (Figure (c)) and increased MDA (Fig-
ure (b)) levels (𝑝 < 0.01 versus CON). Pretreatment with
NAC or NACA signicantly prevented this (Figure , 𝑝<
0.05 and 𝑝 < 0.01 versus CIN, resp.). At equimolar concen-
trations, NACA was more eective than NAC at preserving
SOD, MDA, and GSH levels (𝑝 < 0.05 versus CIN+NAC,
Figures (a)–(c)).
3.6. Eects of NAC and NACA on P38 MAPK Phosphorylation.
Activation of p MAPK was conrmed by a signicant
increase in phospho-p MAPK levels as detected by western
blotting and IHC (Figures (b) and (g), 𝑝 < 0.01 ver-
sus CON). Pretreatment with NAC or NACA signicantly
prevented the CIN-induced p MAPK activation in kidney
(Figures (c), (d), (e), and (g), 𝑝 < 0.01 versus CIN).
Again,NACAwasmoreeectivethanNACatthesamedose
(Figures (f) and (h), 𝑝 < 0.05 versus CIN+NAC).
3.7. Eects of NAC and NACA on ASK1 Phosphorylation. We
next examined whether ASK, a potential upstream signal of
p MAPK, was involved in CIN pathogenesis. As shown in
Figures (a) and (b), phospho-ASK level was upregulated
in CIN rats (𝑝 < 0.01 versus CON). Pretreatment with NAC
(𝑝 < 0.01 versus CIN) or, more eectively, NACA (Figures
(a) and (b), 𝑝 < 0.05 versus CIN+NAC) inhibited this
activation.
3.8. Eects of NAC and NACA on Trx1 mRNA and Protein
Expressions. In order to deduce the role of Trx in CIN, Trx
mRNA and protein expressions were evaluated by QPCR and
western blotting, respectively. As shown in Figure , Trx
protein expression (c, d) and mRNA level (e) were decreased
in renal cortex following CIN induction (𝑝 < 0.01 CON
Oxidative Medicine and Cellular Longevity
0
100
200
300
400
500
600
SOD (U/mg protein)
Con CIN CIN+
NAC
CIN+
NACA1
CIN+
NACA2
∗∗
## ##
##&
(a)
0
2
4
6
8
MDA (nmol/mg protein)
Con CIN CIN+
NAC
CIN+
NACA1
CIN+
NACA2
∗∗
∗∗## ∗∗## ##&
(b)
0.0
1.0
2.0
3.0
GSH (nmol/mg protein)
Con CIN CIN+
NAC
CIN+
NACA1
CIN+
NACA2
∗∗## ∗∗##
∗∗
∗∗##&
(c)
F : NACA inhibited CM-induced indicators of oxidative stress in kidney. Exposure to CM signicantly attenuated renal SOD (a) and
GSH levels and increased MDA (b) levels. Pretreatment with NAC or NACA blocked the CIN-induced changes. Figures are representative
of  to  rats in each group. e values are means ±SD (𝑛=5). 𝑝< 0.05 versus CON, ∗∗ 𝑝< 0.01 versus CON, ##𝑝< 0.01 versus CIN, and
&𝑝< 0.05 versus CIN+NAC.
versus CIN). However, the downregulated expressions of Tr x
were markedly blocked by NAC and NACA (𝑝 < 0.01 versus
CIN).
4. Discussion
Although the mechanisms underlying CIN have not been
fully claried, several studies have shown that ROS-induced
oxidative stress and direct cytotoxicity of contrast media are
important in the pathogenesis of the disease [, , , ].
Here, we conrm the importance of oxidative stress and
demonstrate that the novel antioxidant NACA, an amide
derivate of NAC with better tissue penetration, oers more
eective prevention against CIN. Furthermore, we show that
Txr/ASK signal act as the upstream modulator of the p
MAPK and present a potential drug target for prevention.
Toourknowledge,thisistherstreportdemonstratingthat
Trx/ASK signal are involved in CIN.
e oxidative stress induced in rats with CIN was evident
by drastically decreased levels of SOD and GSH, as well as
an increased level of MDA, which is consistent with our
previous data [, ] and other reports [, ]. e ndings
that NACA consistently exhibited better renoprotection than
NAC may be related to its better membrane penetration.
However, whether other mechanisms of action are involved
is not clear and needs further studies. Overall, the present
ndings conrmed again that oxidative stress is an important
factor in the pathogenesis of CIN.
Since there is signicant inconsistency in the guidelines
regarding the benet of NAC to reduce the risk for CIN [,
], large-scale, randomized clinical trials that are adequately
powered have been proposed to determine the eectiveness
of NAC for prevention of CIN. Our present ndings suggest
that consideration should be given to the use of NACA as a
result of its superior renoprotective function relative to NAC.
CIN, also called contrast-induced acute kidney injury
(CI-AKI), is typically dened by an increase in serum cre-
atinine aer intravascular administration of contrast media,
but serum creatinine is a late and insensitive indicator of AKI
[, –]. us, several additional biomarkers have been
investigated in order to improve both prediction and diag-
nosis of AKI. Our previous study rst reported that urinary
𝛾-glutamyl transpeptidase (UGGT) has good sensitivity in
early detection of contrast-induced acute renal injury and
thus in early diagnosing AKI []. UGGT as a potential early
diagnosticbiomarkerofAKIhasbeenconrmedinAKI
patients aer liver transplantation []. Furthermore, another
GSH-dependent enzyme present in large amount in liver,
urinary glutathione S-transferases (UGST), has recently been
Oxidative Medicine and Cellular Longevity
(a) (b) (c) (d) (e)
Area density of phospho-
0.0
0.1
0.2
0.3
p38 MAPK in kidney
Con CIN CIN+
NAC
CIN+
NACA1
CIN+
NACA2
∗∗
∗∗## ∗∗##
##&
(f)
P-p38 MAPK
p38 MAPK
43 kd
43 kd
Con CIN CIN+CIN+CIN+
NAC NACA1NACA2
(g)
Con CIN CIN+
NAC
CIN+
NACA1
CIN+
NACA2
∗∗
##
##
##&
Relative density (phospho-p38
0.0
0.2
0.4
0.6
0.8
1.0
1.2
MAPK/p38 MAPK)
(h)
F : Eects of NACA on p MAPK phosphorylation in kidney. IHC staining of phospho-p MAPK in CON (a), CIN (b), NAC+CIN
(c), NACA+CIN (d), and NACA+CIN (e) rats, respectively (gures are representative of  to  rats in each group). Note positive stained area
(yellow) of IHC staining (arrow). (f) Semiquantitative analysis of phosphor-p MAPK expression in kidneys with IHC. (g) Phospho-p
MAPK and total-p MAPK expressions by western blotting (𝑛=3each). (h) Relative densitometry analysis of the ratio of phospho-p
MAPK to total-p MAPK. Data are shown as means ±SD (𝑛=3each). 𝑝< 0.05 versus CON, ∗∗𝑝< 0.01 versus CON, ##𝑝< 0.01 versus
CIN, and &𝑝 < 0.05 versus CIN+NAC.
identied as a biomarker of AKI []. Subsequent studies
should address usefulness of these and other biomarkers in
terms of sensitivity and early detection of kidney injury.
Previous work from our laboratory and others has con-
rmed that contrast-induced apoptotic cell death via p
MAPK pathway is an important pathogenic mechanism in
CIN[,,,].ereisevidencethatpMAPKactivation
is associated with renal injury, which highlights p MAPK
pathway as an attractive therapeutic target. However, the
potential pathway by which p MAPK is activated in CIN
isstillnotwelldenedsofar.
