Hydroxybutyrate Attenuates Renal Ischemia-Reperfusion Injury Through Its Anti-Pyroptotic Effects

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

www.kidney-international.

org basic research

b-hydroxybutyrate attenuates renal


ischemia-reperfusion injury through
its anti-pyroptotic effects OPEN

Takaya Tajima1, Ayumi Yoshifuji1, Ayumi Matsui1, Tomoaki Itoh1, Kiyotaka Uchiyama1, Takeshi Kanda1,
Hirobumi Tokuyama1, Shu Wakino1 and Hiroshi Itoh1
1
Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan

Ketone bodies including b-hydroxybutyrate (b-OHB) have


been shown to protect against ischemic tissue injury when Translational Statement
present at low concentrations. We evaluated the impact of
b-OHB on renal ischemia/reperfusion injury (IRI). Mice were In this study, endogenous ketone body, b-hydrox-
treated with a continuous infusion of b-OHB using an ybutyrate is shown to alleviate renal ischemia-reperfu-
sion injury through its antipyroptotic effects. Its effects
osmotic mini-pump before and after IRI. We also tested the
were dependent on forkhead transcription factor O3
effects of increasing endogenous serum b-OHB levels by
induction and associated with epigenetic gene regula-
fasting. Renal IRI was attenuated by b-OHB treatment tion through increased histone acetylation. Dietary or
compared to saline control, with similar results in the pharmacological approaches to elevate b-hydrox-
fasting condition. b-OHB treatment reduced the number of ybutyrate levels hold promise in reducing the severity of
terminal deoxynucleotidyl transferase–mediated dUTP nick acute kidney injury including postsurgery acute kidney
end-labeling (TUNEL)-positive cells and increased injury and ensued chronic kidney disease progression as
expression of forkhead transcription factor O3 (FOXO3), an well. Of note, because the involvement of epigenetic
upstream regulator of pyroptosis. Although b-OHB gene modulation, priming gene expressions by
treatment did not impact markers of apoptosis, it b-hydroxybutyrate before renal insults including surgery
decreased the expression of caspase-1 and can protect the kidney against acute kidney injury. Novel
therapy, the pretreatment with b-hydroxybutyrate, can
proinflammatory cytokines, indicating that b-OHB blocked
be a novel prophylactic strategy against acute kidney
pyroptosis. In a human proximal tubular cell line exposed
injury.
to hypoxia and reoxygenation, b-OHB reduced cell death in
a FOXO3-dependent fashion. Histone acetylation was
decreased in kidneys exposed to IRI and in proximal
etone bodies are small, lipid-derived molecules that
tubular cells exposed to hypoxia and reoxygenation, and
this effect was ameliorated by b-OHB through the
inactivation of histone deacetylases. In vitro, b-OHB
treatment restored histone acetylation at the FOXO3
K serve as a circulating energy source for tissues in times
of fasting or prolonged exercise.1 The term "endoge-
nous ketone bodies" refers to 3 molecules: acetoacetate, b-
hydroxybutyrate (b-OHB), and acetone. Most ketone bodies
promoter. Consistent with epigenetic molecular effects, the
are produced in the liver, although smaller amounts may be
renoprotective effects of b-OHB were still observed when
produced in other tissues.2 The level of b-OHB in blood in-
the continuous infusion was stopped at the time of IRI.
creases to 1 to 2 mmol/l while fasting, during which the liver
Thus, b-OHB attenuates renal IRI through anti-pyroptotic
switches to fatty acid oxidation,2,3 and to even higher concen-
effects, likely mediated by an epigenetic effect on FOXO3
trations during prolonged periods of fasting (6–8 mmol/l)4
expression.
and in diabetic ketoacidosis (>25 mmol/l).5 Many studies
Kidney International (2019) -, -–-; https://doi.org/10.1016/
j.kint.2018.11.034
have shown that ketosis is associated with accelerated aging.
Thus, increased ketone levels in the body cause age-related
KEYWORDS: acute kidney injury; cell death; cell survival; inflammation;
ischemia reperfusion metabolic diseases.1 However, accumulating evidence has
Copyright ª 2019, International Society of Nephrology. Published by shown that ketone bodies in low concentrations have benefi-
Elsevier Inc. This is an open access article under the CC BY-NC-ND license cial effects. Some studies have shown that b-OHB, at low con-
(http://creativecommons.org/licenses/by-nc-nd/4.0/). centrations, has antioxidative and anti-inflammatory effects6
and that exogenous b-OHB has therapeutic effects in stress
Correspondence: Shu Wakino, Department of Internal Medicine, School of conditions, such as hemorrhagic shock,7,8 extensive burns,9
Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, and cerebral hypoxia, anoxia, and ischemia.10 One of the pu-
Japan. E-mail: shuwakino@z8.keio.jp tative mechanisms where ketone has a protective role against
Received 16 April 2018; revised 16 November 2018; accepted 21 tissue injuries is its effects on pyroptosis.11 Pyroptosis is a
November 2018 unique type of programmed cell death that is distinct from

Kidney International (2019) -, -–- 1


basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

Figure 1 | The effects of b–hydroxybutyrate (b-OHB) on renal function and histology. (a) b-OHB plasma levels were measured 24 hours
before and after sham or ischemia-reperfusion (IR) operation. (b,c) Blood urea nitrogen levels (b) and serum levels of (continued)

