ArticlePDF Available

Rhabdomyolysis-Induced AKI Was Ameliorated in NLRP3 KO Mice via Alleviation of Mitochondrial Lipid Peroxidation in Renal Tubular Cells

Authors:

Abstract and Figures

Introduction: A recent study showed that early renal tubular injury is ameliorated in Nod-like receptor pyrin domain-containing protein 3 (NLRP3) KO mice with rhabdomyolysis-induced acute kidney injury (RIAKI). However, the precise mechanism has not been determined. Therefore, we investigated the role of NLRP3 in renal tubular cells in RIAKI. Methods: Glycerol-mediated RIAKI was induced in NLRP3 KO and wild-type (WT) mice. The mice were euthanized 24 h after glycerol injection, and both kidneys and plasma were collected. HKC-8 cells were treated with ferrous myoglobin to mimic a rhabdomyolytic environment. Results: Glycerol injection led to increase serum creatinine, aspartate aminotransferase (AST), and renal kidney injury molecule-1 (KIM-1) level; renal tubular necrosis; and apoptosis. Renal injury was attenuated in NLRP3 KO mice, while muscle damage and renal neutrophil recruitment did not differ between NLRP3 KO mice and WT mice. Following glycerin injection, increases in cleaved caspase-3, poly (ADP-ribose) polymerase (PARP), and a decrease in the glutathione peroxidase 4 (GPX-4) level were observed in the kidneys of mice with RIAKI, and these changes were alleviated in the kidneys of NLRP3 KO mice. NLRP3 was upregulated, and cell viability was suppressed in HKC-8 cells treated with ferrous myoglobin. Myoglobin-induced apoptosis and lipid peroxidation were significantly decreased in siNLRP3-treated HKC-8 cells compared to ferrous myoglobin-treated HKC-8 cells. Myoglobin reduced the mitochondrial membrane potential and increased mitochondrial fission and reactive oxygen species (ROS) and lipid peroxidation levels, which were restored to normal levels in NLRP3-depleted HKC-8 cells. Conclusions: NLRP3 depletion ameliorated renal tubular injury in a murine glycerol-induced acute kidney injury (AKI) model. A lack of NLRP3 improved tubular cell viability via attenuation of myoglobin-induced mitochondrial injury and lipid peroxidation, which might be the critical factor in protecting the kidney.
Content may be subject to copyright.
International Journal of
Molecular Sciences
Article
Rhabdomyolysis-Induced AKI Was Ameliorated in
NLRP3 KO Mice via Alleviation of Mitochondrial
Lipid Peroxidation in Renal Tubular Cells
Seok Jong Song , Su-mi Kim , Sang-ho Lee, Ju-Young Moon, Hyeon Seok Hwang, Jin Sug Kim,
Seon-Hwa Park, Kyung Hwan Jeong * and Yang Gyun Kim *
Division of Nephrology, Department of Internal Medicine, Kyung Hee University College of Medicine,
Seoul 05278, Korea; testwar-01@hanmail.net (S.J.S.); miya26@nate.com (S.-m.K.); lshkidney@khu.ac.kr (S.-h.L.);
kidmjy@hanmail.net (J.-Y.M.); hwanghsne@gmail.com (H.S.H.); jinsuk0902@hanmail.net (J.S.K.);
01love14@hanmail.net (S.-H.P.)
*Correspondence: aprilhwan@naver.com (K.H.J.); apple8840@hanmail.net (Y.G.K.)
These authors contributed equally to this work.
Received: 21 October 2020; Accepted: 10 November 2020; Published: 13 November 2020


Abstract:
Introduction: A recent study showed that early renal tubular injury is ameliorated in
Nod-like receptor pyrin domain-containing protein 3 (NLRP3) KO mice with rhabdomyolysis-induced
acute kidney injury (RIAKI). However, the precise mechanism has not been determined. Therefore,
we investigated the role of NLRP3 in renal tubular cells in RIAKI. Methods: Glycerol-mediated
RIAKI was induced in NLRP3 KO and wild-type (WT) mice. The mice were euthanized 24 h after
glycerol injection, and both kidneys and plasma were collected. HKC-8 cells were treated with
ferrous myoglobin to mimic a rhabdomyolytic environment. Results: Glycerol injection led to
increase serum creatinine, aspartate aminotransferase (AST), and renal kidney injury molecule-1
(KIM-1) level; renal tubular necrosis; and apoptosis. Renal injury was attenuated in NLRP3 KO
mice, while muscle damage and renal neutrophil recruitment did not dier between NLRP3 KO
mice and WT mice. Following glycerin injection, increases in cleaved caspase-3, poly (ADP-ribose)
polymerase (PARP), and a decrease in the glutathione peroxidase 4 (GPX-4) level were observed
in the kidneys of mice with RIAKI, and these changes were alleviated in the kidneys of NLRP3
KO mice. NLRP3 was upregulated, and cell viability was suppressed in HKC-8 cells treated
with ferrous myoglobin. Myoglobin-induced apoptosis and lipid peroxidation were significantly
decreased in siNLRP3-treated HKC-8 cells compared to ferrous myoglobin-treated HKC-8 cells.
Myoglobin reduced the mitochondrial membrane potential and increased mitochondrial fission and
reactive oxygen species (ROS) and lipid peroxidation levels, which were restored to normal levels
in NLRP3-depleted HKC-8 cells. Conclusions: NLRP3 depletion ameliorated renal tubular injury
in a murine glycerol-induced acute kidney injury (AKI) model. A lack of NLRP3 improved tubular
cell viability via attenuation of myoglobin-induced mitochondrial injury and lipid peroxidation,
which might be the critical factor in protecting the kidney.
Keywords: rhabdomyolysis; NLRP3; acute kidney injury; myoglobin
1. Introduction
Rhabdomyolysis is caused by the dissolution of skeletal muscle due to various factors, such as
trauma, drug toxicity, infection, excessive exercise, and genetic defects [
1
,
2
]. Generally, 10–40% of
cases of rhabdomyolysis lead to acute kidney injury (AKI) [
1
,
3
,
4
]. Nevertheless, there is no specific
treatment targeting the pathogenesis of rhabdomyolysis. Intracellular materials, including myoglobin,
Int. J. Mol. Sci. 2020,21, 8564; doi:10.3390/ijms21228564 www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2020,21, 8564 2 of 13
electrolytes, and sarcoplasmic proteins, leak into the systemic circulation following rhabdomyolysis [
1
,
3
,
4
].
Among these materials, released myoglobin is freely filtered by glomeruli and reabsorbed by renal
tubular cells via endocytosis, and the presence of myoglobin is indicated by red–brown urine [
1
].
Free myoglobin is not only directly toxic to renal epithelial cells but also acts as a mediator of
inflammation. Concentrated myoglobin in renal tubules leads to tubular obstruction when it precipitates
with Tamm-Horsfall protein [
5
]. The binding of oxygen to ferrous oxide derived from myoglobin
can lead to the generation of a hydroxyl radical, which can cause mitochondrial damage and
tubular apoptosis [
6
]. Additionally, myoglobin contains a peroxidase-like enzyme that initiates
lipid peroxidation and generates isoprostanes [
7
]. Myoglobin-derived lipid peroxidation and iron
accumulation injures the kidney by escalating ferroptosis during rhabdomyolysis [
8
]. However,
there is limited information regarding ferroptosis as a critical pathogenesis of rhabdomyolysis.
Damage-associated molecular patterns (DAMPs) can activate NOD-like receptor family proteins and the
pyrin domain containing-3 (NLRP3) inflammasome in the absence of pathogens [
9
,
10
]. Several studies
have suggested that in rhabdomyolysis, myoglobin can mediate AKI by activating the NLRP3
inflammasome [
11
,
12
]. One study demonstrated that glycerol-mediated rhabdomyolysis-induced
acute kidney injury (RIAKI) is ameliorated in NLRP3 knockout (KO) mice compared to control
mice [
13
]. However, the underlying mechanisms linking NLRP3 to renal injury remain unknown.
Renal tubular injury is preceded by immune cell infiltration into the kidney 24 h after glycerol
injection, and the depletion of NLRP3 mitigates the renal inflammatory response in a murine RIAKI
model [
13
]. We previously confirmed that renal tubular epithelial cells (TECs) do not express cleaved
caspase-1 or release IL-1
β
[
14
]. The combination of NLRP3 agonists, such as lipopolysaccharide
(LPS), and ATP, nigericin, or monosodium urate fails to induce IL-1
β
release by renal TECs [
15
].
Therefore, the depletion of inflammasome-independent NLRP3 in renal TECs could contribute to
attenuation of renal tubular injury in glycerol-induced RIAKI. However, there has been no study
testing this hypothesis. We reported that NLRP3 depletion decreases mitochondrial damage and
apoptosis in hypoxia; in contrast, overexpression of NLRP3 is sucient to elevate mitochondrial
reactive oxygen species (ROS) levels even under normoxic condition [
14
]. Additionally, when it
forms a complex with ASC and caspase-8 in mitochondria, NLRP3 regulates apoptotic cell death
in the renal and gut epithelium [
15
]. This evidence suggests that NLRP3 regulates apoptosis by
modulating mitochondrial function in addition to forming the canonical inflammasome. Therefore,
we hypothesized that inflammasome-independent NLRP3, especially in renal tubular cells, is involved
in mediating renal injury in RIAKI and we attempted to clarify the precise mechanism underlying
myoglobin-induced apoptosis and ferroptosis.
2. Results
2.1. NLRP3 KO Mice Were Protected from RIAKI
Glycerol-mediated RIAKI was established in wild-type (WT) and NLRP3 KO mice.