SinceMAPKscouldberegulatedupstreambytheMAPK
kinase kinase kinase (MAPK) and p MAPK could act as
the target of ASK, we investigated the eect of contrast media
on activation of ASK, a member of MAPK family [, ].
e present data demonstrated increased ASK phosphory-
lation in CIN, which could be blocked by NACA and NAC.
Our present data indicated clearly that contrast media could
activate the stress/death signaling mitogen-activated protein
kinase (MAPK) phosphorylation cascade and thus conrmed
that ASK/p MAPK could be a potential drug target for
preventing CIN. e study of Ma et al. has also identied
ASK as a potential therapeutic target in renal brosis [].
Reduced Trx could be a direct inhibitor or negative
regulatorofASK,whileROSstimulationcoulddissociate
Trx from Trx/ASK complex and lead to ASK activation
andinturnresultinthephosphorylationofitsdownstream
substrate p MAPK [, ]. As mentioned above, we
previously reported that contrast media exposure directly
increased cellular oxidation and induced p AMPK/iNOS
Oxidative Medicine and Cellular Longevity
P-ASK 1
ASK1
155 kd
155 kd
Con CIN CIN+CIN+CIN+
NAC NACA1NACA2
(a)
Con CIN CIN+CIN+CIN+
NAC NACA1NACA2
Relative density (phospho-
0.0
0.2
0.4
0.6
0.8
1.0
ASK1/ASK1)
## ## ##&
∗∗
(b)
Con CIN CIN+CIN+CIN+
NAC NACA1NACA2
Trx1
Beta-actin
12 kd
(c)
Con CIN CIN+CIN+CIN+
NAC NACA1NACA2
∗∗##
∗∗##
∗∗##
∗∗
0.0
0.3
0.5
0.8
1.0
1.3
Relative density (Trx1/actin)
(d)
Con CIN CIN+CIN+CIN+
NAC NACA1NACA2
##
##
##
∗∗
0.00
0.25
0.50
0.75
1.00
1.25
Relative Trx1mRNA expression
(e)
F : NACA blocked CM-induced p MAPK activation by inhibiting ASK phosphorylation and upregulating Trx mRNA and protein
expression. (a) Phospho-ASK and ASK expressions by western blotting (𝑛=3each). (b) Relative densitometry analysis of the ratio of
phospho-ASK to ASK. (c) Trx expression by western blotting (𝑛=3each). (d) Relative densitometry analysis of the ratio of Trx to beta-
actin. (e) QPCR analysis for Trx mRNA in renal cortex. Data are shown as means ±SD (𝑛=3each). 𝑝< 0.05 versus CON, ∗∗ 𝑝< 0.01
versus CON, ## 𝑝< 0.01 versus CIN, and &𝑝< 0.05 versus CIN+NAC.
pathway mediated apoptosis in renal tubular cells in vitro
and vivo [, ]. Additionally, our present data further clearly
indicated that contrast media exposure downregulated Trx
and increased ASK phosphorylation in CIN rat model. us
we could speculate that the more complete signal mechanism
for CIN should include the following key steps (Figure );
rst, contrast medium exposure increased ROS production
of kidney, resulting in suppression of Trx, which lead to
Trx/ASK complex dissociation to facilitate the activation of
ASK. is results in the downstream activation of its sub-
strate p MAPK and an imbalance of pro- and antiapoptotic
members of the Bcl- family and nally induced apoptotic
cell death. Interestingly, such a pathologic process could be
eciently inhibited by NACA through its antioxidant activity
and by upregulating Trx.
5. Conclusion
Summarily, based on our present and previous studies, we
demonstrated that NACA is more eective than NAC in
preventing CIN both in vivo and in vitro and identied the
Oxidative Medicine and Cellular Longevity
Contrast media Iohexol
ASK1
Trx1
ASK1
FoxO1
p-ASK1
p-p38
Trx1
Cytoskeleton disorganization
(Ps externalization)
NACA
caspase 9
Pro-caspase 9
Pro-caspase 3
caspase 3
Mitochondria
Bcl-2
Mcl-1
Bax
Nucleus
ROS
iNOS
NO
Cyt c
F : Schematic diagram illustrates the signal mechanism for CIN and the renoprotection of NACA. Contrast media exposure increases
ROS production in kidney, resulting in suppression of Trx and thus dissociating the Trx/ASK complex to facilitate the activation of ASK
(phosphorylation). Subsequently, activated ASK results in the downstream activation of its substrate p MAPK, an imbalance of pro- and
antiapoptotic members of the Bcl- family, and nally induces apoptosis. NACA eciently inhibits such a pathologic process through its
antioxidant activity and by upregulating Trx. p, phosphorylation. Ps, phosphatidylserine.
underlying mechanisms including suppression of oxidative
stress, upregulating Trx and in turn inhibiting ASK/p
MAPK pathway, and preventing renal tubular cell apoptosis.
Competing Interests
e authors declare that there is no conict of interests
regarding the publication of this paper.
Acknowledgments
e authors would like to thank Dr. Glenn Goldstein for
providing N-acetylcysteine amide. is work was supported
by National Natural Science Foundation of China (Grants
 and ) and sponsored by Shanghai Pujiang
Program.
References
[] J. J. Keaney, C. M. Hannon, and P. T. Murray, “Contrast-induced
acute kidney injury: how much contrast is safe?” Nephrology
Dialysis Transplantation,vol.,no.,pp.,.
[]H.Li,C.Wang,C.Liu,R.Li,M.Zou,andG.Cheng,
Ecacyofshort-termstatintreatmentforthepreventionof
contrast-induced acute kidney injury in patients undergoing
coronary angiography/percutaneous coronary intervention: a
meta-analysis of  randomized controlled trials,American
Journal of Cardiovascular Drugs,vol.,no.,pp.,.
[]R.J.Owen,S.Hiremath,A.Myers,M.Fraser-Hill,andB.
J. Barrett, “Canadian association of radiologists consensus
guidelines for the prevention of contrast-induced nephropathy:
update ,Canadian Association of Radiologists Journal,vol.
,no.,pp.,.
[] R. Rear, R. M. Bell, and D. J. Hausenloy, “Contrast-induced
nephropathy following angiography and cardiac interventions,
Heart,vol.,no.,pp.,.
[] R. M. Bell, R. Rear, J. Cunningham, A. Dawnay, and D. M.
Yellon, “Eect of remote ischaemic conditioning on contrast-
induced nephropathy in patients undergoing elective coronary
angiography (ERICCIN): rationale and study design of a ran-
domised single-centre, double-blind placebo-controlled trial,
Clinical Research in Cardiology,vol.,no.,pp.,.
[] X.Gong,G.Celsi,K.Carlsson,S.Norgren,andM.Chen,“N-
acetylcysteine amide protects renal proximal tubular epithelial
cells against iohexol-induced apoptosis by blocking p MAPK
and iNOS signaling,American Journal of Nephrology,vol.,
no. , pp. –, .
[] X. Gong, Q. Wang, X. Tang et al., “Tetramethylpyrazine pre-
vents contrast-induced nephropathy by inhibiting p MAPK
and FoxO signaling pathways,American Journal of Nephrol-
ogy,vol.,no.,pp.,.
[] T. B. Sterenborg, T. P. Menting, Y. de Waal et al., “Remote
ischemic preconditioning to reduce contrast-induced nephrop-
athy: study protocol for a randomized controlled trial,Trial s ,
vol. , article , .