2 Kidney International (2019) -, -–-


T Tajima et al.: Effects of b-hydroxybutyrate on IRI basic research

apoptosis and necrosis.12,13 Caspase-1 is a key molecule that P < 0.01) (Figure 1b). Serum creatinine level also significantly
leads to the induction of pyroptosis. Proinflammatory factors, increased to 0.43  0.09 mg/dl in the phosphate-buffered
such as pro-interleukin (IL)-1b and pro-IL-18, can be acti- saline–treated IR group compared with that in the
vated by caspase-1 cleavage and trigger or aggravate inflam- phosphate-buffered saline–treated sham group (0.14  0.01
matory responses.14–16 Therefore, pyroptosis may not only mg/dl, P < 0.01). Treatment with b-OHB reduced the level of
lead to cell death, but it may also play an important role in serum creatinine in IR-injured mice (0.24  0.02 mg/dl, P <
the cascade of reactions that lead to damaged surrounding tis- 0.05) (Figure 1c). In histological findings, IR-injured kidneys
sues.17 One of the upstream molecules of pyroptosis is forkhead exhibited brush border loss, vacuolization, and desquamation
transcription factor O3 (FOXO3). FOXO3 is a major transcrip- of epithelial cells in renal tubules. Tubular injuries were
tion factor that has functions associated with cell cycle arrest, assessed by tubule-interstitial injury score both in hematox-
oxidative scavenging, cell proliferation, survival, and death.18,19 ylin and eosin section (Figure 1d) and in periodic acid–Schiff
FOXO3 upregulates the expression of apoptosis repressor with section (Figure 1e) as described in the Materials and Methods
caspase recruitment domain (ARC), which leads to section. Both assays revealed that IR significantly increased
downregulation of the expression of caspase-1 and downstream tubule-interstitial injury scores. The tubule-intestinal injury
proinflammatory factors for pyroptosis.20–22 It has been re- assessed by these scores was significantly attenuated by the
ported that b-OHB treatment almost completely blocked pyrop- treatment with b-OHB (Figure 1d and e).
tosis induced by adenosine triphosphate and monosodium urate
crystals in macrophages,11 although the effects on renal protec- The effects of 2-day fasting and b-OHB on renal function in
tion through the regulation of pyroptosis are scarcely reported. mice with IR injury
In this study, we explored the effects of b-OHB on acute Because fasting condition is well known to increase serum
renal injuries induced by ischemia-reperfusion (IR) insults levels of b-OHB endogenously, we tested the effects of fasting
and elucidated the molecular mechanisms behind this effect. on the renal function with IR injuries. Male C57BL/6J mice
We demonstrated b-OHB has an antipyroptotic potency and were either fasted for 2 days or fed ad libitum (nonfasted)
protective function against renal IR injuries. preceding surgery. After 2 days fasting, the body weight
significantly decreased as compared to that in nonfasted
RESULTS group (Figure 2a). The plasma levels of b-OHB increased in
The effects of b-OHB on renal function and renal histology in the fasted sham and the fasted IR groups as compared to
mice with IR injury those in nonfasted sham and nonfasted IR groups (Figure 2b).
We used implanted osmotic pumps to administer exogenous The level of blood urea nitrogen and creatinine significantly
b-OHB or phosphate-buffered saline. The plasma levels of b- increased in the nonfasted IR group compared with that in
OHB increased to 1.28  0.34 mmol/l in the b-OHB-treated the nonfasted sham group. Fasting preconditioning reduced
sham group versus 0.33  0.05 mmol/l in phosphate-buffered the levels of both blood urea nitrogen and creatinine in IR-
saline–treated sham group (P < 0.01), and to 1.35  0.36 injured mice (Figure 2c and d, respectively). These data
mmol/l in the b-OHB-treated IR group versus 0.31  0.04 indicated that fasting reversed renal dysfunction in IR-injured
mmol/l in phosphate-buffered saline–treated IR group (P < mouse kidneys, which can be associated with the increase in
0.01), 24 hours after the administration of b-OHB. Twenty- serum b-OHB levels. We further examined an additive effect
four hours after the operation, plasma levels of b-OHB by fasting and continuous infusion of b-OHB. The fasting
were sustained at 1.40  0.28 mmol/l in the b-OHB-treated group was fasted for 2 days preoperatively and implanted with
sham group versus 0.37  0.05 mmol/l in phosphate-buffered osmotic pumps to administer exogenous b-OHB or
saline–treated sham group (P < 0.01), and at 1.33  0.32 phosphate-buffered saline for 1 day before surgery. After 2
mmol/l in the b-OHB-treated IR group versus 0.35  0.05 days, the mean weight decreased in fasting groups both with
mmol/l in phosphate-buffered saline–treated IR group (P < and without b-OHB infusion. The administration of b-OHB
0.01) (Figure 1a). The level of blood urea nitrogen signifi- for 1 day did not affect body weight loss (Figure 2e). The
cantly increased to 86.6  9.54 mg/dl in the phosphate- plasma levels of b-OHB increased both in the nonfasted IR
buffered saline–treated IR group compared with that in the with b-OHB group and in the fasted IR group as compared to
phosphate-buffered saline–treated sham group (33.0  1.38 the nonfasted IR group. The level of b-OHB further increased
mg/dl, P < 0.01). b-OHB treatment reduced the level of in the fasted IR with b-OHB group at the operation to the
blood urea nitrogen in IR-injured mice (47.0  3.05 mg/dl, concentration of 4.73  0.24 mmol/l (Figure 2f). The level of
=
Figure 1 | (continued) creatinine (c) in each mice group; phosphate-buffered saline–treated sham-operated mice, b-OHB-treated sham
(shamþb-OHB), phosphate-buffered saline–treated IR-operated mice, and b-OHB-treated IR (IRþb-OHB). (d) Representative kidney section
stained for hematoxylin and eosin in each mice group. Arrowheads indicate inflammatory cells or cell debris. Bar graph in the lower panel
represents semiquantitative analysis of tubule interstitial injury. (e) Representative kidney section stained with periodic acid–Schiff in each mice
group. Arrowheads indicate brush border. Bar graph in the lower panel represents semiquantitative analysis of tubular injury. (d,e) Original
magnification 200. Insets: increased magnification 500. Bars ¼ 50 mm. Insets are more magnified view of tubular cells. Inset bars ¼ 20 mm.
##
P < 0.01 versus sham, **P < 0.01 versus IR, *P < 0.05 versus IR. n ¼ 6 for sham and shamþb-OHB, n ¼ 8 for IR and IRþb-OHB. Bar represents
mean  SEM. To optimize viewing of this image, please see the online version of this article at www.kidney-international.org.

Kidney International (2019) -, -–- 3


basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

Figure 2 | The effects of 2-day fasting and b-hydroxybutyrate (b-OHB) on renal function in mice with ischemia-reperfusion (IR) injury.
(a–d) The effects of 48-hour fasting on IR injury. Body weights (a) and b-OHB plasma levels (b) were measured 48 hours before (continued)

4 Kidney International (2019) -, -–-


T Tajima et al.: Effects of b-hydroxybutyrate on IRI basic research

both blood urea nitrogen and creatinine significantly The effects of b-OHB on pyroptosis in hypoxic HK-2 cells
increased in the nonfasted IR group compared with that in The effects of b-OHB on pyroptosis were examined using a
the nonfasted sham group. Both fasting and the treatment of human proximal tubular cell line, human kidney-2 (HK-2)
b-OHB reduced the level of blood urea nitrogen and creati- cells, which were exposed to hypoxia (5% CO2, 1% O2) for 24
nine in IR-injured mice. However, combined treatment with hours followed by 12 hours of reoxygenation (5% CO2, 21%
fasting and b-OHB treatment failed to show an additive effect O2) with or without b-OHB. Cell viability assay revealed that
on these levels to that of fasting condition and b-OHB 0 to 10 mmol/l b-OHB had little effect on cell viability. Cell
infusion (Figure 2g and h). These data indicated ketone viability decreased in hypoxic-treated HK-2 cells compared
bodies in low concentrations have beneficial effects, on the with that in normoxic HK-2 cells, although cell viability
other hand, high b-OHB level around 4 to 5 mmol/l might significantly decreased in normoxic HK-2 cells by the treat-
not show higher renoprotective effects or possibly may ment with 20 and 40 mmol/l b-OHB. These data indicated
worsen acute kidney injury. the high concentration of b-OHB has cellular toxic effects.
The treatment with b-OHB significantly improved cell
b-OHB suppressed the pyroptosis in IR-injured kidneys viability in hypoxic HK-2 cells up to the concentration of 10
To evaluate the effect of b-OHB on renal cell deaths in IR- mmol/l (Figure 4a), while higher concentration of b-OHB
injured kidneys, we first performed the terminal deoxy- decreased cell viability. To evaluate the effect of b-OHB on
nucleotidyl transferase–mediated dUTP nick end-labeling pyroptosis and apoptosis, we measured the expression of
(TUNEL) assay, the classical method for the identification genes related to pyroptosis and apoptosis activation,
of the apoptotic cells. TUNEL assay showed a significant in- including FOXO3 (Figure 4b), ARC (Figure 4c), caspase-1
crease in TUNEL-positive cell number in kidneys of IR- (Figure 4d), the proinflammatory cytokines IL-1b
injured mice as compared to that of sham-treated mice. (Figure 4e), IL-18 (Figure 4f), Bax (Figure 4g), and Bcl-2
Treatment with b-OHB significantly reduced the number of (Figure 4h). The mRNA expression levels of FOXO3 and
TUNEL-positive cells in IR-injured kidneys (Figure 3a). This ARC significantly decreased in hypoxic HK-2 cells as
suggested that b-OHB has renal protective roles in IR injury compared to those in normoxic HK-2 cells. These decreases
by its antiapoptotic effects. However, the expressions of were ameliorated by treatment with b-OHB at the dose of 10
apoptotic molecular markers, neither Bax (Figure 3b and c) mmol/l (Figure 4b and c, respectively). The mRNA expres-
nor Bcl-2 (Figure 3d and e) were affected by the b-OHB sion levels of caspase-1, IL-1b, and IL-18 significantly
treatment. We focused on another type of cell death, pyrop- increased in hypoxic HK-2 cells as compared to those in
tosis, and explored the expressions of genes related to normoxic HK-2 cells. These increases were attenuated by the
pyroptosis, including FOXO3, ARC, caspase-1, and the treatment with b-OHB at the dose of 10 mmol/l (Figure 4d, e,
proinflammatory cytokines IL-1b and IL-18. The mRNA and f, respectively). The mRNA expression levels of Bax were
expression and protein expression levels of FOXO3 signifi- significantly increased and those of Bcl-2 were significantly
cantly decreased in IR-injured kidneys compared with those decreased in hypoxic HK-2 cells as compared to those in
in kidneys of sham-treated mice (Figure 3f and g, respec- normoxic HK-2 cells. These increases or decreases were not
tively). Treatment with b-OHB increased these expression affected by treatment b-OHB (Figure 4g and h, respectively).
levels of FOXO3 in IR-injured kidneys. The mRNA expression These data indicated that these in vivo effects of treatment
level of ARC (Figure 3h) significantly decreased in IR-injured with b-OHB in renal IR injury can be due to the direct
kidneys as compared with this level in the kidneys of sham- antipyroptotic action on renal proximal tubular cells.
treated mice. Treatment with b-OHB increased the mRNA In IR injury, reperfusion insult following ischemia is very
expression level of ARC in IR-injured kidneys. Consistently, critical due to the increase in oxidative stress followed by the
the mRNA expression levels of caspase-1 (Figure 3i), IL-1b induction of inflammatory response and cell death. To
(Figure 3j), and IL-18 (Figure 3k) significantly increased explore the effect of b-OHB on the oxidative stress, we treated
in IR-injured kidneys as compared with those in kidneys them with H2O2, which is the main culprit in the patho-
of sham-treated mice. Treatment with b-OHB decreased genesis of IR injury.23 The mRNA expression level of FOXO3
the mRNA expression levels of these genes in IR-injured significantly decreased in H2O2-treated HK-2 cells at the
kidneys. concentration of 2 mmol/l (Figure 4i). Cell viability decreased