Serum creatinine
levels were increased 24 h after glycerol injection in mice of both genotypes and were lower in NLRP3
KO mice than in WT mice (Figure 1A). However, RIAKI-induced elevation of aspartate aminotransferase
(AST) and alanine transferase (ALT) was not significantly dierent between WT and NLRP3 KO mice,
suggesting that NLRP3 depletion did not prohibit glycerol-induced muscle damage but protected the
kidney from RIAKI (Figure 1B,C). Periodic acid-schi(PAS) and terminal deoxynucleotidyl transferase
dUTP nick end labeling (TUNEL) staining showed that glycerol-induced tubular necrosis and apoptosis
were decreased in NLRP3 KO mice compared to WT mice (Figure 1D,F,G,I). However, there was
no dierence in the number of Ly6B-positive intrarenal cells between WT and NLRP3 KO mice
(Figure 1E,H). NLRP3 expression was increased in the kidneys of mice with RIAKI compared to
control mice (Figure 2A,B). Kidney injury molecule-1 (KIM-1) and neutrophil gelatinase associated
lipocalin (NGAL), markers of proximal tubular injury, were augmented in the kidneys of WT mice
with RIAKI, whereas this increase was significantly mitigated in the kidneys of NLRP3 KO mice
Int. J. Mol. Sci. 2020,21, 8564 3 of 13
with RIAKI. In particular, in the kidneys of mice with RIAKI, glutathione peroxidase 4 (GPX4) was
downregulated, showing that ferroptosis was induced. Interestingly, the expression of ferroptotic
markers was decreased in the kidneys of NLRP3-depleted mice with RIAKI compared with those of WT
mice with RIAKI. The expression of apoptotic markers, such as cleaved caspase-3 and poly (ADP-ribose)
polymerase (PARP), was elevated in the kidney following glycerol injection, whereas NLRP3 depletion
reduced the expression of these markers. NLRP3 depletion decreased the mRNA expression of
inflammatory cytokines, including TNF-
α
, IL-1
β
, and IL-6, induced by rhabdomyolysis (Figure 2C–E).
Figure 1.
Glycerol-induced renal injury was attenuated in NLRP3 KO mice. Glycerol was
intramuscularly injected into WT and NLRP3 KO mice, and the mice were euthanized 24 h after
injection. (
A
C
) The plasma levels of creatinine, AST, and ALT were measured. (
D
) PAS-stained
(200
×
magnification), (
E
) Ly6B-stained (400
×
magnification), (
F
) TUNEL-stained renal cortices of sham
and glycerol-injected WT and NLRP3 KO mice (200
×
magnification). (
G
) Tubular necrosis was scored
in 10 randomly selected fields. (
H
) Ly6B-positive cells were counted in high-power fields. (
I
) Green
fluorescent TUNEL-positive puncta were counted. We used 6 mice per group. NLRP3, NOD-like
receptor family proteins and the pyrin domain containing-3; AST, aspartate aminotransferase; ALT,
alanine transferase; PAS, periodic acid-schi; TUNEL, TdT-mediated dUTP nick end labeling, * p<0.05
vs. sham mice, # p<0.05 vs. WT mice.
Int. J. Mol. Sci. 2020,21, 8564 4 of 13
Figure 2.
RIAKI led to increases in tubular injury, apoptosis, ferroptosis, and inflammation in the
kidney, which were abrogated in the kidneys of NLRP3 KO mice. (
A
) Western blot analysis of NLRP3,
KIM-1, NGAL, ACSL4, GPX4, cleaved caspase-3, and PARP expression in the kidneys of sham and
glycerol-injected WT and NLRP3 KO mice. (
B
) Densitometric analysis of the Western blots. (
C
E
)
RT-PCR analysis of IL-6, IL-1
β
, and TNF-
α
expression in sham and glycerol-injected WT and NLRP3
KO mice. KIM-1, kidney injury molecule-1; NGAL, neutrophil gelatinase associated lipocalin; ACSL4,
acyl-CoA synthetase long-chain family; GPX4, glutathione peroxidase 4; PARP, poly (ADP-ribose)
polymerase; IL-6, interleukine-6; IL-1
β
, interleukine-1
β
; TNF-
α
, tumor necrosis factor-
α
. We used
6 mice per group. * p<0.05 vs. sham mice, # p<0.05 vs. WT mice.
2.2. NLRP3 Depletion in Renal Tubular Cells Mitigated Myoglobin-Induced Ferroptosis and Apoptosis
In vivo
studies suggest that the renal protective eect of NLRP3 depletion mainly originates
from alleviation of renal tubular injury; thus, experiments were performed on tubular cells. Ferrous
myoglobin was applied to HKC-8 cells to induce an
in vitro
model of rhabdomyolysis. Since reduced
myoglobin (Fe
2+
) has been reported to be cytotoxic, ascorbic acid was added to metmyoglobin (Fe
3+
)
to produce ferrous myoglobin [
16
]. Cell viability decreased as the myoglobin concentration increased
to 5 mg/mL, and approximately 60% of 5 mg/mL myoglobin-treated cells survived (Figure 3A).
At 1, 5, and 10 mg/mL, ferrous myoglobin similarly increased NLRP3 expression in HKC-8 cells.
Since ferroptosis and apoptosis in the kidney are the primary manifestations of RIAKI, the expression of
markers related to these forms of cells death were analyzed in HKC-8 cells. Ferrous myoglobin induced
an increase in ACSL4 expression, a decrease in GPX4 expression and upregulation of cleaved caspase-3
and PARP expression in HKC-8 cells (Figure 3B,C). This result suggested that rhabdomyolysis-mediated
renal tubular cells underwent ferroptosis and apoptosis. These changes were reversed in siNLRP3-treated
HKC-8 cells, although the level of GPX4 was not significantly altered (Figure 3D,E).
Int. J. Mol. Sci. 2020,21, 8564 5 of 13
Figure 3.
NLRP3-depleted HKC-8 cells were protected from ferrous myoglobin-induced apoptosis and
ferroptosis. (
A
) HKC-8 cells were treated with various doses of myoglobin for 24 h, and cell viability
was measured by the MTT assay. (
B
,
C
) The expression of NLRP3, ASCL4, GPX4, cleaved caspase-3,
and PARP in HKC-8 cells treated with dierent doses of myoglobin was analyzed by immunoblotting
and densitometric analysis. (
D
,
E
) Western blot analysis of NLRP3, ASCL4, GPX4, cleaved caspase-3,
and PARP expression in HKC-8 cells transfected with or without siNLRP3 and treated with or without
1 mg/mL or 5 mg/mL myoglobin and densitometric analysis. We repeated 3 experiments in the
respective sections. * p<0.05 vs. Mb-untreated cells, # p<0.05 vs. siNLRP3-untreated cells.
2.3. NLRP3 Depletion in Renal Tubular Cells Attenuated Myoglobin-Induced Mitochondrial Injury
Previously, we showed that inflammasome-independent NLRP3 regulates apoptosis in TECs
by interacting with mitochondria and mediating mitochondrial ROS production [
14
]. Therefore,
we decided to examine whether mitochondrial damage under myoglobin stimulation varies depending
on the presence or absence of NLRP3. The mitochondrial membrane potential (JC-1) was markedly
reduced following ferrous myoglobin stimulation, and this decrease was reversed in siNLRP3-treated
HKC-8 cells (Figure 4A,B). Myoglobulin stimulation increased the expression of the mitochondrial
fission protein dynamin-related protein 1 (DPR1) and decreased the expression of the mitochondrial
fusion protein Mitofusin 1 (MFN1) (Figure 4C,D). Additionally, the mitochondrial changes were
attenuated in siNLRP3-treated HKC-8 cells.
Int. J. Mol. Sci. 2020,21, 8564 6 of 13
Figure 4.
NLRP3-depleted HKC-8 cells were protected from myoglobin-induced mitochondrial injury.
HKC-8 cells transfected with or without siNLRP3 were treated with control or myoglobin for 24 h.
(
A
,
B
) Flow cytometry analysis of JC-1-stained cells for the detection of mitochondrial membrane
potential changes and graphs showing fluorescence. (
C
,
D
) Western blot pictures showing DRP1 and
MFN1 expression and densitometric analysis. DRP1, dynamin-related protein 1; MFN1, mitofusin 1.
We repeated 3 experiments in the respective sections. * p<0.05 vs. Mb-untreated cells, # p<0.05 vs.
siNLRP3-untreated cells.
2.4. NLRP3 Depletion Contributed to Decreasing Myoglobin-Induced ROS Levels and Lipid Peroxidation
Next, we evaluated ROS and lipid peroxidation levels in myoglobin-induced renal tubular
cells. BODIPY staining was increased, and BODIPY was colocalized with MitoTracker in HKC-8
cells following myoglobin administration (Figure 5A). Myoglobin-induced lipid peroxidation was
significantly attenuated in siNLRP3-treated HKC-8 cells compared to ferrous myoglobin-treated HKC-8
cells. Malondialdehyde (MDA) production was also augmented in ferrous myoglobin-stimulated
HKC-8 cells, whereas it was significantly decreased in NLRP3-depleted HKC-8 cells (Figure 5B).
The total ROS concentration, as determined by DCFH-DA, was increased in myoglobin-stimulated
HKC-8 cells and markedly reduced in siNLRP3-treated HKC-8 cells (Figure 5D). 4-hydroxynonenal
(4-HNE) expression was increased in response to myoglobin and decreased in siNLRP3-treated HKC-8
cells (Figure 5E,F). In particular, 4-HNE was upregulated following NLRP3 pCMV6-induced NLRP3
overexpression, even in the absence of stimulation. These results suggested that NLRP3 could be
involved in regulating lipid peroxidation in renal tubular cells under myoglobin stimulation.
Figure 5. Cont.
Int. J. Mol. Sci. 2020,21, 8564 7 of 13
Figure 5.
NLRP3-depleted HKC-8 cells were protected from myoglobin-induced ROS production.
HKC-8 cells transfected with or without siNLRP3 were treated with control or myoglobin for 24 h.
(
A
) Confocal microscopy (63
×
magnification) and (
B
) analysis of MDA production for the evaluation of
lipid peroxidation induced by myoglobin in HKC-8 cells. (
C
,
D
) Flow cytometry of DCFH-DA-stained
cells for the detection of ROS production induced by myoglobin in HKC-8 cells. (
E
,
F
) Immunoblotting
for NLRP3 and 4-HNE in HKC-8 cells and densitometric analysis. MDA, malondialdehyde; 4-HNE,
4-hydroxynonenal. We repeated 3 experiments in the respective sections. * p<0.05 vs. Mb-untreated
cells, # p<0.05 vs. siNLRP3-untreated cells.