[]J.A.Neyra,S.Shah,R.Mooney,G.Jacobsen,J.Yee,andJ.
E. Novak, “Contrast-induced acute kidney injury following
coronary angiography: a cohort study of hospitalized patients
with or without chronic kidney disease,Nephrology Dialysis
Transplanta tio n,vol.,no.,pp.,.
 Oxidative Medicine and Cellular Longevity
[] J. Kooiman, Y. W. J. Sijpkens, J.-P. P. M. De Vries et al., “A
randomized comparison of -h sodium bicarbonate hydration
versus standard peri-procedural saline hydration in patients
with chronic kidney disease undergoing intravenous contrast-
enhanced computerized tomography,Nephrology Dialysis
Transplanta tio n,vol.,no.,pp.,.
[] N. Lameire, J. A. Kellum, P. Aspelin et al., “Contrast-induced
acute kidney injury and renal support for acute kidney injury:
a KDIGO summary (part ),Critical Care,vol.,no.,article
, .
[] K. A. Carver and D. Yang, “N-acetylcysteine amide protects
against oxidative stress–induced microparticle release from
human retinal pigment epithelial cells,Investigative Ophthal-
mology and Visual Science,vol.,no.,pp.,.
[] M. Paul, M. Hemshekhar, R. M. ushara et al., “Methotrexate
promotes platelet apoptosis via JNK-mediated mitochondrial
damage: alleviation by N-acetylcysteine and N-acetylcysteine
amide,PLoS ONE,vol.,no.,ArticleIDe,.
[]T.Benterud,M.B.Ystgaard,S.Manueldasetal.,“N-
acetylcysteine amide exerts possible neuroprotective eects in
newborn pigs aer perinatal asphyxia,Neonatology, vol. , no.
, pp. –, .
[] K. Sunitha, M. Hemshekhar, R. M. ushara et al., “N-
Acetylcysteine amide: a derivative to fulll the promises of N-
Acetylcysteine,Free Radical Research,vol.,no.,pp.,
.
[]X.Gong,G.Celsi,K.Carlsson,andS.Norgren,“Protective
eects of N-acetylcysteine amide (NACA) on gentamicin-
induced apoptosis in LLC-PK cells,Renal Failure,vol.,no.
, pp. –, .
[] M. Andreucci, G. Lucisano, T. Fagaet al., “Dierential activation
of signaling pathways involved in cell death, survival and
inammation by radiocontrast media in human renal proximal
tubular cells,Toxicological Sciences,vol.,no.,pp.,
.
[] C. Quintavalle, M. Brenca, F. De Micco et al., “In vivo and
in vitro assessment of pathways involved in contrast media-
induced renal cells apoptosis,Cell Death and Disease,vol.,
no. , article no. e, .
[] F.Y.Ma,G.H.Tesch,andD.J.Nikolic-Paterson,“ASK/p
signaling in renal tubular epithelial cells promotes renal brosis
in the mouse obstructed kidney,American Journal of Physiolog y
-RenalPhysiology,vol.,no.,pp.FF,.
[] F.Y.Ma,G.H.Tesch,andD.J.Nikolic-Paterson,“ASK/psig-
naling in renal tubular epithelial cells promotes renal brosis in
the mouse obstructed kidney,American Journal of Physiolog y—
Renal Physiology,vol.,no.,pp.FF,.
[] W. Hao, T. Takano, J. Guillemette, J. Papillon, G. Ren, and A.
V. Cybulsky, “Induction of apoptosis by the Ste-like kinase
SLK, a germinal center kinase that activates apoptosis signal-
regulating kinase and p,” Journal of Biological Chemistry,vol.
, no. , pp. –, .
[] R. M. Hashem, K. M. Hassanin, L. A. Rashed, M. O. Mah-
moud, and M. G. Hassan, “Eect of silibinin and vitamin
E on the ASK-p MAPK pathway in D-galactosamine/
lipopolysaccharide induced hepatotoxicity,Experimental Biol-
ogy and Medicine,vol.,no.,pp.,.
[] Q.-Q. Wu, M. Xu, Y. Yuan et al., “Cathepsin B deciency atten-
uates cardiac remodeling in response to pressure overload via
TNF-𝛼/ASK/JNK pathway,AmericanJournalofPhysiology
Heart and Circulatory Physiology,vol.,no.,pp.H
H, .
[] Y. Huang, L. Wu, J. Wu, Y. Li, and L. Hou, “Cellular FLICE-
like inhibitory protein protects against cardiac hypertrophy by
blocking ASK/p signaling in mice,Molecular and Cellular
Biochemistry,vol.,no.-,pp.,.
[] K. Shen, F. Lu, J. Xie et al., “Cambogin exerts anti-proliferative
and pro-apoptotic eects on breast adenocarcinoma through
the induction of NADPH oxidase  and the alteration of
mitochondrial morphology and dynamics,Oncotarget,vol.,
no. , pp. –, .
[] R. Jin, Y. Gao, S. Zhang et al., “Trx/TrxR system regulates post-
selected DP thymocytes survival by modulating ASK-JNK/p
MAPK activities,Immunology and Cell Biology,vol.,no.,
pp. –, .
[] Y. Agmon, H. Peleg, Z. Greenfeld, S. Rosen, and M. Brezis,
“Nitric oxide and prostanoids protect the renal outer medulla
from radiocontrast toxicity in the rat,Journal of Clinical
Investigation,vol.,no.,pp.,.
[] Y. Yokomaku, T. Sugimoto, S. Kume et al., “Asialoerythropoietin
prevents contrast-induced nephropathy,Journal of the Ameri-
can Society of Nephrology,vol.,no.,pp.,.
[] H. T. Lee, M. Jan, C. B. Soo et al., “A adenosine receptor knock-
out mice are protected against acute radiocontrast nephropathy
in vivo,American Journal of Physiology—Renal Physiology,vol.
, no. , pp. F–F, .
[]X.Gong,V.N.Ivanov,andT.K.Hei,“,,,-Tetrameth-
ylpyrazine (TMP) down-regulated arsenic-induced heme
oxygenase- and ARS expression by inhibiting Nrf, NF-𝜅B,
AP- and MAPK pathways in human proximal tubular cells,
Archives of Toxicology, vol. , no. , pp. –, .
[] X. Gong, V. N. Ivanov, M. M. Davidson, and T. K. Hei,
“Tetramethylpyrazine (TMP) protects against sodium arsenite-
induced nephrotoxicity by suppressing ROS production, mito-
chondrial dysfunction, pro-inammatory signaling pathways
and programed cell death,Archives of Toxicology,vol.,no.
,pp.,.
[] L. Tongqiang, L. Shaopeng, Y. Xiaofang et al., “Salvianolic acid
B prevents iodinated contrast media-induced acute renal injury
in rats via the PIK/Akt/Nrf pathway,Oxidative Medicine and
Cellular Longevity,vol.,ArticleID,pages,.
[] M. Buyuklu, F. M. Kandemir, M. Ozkaraca, T. Set, E. M.
Bakirci, and E. Topal, “Protective eect of curcumin against
contrast induced nephropathy in rat kidney: what is happening
to oxidative stress, inammation, autophagy and apoptosis?”
European Review for Medical and Pharmacological Sciences,vol.
,no.,pp.,.
[] A.Tasanarong,S.Kongkham,andA.Itharat,“Antioxidanteect
of Phyllanthus emblica extract prevents contrast-induced acute
kidney injury,BMC Complementary and Alternative Medicine,
vol. , article , .