=
Figure 2 | (continued) and at the time of the sham or IR operation. Blood urea nitrogen levels (c) and serum levels of creatinine
(d) were measured 24 hours after the sham or IR operation. ##P < 0.01 versus nonfasted sham-operated mice (shamþnonfasted),
#
P < 0.05 versus shamþnonfasted, **P < 0.01 versus nonfasted IR-operated mice (IRþnonfasted), and *P < 0.05 versus IRþnonfasted. n ¼ 3
for shamþnonfasted and shamþfasted; n ¼ 4 for IRþnonfasted and IRþfasted. (e–h) The combined effects of b-OHB treatment and
fasting on IR injury. Body weights (e) and b-OHB plasma levels (f) were measured 48 hours before and at the time of the sham or IR operation.
##
P < 0.01 versus phosphate-buffered saline–treated nonfasted mice with IR operation (IRþnonfasted), #P < 0.05 versus IRþnonfasted,
and **P < 0.01 versus b-OHB–treated nonfasted mice with IR operation (IRþnonfastedþb-OHB). Blood urea nitrogen levels (g) and serum
levels of creatinine (h) were measured 24 hours after the sham or IR operation. ##P < 0.01 versus phosphate-buffered saline–treated
nonfasted mice with sham operation (shamþnonfasted), **P < 0.01 versus IRþnonfasted, *P < 0.05 versus IRþnonfasted,
$
P < 0.05 versus IRþnonfastedþb-OHB, ¶¶P < 0.01 versus phosphate-buffered saline–treated fasted mice with IR operation (IRþfasted),
and ¶P < 0.05 versus IRþfasted. n ¼ 6 for each mouse group. Bar represents mean  SEM.
Kidney International (2019) -, -–- 5
basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

Figure 3 | The effect of b-hydroxybutyrate (b-OHB) on pyroptosis in ischemia-reperfusion (IR) injured kidneys. (a) Representative results
of terminal deoxynucleotidyl transferase–mediated dUTP nick end-labeling (TUNEL) staining of the kidney section in each mice group.
Arrowheads indicate TUNEL-positive cells. Bars ¼ 50 mm. Semiquantitative analysis of TUNEL-positive cells is shown in right panel. Original
magnification 200. (b–k) The quantitative real-time polymerase chain reaction analysis of mRNA expressions and Western blot analysis of
markers related to apoptosis and pyroptosis activation including Bax (b,c), Bcl-2 (d,e), forkhead transcription (continued)

6 Kidney International (2019) -, -–-


T Tajima et al.: Effects of b-hydroxybutyrate on IRI basic research

in H2O2-treated HK-2 cells compared with that in control b-OHB upregulated histone acetylation in IR-injured kidneys
HK-2 cells at 3 hours and 6 hours after the stimulation with and hypoxic HK-2 cells
H2O2. The treatment with b-OHB significantly improved cell b-OHB was recently reported as an endogenous inhibitor of
viability in H2O2-treated HK-2 cells at the dose of 10 mmol/l several histone deacetylases (HDACs), including classes I and
(Figure 4j). To evaluate the effect of b-OHB on pyroptosis in IIa,24 which usually function as gene silencers. It has been
H2O2-treated HK-2 cells, we measured the expression of reported that the treatment of kidney cells with b-OHB
genes related to pyroptosis activation, including FOXO3, increased lysine-9 acetylation in histone H3 (AcH3K9) at the
ARC, caspase-1, and the proinflammatory cytokines IL-1b FOXO3 promoter and caused upregulation of mRNA
and IL-18 (Figure 4k). The mRNA expression levels of expression of FOXO3.24 We supposed that the HDAC inhi-
FOXO3 and ARC significantly decreased in H2O2-treated bition by b-OHB contributed to FOXO3 gene regulation.
HK-2 cells as compared to those in control HK-2 cells. These Immunoblotting analysis using antibodies against AcH3K9
decreases were ameliorated by treatment with b-OHB at the revealed that in IR-injured kidneys, AcH3K9 levels decreased
dose of 10 mmol/l. The mRNA expression levels of caspase-1, compared with those in the kidneys of sham-treated mice.
IL-1b, and IL-18 significantly increased in H2O2-treated Treatment with b-OHB increased AcH3K9 levels in both
HK-2 cells as compared to those in control HK-2 cells. These sham-treated and IR-injured kidneys (Figure 7a). The alter-
increases were attenuated by the treatment with b-OHB at the ation of AcH3K9 levels implied the changes in the activities of
dose of 10 mmol/l. HDACs or histone acetyltransferases (HATs). Both HDAC and
HAT activities decreased in IR-injured kidneys as compared to
The effects of FOXO3 gene knockdown on pyroptotic-related those in sham-treated mice, indicating that the decrease in
genes expression in HK-2 cells
HAT activity played a more significant role in the decreased
Our data suggested that the downregulation of FOXO3 levels of AcH3K9 in IR-injured kidney. The treatment with b-
could contribute to trigger pyroptosis pathway in hyp- OHB further decreased HDACs activities in IR-injured kid-
oxic HK-2 cells. We first examined the effects of FOXO3 neys. On the other hand, b-OHB did not affect HAT activity
expression on pyroptotic-related genes by using gene in IR-injured kidneys (Figure 7b). These data indicated that
silencing of FOXO3. Transfection with small, interfering the reversal of AcH3K9 levels by b-OHB treatment was
RNA (siRNA) targeted against the human FOXO3 gene attributed to its inhibitory effects on HDAC activity. We
downregulated FOXO3 mRNA expression in HK-2 cells further examined this effect of b-OHB on the acetylation of
with or without cellular hypoxic insults by using hyp- H3K9 in HK-2 cells subject to hypoxic insult in hypoxia
oxia incubator (Figure 5a). This siRNA downregulated incubator. We found that histone acetylation levels decreased
the mRNA expressions of ARC (Figure 5b) and up- in hypoxic HK-2 cells as compared to those in normoxic HK-
regulated pyroptosis-related genes caspase-1 (Figure 5c), 2 cells. Treatment with b-OHB at the dose of 10 mmol/l
IL-1b (Figure 5d), and IL-18 (Figure 5e) in HK-2 cells with increased the histone acetylation levels in hypoxic HK-2 cells
or without cellular hypoxic insults. These data indicated (Figure 7c). We subsequently investigated the activities of
that a set of pyroptotic genes is constitutively regulated by HDACs and HATs in HK-2 cells. Both HDAC and HAT activities
the expression levels of FOXO3 in HK-2 cells. decreased in hypoxic HK-2 cells compared with those in nor-
The effects of FOXO3 knockdown on b-OHB mediated moxic HK-2 cells. Treatment with b-OHB at the dose of 10
antipyroptosis effects in hypoxic HK-2 cells mmol/l decreased HDAC activities in hypoxic HK-2 cells, while
We next investigate the potential mechanism of b-OHB- HAT activities were not altered by b-OHB at any dosage of 1 to
mediated antipyroptotic gene regulation. We transfected 10 mmol/l (Figure 7d) in hypoxic HK-2 cells. Finally, chromatin
FOXO3 siRNA into HK-2 cells under hypoxic conditions by immunoprecipitation analysis of the FOXO3 promoter with
hypoxia incubator with the treatment with b-OHB. With this distinct primer pairs for FOXO3 promoter revealed decreased
siRNA, the upregulation of ARC and downregulation of histone H3K9 acetylation at FOXO3 promoter in hypoxic HK-2
caspase-1, IL-1b, and IL-18 by the treatment with b-OHB in cells as compared to those in normoxic HK-2 cells. Treatment
hypoxic HK-2 cells were abrogated (Figure 6a to d, respec- with b-OHB at the dose of 10 mmol/l increased histone H3K9
tively). Finally, the protective effects on hypoxic cell viability acetylation at FOXO3 promoter in hypoxic HK-2 cells
by b-OHB were also blocked by the transfection of siRNA for (Figure 7e). Combined with the previous reports that AcH3K9
FOXO3 gene (Figure 6e). These data indicated that the protec- levels at the FOXO3 promoter regulates FOXO3 gene expres-
tive effects by b-OHB on renal tubular cell was through FOX- sion,24 our data suggested that FOXO3-dependent antipyroptotic
O3-dependent regulatory mechanism of pyroptosis pathway. action by b-OHB might result from the inhibition of HDAC