3. Discussion
In this experiment, we demonstrated the following: (1) renal damage caused by rhabdomyolysis
is mitigated in NLRP3-depleted mice compared to WT mice via alleviation of renal inflammation,
apoptosis, and ferroptosis despite no dierence in muscle damage; (2) myoglobin-induced apoptosis
and lipid peroxidation are attenuated in NLRP3-depleted tubular cells; (3) depletion of NLRP3 in
renal tubular cells improves mitochondrial damage and alters mitochondrial biogenesis induced by
myoglobin; and (4) the preservation of mitochondria following myoglobin simulation might contribute
to alleviating lipid peroxidation. Several studies have suggested that NLRP3 can aggravate renal
injury in the pathogenesis of RIAKI; nevertheless, the precise mechanisms are not yet clear [
11
,
13
].
Our study suggested that reduction in mitochondrial damage and lipid peroxidation in renal tubules
is the main contributor to the protection of the kidneys in NLRP3 KO mice with RIAKI. NLRP3
is known to mediate renal injury via inflammasome formation in various renal diseases, such as
RIAKI, ischemic reperfusion injury (IRI), contrast-induced AKI, and unilateral ureter obstruction
nephropathy [
17
19
]. These renal diseases can be ameliorated by NLRP3 inflammasome inhibitors or
genetic deletion of inflammasome components [
20
22
]. However, it has been shown that caspase-1
inhibitors do not improve renal tubular apoptosis in rats with RIAKI [
11
]. Additionally, tubular
apoptosis following IRI is decreased in NLRP3 KO mice compared with WT mice, whereas WT mice
engrafted with NLRP3 KO bone marrow fail to show improvement [
17
]. Previously, we confirmed
that in renal tubular cells, NLRP3 regulates mitochondrial damage [
14
]. Similarly, NLRP3 KO renal
tubular cells attenuate apoptosis in mitochondria caused by TNF
α
/CHX [
15
]. Consistently, our data
elucidated that the absence of NLRP3 in renal tubular cells mitigates tubular apoptosis and lipid
peroxidation in response to myoglobin stimulation. The fundamental cause of this protective eect
was assumed to be connected to the preservation of mitochondria. Tubular cell damage without an
increase in the number of intrarenal immune cells is the main manifestation during the early phase
of RIAKI [
13
]. Thus, the elimination of NLRP3 from renal tubular cells could be a critical factor
in improving renal function via mitochondrial protection during early RIAKI. The results showing
that myoglobin-induced apoptosis and lipid peroxidation were mitigated in NLRP3-depleted renal
tubular cells supported our suggestion. Our experiments also demonstrated that glycerol injection
led to an increase in the levels of inflammatory cytokines, such as TNF
α
, IL-
β
, and IL-6, in the
mouse kidney. Inflammation is the main pathogenic feature of AKI induced by rhabdomyolysis,
and previous studies have shown that pattern recognition receptors such as TLR4 and NLRP3 are
upregulated in the kidney in RIAKI [
13
,
23
,
24
]. We found that inflammatory cytokine levels were
decreased in the kidneys of NLRP3-depleted mice compared with those of WT mice, which is in
line with a previous study [
13
]. Nevertheless, the number of intrarenal immune cells were not
Int. J. Mol. Sci. 2020,21, 8564 8 of 13
increased in the kidney 24 h following glycerol injection. Therefore, we assume that the main factor
in decreasing inflammation is attenuation of inflammatory responses in renal tubular cells rather
than renal immune cells. NLRP3 in nonimmune cells is involved in potentiating mitochondrial ROS
levels and ultimately augmenting fibrosis under TGF-
β
stimulation [
25
,
26
]. In addition, NLRP3
KO fibroblasts recruit more anti-inflammatory macrophages than inflammatory macrophages under
TGF-
β
stimulation, ultimately leading to a decrease in inflammation [
27
]. These data suggest that
the absence of NLRP3 in nonimmune renal resident cells may be a principal factor in attenuating
renal inflammation during the early phase of RIAKI. This study showed that ferroptosis occurs in
glycerol-induced AKI, as ASCL4 expression was upregulated and GPX4 expression was downregulated
in the glycerol-injected kidney. Two main factors that lead to ferroptosis are an inability to reduce
lipid peroxide levels due to the absence or inactivation of GPX4 and increased formation of lipid
peroxides [
28
]. Oxyferrous myoglobin is converted to metmyoglobin (MetMb, Fe
3+
) via auto-oxidation
and can catalytically accelerate lipid peroxidation [
29
]. The administration of antioxidant agents
preserves renal function in glycerol-induced rhabdomyolysis mice [
30
,
31
]. Ferroptosis inhibitors
eectively ameliorate renal structure and function, while the inhibition of apoptosis or necroptosis does
not inhibit renal injury in glycerol-induced AKI [
8
]. Our study demonstrated that NLRP3 depletion
decreased lipid peroxidation and ferroptosis in the kidney during the early phase of RIAKI. The levels
of lipid peroxides, including MDA and 4-HNE, and ferroptosis inducers, such as ASCL4, were reduced
in NLRP3-deficient renal tubular cells in response to myoglobin stimulation, whereas GPX4 expression
was not changed. It is not clear how NLRP3 absence contributes to inhibiting lipid peroxidation.
In our study, NLRP3-depleted tubular cells mitigated mitochondrial dysfunction and enhanced
mitochondrial biogenesis. Phospholipids in mitochondrial membranes are the primary targets of ROS
attack, which results in lipid peroxidation [
32
]. ROS produced by rhabdomyolysis may extend to form
oxidized mitochondrial lipids and are connected to mitochondrial dysfunction. Myoglobin-induced
mitochondrial dysfunction and altered mitochondrial biogenesis were reversed when NLRP3 was
depleted. Therefore, we speculate that decreases in mitochondrial dysfunction and lipid peroxide levels
following NLRP3 depletion can lead to a decrease in the induction of ferroptosis. Correspondingly,
NLRP3 overexpression caused an increase in 4-HNE levels, even in the absence of stimulation. However,
iron-dependent ferroptosis induced by myoglobin might not be fully blocked in NLRP3-deficient
tubular cells, and, thus, GPX4 expression was not decreased. Recent evidence suggests that ACSL4 and
activated NLRP3 are similarly localized in the mitochondria-associated membrane, which is located at
the junction between the endoplasmic reticulum and mitochondria [
33
,
34
]. Therefore, more research is
needed to determine whether the interaction between NLRP3 and ACSL4 is connected to the regulation
of lipid peroxidation and ferroptosis. This study has limitations. We used conventional NLRP3 KO
mice in the animal experiments since tubular cell-specific NLRP3 KO mice were not available. Thus,
we cannot thoroughly conclude that this renal protective eect originates solely from the depletion
of inflammasome-independent NLRP3 in renal tubular cells. Further studies using cell-specific KO
mice and NLRP3 inhibitors are necessary for clarification. In addition, we could not clarify the
mechanisms by which myoglobin stimulates renal tubular cells in this study. Nevertheless, this study
confirmed for the first time that ferrous myoglobin upregulated NLRP3 expression in renal tubular cells.
In summary, NLRP3 depletion during rhabdomyolysis protects against renal injuries via attenuation of
myoglobin-induced mitochondrial dysfunction and subsequent lipid peroxidation in renal tubular
cells. This study shows that ferroptosis may be the critical factor in inducing renal injury in RIAKI
and that inflammasome-independent NLRP3 in renal tubules is associated with the regulation of lipid
peroxidation. NLRP3 may be a new therapeutic target for rhabdomyolysis.
Int. J. Mol. Sci. 2020,21, 8564 9 of 13
4. Material and Methods
4.1. Animal Models
All animal experiments were performed according to the guidelines, care, and use of Experiments
Animals Committee of Kyunghee University Hospital at Gangdong (approval number: KHNMC
AP2018-004, 1 February 2018). C57BL/6J WT mice were purchased from Jun Bio.inc. (Suwon, Korea).
RGEN/Cas9 NLRP3 KO mice were established from Macrogen (Seoul, Korea). The establishment of
NLRP3 KO mice was precisely described in the previous study [
14
]. The animals were maintained on
a 12 h dark/light cycle with free access to food and water. For the purpose of inducing rhabdomyolysis,
the two hind limbs of the animals were intramuscularly injected with 5 mL/kg of 50% glycerol in
8–10-week-old male mice. Water was withheld for 24 h before the glycerol injection to increase the
incidence of renal injury.
4.2. Blood Chemistry
Serum levels of creatinine, blood urea nitrogen (BUN), AST, and ALT were measured by using Vet
test 8008 (IDEXX, Ludwigsburg, Germany) according to the manufacturer’s instructions.
4.3. Histopathology
Kidneys were fixed in 10% neutral buered formalin, embedded in paran, cut into 4
µ
m sections,
and stained with PAS reagent. Tissue sections were viewed by a light microscope at
×
200 magnification.
For semiquantitative analysis, 10 dierent fields at the corticomedullary junction from each group
were randomly selected. Necrotic tubules were scored on a scale of 0 to 5 based on the percentage of
necrosis area as follows: 0, absent; 1, 1–25%; 2, 26–50%; 3, 51–75%; 4, 76–99%; and 5, 100%.
4.4. TdT Mediated dUTP Nick End Labeling (TUNEL) Assay
Apoptosis in renal tissues was identified by TUNEL assay with an in situ Cell Death Detection kit
following the manufacturer’s instructions (Roche Applied Science, Indianapolis, IN, USA). The number
of apoptotic cells were counted under a fluorescence microscope at
×
200 magnification. At least
ten areas at the corticomedullary junction in the sections from dierent mice of each group were
determined and averaged.
4.5. Real-Time PCR
Total RNA was extracted from renal tissues using a QIAzol
®
Lysis Reagent (QIAGEN,
Hilden, Germany) according to the manufacturer’s instructions. The RNA concentration and purity
were confirmed with Nanodrop 2000. Real-time RT-PCR analysis was performed using the Step One
Plus Real-Time PCR System (Applied Biosystems, Foster City, CA, USA) to detect mRNA expression
of IL-6, TNF-
α
, and IL-1
β
. Each sample was processed in duplicate in separate tubes to quantify target
gene expression and the results were normalized to 18S expression.