[] N. Wang, R.-B. Wei, Q.-P. Li et al., “Renal protective eect of
probucol in rats with contrast-induced nephropathy and its
underlying mechanism,Medical Science Monitor,vol.,pp.
–, .
[] M. Lichosik, A. Jung, K. Jobs, A. Mierzejewska, R. Zdanowski,
and B. Kalicki, “Interleukin  and neutrophil-gelatinase
associated lipocalin in assessment of the risk of contrast-
induced nephropathy in children,Central European Journal of
Immunology,vol.,no.,pp.,.
[] G. F. Lehner, L. G. Forni, and M. Joannidis, “Oliguria and
biomarkers of acute kidney injury: star struck lovers or
strangers in the night?” Nephron,vol.,no.,pp.,
.
Oxidative Medicine and Cellular Longevity 
[] S. S. Waikar and J. V. Bonventre, “Creatinine kinetics and
the denition of acute kidney injury,Journal of the American
Society of Nephrology,vol.,no.,pp.,.
[] P. Marcelino, I. Tavares, D. Carvalho et al., “Is urinary
𝛾-glutamyl transpeptidase superior to urinary neutrophil
gelatinase-associated lipocalin for early prediction of acute
kidney injury aer liver transplantation?” Tra nsp lanta tio n Pro -
ceedings,vol.,no.,pp.,.
[] K. H. Shu, C. Wang, C. Wu et al., “Urinary 𝜋-glutathione S-
transferase predicts advanced acute kidney injury following
cardiovascular surgery,Scientic Reports,vol.,article,
.
[] A. Ray, N. Sehgal, S. Karunakaran, G. Rangarajan, and V.
Ravindranath, “MPTP activates ASK-p MAPK signaling
pathway through TNF-dependent Trx oxidation in parkinson-
ism mouse model,FreeRadicalBiologyandMedicine,vol.,
Article ID , pp. –, .
... However, accumulated reactive oxygen species ROS promote TXNIP binding to the endogenous antioxidant thioredoxin (Thioredoxin , TRX) and inhibiting the antioxidant activity of the protein by exchanging disul de bonds with Trx , thus participating in the pathogenesis of various diseases such as cancer, autoimmune diseases and diabetes (27). Thioredoxin 1 (Trx1) is localized in the cytoplasm, and reduced expression of Trx1 is a direct inhibitor or negative regulator of Apoptosis signalregulating kinase 1 (ASK1) (28, 29), which would activate ASK1, thereby promoting its downstream substrate p38 mitogen-activated protein kinase (p38 mitogen-activated protein kinase, p38 MAPK) phosphorylation, ultimately leading to the onset of apoptosis in renal tubular cells (30). Studies have shown that glucose can regulate the expression of TXNIP through histone acetylase, thus promoting the development of DKD (31). ...
... Previous evidences have been proved p38MAPK activation is related to renal injury, such as iohexolinduced (30,47,48) or diabetes-induced(49-52) apoptotic cell death. MAPK kinase (MAPK3k) acts as an upstream regulator of MAPKs. ...
... Apoptosis signal-regulating kinase 1 (ASK1) is a widely expressed redoxsensitive serine threonine kinase. Studies have shown that activation of ASK1 (as a member of the MAP3K family) can target its substrate p38 MAPK, which is involved in the development of renal brosis (53), glomerular injury (54), tubular epithelial cell apoptosis, and in ammation (30). Recently, a new study found a signi cant increase in the expression of ASK1 and P38 in patients' venous tissues in maintenance hemodialysis patients, and p38MAPK signaling pathway may be involved in failure of autogenous arteriovenous stula caused by stenosis (55). ...
Preprint
Full-text available
Background Contrast-induced acute kidney injury (CIAKI) is the third most common cause of hospital-acquired acute kidney injury. Diabetes mellitus (DM) is a major risk factor for CIAKI. Nevertheless, the mechanism of its role in diabetes susceptibility to CIAKI remains unclear. This study aimed to explore the role played by Histone deacetylase 9 (HDAC9) during the susceptibility of diabetic model to CIAKI. Methods Both in vitro and in vivo model of diabetes were induced by treating human renal tubular epithelial cells (HK-2) with high glucose (HG, 50mM) and by feeding mice with a high-fat diet (HFD) followed by intraperitoneal injection of streptozotocin (STZ), respectively. CIAKI mice models were constructed by contrast-media (iohexol), and iohexol also was treated HK-2 cells. Then, BRD-4354 (an inhibitor of HDAC9) was added into treated cells and mice. Finally, knockdown HDAC9 in HK-2 cultured with HG, and iohexol was added. The pathological changes, oxidative stress and apoptosis levels in mice kidney tissues were assessed. Meanwhile, cellular reactive oxygen species and the activity of HK-2 cells was measured. Western blot was used to determine the expression of HDAC9 and TXNIP/Trx1/P-ASK1/p38MAPK signaling pathways in cells and kidneys. Results HDAC9 was increased in both diabetic kidney tissues and HG-induced HK-2 cells. In vitro experiment indicated that HK-2 exposed to HG attenuated further damage to apoptosis and oxidative stress by iohexol via knockdown and inhibiting HDAC9. In vivo assay revealed that BRD-4354 reduced diabetic mice sensitivity to CI-AKI. Mechanically, HDAC9 could activate TXNIP/Trx1/P-ASK1/p38MAPK signaling pathway involving in the susceptibility of diabetes to CIAKI. Conclusion HDAC9 promotes the sensitivity of diabetes to CIAKI; and may be involved in oxidative stress and apoptosis through regulation of the TXNIP/Trx1/P-ASK1/p38MAPK pathway.
... However, accumulated reactive oxygen species ROS promote TXNIP binding to the endogenous antioxidant thioredoxin (Thioredoxin , TRX) and inhibiting the antioxidant activity of the protein by exchanging disul de bonds with Trx , thus participating in the pathogenesis of various diseases such as cancer, autoimmune diseases and diabetes (27). Thioredoxin 1 (Trx1) is localized in the cytoplasm, and reduced expression of Trx1 is a direct inhibitor or negative regulator of Apoptosis signalregulating kinase 1 (ASK1) (28, 29), which would activate ASK1, thereby promoting its downstream substrate p38 mitogen-activated protein kinase (p38 mitogen-activated protein kinase, p38 MAPK) phosphorylation, ultimately leading to the onset of apoptosis in renal tubular cells (30). Studies have shown that glucose can regulate the expression of TXNIP through histone acetylase, thus promoting the development of DKD (31). ...
... Previous evidences have been proved p38MAPK activation is related to renal injury, such as iohexolinduced (30,47,48) or diabetes-induced(49-52) apoptotic cell death. MAPK kinase (MAPK3k) acts as an upstream regulator of MAPKs. ...
... Apoptosis signal-regulating kinase 1 (ASK1) is a widely expressed redoxsensitive serine threonine kinase. Studies have shown that activation of ASK1 (as a member of the MAP3K family) can target its substrate p38 MAPK, which is involved in the development of renal brosis (53), glomerular injury (54), tubular epithelial cell apoptosis, and in ammation (30). Recently, a new study found a signi cant increase in the expression of ASK1 and P38 in patients' venous tissues in maintenance hemodialysis patients, and p38MAPK signaling pathway may be involved in failure of autogenous arteriovenous stula caused by stenosis (55). ...