=
Figure 3 | (continued) factor O3 (FOXO3) (f,g), apoptosis repressor with caspase recruitment domain (ARC) (h), caspase-1 (i), as well
as the proinflammatory cytokines interleukin-1b (IL-1b) (j) and IL-18 (k) in each mouse group. The experimental groups consist of
phosphate-buffered saline–treated mice with sham operation (n ¼ 6), b-OHB–treated sham mice (shamþb-OHB) (n ¼ 6),
phosphate-buffered saline–treated mice with IR injury (n ¼ 8), and b-OHB–treated IR mice (IRþb-OHB) (n ¼ 8). ##P < 0.01 versus sham,
#
P < 0.05 versus sham, **P < 0.01 versus IR, *P < 0.05 versus IR. Bar represents mean  SEM. GAPDH, glyceraldehyde-3-phosphate
dehydrogenase. To optimize viewing of this image, please see the online version of this article at www.kidney-international.org.

Kidney International (2019) -, -–- 7


basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

Figure 4 | The effect of b-hydroxybutyrate (b-OHB) on pyroptosis in hypoxic and H2O2-treated human kidney-2 (HK-2) cells. (a) Cell
viability in normoxic and hypoxic HK-2 cells treated with increasing concentrations of b-OHB. ##P < 0.01 versus normoxic HK-2 (continued)

8 Kidney International (2019) -, -–-


T Tajima et al.: Effects of b-hydroxybutyrate on IRI basic research

Figure 5 | The effect of forkhead transcription factor O3 (FOXO3) knockdown on pyroptosis related genes in human kidney-2 (HK-2)
cells. HK-2 cells were transfected with small, interfering RNAs (siRNAs) specific for the human FOXO3 gene or negative control siRNA and subject
to hypoxic insults by using hypoxia incubator. The quantitative real-time polymerase chain reaction analysis of mRNA expression levels of
FOXO3 (a), apoptosis repressor with caspase recruitment domain (ARC) (b), caspase-1 (c), interleukin-1b (IL-1b) (d), and IL-18 (e). ##P < 0.01 versus
normoxic HK-2 cells with control siRNA, #P < 0.05 versus normoxic HK-2 cells with control siRNA, **P < 0.01 versus hypoxic HK-2 cells with
control siRNA. *P < 0.05 versus hypoxic HK-2 cells with control siRNA. n ¼ 5 in each experimental group. Bar represents mean  SEM.

activity and the amelioration of the decreased acetylation of could sustain even if continuous infusion is stopped and
H3K9 by IR insults. without the infusion of b-OHB just after renal IR insult. We
continuously infused b-OHB just for 24 hours by the time of
The renoprotective effects of b-OHB in the case that IR (24-hour group) and stopped infusing, while in the orig-
continuous infusion is stopped after IR insult inal experiments, we infused b-OHB for 24 hours before and
Considering epigenetic effects by b-OHB, we hypothesized after the time of IR, that is, for 48 hours in total (48-hour
that the effects of b-OHB on pyroptosis in IR-injured kidneys group). The plasma levels of b-OHB increased both in the

=
Figure 4 | (continued) cells, **P < 0.01 versus hypoxic HK-2 cells without b-OHB, *P < 0.05 versus hypoxic HK-2 cells without b-OHB, ¶¶P < 0.01
versus hypoxic HK-2 cells with 10 mmol/l (mM) of b-OHB, ¶P < 0.05 versus hypoxic HK-2 cells with 10 mM of b-OHB. The quantitative
real-time polymerase chain reaction analysis of mRNA expression levels of forkhead transcription factor O3 (FOXO3) (b), apoptosis repressor
with caspase recruitment domain (ARC) (c), caspase-1 (d), interleukin-1b (IL-1b) (e), IL-18 (f), Bax (g), and Bcl-2 (h) in hypoxic HK-2 cells. ##P < 0.01
versus normoxic HK-2 cells, **P < 0.01 versus hypoxic HK-2 cells without b-OHB, *P < 0.05 versus hypoxic HK-2 cells without b-OHB. n ¼ 5 in
each experimental group. (i) Time-dependent FOXO3 mRNA expression in HK-2 cells treated with 2 mM of H2O2. ##P < 0.01 versus control HK-2 cells,
**P < 0.01 versus H2O2-treated HK-2 cells for 3 hours, *P < 0.05 versus H2O2-treated HK-2 cells for 3 hours. n ¼ 4 in each experimental group.
(j) The effects of b-OHB (10 mM) on cell viability in H2O2 (2 mM)-treated HK-2 cells. ##P < 0.01 versus control HK-2 cells, **P < 0.01 versus. H2O2
(2 mM)-treated HK-2 cells, *P < 0.05 versus H2O2 (2 mM)-treated HK-2 cells. n ¼ 5 in each experimental group. (k) The effects of b-OHB (10 mM)
on mRNA expression levels of FOXO3, ARC, caspase-1, IL-1b, and IL-18 in H2O2-treated HK-2 cells. ##P < 0.01 versus control HK-2 cells, *P < 0.05
versus H2O2 (2 mM)-treated HK-2 cells. n ¼ 5 in each experimental group. Bar represents mean  SEM.

Kidney International (2019) -, -–- 9


basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

Figure 6 | The effect of forkhead transcription factor O3 (FOXO3) knockdown on b-hydroxybutyrate (b-OHB)–mediated antipyroptosis
in hypoxic human kidney-2 (HK-2) cells. The quantitative real-time polymerase chain reaction analysis of mRNA expression levels of apoptosis
repressor with caspase recruitment domain (ARC) (a), caspase-1 (b), interleukin-1b (IL-1b) (c), and IL-18 (d), as well as cell viability (e) in
b-OHB–treated hypoxic HK-2 cells transfected with small, interfering RNAs (siRNAs) specific for the human FOXO3 gene or negative control
siRNA. $$P < 0.01 versus hypoxic HK-2 cells, $P < 0.05 versus hypoxic HK-2 cells, ##P < 0.01 versus b-OHB (10 mmol/l [mM])–treated hypoxic
HK-2 cells, **P < 0.01 versus b-OHB (10 mM)–treated hypoxic HK-2 cells transfected with control siRNA. n ¼ 5 in each experimental group.
Bar represents mean  SEM.