4.6. Western Blot
Kidney tissue and cells were lysed in an ice-cold lysis buer (PRO-PREP
Protein Extraction
solution, INTRON, Seongnam-si, Korea). Proteins were separated with 10% PAGE and electroblotted
onto a PVDF membrane (BIO-RAD, Seoul, Korea). The membrane was incubated with primary antibody
raised against NLRP3 (NOVUS, Centennial, CO, USA), KIM-1 (Abcam,
Cambridge, MA, USA
),
NGAL (Abcam, Cambridge, MA, USA), ACSL4 (Santa cruz, Dallas, TX, USA), GPX4 (Abcam,
Cambridge, MA, USA
), Cleaved caspase-3 (Cell signaling, Danvers, MA, USA), PARP (Cell sinaling,
Danvers, MA, USA), dynamin-1-like protein (DRP1) (Abcam, Cambridge, MA, USA), Mitofusin1
(Abcam, Cambridge, MA, USA),
β
-actin (Santa cruz, Dallas, TX, USA), GAPDH (Cell Signaling, Danvers,
MA, USA) (1:1000) and, subsequently, with horseradish peroxidase-conjugated goat anti-rabbit
Int. J. Mol. Sci. 2020,21, 8564 10 of 13
or mouse immunoglobulin G (1:10,000, BETHYL). The immunoreactive bands were detected by
chemiluminescence (ECL, Advansta, CA, USA).
β
-actin and GAPDH were used as internal controls of
cells and tissues.
4.7. Cell Culture
The human renal proximal tubular epithelial cell line HKC8 was obtained from Dr. L. Rausen
(Johns Hopkins University, Baltimore, MD, USA) and was maintained in Dulbecco’s Modified
Eagle Medium supplemented with Ham’s F12 medium (DMEM/F12; Thermo Fisher Scientific,
Waltham, MA, USA
). DMEM/F12 was supplemented with 10% fetal bovine serum and 1%
penicillin/streptomycin (WelGENE, Daegu, Korea). Myoglobin from equine heart muscle (M5696,
Sigma, St. Louis, MO, USA) and 2 mM ascorbic acid (Sigma, St. Loiuse, MO, USA) was dissolved in
medium and prepared with ferrous myoglobin right obefore
in vitro
experiment. The concentration
of myoglobin was adjusted to 200 mM with ascorbic acid 2 mM.
4.8. Small Interfering RNA Knockdown Experiments
Duplex small interfering RNAs (siRNAs) targeting NLRP3 (ORIGENE, Rockville, MD, USA)
and a control siRNA were purchased from Bioneer Inc. (Seoul, Korea). HKC8 cells were transfected
using Lipofectamine 2000 (Invitrogen, Waltham, MA, USA), after which these cells were utilized for
functional studies 24 h later, and knockdown eciency was assessed by Western blot analysis using
NLRP3 and GAPDH antibodies.
4.9. MTT Assay
After myoglobin (with or without L-ascorbic acid 2 mM) treatment, the cell viability of HKC8 cells
plated in 24-well plates was detected by using 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium
bromide (MTT) assay. Briefly, the cells were treated with MTT and incubated at 37
C for 4 h.
The supernatant was removed, and the dye was dissolved with 200
µ
L of DMSO and shaken on
an orbital shaker for 10 min in the dark. The optical density (OD) was recorded at 570 nm using a
spectrophotometer. Each experiment was performed in triplicate.
4.10. Flow Cytometric Analysis
Mitochondrial membrane potential
∆Ψm
was measured by the sensitive and relatively
mitochondrion-specific lipophilic cationic probe fluorochrome 5,5
0
,6,6
0
-tetrachloro-1,1
0
,3,3
0
-
tetraethylbenzimidazoly-carbocyanine iodide (JC-1) (Molecular Probes, Eugene, OR, USA). Briefly,
HKC8 cells were incubated with JC-1 (5
µ
mol/L) at 37
C for 20 min and examined by flow
cytometry (BD FACSCaliber Flow Cytometry, San Jose, CA, USA). Intracellular ROS was measured by
2
0
,7
0
-dichlorofluorescin diacetate (DCF-DA) (Sigma, St. Louis, MO, USA). Briefly, HKC8 cells were
incubated with DCF-DA (5
µ
M) at 37
C for 30 min and examined by flow cytometry (BD FACSCaliber
Flow Cytometry, San Jose, CA, USA).
4.11. Immunocytochemistry
Cells were fixed with 4% paraformaldehyde after being stained to BODIPY (Thermo Fisher
Scientific, Waltham, MA, USA) and Mitotracker (Thermo Fisher Scientific, Waltham, MA, USA).
The nuclei were stained with DAPI (1:1000, Invitrogen). The slides were mounted with Fluorescence
Mounting Medium (Dako), and images were acquired by a confocal microscope (Carl Zeiss LSM 700,
Jena, Germany). Z-stack of images were projected into one plane (maximum intensity projection).
Int. J. Mol. Sci. 2020,21, 8564 11 of 13
4.12. Lipid Peroxidation Product Assay
Lipid peroxidation product malondialdehyde (MDA) in HKC-8 cells (2
×
10
6
cells) was measured
using a commercial MDA kit (Abcam, Cambridge, MA, USA). The spectrophotometric absorbance was
assessed at 532 nm in accordance with the manufacturer’s instructions.
4.13. Statistical Analysis
Statistical analyses were conducted using SPSS software (version 20 SPSS, Inc., Chicago, IL, USA)
and GraphPad Prism 5.0. Descriptive data were presented as mean
±
SEM. Results were analyzed
using the Kruskal–Wallis nonparametric test for multiple comparisons and Mann–Whitney test for
two objects; pvalues <0.05 were considered statistically significant.
Author Contributions:
Conceptualization, Y.G.K. and S.-h.L.; methodology, S.J.S.; software, J.-Y.M.; validation,
S.-m.K., and S.-H.P.; formal analysis, H.S.H.; investigation, J.S.K.; resources, S.J.S.; data curation, S.-m.K.;
writing—original draft preparation, Y.G.K.; writing—review and editing, K.H.J.; visualization, S.-m.K.; supervision,
S.-h.L.; project administration, Y.G.K.; funding acquisition, K.H.J. All authors have read and agreed to the published
version of the manuscript.
Funding: This research was supported by National Research Foundation of Korea (2020R1H1A2010539).
Conflicts of Interest:
The authors declare no conflict of interest. The funders had no role in the design of the
study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to
publish the results.
References
1.
Bosch, X.; Poch, E.; Grau, J.M. Rhabdomyolysis and acute kidney injury. N. Engl. J. Med.
2009
,361, 62–72.
[CrossRef]
2.
Petejova, N.; Martinek, A. Acute kidney injury due to rhabdomyolysis and renal replacement therapy:
A critical review. Crit. Care 2014,18, 224. [CrossRef]
3.
Chatzizisis, Y.S.; Misirli, G.; Hatzitolios, A.I.; Giannoglou, G.D. The syndrome of rhabdomyolysis:
Complications and treatment. Eur. J. Intern. Med. 2008,19, 568–574. [CrossRef]
4.
Chander, V.; Chopra, K. Molsidomine, a nitric oxide donor and L-arginine protects against
rhabdomyolysis-induced myoglobinuric acute renal failure. Biochim. Biophys. Acta
2005
,1723, 208–214.
[CrossRef] [PubMed]
5.
Zager, R.A. Studies of mechanisms and protective maneuvers in myoglobinuric acute renal injury. Lab. Investig.
1989,60, 619–629. [PubMed]
6.
Zager, R.A.; Foerder, C.A. Eects of inorganic iron and myoglobin on
in vitro
proximal tubular lipid
peroxidation and cytotoxicity. J. Clin. Investig. 1992,89, 989–995. [CrossRef] [PubMed]
7.
Reeder, B.J.; Wilson, M.T. Hemoglobin and myoglobin associated oxidative stress: From molecular
mechanisms to disease States. Curr. Med. Chem. 2005,12, 2741–2751. [CrossRef]
8.
Guerrero-Hue, M.; Garcia-Caballero, C.; Palomino-Antolin, A.; Rubio-Navarro, A.; Vazquez-Carballo, C.;
Herencia, C.; Martin-Sanchez, D.; Farre-Alins, V.; Egea, J.; Cannata, P.; et al. Curcumin reduces renal damage
associated with rhabdomyolysis by decreasing ferroptosis-mediated cell death. FASEB J.
2019
,33, 8961–8975.
[CrossRef]
9.
Shen, H.; Kreisel, D.; Goldstein, D.R. Processes of sterile inflammation. J. Immunol.
2013
,191, 2857–2863.
[CrossRef]
10.
Chen, G.Y.; Nunez, G. Sterile inflammation: Sensing and reacting to damage. Nat. Rev. Immunol.
2010
,
10, 826–837. [CrossRef]
11.
Homsi, E.; Janino, P.; de Faria, J.B. Role of caspases on cell death, inflammation, and cell cycle in
glycerol-induced acute renal failure. Kidney Int. 2006,69, 1385–1392. [CrossRef] [PubMed]
12.
Homsi, E.; Ribeiro-Alves, M.A.; Lopes de Faria, J.B.; Dias, E.P. Interleukin-6 stimulates tubular regeneration
in rats with glycerol-induced acute renal failure. Nephron 2002,92, 192–199. [CrossRef] [PubMed]
13.
Komada, T.; Usui, F.; Kawashima, A.; Kimura, H.; Karasawa, T.; Inoue, Y.; Kobayashi, M.; Mizushina, Y.;
Kasahara, T.; Taniguchi, S.; et al. Role of NLRP3 Inflammasomes for Rhabdomyolysis-induced Acute Kidney
Injury. Sci. Rep. 2015,5, 10901. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2020,21, 8564 12 of 13
14.
Kim, S.M.; Kim, Y.G.; Kim, D.J.; Park, S.H.; Jeong, K.H.; Lee, Y.H.; Lim, S.J.; Lee, S.H.; Moon, J.Y.
Inflammasome-Independent Role of NLRP3 Mediates Mitochondrial Regulation in Renal Injury.
Front. Immunol. 2018,9, 2563. [CrossRef] [PubMed]
15.
Chung, H.; Vilaysane, A.; Lau, A.; Stahl, M.; Morampudi, V.; Bondzi-Simpson, A.; Platnich, J.M.; Bracey, N.A.;
French, M.C.; Beck, P.L.; et al. NLRP3 regulates a non-canonical platform for caspase-8 activation during
epithelial cell apoptosis. Cell Death Dier. 2016,23, 1331–1346. [CrossRef] [PubMed]
16.