Preprint
Full-text available
Background Contrast-induced acute kidney injury (CIAKI) is the third most common cause of hospital-acquired AKI. Diabetes mellitus (DM) is a major risk factor for CIAKI. Nevertheless, the mechanism of its role in diabetes susceptibility to CIAKI remains unclear. This study aimed to explore the role played by HDAC9 during the susceptibility of diabetic model to CIAKI. Methods Both in vitro and in vivo model of diabetes were induced by treating human renal tubular epithelial cells (HK-2) with high glucose (HG, 50mM) and by feeding mice with a high-fat diet (HFD) followed by intraperitoneal injection of streptozotocin (STZ), respectively. CIAKI mice models were constructed by contrast-media (iohexol), and iohexol also was treated HK-2 cells. Then, BRD-4354 (an inhibitor of HDAC9) was added into treated cells and mice. Finally, knockdown HDAC9 in HK-2 cultured with HG, and iohexol was added. The pathological changes, oxidative stress and apoptosis levels in mice kidney tissues were assessed. Meanwhile, cellular reactive oxygen species and the activity of HK-2 cells was measured. Western blot was used to determine the expression of HDAC9 and TXNIP/Trx1/P-ASK1/p38MAPK signaling pathways in cells and kidneys. Results HDAC9 was increased in both diabetic kidney tissues and HGinduced HK-2 cells. In vitro experiment indicated that HK-2 exposed to HG attenuated further damage to apoptosis and oxidative stress by iohexol via knockdown and inhibiting HDAC9. In vivo assay revealed that BRD4354 reduced diabetic mice sensitivity to CI-AKI. Mechanically, HDAC9 could activate TXNIP/Trx1/P-ASK1/p38MAPK signaling pathway involving in the susceptibility of diabetes to CIAKI. Conclusion HDAC9 promotes the sensitivity of diabetes to CIAKI; and may be involved in oxidative stress and apoptosis through regulation of the TXNIP/Trx1/P-ASK1/p38MAPK pathway.
... Oxidative stress is an important driving mechanism of CIN and is the pathological basis of antioxidant therapy for CIN [18]. In previous studies, we demonstrated the involvement of ROS in localized renal injury in a CIN model, indicated by a dramatic decrease in SOD and GSH and an increase in MDA in renal tissues [19]. Therefore, we hypothesize that the alleviation of CIN renal injury by TMP is accompanied by an attenuation of lipid peroxidation. ...
Article
Full-text available
Contrast-induced nephropathy (CIN) is a leading cause of hospital-acquired acute kidney injury (AKI). Recently ferroptosis was reported to be crucial for AKI pathogenesis. Our previous studies indicated antioxidant tetramethylpyrazine (TMP) prevent CIN in vivo. However, whether ferroptosis is involved in TMP nephroprotective mechanism against CIN is unclear. In present study we investigated the role of renal tubular epithelial cell ferroptosis in TMP reno-protective effect against CIN and the molecular mechanisms by which TMP regulates ferroptosis. Classical contrast-medium Iohexol was used to construct CIN models in rats and HK-2 cells. Results showed tubular cell injury was accompanied by ferroptosis both in vivo and in vitro, including the typical features of ferroptosis, Fe2+ accumulation, lipid peroxidation and decreased glutathione peroxidase 4 (GPX4). Ferroptosis inhibition by classic inhibitors Fer-1 and DFO promoted cell viability and reduced intracellular ROS production. Additionally, TMP significantly inhibited renal dysfunction, reduced AKI biomarkers, prevented ROS production, inhibited renal Fe2+ accumulation and increased GPX4 expression. Expressions of various proteins associated with iron ion metabolism, including transferrin receptor (TFRC), divalent metal transporter 1, iron-responsive element binding protein 2, ferritin heavy chain 1, ferroportin 1, and heat shock factor binding protein 1, were examined using mechanistic analyses. Among these, TFRC changes were the most significant after TMP pretreatment. Results of siRNA knockdown and plasmid overexpression of TFRC indicated TFRC is essential for TMP to alleviate ferroptosis and reduce LDH release, Fe2+ accumulation and intracellular ROS. Our findings provide crucial insights about the potential of TMP in treating AKI associated with ferroptosis.
... Meanwhile, 8) NAC can reduce tubular cell injury caused by contrast media, which can help prevent CI-AKI. Finally, 9) NAC can reduce renal tubular cell apoptosis caused by contrast media, which can help prevent CI-AKI (13)(14)(15)(16)(17). ...
Article
Contrast-induced acute kidney injury (CI-AKI) is a potential complication of medical imaging procedures that use contrast media. It is important to identify and manage risk factors for contrast-induced nephropathy and to monitor patients for signs of renal damage after contrast administration. N-acetylcysteine (NAC) can prevent CI-AKI through multiple mechanisms of action, including reducing oxidative stress, improving renal hemodynamics, reducing inflammation, reducing apoptosis and fibrosis, reducing oxidative stress-induced DNA damage, reducing tubular cell injury, and reducing renal tubular cell apoptosis. However, the exact mechanisms of action may vary based on the specific study or context. Further research is needed to fully elucidate the molecular mechanisms of NAC in preventing CI-AKI.
... With the advancement of clinical treatment for CIN, drugs such as N-acetylcysteine (NAC), 7 vitamin C 8 and statins 9 have been utilized. However, there is no strong evidence to prove that any drug can reduce the occurrence of CIN. ...
Article
Full-text available
Contrast‐induced nephropathy (CIN) is a condition that causes kidney damage in patients receiving angiography with iodine‐based contrast agents. This study investigated the potential protective effects of berberine (BBR) against CIN and its underlying mechanisms. The researchers conducted both in vivo and in vitro experiments to explore BBR's renal protective effects. In the in vivo experiments, SD rats were used to create a CIN model, and different groups were established. The results showed that CIN model group exhibited impaired renal function, severe damage to renal tubular cells and increased apoptosis and ferroptosis. However, BBR treatment group demonstrated improved renal function, decreased apoptosis and ferroptosis. Similar results were observed in the in vitro experiments using HK‐2 cells. BBR reduced ioversol‐induced apoptosis and ferroptosis, and exerted its protective effects through Akt/Foxo3a/Nrf2 signalling pathway. BBR administration increased the expression of Foxo3a and Nrf2 while decreasing the levels of p‐Akt and p‐Foxo3a. In conclusion, this study revealed that BBR effectively inhibited ioversol‐induced apoptosis and ferroptosis in vivo and in vitro. The protective effects of BBR were mediated through the modulation of Akt/Foxo3a/Nrf2 signalling pathway, leading to the alleviation of CIN. These findings suggest that BBR may have therapeutic potential for protecting against CIN in patients undergoing angiography with iodine‐based contrast agents.
... Previous studies have reported that NAC, a precursor of GSH, inhibits the activation of p38 [24,25], suggesting that GSH acts on upstream signaling pathways for p38 inhibition. Additionally, transaldolase deficiency in mouse livers was reported to result in GSH depletion [9]. ...
Article
Full-text available
In atherosclerosis, macrophage-derived foam cell formation is considered to be a hallmark of the pathological process; this occurs via the uptake of modified lipoproteins. In the present study, we aim to determine the role of transaldolase in foam cell formation and atherogenesis and reveal the mechanisms underlying its role. Bone marrow-derived macrophages (BMDMs) isolated from mice successfully form foam cells after treatment with oxidized low-density lipoprotein (80 μg/mL). Elevated transaldolase levels in the foam cell model are assessed by quantitative polymerase chain reaction and western blot analysis. Transaldolase overexpression and knockdown in BMDMs are achieved via plasmid transfection and small interfering RNA technology, respectively. We find that transaldolase overexpression effectively attenuates, whereas transaldolase knockdown accelerates, macrophage-derived foam cell formation through the inhibition or activation of cholesterol uptake mediated by the scavenger receptor cluster of differentiation 36 (CD36) in a p38 mitogen-activated protein kinase (MAPK) signaling-dependent manner. Transaldolase-mediated glutathione (GSH) homeostasis is identified as the upstream regulator of p38 MAPK-mediated CD36-dependent cholesterol uptake in BMDMs. Transaldolase upregulates GSH production, thereby suppressing p38 activity and reducing the CD36 level, ultimately preventing foam cell formation and atherosclerosis. Thus, our findings indicate that the transaldolase-GSH-p38-CD36 axis may represent a promising therapeutic target for atherosclerosis.