24-hour group and the 48-hour group as compared with that in the 24-hour group and the 48-hour group. Consistently,
in phosphate-buffered saline–treated IR group. However, 24 the mRNA expression levels of caspase-1 (Figure 8f), IL-1b
hours after IR insults, the plasma levels of b-OHB in the 24- (Figure 8g), and IL-18 (Figure 8h) significantly increased in
hour group were dropped to the level in phosphate-buffered IR-injured kidneys as compared with those in kidneys of
saline–treated IR group (Figure 8a). Twenty-four hours after sham-treated mice. Treatment with b-OHB decreased the
IR insult, the levels of both blood urea nitrogen and creatinine mRNA expression levels of these genes both in the 24-hour
significantly increased in the phosphate-buffered saline– group and the 48-hour group. Immunoblotting analysis
treated IR group compared with that in the phosphate- revealed that in IR-injured kidneys, AcH3K9 levels
buffered saline–treated sham group. The treatment with decreased compared with those in those of sham-treated
b-OHB significantly reduced the level of both blood urea mice. Treatment with b-OHB increased AcH3K9 levels
nitrogen and creatinine both in the 24 hour and the 48 hour both in the 24-hour group and the 48-hour group
groups (Figure 8b and c, respectively). The mRNA expression (Figure 8i). These data indicated that the effects of b-OHB
levels of FOXO3 (Figure 8d) and ARC (Figure 8e) significantly were observed even if continuous infusion is stopped after
decreased in IR-injured kidneys as compared with those in the IR insult and that the priming FOXO3-dependent pathway
kidneys of sham-treated mice. Treatment with b-OHB through epigenetic effects have long-term effects of renal
increased the mRNA expression levels of these genes both protection.

10 Kidney International (2019) -, -–-


T Tajima et al.: Effects of b-hydroxybutyrate on IRI basic research

Figure 7 | The effects of b-hydroxybutyrate (b-OHB) on histone acetylation and the activities of histone deacetylases (HDACs)/histone
acetyltransferases (HATs) in ischemic-reperfusion (IR)–injured kidneys and hypoxic human kidney-2 (HK-2) cells. (a) The expression
of acetyl-histone H3 and histone H3 in each mouse group; phosphate-buffered saline–treated mice with sham operation (n ¼ 6),
b-OHB–treated sham (shamþb-OHB) (n ¼ 6), phosphate-buffered saline–treated mice with IR injury (n ¼ 8), and b-OHB–treated IR (IRþb-OHB)
(n ¼ 8). The lower panel represents the quantification of band intensity. (b) HDAC activities (left panel) and HAT activities (right panel) in
each mouse group. (a,b) ##P < 0.01 versus sham, #P < 0.05 versus sham, *P < 0.05 versus IR. (c) The expression of acetyl-histone H3 in (continued)

Kidney International (2019) -, -–- 11


basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

DISCUSSION Most investigators agree that a normal serum level of b-


In the present study, we have demonstrated that b-OHB OHB is less than 0.5 mmol/l; a level greater than 1.0 mmol/l
reversed renal dysfunction and reduced pyroptosis in IR- can be defined as hyperketonemia, and a level greater than 3.0
injured mouse kidneys. The renoprotective effects of b- mmol/l can be defined as ketoacidosis.30 Metabolic acidosis is
OHB on renal IR injury may be mediated via antipyroptotic an independent risk factor for the development of acute
mechanisms, the effects of which were dependent on FOXO3 kidney injury.31 Ketoacidosis is also shown to be associated
upregulation and were associated with increased H3K9 acet- with significant mortality and morbidity.32 Therefore, higher
ylation (Figure 9). b-OHB might worsen renal function in IR-injured mouse
We first tried to develop an appropriate experimental kidneys and optimal serum levels lower than approximately
model in which plasma b-OHB levels were maintained at the 3.0 mmol/l are required for tissue protection.
effective levels for organ protection. For this purpose, we We revealed pyroptosis is among the mechanisms for acute
implanted mice with an i.p. pump (model 2001D; ALZET kidney injury by IR injury models. Pyroptosis is characterized
Osmotic Pumps, DURECT Corp., Cupertino, CA) delivering by rapid plasma membrane rupture and the release of
b-OHB, to increase b-OHB levels to approximately 1.3 proinflammatory intracellular contents and is morphologi-
mmol/l in the sham group 24 hours after administration and cally and mechanistically distinct from other forms of cell
to 1.4 mmol/l at 48 hours after administration. A previous death.33–37 Several studies have indicated that pyroptosis
study showed that plasma b-OHB levels increased to 1.2  contributes to infectious diseases, nervous system disorders,
0.1 mmol/l with administration of b-OHB via an i.p. pump and atherosclerosis.38–40 Pyroptosis is sometimes followed by
for 24 hours and enhanced the resistance of the mouse kidney the induction of inflammasome formation, which is also
to oxidative stress by paraquat.24 Another study demonstrated pathogenic for the development of renal tissue damages in IR
that b-OHB prevented neural damage in a mouse stroke injuries. During the development and progression of IR in-
model by subcutaneous treatment at a dosage of 8 mg/h, sults, renal cell death and inflammation may influence the
which we used in this study.25 Finally, the protocol used in severity and prognosis of IR.41–43 Thus, upregulated levels of
this study facilitated sufficient delivery of b-OHB over a IL-1 family members such as IL-1b and IL-18 in the kidney
longer period and revealed the detailed mechanisms of action tissues and urine may serve as a prognostic factor for the
besides antipyroptotic effects. To confirm this renoprotective progression of IR injury.17,44 Moreover, in IL-18 knockout
effect, we investigated the effects of preoperative fasting on mice, reduced tubular damage and improved renal function
renal IR injury and the plasma b-OHB levels in male C57BL/ in IR injury were observed.45 By inhibiting pyroptosis, ketone
6J mice. Previous research showed that applying short periods treatment reduced the inflammatory changes in IR injuries as
of dietary restriction (2 and 4 weeks with 30% dietary re- well as decreased the creatinine levels and ameliorated renal
striction) as well as 3 days of fasting, conveyed strong pro- dysfunction.
tection against morbidity and mortality induced by IR injury In the present study, b-OHB decreased TUNEL-positive
in the kidney.26–28 In our experiments, 2 days fasting had a cells in IR-injured kidneys. Although we investigated the ex-
significant improved renal dysfunction in IR-injured mouse pressions of apoptotic molecular markers, Bax and Bcl-2,
kidneys, which was associated with plasma b-OHB levels neither Bax nor Bcl-2 were affected by the b-OHB treat-
increased to approximately 2.0 mmol/l, which is similar to ment. Therefore, we explored and focused on another
serum levels in our b-OHB infusion experiments (Figure 1a). mechanism of cell death, pyroptosis, because pyroptotic cells
However, the effects of b-OHB seem to be complex, because also show positive TUNEL staining.21 Increasing evidences
raising b-OHB up to 5 mmol/l by the combination of 2 days have also shown that a necrosis-induced tissue injury pathway
of fasting and continuous infusion failed to show the recovery rather than an apoptosis-induced pathway, has an important
from IR injury. The toxic effects we have shown in in vitro role at the early stage of pathological progression of various
experiments (Figure 4a) might cancel the tissue protective types of acute kidney injury.46 In necrosis-induced tissue
effects. Consistently with our study, previous study also damage pathway, necroptosis is a highly orchestrated process,
showed b-OHB impaired the cell viability of mouse bone where the necrotized cells release danger-associated molecular
marrow–derived macrophages at a concentration of 20 patterns.47 Subsequently, danger-associated molecular patterns
mmol/l, while it did not impair the cell viability at a con- activate the innate immunity to produce more inflammatory
centration of 10 mmol/l increased cellular proliferation.29 cytokines including IL-1b and IL-18, which in turn aggravate