Zager, R.A.; Burkhart, K. Myoglobin toxicity in proximal human kidney cells: Roles of Fe, Ca2+, H2O2,
and terminal mitochondrial electron transport. Kidney Int. 1997,51, 728–738. [CrossRef] [PubMed]
17.
Shigeoka, A.A.; Mueller, J.L.; Kambo, A.; Mathison, J.C.; King, A.J.; Hall, W.F.; Correia Jda, S.; Ulevitch, R.J.;
Homan, H.M.; McKay, D.B. An inflammasome-independent role for epithelial-expressed Nlrp3 in renal
ischemia-reperfusion injury. J. Immunol. 2010,185, 6277–6285. [CrossRef]
18.
Shen, J.; Wang, L.; Jiang, N.; Mou, S.; Zhang, M.; Gu, L.; Shao, X.; Wang, Q.; Qi, C.; Li, S.; et al. NLRP3
inflammasome mediates contrast media-induced acute kidney injury by regulating cell apoptosis. Sci. Rep.
2016,6, 34682. [CrossRef]
19.
Yamagishi, H.; Yokoo, T.; Imasawa, T.; Mitarai, T.; Kawamura, T.; Utsunomiya, Y. Genetically modified bone
marrow-derived vehicle cells site specifically deliver an anti-inflammatory cytokine to inflamed interstitium
of obstructive nephropathy. J. Immunol. 2001,166, 609–616. [CrossRef]
20.
Gong, W.; Mao, S.; Yu, J.; Song, J.; Jia, Z.; Huang, S.; Zhang, A. NLRP3 deletion protects against renal fibrosis
and attenuates mitochondrial abnormality in mouse with 5/6 nephrectomy. Am. J. Physiol. Renal Physiol.
2016,310, F1081–F1088. [CrossRef]
21.
Ludwig-Portugall, I.; Bartok, E.; Dhana, E.; Evers, B.D.; Primiano, M.J.; Hall, J.P.; Franklin, B.S.; Knolle, P.A.;
Hornung, V.; Hartmann, G.; et al. An NLRP3-specific inflammasome inhibitor attenuates crystal-induced
kidney fibrosis in mice. Kidney Int. 2016,90, 525–539. [CrossRef] [PubMed]
22.
Shahzad, K.; Bock, F.; Dong, W.; Wang, H.; Kopf, S.; Kohli, S.; Al-Dabet, M.M.; Ranjan, S.; Wolter, J.; Wacker, C.;
et al. Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy.
Kidney Int. 2015,87, 74–84. [CrossRef] [PubMed]
23.
Huang, R.S.; Zhou, J.J.; Feng, Y.Y.; Shi, M.; Guo, F.; Gou, S.J.; Salerno, S.; Ma, L.; Fu, P. Pharmacological
Inhibition of Macrophage Toll-like Receptor 4/Nuclear Factor-kappa B Alleviates Rhabdomyolysis-induced
Acute Kidney Injury. Chin. Med. J. 2017,130, 2163–2169. [CrossRef] [PubMed]
24.
Kundert, F.; Platen, L.; Iwakura, T.; Zhao, Z.; Marschner, J.A.; Anders, H.J. Immune mechanisms in the
dierent phases of acute tubular necrosis. Kidney Res. Clin. Pract. 2018,37, 185–196. [CrossRef]
25.
Wang, W.; Wang, X.; Chun, J.; Vilaysane, A.; Clark, S.; French, G.; Bracey, N.A.; Trpkov, K.; Bonni, S.; Du, H.J.;
et al. Inflammasome-independent NLRP3 augments TGF-beta signaling in kidney epithelium. J. Immunol.
2013,190, 1239–1249. [CrossRef]
26.
Bracey, N.A.; Gershkovich, B.; Chun, J.; Vilaysane, A.; Meijndert, H.C.; Wright, J.R., Jr.; Fedak, P.W.; Beck, P.L.;
Muruve, D.A.; Du, H.J. Mitochondrial NLRP3 protein induces reactive oxygen species to promote Smad
protein signaling and fibrosis independent from the inflammasome. J. Biol. Chem.
2014
,289, 19571–19584.
[CrossRef]
27.
Anders, H.J.; Suarez-Alvarez, B.; Grigorescu, M.; Foresto-Neto, O.; Steiger, S.; Desai, J.; Marschner, J.A.;
Honarpisheh, M.; Shi, C.; Jordan, J.; et al. The macrophage phenotype and inflammasome component NLRP3
contributes to nephrocalcinosis-related chronic kidney disease independent from IL-1-mediated tissue injury.
Kidney Int. 2018,93, 656–669. [CrossRef]
28.
Lei, P.; Bai, T.; Sun, Y. Mechanisms of Ferroptosis and Relations with Regulated Cell Death: A Review.
Front. Physiol. 2019,10, 139. [CrossRef]
29.
Reeder, B.J.; Sharpe, M.A.; Kay, A.D.; Kerr, M.; Moore, K.; Wilson, M.T. Toxicity of myoglobin and haemoglobin:
Oxidative stress in patients with rhabdomyolysis and subarachnoid haemorrhage. Biochem. Soc. Trans.
2002
,
30, 745–748. [CrossRef]
30.
Kim, J.H.; Lee, S.S.; Jung, M.H.; Yeo, H.D.; Kim, H.J.; Yang, J.I.; Roh, G.S.; Chang, S.H.; Park, D.J.
N-acetylcysteine attenuates glycerol-induced acute kidney injury by regulating MAPKs and Bcl-2 family
proteins. Nephrol. Dial. Transplant. 2010,25, 1435–1443. [CrossRef]
Int. J. Mol. Sci. 2020,21, 8564 13 of 13
31.
Nishida, K.; Watanabe, H.; Ogaki, S.; Kodama, A.; Tanaka, R.; Imafuku, T.; Ishima, Y.; Chuang, V.T.;
Toyoda, M.; Kondoh, M.; et al. Renoprotective eect of long acting thioredoxin by modulating oxidative
stress and macrophage migration inhibitory factor against rhabdomyolysis-associated acute kidney injury.
Sci. Rep. 2015,5, 14471. [CrossRef] [PubMed]
32.
Xiao, M.; Zhong, H.; Xia, L.; Tao, Y.; Yin, H. Pathophysiology of mitochondrial lipid oxidation: Role
of 4-hydroxynonenal (4-HNE) and other bioactive lipids in mitochondria. Free Radic. Biol. Med.
2017
,
111, 316–327. [CrossRef] [PubMed]
33.
Missiroli, S.; Patergnani, S.; Caroccia, N.; Pedriali, G.; Perrone, M.; Previati, M.; Wieckowski, M.R.; Giorgi, C.
Mitochondria-associated membranes (MAMs) and inflammation. Cell Death Dis.
2018
,9, 329. [CrossRef]
[PubMed]
34.
Doll, S.; Proneth, B.; Tyurina, Y.Y.; Panzilius, E.; Kobayashi, S.; Ingold, I.; Irmler, M.; Beckers, J.; Aichler, M.;
Walch, A.; et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol.
2017,13, 91–98. [CrossRef] [PubMed]
Publisher’s Note:
MDPI stays neutral with regard to jurisdictional claims in published maps and institutional
aliations.
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... Then, myoglobin circulates to the kidneys and filters freely from the glomerulus into the tubules [45], where it is reabsorbed by proximal renal tubule cells via endocytic receptors, such as megalin [47]. Noteworthily, proximal renal tubular cells are particularly susceptible to injury from nephrotoxic myoglobin [48][49][50]. Direct cytotoxic effects associated with myoglobin have been ascribed to oxidative injury [46]. Moreover, myoglobin binds to the Tam-Horsfall protein and forms a precipitate in the distal renal tubules [45], which forms pigmented granular casts that cause tubular obstruction [51], that will lead to increased pressure within the kidney tubules, exceeding the interstitial pressure. ...
... Further studies are needed to explore the mechanism of STING activation in wasp venom-induced AKI. Nonetheless, previous studies indicate that STING activation is associated with mitochondrial DNA (mtDNA) leakage from the kidney and its release in urine [42,66], a process that is also associated with myoglobin-induced mitochondrial lipid peroxidation, oxidative stress of mitochondria, and mitochondria apoptosis [49,67,68]. Additionally, myoglobin can impair the mitochondrial membrane potential and increase the levels of reactive oxygen species and lipid peroxidation in mitochondria of the renal tubular epithelial cells [49]. ...
... Nonetheless, previous studies indicate that STING activation is associated with mitochondrial DNA (mtDNA) leakage from the kidney and its release in urine [42,66], a process that is also associated with myoglobin-induced mitochondrial lipid peroxidation, oxidative stress of mitochondria, and mitochondria apoptosis [49,67,68]. Additionally, myoglobin can impair the mitochondrial membrane potential and increase the levels of reactive oxygen species and lipid peroxidation in mitochondria of the renal tubular epithelial cells [49]. This mechanism is supported by the activation of markers of oxidative stress (malondialdehyde and superoxide dismutase), which was described in rats with wasp venom-induced AKI [41]. ...
Article
Full-text available
Wasp venom can trigger local and systemic reactions, with the kidneys being commonly affected, potentially causing acute kidney injury (AKI). Despite of the recent advances, our knowledge on the underlying mechanisms of toxicity and targeted therapies remain poor. AKI can result from direct nephrotoxic effects of the wasp venom or secondary rhabdomyolysis and intravascular hemolysis, which will release myoglobin and free hemoglobin. Inflammatory responses play a central role in these pathological mechanisms. Noteworthily, the successful establishment of a suitable experimental model can assist in basic research and clinical advancements related to wasp venom-induced AKI. The combination of therapeutic plasma exchange and continuous renal replacement therapy appears to be the preferred treatment for wasp venom-induced AKI. In addition, studies on cilastatin and varespladib for wasp venom-induced AKI treatment have shown their potential as therapeutic agents. This review summarizes the available evidence on the mechanisms and treatment of wasp venom-induced AKI, with a particular focus on the role of inflammatory responses and potential targets for therapeutic drugs, and, therefore, aiming to support the development of clinical treatment against wasp venom-induced AKI.