... This in vitro cell model with CM being removed by medium replacement may also be useful for exploring potential treatments for CIN since previous studies only focused on the effect of medication compared to no treatment [27][28][29][30][31][32]. However, as the standard management in clinical settings, hydration has seldom been used as a control group in laboratory studies [11,[33][34][35][36]. CIN was initially regarded as a spontaneous recovery process among healthy people. ...
Article
Full-text available
Contrast-induced nephropathy (CIN) is one of the most common causes of acute kidney injury (AKI). However, management is still limited, and the cellular response to radiocontrast removal for CIN remains unclear. This study aimed to explore the latent effects of iohexol in cultured renal tubular cells with or without the removal of iohexol by medium replacement. HK2 renal tubular cells were subcultured 24 h before use in CIN experiments. Three treatment groups were established: the control, a radiocontrast (iohexol)-only group at 75 mg I/mL (I-75), and iohexol exposure for 24 h with culture medium replacement (I-75/M). Cell cycle arrest, fibrogenic mediator assays, cell viability, cell function, and cell-cycle-related protein expression were compared between groups. Iohexol induced numerous changes in HK2 renal tubular cells, such as enlarged cell shape, cell cycle arrest, increased apoptosis, and polyploidy. Iohexol inhibited the expression of cyclins, CDKs, ZO-1, and E-cadherin but conversely enhanced the expression of p21 and fibrosis-related genes, including TGF-β1, CTGF, collagen I, collagen III, and HIF-1α within 60 hr after the exposure. Except for the recovery from cell cycle arrest and cell cycle gene expression, notably, the removal of iohexol by medium replacement could not fully recover the renal tubular cells from the formation of polyploid cells, the adhesion or spreading, or the expression of fibrosis-related genes. The present study demonstrates, for the first time, that iohexol exerts latent cytotoxic effects on cultured renal tubular cells after its removal, suggesting that these irreversible cell changes may cause the insufficiency of radiocontrast reduction in CIN, which is worth investigating further.
... Chlorogenic acid (CGA) is a phenolic compound widely found in fruits, vegetables, coffee, and tea, and it also has well-characterized antioxidant and anti-inflammatory properties [34,35]. Indeed, the antioxidant and anti-inflammatory properties of NAC [36,37] and CGA [38,39] have been demonstrated to promote nephroprotection. Few studies have recently linked the inflammasome pathway to their hepatoprotective effect [40,41]. ...
Article
Full-text available
Background: Cisplatin (Cp) is an antineoplastic agent with a dose-limiting nephrotoxicity. Cp-induced nephrotoxicity is characterized by the interplay of oxidative stress, inflammation, and apoptosis. Toll-4 receptors (TLR4) and NLPR3 inflammasome are pattern-recognition receptors responsible for activating inflammatory responses and are assigned to play a significant role with gasdermin (GSDMD) in acute kidney injuries. N-acetylcysteine (NAC) and chlorogenic acid (CGA) have documented nephroprotective effects by suppressing oxidative and inflammatory pathways. Therefore, the current study aimed to investigate the contribution of the upregulation of TLR4/inflammasomes/gasdermin signaling to Cp-induced nephrotoxicity and their modulation by NAC or CGA. Methods: A single injection of Cp (7 mg/kg, i.p.) was given to Wistar rats. Rats received either NAC (250 mg/kg, p.o.) and/or CGA (20 mg/kg, p.o.) one week before and after the Cp injection. Results: Cp-induced acute nephrotoxicity was evident by the increased blood urea nitrogen and serum creatinine and histopathological insults. Additionally, nephrotoxicity was associated with increased lipid peroxidation, reduced antioxidants, and elevated levels of inflammatory markers (NF-κB and TNF-α) in the kidney tissues. Moreover, Cp upregulated both TLR4/NLPR3/interleukin-1beta (IL-1β) and caspase-1/GSDMD-signaling pathways, accompanied by an increased Bax/BCL-2 ratio, indicating an inflammatory-mediated apoptosis. Both NAC and/or CGA significantly corrected these changes. Conclusions: This study emphasizes that inhibition of TLR4/NLPR3/IL-1β/GSDMD might be a novel mechanism of the nephroprotective effects of NAC or CGA against Cp-induced nephrotoxicity in rats.
... However, with the development of research in the field of pharmacology, NAC which acts as an antioxidant by reducing or even preventing oxidative stress and as a scavanger of reactive oxygen species is also used to protect kidney function and is used in the field of cardiology (Samuni et al., 2013;Pedre et al., 2021). Based on a study conducted on animals, present a form of NAC amide (NACA) which has better activity as a renoprotective agent when compared to NAC itself by upregulating thioredoxin-1 and inhibition of signal-regulating kinase 1 (ASK1) which is an activator of the p38MAPK pathway in inhibiting apoptosis of renal cells, consequently it can significantly reduce serum creatinine elevation, blood urea nitrogen, biomarkers of AKI, and prevent histologic changes resulting from renal tubular injuries (Gong et al., 2016). A pharmacokinetic study of NAC, showed that there was a significant decrease in NAC clearance of up to 90% when oral NAC was administered to patients with end-stage renal disease (ESRD), although plasma NAC levels increased dose-related (Nolin et al., 2010). ...
Article
Full-text available
The incidence of acute kidney injury (AKI) post-cardiopulmonary bypass (CPB) can cause an increase in the rate of renal replacement therapy (RRT) and mortality rate. Compared to brain and liver damage post-CPB, AKI has the highest incidence of 83%. Based on this phenomenon, various efforts have been made to reduce the incidence of AKI post-CPB, both pharmacologically and non-pharmacologically interventions. The purpose of this review is to emphasize several renal protector agents which under optimal conditions can provide significant benefits in reducing the incidence of AKI post-CPB. This article was obtained by conducting a study on several kinds of literature, including the original article, RCT study, systematic review and meta-analysis, and other review articles. There are five renal protector agents that are the focus of this article, those are fenoldopam which effectively works to prevent the incidence of AKI post-CPB, while furosemide has shown satisfactory results in patients with decreased renal function when administered in the Renal Guard (RG) system, mannitol, and nitric oxide, both of these can also effectively reduce the incidence of AKI post‐CPB by controlling its blood concentration and timing of administration, and another form of N-Acetylcysteine, namely N‐Acetylcysteine amide has better activity as a renoprotective agent than N‐Acetylcysteine itself. The benefits of these agents can be obtained by developing devices that can control drug levels in the blood and create optimal conditions for drugs during the use of a CPB machine.