=
Figure 7 | (continued) hypoxic HK-2 cells with or without increasing concentrations of b-OHB. The lower graph represents the ratio of
acetyl histone H3 to histone H3. (d) The activities of HDACs (left panel) and HATs (right panel) in hypoxic HK-2 cells treated with b-OHB. (c,d)
##
P < 0.01 versus normoxic HK-2 cells, **P < 0.01 versus hypoxic HK-2 cells without b-OHB, *P < 0.05 versus hypoxic HK-2 cells without
b-OHB. n ¼ 5 in each group. (e) Chromatin immunoprecipitation assay for acetylated H3K9 levels in the forkhead transcription
factor O3 (FOXO3) promoter. The percentage of input DNA values were enrichment levels of immunoprecipitated promoter
fragments determined by quantitative polymerase chain reaction normalized to the level of input (nonimmunoprecipitated) DNA. #P < 0.05
versus normoxic HK-2 cells, **P < 0.01 versus hypoxic HK-2 cells without b-OHB. n ¼ 4 in each group. Bar represents mean  SEM.
AcH3K9, lysine-9 acetylation in histone H3; mM, mmol/l. To optimize viewing of this image, please see the online version of this article at www.
kidney-international.org
12 Kidney International (2019) -, -–-
T Tajima et al.: Effects of b-hydroxybutyrate on IRI basic research

Figure 8 | The effects of b-hydroxybutyrate (b-OHB) on pyroptosis in ischemic-reperfusion (IR)–injured kidneys in the case that
continuous infusion is stopped just after renal ischemic insult. (a) b-OHB plasma levels were measured 24 hours before and (continued)

Kidney International (2019) -, -–- 13


basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

Figure 9 | Schema depicting the mechanisms for renoprotection by b-hydroxybutyrate (b-OHB) against ischemia-reperfusion (IR)
injury. In IR injuries, both histone acetyltransferase (HAT) and histone deacetylase (HDAC) activities decreased although the decrease in HAT
activity was more responsible for decreased levels of acetylated H3K9. These changes might downregulate forkhead transcription factor O3
(FOXO3) and apoptosis repressor with caspase recruitment domain (ARC) leading to upregulate caspase-1, interleukin-1b (IL-1b), and IL-18.
These gene alterations resulted in pyroptosis and renal tubular injuries after IR insults. The treatment with b-OHB inhibited HDAC activity and
further reduced the HDAC activity in the IR-injured kidney and ameliorated the decreased levels of acetylated H3K9. This effect by b-OHB
reversed the pyroptosis gene alterations in the IR-injured kidney and ameliorated renal dysfunction.

necrosis and triggers more inflammatory reactions.48,49 We did trigger of apoptotic or pyroptotic cell death as evidenced by
not investigate the expressions of necroptotic molecular markers, the present data showing that gene-silencing of FOXO3 by
including danger-associated molecular patterns. However, there siRNA directly regulated pyroptosis-related genes (Figure 5a
is possibility that the renoprotective effects of b-OHB may also to e). Our data have shown that the treatment with b-OHB
contribute to antinecroptotic mechanisms by decreasing the increased FOXO3 expression both in ischemic kidney
proinflammatory cytokines including IL-1b and IL-18 through (Figure 3f and g) and in hypoxic HK-2 cells (Figure 4b). It is
the upregulation of FOXO3. also reported that the treatment of kidney cells with b-OHB
FOXO3 is involved in the inhibition of cell death and the increased lysine-9 acetylation in histone H3 at the FOXO3
promotion of cellular growth in ischemic renal diseases.50 promoter and caused upregulation of mRNA expression of
Especially, downregulation of FOXO3 can be a primary FOXO3.24 In these backgrounds, we examined the effects by
=
Figure 8 | (continued) after the sham or IR operation in each mouse group; phosphate-buffered saline–treated sham-operated mice,
phosphate-buffered saline–treated IR-operated mice, IR where b-OHB treatment was stopped just after IR operation (IRþb-OHB [24 h]), and IR
treated with b-OHB all through the experimental period (IRþb-OHB [48 h]). Blood urea nitrogen levels (b) and serum levels of creatinine (c) were
measured 24 hours after the sham or IR operation. The quantitative real-time polymerase chain reaction analysis of mRNA expressions of
markers related to pyroptosis activation including forkhead transcription factor O3 (FOXO3) (d), apoptosis repressor with caspase recruitment domain
(ARC) (e), caspase-1 (f), as well as the proinflammatory cytokines interleukin-1b (IL-1b) (g) and IL-18 (h). (i) Western blot analysis of acetyl-histone
H3 and histone H3 in each mice group. The lower panel represents the quantification of band intensity. ##P < 0.01 versus sham, **P < 0.01
versus IR, *P < 0.05 versus IR, $$P < 0.01 versus IRþb-OHB (24 h), and $P < 0.05 versus IRþb-OHB (24 h). n ¼ 4 for each mouse group. Bar
represents mean  SEM. AcH3K9, lysine-9 acetylation in histone H3; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