... Proximal renal tubular cells have high mitochondrial content and are particularly susceptible to injury from the nephrotoxic myoglobin due to rhabdomyolysis [11][12][13]. Mitochondria are the primary target of rhabdomyolysis in renal tubular cells, and rhabdomyolysis-induced renal injury involves a direct interaction between myoglobin and mitochondria [14,15]. Maekawa et al. indicated that the inflammatory response in AKI caused by nephrotoxic substances is induced by the activation of endogenous substances, such as DNA, from damaged mitochondria [16]. ...
... Maekawa et al. indicated that the inflammatory response in AKI caused by nephrotoxic substances is induced by the activation of endogenous substances, such as DNA, from damaged mitochondria [16]. Myoglobin causes mitochondrial dysfunction and damage in renal tubular cells [12]. DNA from damaged mitochondria activates cyclic GMP-AMP synthase (cGAS), which promotes stimulator of interferon genes (STING) and active tank-binding kinase 1 (TBK1) [17,18]. ...
Article
Full-text available
Objective Recent evidence suggests a key role of the inflammatory responses in wasp venom-induced acute kidney injury (AKI). However, the potential regulatory mechanisms underlying the inflammatory responses in wasp venom-induced AKI remain unclear. STING reportedly plays a critical role in other AKI types and is associated with inflammatory responses and diseases. We aimed to investigate the involvement of STING in inflammatory responses associated with wasp venom-induced AKI. Methods The role of the STING signaling pathway in wasp venom-induced AKI was studied in vivo using a mouse model of wasp venom-induced AKI with STING knockout or pharmacological inhibition and in vitro using human HK2 cells with STING knockdown. Results STING deficiency or pharmacological inhibition markedly ameliorated renal dysfunction, inflammatory responses, necroptosis, and apoptosis in wasp venom-induced AKI in mice. Moreover, STING knockdown in cultured HK2 cells attenuated the inflammatory response, necroptosis, and apoptosis induced by myoglobin, the major pathogenic factor in wasp venom-induced AKI. Urinary mitochondrial DNA upregulation has also been observed in patients with wasp venom-induced AKI. Conclusions STING activation mediates the inflammatory response in wasp venom-induced AKI. This may offer a potential therapeutic target for the management of wasp venom-induced AKI.
... This disorder may lead to cardiac arrhythmia and should be treated promptly to prevent complications. 15,16 Various in vivo and ex vivo experimental models using laboratory animals have demonstrated that glycerol injection can induce acute renal failure through rhabdomyolysis. 15,17 Glycerol leads to the disintegration of muscle cell membranes, releasing iron-containing proteins into the extra-cellular environment. ...
Article
Full-text available
Studies conducted on animal models have shown that the administration of glycerol can lead to kidney tissue damage and impaired renal function. This is believed to be caused by oxidative stress and inflammation, which in turn can result in elevated levels of blood urea nitrogen (BUN) and creatinine. These metabolites are commonly used as indicators of renal function. The aim of the current experimental research was to investigate the protective efficacy of ellagic acid in a rat model of rhabdomyolysis induced by glycerol. Sixty healthy adult male Wistar rats weighing between 250 - 300 g were divided into five equal groups including control, rhabdomyolysis (administered 8.00 mL kg⁻¹ of glycerol), and three rhabdomyolysis plus various doses of ellagic acid (25.00, 50.00 and 100 mg kg⁻¹ per day; 72 hr after receiving glycerol for 14 days successively) groups. Serum levels of BUN, creatinine, lactate dehydrogenase, alkaline phosphatase, electrolytes and inflammatory cytokines were evaluated in all rats. Histopathological studies were also performed on kidney tissues from all groups. The administration of ellagic acid resulted in a significant increase in renal function biomarkers compared to the rats with acute kidney injury. This increase was consistent with notable reductions in tumor necrosis factor-α levels and increases in interleukin-10 levels observed in blood samples. Furthermore, the improvement in histopathological indices observed in rats received ellagic acid confirmed its nephroprotective role. The results of the current experimental study suggest that ellagic acid can improve kidney damage following glycerol injection, potentially by modulating the inflammatory process.
... Notably, previous research generally suggests that ROS production, lysosome breakdown, and MD can induce NLRP3 inflammasome activation (4). Related reports indicate that mitophagy can be used as an intervention target for NLRP3 inflammasome activation and that promoting mitophagy inhibits the activation of NLRP3 inflammasome (46). Tang et al. confirmed that PINK1-/ parkin-mediated activation of mitophagy protects against renal ischemia-reperfusion-induced renal injury via the reduction of MD, ROS production, and inflammatory response (47). ...
Article
Full-text available
Background Irradiation (IR) promotes inflammation and apoptosis by inducing oxidative stress and/or mitochondrial dysfunction (MD). The kidneys are rich in mitochondria, and mitophagy maintains normal renal function by eliminating damaged mitochondria and minimizing oxidative stress. However, whether astragaloside IV (AS-IV) can play a protective role through the mitophagy pathway is not known. Methods We constructed a radiation injury model using hematoxylin and eosin (HE) staining, blood biochemical analysis, immunohistochemistry, TdT-mediated dUTP nick end labeling (TUNEL) staining, ultrastructural observation, and Western blot analysis to elucidate the AS-IV resistance mechanism for IR-induced renal injury. Results IR induced mitochondrial damage; the increase of creatinine (SCr), blood urea nitrogen (BUN) and uric acid (UA); and the activation of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome and apoptosis in renal tissue. AS-IV administration attenuated the IR-induced MD and reactive oxygen species (ROS) levels in the kidney; enhanced the levels of mitophagy-associated protein [PTEN-induced putative kinase 1 (PINK1)], parkin proteins, and microtubule-associated protein 1 light 3 (LC3) II/I ratio in renal tissues; diminished NLRP3 inflammasome activation-mediated proteins [cleaved cysteinyl aspartate-specific proteinase-1 (caspase-1), interleukin-1β (IL-1β)] and apoptosis-related proteins [cleaved caspase-9, cleaved caspase-3, BCL2-associated X (Bax)]; reduced SCr, BUN, and UA levels; and attenuated the histopathological alterations in renal tissue. Conversely, mitophagy inhibitor cyclosporin A (CsA) suppressed the AS-IV-mediated protection of renal tissue. Conclusions AS-IV can strongly diminish the activation and apoptosis of NLRP3 inflammasome, thus attenuating the renal injury induced by radiation by promoting the PINK1/parkin-mediated mitophagy. These findings suggest that AS-IV is a promising drug for treating IR-induced kidney injury.
... [41] Another kidney insult is mediated by intact heme (Fe 2+ -heme), which is oxidized by lipid peroxidase to generate Fe 3+ -heme, which could induce lipid peroxidation through redox action and cause oxidative damage to the kidney. [41][42][43] ...
Article
Full-text available
Natural disasters are unpredictable and thousands of people are affected yearly. Currently, this risk persists, given the large population living in risk areas prone to suffering another seismic event. Generally, on-site mortality is high and occurs immediately from massive trauma or asphyxia. After surviving the first event, extricated patients are at risk of developing crush syndrome caused by direct physical trauma and compression of the human body with lesions in different tissues. This could lead to several systemic complications, including acute kidney injury (AKI), sepsis, acute respiratory distress syndrome, bleeding, hypovolemic shock, arrhythmias, electrolyte disturbances and disseminated intravascular coagulation. Hence, AKI in this scenario can occur due to many causes, such as rhabdomyolysis, direct renal trauma, hypovolemia and hemodynamic alterations. The most important measure to reduce crush syndrome mortality and prevent the development of crush syndrome related AKI in disaster situations is the immediate start of treatment. Nevertheless, despite optimal therapy delivery, these previous efforts might not suffice the development and progression of AKI, consequently, the indication of extracorporeal blood purification techniques. This narrative review provides a focused overview of crush syndrome-related AKI, including etiology, mechanisms, diagnosis, current treatment, removal of myoglobin and their limitations.
... It represents the leading cause of death following earthquakes. Glycerol-induced rhabdomyolysis in rats is similar to human rhabdomyolysis in terms of histological features and cytokine profile [27]. Inflammation and oxidative damage have been shown to be responsible for AKI induced by glycerol which mimics the human situation of RIAKI [9]. ...
Article
Full-text available
Background: Crush syndrome or what is also known as traumatic rhabdomyolysis, is the leading cause of death following extrication from structural collapse due to earthquakes. Rhabdomyolysis is one of the most common reasons for acute kidney injury (AKI). The present study was designed to investigate the potential curative effect of wheat grass (WG) in acute kidney injury induced by glycerol (rat model of crush syndrome). Methods: Following a 24-hour period of water deprivation, male rats were randomly divided into 4 groups (6 rats each): The first group received an intramuscular (IM) injection of an equivalent volume of 0.9% saline into the hind limbs in divided dosages. The second group received IM injection of a single dose of 50% v/v glycerol in 0.9% saline (10 mL/kg), in equally divided doses to both hind limbs. The third group was injected a single dose of 50% v/v glycerol in 0.9% saline (10 mL/kg), into the hind limbs in divided dosages; then, the animals were administered WG (75 mg/kg, p.o.) twice per day for 3 successive days. The fourth group received only WG (75 mg/kg, p.o.) twice per day for 3 successive days. At the end of the experiments, animals were sac-rificed; blood and kidneys were collected. Interleukin-6 (IL-6), interleukin-18 (IL-18), neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule 1 (KIM-1), glutathione S-transferase (GST), and catalase (CAT) were measured using enzyme-linked immunosorbent assay (ELISA). Superoxide dismutase (SOD), serum creatinine (SCr), blood urea nitrogen (BUN), and creatine kinase (CK) were estimated using assay kits. Nuclear factor kappa B (NF-κB) was measured using immunohistochemical analysis. Using thiobarbituric acid reactive substances (TBARS), the renal level of malondialdehyde (MDA) was determined. Histopathological examination was also performed. Results: The levels of BUN, SCr, the rhabdomyolysis marker CK, the oxidative damage marker MDA, and the inflammatory markers IL-6, IL-18, and NF-κB as well as the tubular injury markers KIM-1 and NGAL are increased in glycerol administration group. All these markers were significantly attenuated in those animals when treated with WG. Also, treatment with WG significantly improved GST, SOD, and CAT activities in glycerol-treated animals. In addition, histopathological changes induced by glycerol in renal tissue were highly improved in animals given WG. Conclusions: The current findings demonstrate that WG has the ability to attenuate acute kidney injury secondary to rhabdomyolysis induced by glycerol in rats by modulating oxidative stress, as well as inflammatory and rhabdomyolysis markers.