Article
Trimethylamine N-oxide (TMAO) is associated with overall mortality in patients with chronic kidney disease (CKD). Previous findings suggest that P. frutescens (L.) can alleviate renal injury, but its effects and mechanisms underlying alleviation of TMAO-induced kidney damage remain unclear. In this study, a TMAO injury model, in vivo and in vitro, was established to clarify the effects and mechanisms of P. frutescens in alleviating TMAO-induced kidney injury. The results show that TMAO (60 mM/L) can induce the activation of apoptosis signal-regulating kinase 1 (ASK1)-c-Jun N-terminal kinase (JNK), thus aggravating downstream cell apoptosis in vitro. The study also found that P. frutescens aqueous extract (PFAE) (5 mg/mL) can inhibit TMAO-induced apoptosis by downregulating ASK1-JNK phosphorylation. In the in vivo experiments, it was demonstrated that TMAO can increase the levels of blood urea nitrogen and cystatin C, aggravating renal tubular epithelial apoptosis. The results also show that PFAE can reduce TMAO-induced renal damage by inhibiting ASK1-JNK phosphorylation in vivo. Our findings confirmed that P. frutescens can alleviate TMAO-induced renal tubule apoptosis by regulating ASK1-JNK phosphorylation, indicating that P. frutescens may be an effective treatment for alleviating TMAO damage in CKD.
Article
Full-text available
Urinary biomarkers augment the diagnosis of acute kidney injury (AKI), with AKI after cardiovascular surgeries being a prototype of prognosis scenario. Glutathione S-transferases (GST) were evaluated as biomarkers of AKI. Urine samples were collected in 141 cardiovascular surgical patients and analyzed for urinary alpha-(α-) and pi-(π-) GSTs. The outcomes of advanced AKI (KDIGO stage 2, 3) and all-cause in-patient mortality, as composite outcome, were recorded. Areas under the receiver operator characteristic (ROC) curves and multivariate generalized additive model (GAM) were applied to predict outcomes. Thirty-eight (26.9%) patients had AKI, while 12 (8.5%) were with advanced AKI. Urinary π-GST differentiated patients with/without advanced AKI or composite outcome after surgery (p < 0.05 by generalized estimating equation). Urinary π-GST predicted advanced AKI at 3 hrs post-surgery (p = 0.033) and composite outcome (p = 0.009), while the corresponding ROC curve had AUC of 0.784 and 0.783. Using GAM, the cutoff value of 14.7 μg/L for π-GST showed the best performance to predict composite outcome. The addition of π-GST to the SOFA score improved risk stratification (total net reclassification index = 0.47). Thus, urinary π-GST levels predict advanced AKI or hospital mortality after cardiovascular surgery and improve in SOFA outcome assessment specific to AKI.
Article
Full-text available
Cambogin, a bioactive polycyclic polyprenylated acylphoroglucinol (PPAP) derived from the Garcinia genus, possesses proapoptotic effect in medulloblastoma and breast cancer cells. We have previously demonstrated that the proapoptotic effect of cambogin is driven by the production of reactive oxygen species (ROS). Here we have shown that the inhibitory effect of cambogin on cell proliferation is associated with the loss of mitochondrial transmembrane potential (ΔΨm) and mitochondrial fragmentation. Cambogin also promotes the mutual complex formation of the membrane-bound subunit p22phox of NADPH oxidase 1 (NOX1), as well as the phosphorylation of the cytosolic subunit p47phox, subsequently enhancing membrane-bound NOX1 activity, which leads to increases in intracellular and mitochondrial levels of O2.- and H2O2. Pharmacological inhibition of NOX1 using apocynin (pan-NOX inhibitor), ML171 (NOX1 inhibitor) or siRNA against NOX1 prevents the increases in O2.- and H2O2 levels and the anti-proliferative effect of cambogin. Antioxidants, including SOD (superoxide dismutase), CAT (catalase) and EUK-8, are also able to restore cell viability in the presence of cambogin. Besides, cambogin increases the dissociation of thioredoxin-1 (Trx1) from ASK1, switching the inactive form of ASK1 to the active kinase, subsequently leads to the phosphorylation of JNK/SAPK, which is abolished upon ML171 treatment. The proapoptotic effect of cambogin in breast cancer cells is also aggravated upon knocking down Trx1 in MCF-7 cells. Taken in conjunction, these data indicate that the anti-proliferative and pro-apoptotic effect of cambogin is mediated via inducing NOX1-dependent ROS production and the dissociation of ASK1 and Trx1.
Article
Full-text available
Contrast-induced acute renal injury (CI-AKI) has become a common cause of hospital-acquired renal failure. However, the development of prophylaxis strategies and approved therapies for CI-AKI is limited. Salvianolic acid B (SB) can treat cardiovascular-related diseases. The aim of the present study was to assess the effect of SB on prevention of CI-AKI and explore its underlying mechanisms. We examined its effectiveness of preventing renal injury in a novel CI-AKI rat model. Compared with saline, intravenous SB pretreatment significantly attenuated elevations in serum creatinine and the histological changes of renal tubular injuries, reduced the number of apoptosis-positive tubular cells, activated Nrf2, and lowered the levels of renal oxidative stress induced by iodinated contrast media. The above renoprotection of SB was abolished by the PI3K inhibitor (wortmannin). In HK-2 cells, SB activated Nrf2 and decreased the levels of oxidative stress induced by hydrogen peroxide and subsequently improved cell viability. The above cytoprotection of SB was blocked by the PI3K inhibitor (wortmannin) or siNrf2. Thus, our results demonstrate that, due to its antioxidant properties, SB has the potential to effectively prevent CI-AKI via the PI3K/Akt/Nrf2 pathway.
Article
Full-text available
CIN represents a significant clinical and health economic problem that may be under-recognised through limitations in the currently available biomarkers. Although often a transient injury, CIN may progress to significant persistent renal impairment, ESRF and adverse cardiovascular outcomes. There are a number of recognised risk factors, although the prediction of CIN, particularly prior to contrast administration, remains challenging. Current interventions are largely centred on the avoidance of dehydration, the withdrawal of nephrotoxic agents and minimisation of contrast load, which has limited efficacy in preventing CIN in vulnerable patients. The unmet clinical need in CIN therefore resides in accurate prediction, effective intervention and rapid detection to prevent adverse cardiorenal outcomes. Each of these areas, particularly predictive risk scoring systems, innovative pharmacological and mechanical interventions and novel biomarkers are currently the subject of intensive research and development that may lead to the future development effective strategies to mitigate the risk of CIN.
Article
Full-text available
Abstract Purpose: Oxidative stress is a major factor involved in retinal pigment epithelium (RPE) apoptosis that underlies AMD. Drusen, extracellular lipid- and protein-containing deposits, are strongly associated with the development of AMD. Cell-derived microparticles (MPs) are small membrane-bound vesicles shed from cells. The purpose of this study was to determine if oxidative stress drives MP release from RPE cells, to assess whether these MPs carry membrane complement regulatory proteins (mCRPs: CD46, CD55, and CD59), and to evaluate the effects of a thiol antioxidant on oxidative stress–induced MP release. Methods: Retinal pigment epithelium cells isolated from human donor eyes were cultured and treated with hydrogen peroxide (H2O2) to induce oxidative stress. Isolated MPs were fixed for transmission electron microscopy or processed for component analysis by flow cytometry, Western blot analysis, and confocal microscopy. Results: Transmission electron microscopy showed that MPs ranged in diameter from 100 to 1000 nm. H2O2 treatment led to time- and dose-dependent elevations in MPs with externalized phosphatidylserine and phosphatidylethanolamine, known markers of MPs. These increases were strongly correlated to RPE apoptosis. Oxidative stress significantly increased the release of mCRP-positive MPs, which were prevented by a thiol antioxidant, N-acetylcysteine amide (NACA). Conclusions: This is the first evidence that oxidative stress induces cultured human RPE cells to release MPs that carry mCRPs on their surface. The levels of released MPs are strongly correlated with RPE apoptosis. N-acetylcysteine amide prevents oxidative stress–induced effects. Our findings indicate that oxidative stress reduces mCRPs on the RPE surface through releasing MPs.