14 Kidney International (2019) -, -–-


T Tajima et al.: Effects of b-hydroxybutyrate on IRI basic research

b-OHB on the expression of FOXO3, its FOXO3 dependency, In conclusion, b-OHB can alleviate renal IR injury
and the effects by b-OHB on H3K9 acetylation levels in through its antipyroptotic effects. Its effects were dependent
hypoxic injury. Our data suggested that antipyroptotic action on FOXO3 induction and associated with increased H3K9
by b-OHB is performed through its HDAC inhibitory acetylation. Dietary or pharmacological approaches to elevate
epigenetic action. b-OHB levels, hold promise in reducing the severity of acute
Epigenetic gene regulatory mechanism is characterized by kidney injury and ensued CKD progression as well.
its long-term effects and the present study showed the remote
effects of b-OHB on renal function after the end of ischemic MATERIALS AND METHODS
insult. A recent study revealed a long-term and remote effect Mice and drugs
by the treatment with b-OHB. The i.p. injection of b-OHB All the experiments were performed in 10-week-old male C57BL/6J
during an ischemic attack of the brain can provide favorable mice weighing 21 to 30 g each (Jackson Laboratory, Bar Harbor,
effects on brain function 30 days after the b-OHB adminis- ME). The mice were cared for according to guidelines approved by
tration.51 Another study also reported that 6-hour continuous the Institutional Animal Care and Use Committee of the KEIO
University Medical School (approved number: 15044). IR injury was
infusion of b-OHB ameliorated spinal cord injury 2 weeks
induced as described previously.58 The mice were separated into 4
after the treatment, which is concomitant with the increased
groups: phosphate-buffered saline–treated sham (n ¼ 6), b-OHB-
H3K9 acetylation of the spinal tissues.52 It can be of interest treated sham (n ¼ 6), phosphate-buffered saline–treated IR (n ¼ 8),
to examine the longer-term remote effects than in the present and b-OHB-treated IR (n ¼ 8). Two weeks before the study, the right
study, especially the progression to chronic renal damages kidney was removed through a small flank incision under pento-
after IR injury in this model several days after the termination barbital anesthesia (50 mg/kg i.p.). After a 2-week recovery period,
of b-OHB treatment. We also suppose that pretreatment with the mice were anesthetized with pentobarbital (50 mg/kg i.p.) and
b-OHB primes the epigenetic FOXO3-dependent pyroptotic the left kidney was exposed through a small flank incision. The left
pathway because we observed the acute gene alterations as renal artery and vein were occluded with a nontraumatic clamp (RS-
early as 8 hours after the IR insult (Supplementary Results 5459; Roboz Surgical Instrument Company, Gaithersburg, MD) for
and Figure S1). Therefore, this epigenetic effect explains the 45 minutes. In sham-operated control mice, the kidney was treated
identically except for the clamping stage. For continuous delivery of
mechanism how these relatively small changes in genes
b-OHB (Sigma-Aldrich, St. Louis, MO), osmotic pumps (ALZET
related to pyroptosis pathway explain a dramatic improve-
model 2001D; 8 ml/h, 1 g/ml, dissolved in phosphate-buffered saline)
ment by b-OHB. This priming effects by b-OHB can be an were implanted i.p. 24 hours before surgery and b-OHB was
effective prophylaxis measures for preventing acute kidney administered for 2 days. Control mice were infused with sterile
injury after the various surgical operations. phosphate-buffered saline. Mice were killed 24 hours after the sur-
We found that histone acetylation levels decreased in gical procedure and the blood and kidneys were collected. The
IR-injured kidneys. This result was consistent with that of fasting group was fasted for 2 days preoperatively with no access to
previous studies of IR in other organs and cells.53,54 In the food and unlimited access to water. Fasted animals did not show any
previous report, in response to ischemia, the levels of acety- morbidity due to the fasting. After surgery, these mice had unlimited
lated histone decrease in the proximal tubular cells, probably access to food and water. b-OHB concentrations were determined
as a result of the adenosine triphosphate depletion and the based on a previous study.25 b-OHB plasma levels were measured
with FreeStyle Precision Neo (Abbott, Lake Bluff, IL) by tail vein
decreased in situ HAT activity. After reperfusion, the levels of
puncture at the time of the sham or IR operation in mice and at 24
acetylated histone recover by compensation mechanism in
hours before and after the operation. Tissues were prepared as
part through HDAC downregulation.55 The present data of described previously.59
the decrease in histone acetylation levels after IR insults could
have resulted from the decrease in HAT activity and that Tubular injury score
HDAC downregulation could not fully compensate for the Paraffin-embedded kidney pieces were stained with hematoxylin and
initial decrease in HAT activity. Several studies also reported eosin, and periodic acid–Schiff. In the hematoxylin and eosin sec-
the renoprotective effects by HAT activation and/or HDAC tions, renal cortical vacuolization, peritubular/proximal tubule
inhibition and resultant increased histone acetylation on renal leukocyte infiltration, and proximal tubule simplification were
IR injury.56,57 The treatment with b-OHB recovered this evaluated and scored as follows: 0, normal; 1, mild injury; 2, mod-
reduced histone acetylation probably through its HDAC erate injury; 3, severe injury. The tubulo-interstitial injury score was
inhibitory potency,24 which might contribute to the amelio- defined as described previously.60 Tubular damage (epithelial ne-
ration of renal IR injury. The mechanism in which increased crosis) in periodic acid–Schiff–stained sections was scored as follows:
0, normal; 1, <10%; 2, 10% to 25%; 3, 26% to 75%; 4, >75%.
histone acetylation leads to organ-protective effects in IR
Tubular necrosis was defined as the loss of proximal tubular brush
insults has not been fully elucidated. A recent study also border, blebbing of apical membranes, or intraluminal aggregation
revealed that the expression of one of the HATs, histone of cells and proteins, as described previously.61
acetyltransferase binding to origin recognition complex-1,
regulated cell proliferation and regeneration.56 Although the TUNEL assay
targeted histone is different, it can be surmised that histone TUNEL assay was performed using the in situ Apoptosis Detection
acetylation is associated with epithelial cell growth or regen- Kit (S7100-KIT; EMD Millipore, Temecula, CA), as described
eration, leading to tissue protective effects against IR injuries. previously.62

Kidney International (2019) -, -–- 15


basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

Cell culture and drug treatment polyvinylidene fluoride membrane. Samples with equal amounts of
HK-2, an immortalized proximal tubule epithelial cell line, was pur- purified protein (50 mg) were fractionated using 10% or 12% Tris/
chased from the American Type Culture Collection (Manassas, VA) and glycine sodium dodecylsulfate polyacrylamide gel electrophoresis
cultured. For the hypoxic group, cells were cultured in hypoxia incu- and transferred to a polyvinylidene fluoride membrane. The mem-
bator and exposed to hypoxia (5% CO2, 1% O2, and 94% N2) for 24 branes were then incubated overnight at 4 C with primary anti-
hours followed by 12 hours of reoxygenation (5% CO2, 21% O2, and bodies against AcH3K9 (1:6000; Abcam, Cambridge, MA), histone
74% N2). The stimulation of hypoxia/reoxygenation was performed H3 (1:20000; Abcam), FOXO3 (1:2500; Abcam), Bax (1:5000;
based on a previous study.22 Treatment of cells with hydrogen peroxide Abcam), Bcl-2 (1:2000; Abcam) and glyceraldehyde-3-phosphate
was performed based on a previous study.63,64 b-OHB concentrations dehydrogenase (1:5000; Santa Cruz Biotechnology, Santa Cruz,
were determined based on previous studies24,29,65 and cells were treated CA). The blots were then incubated with a secondary antibody
with b-OHB from 12 hours before hypoxia/reoxygenation or hydrogen (1:10000; Abcam) conjugated to horseradish peroxidase. Immuno-
peroxide stimulation. reactive bands were detected using an ECL detection kit (Amersham
Biosciences, Uppsala, Sweden).
Real-time reverse-transcriptase polymerase chain reaction
Total RNA was extracted from mouse kidney tissue and HK-2 cells HDAC and HAT activity assay
using TRIzol reagent (Invitrogen, Carlsbad, CA). Equal amounts (1 HDAC activity was determined using a fluorometric HDAC assay kit
mg) of total RNA from each sample were converted to cDNA using a (Bio Vision, Milpitas, CA) following the manufacturer’s protocol.
Prime Script RT Reagent Kit with gDNA Eraser (Takara, Ohtsu, HAT activity was determined using a colorimetric HAT activity assay
Japan). Real-time reverse transcriptase polymerase chain reaction kit (Bio Vision) according to the manufacturer’s protocol.
(RT-PCR) was performed using an ABI Step One Plus sequence
detector (Applied Biosystems, Foster City, CA). Sequences of the Chromatin immunoprecipitation analysis
oligo pairs are listed in Table 1. Glyceraldehyde-3-phosphate dehy- Protein-DNA complex were cross-linked with formaldehyde,
drogenase or b-actin housekeeping gene served as an internal control sonicated and immunoprecipitated with antibody conjugated
to normalize the expression levels of the samples. The relative protein A/G agarose beads (Cell Signaling Technology Japan,
expression levels were analyzed by the DDCt method described in the Tokyo, Japan) at 4 C overnight. Antibodies used for chromatin
Applied Biosystems user bulletin.66 immunoprecipitation were normal rabbit IgG (Cell Signaling
Technology Japan), anti-acetyl-H3K9 (Abcam). The immuno-
Gene knockdown of FOXO3 by siRNA precipitated DNA was recovered with proteinase K and purified
Transfection of siRNA against human FOXO3a (SI04916366; Qiagen, with phenol/chloroform precipitation. Precipitated DNA was
Hilden, Germany) and nontarget siRNA (SI03650318; Qiagen) into analyzed with SYBR green Q-PCR. The primer sets used for H3K9
HK-2 cells was done using Lipofectamine RNAiMAX (Invitrogen) at promoter binding assay were as follows: hFOXO3 1K-F
12 hours before hypoxia/reoxygenation stimulation and these cells GGGCACGGATCGTAGAATAA; hFOXO3 1K-R ACGTGGG
were incubated for 48 hours. The HK-2 cells transfected with a CATTTTTGACTTT. The primer arrangement was determined based on
nontarget control siRNA were used as control cells. previous studies.24 The percentage of input DNA values were calculated
as follows: % Input ¼ 2%  2(C[T] 2% Input Sample – C[T] IP Sample), with
Nuclei isolation and Western blot analysis background IgG control values subtracted.
The renal and cellular nuclei were isolated using a nuclei isolation kit
(Sigma-Aldrich). Samples with equal amounts of nuclear protein Cell viability assay
(7 mg) were fractionated using 16.5% Tris/tricine sodium dode- HK-2 cells were incubated in Dulbecco’s modified Eagle’s medium–
cylsulfate polyacrylamide gel electrophoresis and transferred to a F12 medium supplemented with 10% fetal bovine serum with or