... KIM1 and Activin A have been previously studied in AKI, AAV and SLE (10-13), as well as in healthy controls, in whom a reference range of (0.00-398.6 pg./mL) has been proposed for KIM1 (9). We observed comparable levels between subtypes of IIM, indicating a possible area for future exploration into potential pathogenetic mechanisms including myoglobin induced chronic smoldering tubular injury, or the downstream immunological impact of oxidative damage such as the activation of TGF beta pathways (28). Kidney injury has not been extensively explored nor well described in all subtypes of IIMs from a clinical perspective. ...
Article
Full-text available
Introduction The mechanisms leading to chronic kidney disease (CKD) in patients with idiopathic inflammatory myopathies (IIMs) are poorly understood. We assessed the prevalence of subclinical renal injury in patients with IIMs, through elevation in biomarker levels of tubular injury and fibrosis (NGAL, KIM1, Activin A, CD163, and Cys-c), and assessed differences between subtypes of IIMs, and the effect of disease activity and duration. Materials and methods Clinical data, core set measures, sera and urine were prospectively collected from all patients enrolled in the MyoCite cohort from 2017 to 2021. Twenty healthy subjects (HC) and 16 patients with acute kidney injury (AKI) were included as controls. Baseline and follow up data for IIMs were included. Enzyme-linked immunosorbent assay (ELISA) was used to measure urine NGAL (Human Lipocalin-2/NGAL Duoset ELISA, Cat no: DY1757), KIM1 (Human TIM-1/KIM 1/HAVCR Duoset ELISA, Cat.no: DY1750B), Activin A (Human Activin A Duoset ELISA, Cat no: DY338), CD163 (Human CD163 Duoset ELISA,Cat no: DY1607-05), and Cys-c (Human Cystatin C Duoset ELISA, Cat. no.: DY1196) levels, while eGFR (unit mL/min/1.73 m2) was calculated by the Cockcroft-Gault formula and CKD-EPI formula. Results Analysis of 201 visits of 110 adult patients with IIMs indicated higher normalized biomarker levels compared to HCs, and comparable to patients with AKI, with the exception of NGAL, which was higher in the AKI group. Notably 72 (49%) patients with IIMs had eGFR<90; the levels of the 5 biomarkers were comparable between active and inactive IIMs, and different subtypes of IIMs. Similarly, a poor correlation between urine biomarker levels and core set measures of activity and damage was found. Changes in biomarker levels on follow-up did not correlate with eGFR changes. Discussion This exploratory analysis of urinary biomarkers identified low eGFR and elevated biomarkers of CKD in nearly half of the patients with IIMs, comparable to patients with AKI and higher than HCs, indicative of potential renal damage in IIMs that may have a lead to complications in other systems.
Preprint
Full-text available
Acute kidney injury (AKI) is in high prevalence worldwide, but with no therapeutic strategies. Targeting programmed cell death in tubular epithelial cells has been reported to improve a variety of AKI, but the main pathways and mechanisms of programmed cell death are controversial. In further analysis of previous single-cell RNA-seq data, we identified that pyroptosis was primarily responsible to AKI progression, highly relating with ATP depletion. Herein, we found that FAM3A, a mitochondrial protein responsible for ATP synthesis, was decreased and negatively correlated with tubular injury and pyroptosis in both mice and patients with AKI. Knockout of FAM3A further worsened tubular damage and renal function deterioration, increased macrophage and neutrophil infiltration, and facilitated tubular cell pyroptosis in ischemia/reperfusion injury (IRI) model. Conversely, FAM3A overexpression improved kidney injury and alleviated pyroptosis in IRI or cisplatin AKI. Mechanistically, FAM3A depletion suppressed PI3K/AKT/NRF2 signaling, thus leading to mitochondrial dysfunction and mt-ROS accumulation. NLRP3 inflammasome sensed the overloaded mt-ROS and activated Caspase-1. The activated Caspase-1 then cleaved GSDMD, pro-IL-1β, and pro-IL-18 into their mature forms to mediate pyroptosis. The pro-pyroptotic effects of FAM3A depletion were alleviated after treatment whit NRF2 activator, while the anti-pyroptotic function of FAM3A was blocked by deletion of NRF2. Hence, our study provides new mechanisms for AKI progression and demonstrated that FAM3A is a potential therapeutic target for treating AKI.
Article
Contrast-induced acute kidney injury (CI-AKI) is a serious and common complication in patients receiving intravenous iodinated contrast medium (CM). Clinically, congestive heart failure is the most critical risk factor for CI-AKI and always leads to renal congestion for increased central venous pressure and fluid overload. Here, we aimed to investigate a novel CI-AKI rat model based on renal congestion. After the exploratory testing phase, we successfully constructed a CI-AKI rat model by inducing renal congestion by clamping the unilateral renal vein, removing the contralateral kidney, and a single tail vein injection of Iohexol. This novel CI-AKI rat model showed elevated serum creatinine, urea nitrogen, and released tubular injury biomarkers (KIM-1 and NGAL), reduced glomerular filtration rate, and typical pathologic features of CM-induced tubular injury with extensive foamy degeneration, tubular edema, and necrosis. Electron microscopy and confocal laser scanning revealed excessive mitochondrial fission and increased translocation of Drp1 from the cytoplasm to the mitochondrial surface in tubular epithelial cells. As a Drp1 inhibitor, Mdivi-1 attenuated excessive mitochondrial fission and exerted reno-protection against CM injury. Simultaneously, Mdivi-1 alleviated oxidative stress, apoptosis, and inflammatory responses induced by CM toxicity. We concluded that renal congestion exacerbated CM toxicity and presented a novel CI-AKI rat model. Excessive mitochondrial fission plays a crucial role in CM reno-toxicity and is a promising target for preventing and treating CI-AKI.
Article
Full-text available
Acute kidney injury is a common complication of rhabdomyolysis. A better understanding of this syndrome may be useful to identify novel therapeutic targets because there is no specific treatment so far. Ferroptosis is an iron‐dependent form of regulated nonapoptotic cell death that is involved in renal injury. In this study, we investigated whether ferroptosis is associated with rhabdomyolysis‐mediated renal damage, and we studied the therapeutic effect of curcumin, a powerful antioxidant with renoprotective properties. Induction of rhabdomyolysis in mice increased serum creatinine levels, endothelial damage, inflammatory chemokines, and cytokine expression, alteration of redox balance (increased lipid peroxidation and decreased antioxidant defenses), and tubular cell death. Treatment with curcumin initiated before or after rhabdomyolysis induction ameliorated all these pathologic and molecular alterations. Although apoptosis or receptor‐interacting protein kinase (RIPK)3‐mediated necroptosis were activated in rhabdomyolysis, our results suggest a key role of ferroptosis. Thus, treatment with ferrostatin 1, a ferroptosis inhibitor, improved renal function in glycerol‐injected mice, whereas no beneficial effects were observed with the pan‐caspase inhibitor carbobenzoxy‐valyl‐alanyl‐aspartyl‐(O‐methyl)‐fluoromethylketone or in RIPK3‐deficient mice. In cultured renal tubular cells, myoglobin (Mb) induced ferroptosis‐sensitive cell death that was also inhibited by curcumin. Mechanistic in vitro studies showed that curcumin reduced Mb‐mediated inflammation and oxidative stress by inhibiting the TLR4/NF‐κB axis and activating the cytoprotective enzyme heme oxygenase 1. Our findings are the first to demonstrate the involvement of ferroptosis in rhabdomyolysis‐associated renal damage and its sensitivity to curcumin treatment. Therefore, curcumin may be a potential therapeutic approach for patients with this syndrome.—Guerrero‐Hue, M., García‐Caballero, C., Palomino‐Antolín, A., Rubio‐Navarro, A., Vázquez‐Carballo, C., Herencia, C., Martín‐Sanchez, D., Farré‐Alins, V., Egea, J., Cannata, P., Praga, M., Ortiz, A., Egido, J., Sanz, A. B., Moreno, J. A. Curcumin reduces renal damage associated with rhabdomyolysis by decreasing ferroptosis‐mediated cell death. FASEB J. 33,8961–8975 (2019). www.fasebj.org
Article
Full-text available
Ferroptosis is a newly identified form of nonapoptotic regulated cell death (RCD) characterized by iron-dependent accumulation of lipid peroxides. It is morphologically and biochemically different from known types of cell death. Ferroptosis plays a vital role in the treatment of tumors, renal failure, and ischemia reperfusion injury (IRI). Inhibition of glutathione peroxidase 4 (GPX4), starvation of cysteine, and peroxidation of arachidonoyl (AA) trigger ferroptosis in the cells. Iron chelators, lipophilic antioxidants, and specific inhibitor prevent ferroptosis. Although massive researches have demonstrated the importance of ferroptosis in human, its mechanism is not really clear. In this review, we distanced ourselves from this confusion by dividing the mechanisms of ferroptosis into two aspects: processes that facilitate the formation of lipid peroxides and processes that suppress the reduction of lipid peroxides. At the same time, we summarize the relations between ferroptosis and several types of cell death.