Article
Full-text available
Purpose Oxidative stress is a major factor involved in retinal pigment epithelium (RPE) apoptosis that underlies AMD. Drusen, extracellular lipid- and protein-containing deposits, are strongly associated with the development of AMD. Cell-derived microparticles (MPs) are small membrane-bound vesicles shed from cells. The purpose of this study was to determine if oxidative stress drives MP release from RPE cells, to assess whether these MPs carry membrane complement regulatory proteins (mCRPs: CD46, CD55, and CD59), and to evaluate the effects of a thiol antioxidant on oxidative stress–induced MP release. Methods Retinal pigment epithelium cells isolated from human donor eyes were cultured and treated with hydrogen peroxide (H2O2) to induce oxidative stress. Isolated MPs were fixed for transmission electron microscopy or processed for component analysis by flow cytometry, Western blot analysis, and confocal microscopy. Results Transmission electron microscopy showed that MPs ranged in diameter from 100 to 1000 nm. H2O2 treatment led to time- and dose-dependent elevations in MPs with externalized phosphatidylserine and phosphatidylethanolamine, known markers of MPs. These increases were strongly correlated to RPE apoptosis. Oxidative stress significantly increased the release of mCRP-positive MPs, which were prevented by a thiol antioxidant, N-acetylcysteine amide (NACA). Conclusions This is the first evidence that oxidative stress induces cultured human RPE cells to release MPs that carry mCRPs on their surface. The levels of released MPs are strongly correlated with RPE apoptosis. N-acetylcysteine amide prevents oxidative stress–induced effects. Our findings indicate that oxidative stress reduces mCRPs on the RPE surface through releasing MPs.
Article
Oliguria is a common phenomenon that is found in hospitalized patients . Although a rapid reduction in urine excretion rate may reflect a precipitous fall in the glomerular filtration rate, in many cases it may not. Given the common physiological finding of oliguria, we explore the relationship between the functional biomarker of renal injury (oliguria) with the increasing number of markers of renal injury to see if this combination may aid in risk stratification.
Article
Background: Perinatal asphyxia and ensuing reoxygenation change the antioxidant capacity of cells and organs. Objectives: To analyze the neuroprotective effect of the antioxidant N-acetylcysteine amide (NACA) after perinatal hypoxia-reoxygenation with an emphasis on proinflammatory cytokines and the transcription factor NF-κB in the prefrontal cortex of neonatal pigs. Methods: Twenty-nine newborn pigs, aged 12-36 h, were subjected to global hypoxia and hypercapnia. One sham-operated group (n = 5) and 2 experimental groups (n = 12) were exposed to 8% oxygen, until the base excess was -20 mmol/l or the mean arterial blood pressure fell to <20 mm Hg (asphyxia with NACA or saline). The pigs were observed for 9.5 h after hypoxia. Samples of prefrontal cortex and plasma were analyzed. Results: Cortex: there was no significant difference in mRNA expression between the intervention groups regarding IL-1β, IL6, TNFα, MMP2, MMP9 or IL18. Pigs exposed to hypoxia-reoxygenation and treatment with NACA (NACA-pigs) had a significantly lower protein concentration of IL-1β than pigs treated with saline (placebo controls), i.e. 8.8 ± 3.9 versus 16.8 ± 10.5 pg/mg protein (p = 0.02). The activation of the transcription factor NF-κB (measured as the fold-change of phosphorylated p65Ser 536), was reduced in the NACA-pigs when compared to the placebo controls (5.2 ± 4.3 vs. 16.0 ± 13.5; p = 0.02). No difference between the intervention groups regarding brain histopathology or in the levels of 8-oxoguanine measured in the prefrontal cortex were observed. Plasma: the NACA-pigs had a stronger reduction of TNFα in the first 30 min following asphyxia compared with the placebo controls, i.e. 36 (30-44) versus 24 (14-32)% (p = 0.01). Conclusion: The reduced levels of the pivotal inflammatory markers IL-1β and TNFα and the transcription factor NF-κB may indicate that NACA has possible neuroprotective effects after perinatal asphyxia.
Article
Apoptosis signal-regulating kinase 1 (ASK1), a redox-sensor mitogen-activated protein kinase kinase kinase (MAPKKK) that activates p38 MAPK pathways in oxidative stress-induced hepatotoxicity in D-galactosamine/lipopolysaccharide (D-GalN/LPS) model, is a key central pathway in which specific targeting of ASK1 deactivation is of a great therapeutic potential. We tested the effect of silibinin and vitamin E in curative and prophylactic manner of treatment on the negative modulators of ASK1, thioredoxin1 (Trx1), thioredoxin reductase1 (TrxR1), and the protein phosphatase (PP5), whereas they have previously proven to have hepatoprotective effect. Either curative or prophylactic silibinin and vitamin E groups significantly decreased ASK1 and p38 MAPK levels through increasing the gene expression of Trx1, TrxR1, and PP5 to reduce the oxidative stress as demonstrated by decreasing the levels of NADPH oxidase 4 (NOX4), TBARS and conjugated diene with a concomitant increase in the levels of GSH, CAT, and SOD. These results were confirmed by histopathology examination which illustrated progressive degenerative changes of hepatocytes such as hydropic degeneration, vacuolation, pyknosis, karyolysis, and loss of architecture of some cells in D-GalN/LPS treatment, and these features were alleviated with silibinin and vitamin E administration. In conclusion, silibinin and vitamin E decreased ASK1-p38 MAPK pathway through deactivating the upstream signalling ASK1 molecule via increasing the levels of Trx1 and TrxR1 as well as the PP5 to alleviate in D-GalN/LPS induced hepatotoxicity.
Article
Background The results of previous studies have been contradictory in terms of the efficacy of statin treatment in preventing contrast-induced acute kidney injury (CI-AKI) and clinical adverse events (AEs). Objective This meta-analysis was undertaken to assess the role of short-term statin treatment in the prevention of CI-AKI and clinical AEs. Methods We searched the Cochrane Library, EMBASE, and PubMed databases for randomized controlled trials (RCTs) with the development of CI-AKI as a primary outcome. Secondary outcomes were the post-procedural serum creatinine (SCr) level, estimated glomerular filtration rate (eGFR), and development of AEs. We also performed prespecified subgroup analyses. Results A total of 21 RCTs involving 7746 patients were included. Short-term statin treatment significantly reduced the risk of CI-AKI [risk ratio (RR) 0.57; 95 % confident interval (CI) 0.47–0.69; p < 0.00001) and was associated with a lower post-procedural SCr level and a higher eGFR. High-dose statins resulted in a lower incidence of CI-AKI than the lower-dose statins. In addition, the benefit was seen across various subgroups for patients at risk of CI-AKI, statin-naïve patients, and East Asians, regardless of statin type, definition of CI-AKI, use of N-acetylcysteine (NAC) and hydration, and osmolality of contrast. However, there was no significant difference between the two groups in terms of the incidence of AEs. Conclusions The meta-analysis suggests that short-term statin treatment can effectively prevent CI-AKI, and the benefit is also observed in high-risk patients, statin-naïve patients, and an East Asian population. However, the effect of simvastatin for the prevention of CI-AKI, of statins for the prevention of AEs, and whether high-dose statins have a better effect than lower-dose statins are all still uncertain.