Article
Full-text available
The NOD-like receptor family, pyrin domain containing-3 (NLRP3) inflammasome has been implicated in renal inflammation and fibrosis. However, the biological function of inflammasome-independent NLRP3 in non-immune cells is still unclear. We evaluated the role of inflammasome-independent NLRP3 in renal tubular cells and assessed the value of NLRP3 as a therapeutic target for acute kidney injury (AKI). Various renal tubular cell lines and primary cultured tubular cells from NLRP3 knockout (KO) mice were used for in vitro studies. We also tested the role of tubular NLRP3 in AKI with a unilateral ureter obstruction model (UUO). Hypoxia induced significant increase of NLRP3 independent of ASC, caspase-1, and IL-1β. NLRP3 in renal tubular cells relocalized from the cytosol to the mitochondria during hypoxia and bound to mitochondrial antiviral signal protein (MAVS). The deletion of NLRP3 or MAVS in renal tubular cells attenuated mitochondrial reactive oxygen species (ROS) production and depolarization of the mitochondrial membrane potentials under hypoxia. In response to UUO, NLRP3 KO mice showed less fibrosis, apoptosis, and ROS injury than wild type (WT) mice. Compared with WT kidney, mitophagy was up-regulated in NLRP3 KO kidney relative to the baseline and it was protective against AKI. Our results indicate that inflammasome-independent NLRP3 in renal tubular cells plays important role in mitochondrial ROS production and injury by binding to MAVS after hypoxic injury. This mitochondrial regulation in the absence of NLRP3 increases autophagy and attenuates apoptosis after UUO. We suggest that inflammasome-independent NLRP3 could be a therapeutic target of AKI to prevent the progression of chronic kidney disease.
Article
Full-text available
Acute kidney injury is a clinical syndrome that can be caused by numerous diseases including acute tubular necrosis (ATN). ATN evolves in several phases, all of which are accompanied by different immune mechanisms as an integral component of the disease process. In the early injury phase, regulated necrosis, damage-associated molecular patterns, danger sensing, and neutrophil-driven sterile inflammation enhance each other and contribute to the crescendo of necroinflammation and tissue injury. In the late injury phase, renal dysfunction becomes clinically apparent, and M1 macrophage-driven sterile inflammation contributes to ongoing necroinflammation and renal dysfunction. In the recovery phase, M2-macrophages and anti-inflammatory mediators counteract the inflammatory process, and compensatory remnant nephron and cell hypertrophy promote an early functional recovery of renal function, while some tubules are still badly injured and necrotic material is removed by phagocytes. The resolution of inflammation is required to promote the intrinsic regenerative capacity of tubules to replace at least some of the necrotic cells. Several immune mechanisms support this wound-healing-like re-epithelialization process. Similar to wound healing, this response is associated with mesenchymal healing, with a profound immune cell contribution in terms of collagen production and secretion of profibrotic mediators. These and numerous other factors determine whether, in the chronic phase, persistent loss of nephrons and hyperfunction of remnant nephrons will result in stable renal function or progress to decline of renal function such as progressive chronic kidney disease.
Article
Full-text available
The endoplasmic reticulum (ER) and mitochondria are tightly associated with very dynamic platforms termed mitochondria-associated membranes (MAMs). MAMs provide an excellent scaffold for crosstalk between the ER and mitochondria and play a pivotal role in different signaling pathways that allow rapid exchange of biological molecules to maintain cellular health. However, dysfunctions in the ER–mitochondria architecture are associated with pathological conditions and human diseases. Inflammation has emerged as one of the various pathways that MAMs control. Inflammasome components and other inflammatory factors promote the release of pro-inflammatory cytokines that sustain pathological conditions. In this review, we summarize the critical role of MAMs in initiating inflammation in the cellular defense against pathogenic infections and the association of MAMs with inflammation-mediated diseases.
Article
Full-text available
Primary/secondary hyperoxalurias involve nephrocalcinosis-related chronic kidney disease (CKD) leading to end-stage kidney disease. Mechanistically, intrarenal calcium oxalate crystal deposition is thought to elicit inflammation, tubular injury and atrophy, involving the NLRP3 inflammasome. Here, we found that mice deficient in NLRP3 and ASC adaptor protein failed to develop nephrocalcinosis, compromising conclusions on nephrocalcinosis-related CKD. In contrast, hyperoxaluric wild-type mice developed profound nephrocalcinosis. NLRP3 inhibition using the β-hydroxybutyrate precursor 1,3-butanediol protected such mice from nephrocalcinosis-related CKD. Interestingly, the IL-1 inhibitor anakinra had no such effect, suggesting IL-1-independent functions of NLRP3. NLRP3 inhibition using 1,3-butanediol treatment induced a shift of infiltrating renal macrophages from pro-inflammatory (CD45+F4/80+CD11b+CX3CR1+CD206-) and pro-fibrotic (CD45+F4/80+CD11b+CX3CR1+CD206+TGFβ+) to an anti-inflammatory (CD45+F4/80+CD11b+CD206+TGFβ-) phenotype, and prevented renal fibrosis. Finally, in vitro studies with primary murine fibroblasts confirmed the non-redundant role of NLRP3 in the TGF-β signaling pathway for fibroblast activation and proliferation independent of the NLRP3 inflammasome complex formation. Thus, nephrocalcinosis-related CKD involves NLRP3 but not necessarily via intrarenal IL-1 release but rather via other biological functions including TGFR signaling and macrophage polarization. Hence, NLRP3 may be a promising therapeutic target in hyperoxaluria and nephrocalcinosis.
Article
Full-text available
Background: Acute kidney injury (AKI) is the most common and life-threatening systemic complication of rhabdomyolysis. Inflammation plays an important role in the development of rhabdomyolysis-induced AKI. This study aimed to investigate the kidney model of AKI caused by rhabdomyolysis to verify the role of macrophage Toll-like receptor 4/nuclear factor-kappa B (TLR4/NF-κB) signaling pathway. Methods: C57BL/6 mice were injected with a 50% glycerin solution at bilateral back limbs to induce rhabdomyolysis, and CLI-095 or pyrrolidine dithiocarbamate (PDTC) was intraperitoneally injected at 0.5 h before molding. Serum creatinine levels, creatine kinase, the expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6, and hematoxylin and eosin stainings of kidney tissues were tested. The infiltration of macrophage, mRNA levels, and protein expression of TLR4 and NF-κB were investigated by immunofluorescence double-staining techniques, reverse transcriptase-quantitative polymerase chain reaction, and Western blotting, respectively. In vitro, macrophage RAW264.7 was stimulated by ferrous myoglobin; the cytokines, TLR4 and NF-κB expressions were also detected. Results: In an in vivo study, using CLI-095 or PDTC to block TLR4/NF-κB, functional and histologic results showed that the inhibition of TLR4 or NF-κB alleviated glycerol-induced renal damages (P < 0.01). CLI-095 or PDTC administration suppressed proinflammatory cytokine (TNF-α, IL-6, and IL-1β) production and macrophage infiltration into the kidney (P < 0.01). Moreover, in an in vitro study, CLI-095 or PDTC suppressed myoglobin-induced expression of TLR4, NF-κB, and proinflammatory cytokine levels in macrophage RAW264.7 cells (P < 0.01). Conclusion: The pharmacological inhibition of TLR4/NF-κB exhibited protective effects on rhabdomyolysis-induced AKI by the regulation of proinflammatory cytokine production and macrophage infiltration.
Article
Full-text available
Iodinated contrast media serves as a direct causative factor of acute kidney injury (AKI) and is involved in the progression of cellular dysfunction and apoptosis. Emerging evidence indicates that NLRP3 inflammasome triggers inflammation, apoptosis and tissue injury during AKI. Nevertheless, the underlying renoprotection mechanism of NLRP3 inflammasome against contrast-induced AKI (CI-AKI) was still uncertain. This study investigated the role of NLRP3 inflammasome in CI-AKI both in vitro and in vivo. In HK-2 cells and unilateral nephrectomy model, NLRP3 and NLRP3 inflammasome member ASC were significantly augmented with the treatment of contrast media. Moreover, genetic disruption of NLRP3 notably reversed contrast-induced expression of apoptosis related proteins and secretion of proinflammatory factors, similarly to the effects of ASC deletion. Consistent with above results, absence of NLRP3 in mice undergoing unilateral nephrectomy also protected against contrast media-induced renal cells phenotypic alteration and cell apoptosis via modulating expression level of apoptotic proteins. Collectively, we demonstrated that NLRP3 inflammasome mediated CI-AKI through modulating the apoptotic pathway, which provided a potential therapeutic target for the treatment of contrast media induced acute kidney injury.
Article
Mitochondrial lipids are essential for maintaining the integrity of mitochondrial membranes and the proper functions of mitochondria. As the “powerhouse” of a cell, mitochondria are also the major cellular source of reactive oxygen species (ROS). Oxidative stress occurs when the antioxidant system is overwhelmed by overproduction of ROS. Polyunsaturated fatty acids in mitochondrial membranes are primary targets for ROS attack, which may lead to lipid peroxidation (LPO) and generation of reactive lipids, such as 4-hydroxynonenal. When mitochondrial lipids are oxidized, the integrity and function of mitochondria may be compromised and this may eventually lead to mitochondrial dysfunction, which has been associated with many human diseases including cancer, cardiovascular diseases, diabetes, and neurodegenerative diseases. How mitochondrial lipids are oxidized and the underlying molecular mechanisms and pathophysiological consequences associated with mitochondrial LPO remain poorly defined. Oxidation of the mitochondria-specific phospholipid cardiolipin and generation of bioactive lipids through mitochondrial LPO has been increasingly recognized as an important event orchestrating apoptosis, metabolic reprogramming of energy production, mitophagy, and immune responses. In this review, we focus on the current understanding of how mitochondrial LPO and generation of bioactive lipid mediators in mitochondria are involved in the modulation of mitochondrial functions in the context of relevant human diseases associated with oxidative stress.
Article
Ferroptosis is a form of regulated necrotic cell death controlled by glutathione peroxidase 4 (GPX4). At present, mechanisms that could predict sensitivity and/or resistance and that may be exploited to modulate ferroptosis are needed. We applied two independent approaches-a genome-wide CRISPR-based genetic screen and microarray analysis of ferroptosis-resistant cell lines-to uncover acyl-CoA synthetase long-chain family member 4 (ACSL4) as an essential component for ferroptosis execution. Specifically, Gpx4-Acsl4 double-knockout cells showed marked resistance to ferroptosis. Mechanistically, ACSL4 enriched cellular membranes with long polyunsaturated ω6 fatty acids. Moreover, ACSL4 was preferentially expressed in a panel of basal-like breast cancer cell lines and predicted their sensitivity to ferroptosis. Pharmacological targeting of ACSL4 with thiazolidinediones, a class of antidiabetic compound, ameliorated tissue demise in a mouse model of ferroptosis, suggesting that ACSL4 inhibition is a viable therapeutic approach to preventing ferroptosis-related diseases.