Table 1 | The primer sequences used in the real-time PCR experiment


Animal species Gene name Forward Reverse
Mouse GAPDH 50 -TGTGTCCGTCGTGGATCTGA-30 50 -TTGCTGTTGAAGTCGCAGGAG-30
FOXO3 50 -TGCTAAGCAGGCCTCATCTCAA-30 50 -AAGCTGTAAACGGATCACTGTCCAC-30
ARC 50 -AGAATCCCAGGCCTCACCAC-30 50 -GTCAGCCTGCAATGTCTCTACCA-30
Bax 50 -CAGGATGCGTCCACCAAGAA-30 50 -CGTGTCCACGTCAGCAATCA-30
Bcl-2 50 -TGAAGCGGTCCGGTGGATA-30 50 -CAGCATTTGCAGAAGTCCTGTGA-30
Caspase-1 50 -ACTCGTACACGTCTTGCCCTCA-30 50 -CTGGGCAGGCAGCAAATTC-30
IL-1b 50 -TCCAGGATGAGGACATGAGCAC-30 50 -GAACGTCACACACCAGCAGGTTA-30
IL-18 50 -ACCTCCAGCATCAGGACAAAG-30 50 -TGTACAGTGAAGTCGGCCAAAG-30
Human b-actin 50 -TGGCACCCAGCACAATGAA-30 50 -CTAAGTCATAGTCCGCCTAGAAGCA-30
FOXO3 50 -AACACCTGAGAGTCAAGCAGTTGAG-30 50 -GGCAGAGTTAATTCATGGCAACA-30
ARC 50 -CTTTCTGCACCGTCATCTCCTG-30 50 -ACGCAGACTCTCCGCTTTCTTC-30
Bax 50 -GACACCTGAGCTGACCTTGG-30 50 -AGGAAGTCCAGTGTCCAGC-30
Bcl-2 50 -TGGACAACCATGACCTTGGAC-30 50 -GTGCTCAGCTTGGTATGCAGAA-30
Caspase-1 50 -GCCTGTTCCTGTGATGTGGAG-30 50 -TGCCCACAGACATTCATACAGTTTC-30
IL-1b 50 -CCAGGGACAGGATATGGAGCA-30 50 -TTCAACACGCAGGACAGGTACAG-30
IL-18 50 -CTGCCACCTGCTGCAGTCTA-30 50 -TCTACTGGTTCAGCAGCCATCTTTA-30
ARC, apoptosis repressor with caspase recruitment domain; FOXO3, forkhead transcription factor O3; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; IL-1b, interleukin-
1b; PCR, polymerase chain reaction.

16 Kidney International (2019) -, -–-


T Tajima et al.: Effects of b-hydroxybutyrate on IRI basic research

without b-OHB from 12 hours before hypoxia/reoxygenation or 8. Hiraide A, Katayama M, Mizobata Y, et al. Effect of sodium D-3-
hydrogen peroxide stimulation in 96-well culture plates (Falcon, hydroxybutyrate on amino acidemia in hemorrhagic hypotension. Eur
Surg Res. 1991;23:250–255.
Bedford, MA). Cells were incubated with 10 ml of CCK-8 reagent 9. Mizobata Y, Hiraide A, Katayama M, et al. Oxidation of D(-)3-
(Dojindo, Kumamoto, Japan) for 2 hours at 37 C. The color reaction hydroxybutyrate administered to rats with extensive burns. Surg Today.
was measured at 450 nm with a SpectraMax Paradigm Multi-Mode 1996;26:173–178.
Microplate Reader (Molecular Devices, Sunnyvale, CA). 10. Suzuki M, Suzuki M, Sato K, et al. Effect of beta-hydroxybutyrate, a
cerebral function improving agent, on cerebral hypoxia,
anoxia and ischemia in mice and rats. Jpn J Pharmacol. 2001;87:
Statistical analysis 143–150.
Data are expressed as the mean  SEM and were analyzed by one- 11. Deora V, Albornoz EA, Zhu K, et al. The ketone body b-hydroxybutyrate
does not inhibit synuclein mediated inflammasome activation in
way analysis of variance followed by the Bonferroni post hoc test. microglia. J Neuroimmune Pharmacol. 2017;12:568–574.
A P value of <0.05 was considered statistically significant. 12. Lee S, Choi E, Cha MJ, Hwang KC. Looking for pyroptosis-modulating
miRNAs as a therapeutic target for improving myocardium survival.
Mediators Inflamm. 2015;2015:254871.
DISCLOSURE 13. Nie H, Xue X, Li J, et al. Nitro-oleic acid attenuates OGD/R-triggered
All the authors declared no competing interests. apoptosis in renal tubular cells via inhibition of Bax mitochondrial
translocation in a PPAR-g-dependent manner. Cell Physiol Biochem.
2015;35:1201–1218.
ACKNOWLEDGMENTS 14. Jorgensen I, Lopez JP, Laufer SA, et al. IL-1b, IL-18, and eicosanoids
This work was partially supported by grants from the Ministry of promote neutrophil recruitment to pore-induced intracellular traps
Education, Culture, Science, Sports and Technology of Japan following pyroptosis. Eur J Immunol. 2016;46:2761–2766.
(26253053, 16H05315). The authors are grateful to Professor Ikuo 15. Napier BA, Brubaker SW, Sweeney TE, et al. Complement pathway
amplifies caspase-11-dependent cell death and endotoxin-induced
Kimura, Dr. Junki Miyamoto, and Dr. Junichiro Irie for helpful
sepsis severity. J Exp Med. 2016;213:2365–2382.
discussions and to Novartis Pharmaceuticals for helpful support to 16. Pillon NJ, Chan KL, Zhang S, et al. Saturated fatty acids activate caspase-
provide the place to perform research. 4/5 in human monocytes, triggering IL-1b and IL-18 release. Am J Physiol
Endocrinol Metab. 2016;311:E825–E835.
17. Yang JR, Yao FH, Zhang JG, et al. Ischemia-reperfusion induces renal
SUPPLEMENTARY MATERIAL tubule pyroptosis via the CHOP-caspase-11 pathway. Am J Physiol Renal
Supplementary Results. The effect of b-hydroxybutyrate on Physiol. 2014;306:F75–F84.
pyroptosis in IR-injured kidneys at different time point. 18. Wang K, Liu F, Zhou LY, et al. miR-874 regulates myocardial necrosis by
Figure S1. The effects of b-hydroxybutyrate (b-OHB) on pyroptosis in targeting caspase-8. Cell Death Dis. 2013;4:e709.
19. Ferber EC, Peck B, Delpuech O, et al. FOXO3a regulates reactive oxygen
IR-injured kidneys at different time point. (A–E) The quantitative real-
metabolism by inhibiting mitochondrial gene expression. Cell Death
time polymerase chain reaction analysis of mRNA expressions of Differ. 2012;19:968–979.
markers related to pyroptosis activation including forkhead tran- 20. Lu D, Liu J, Jiao J, et al. Transcription factor Foxo3a prevents apoptosis by
scription factor O3 (FOXO3) (A), apoptosis repressor with caspase regulating calcium through the apoptosis repressor with caspase
recruitment domain (ARC) (B), caspase-1 (C), as well as the proin- recruitment domain. J Biol Chem. 2013;288:8491–8504.
flammatory cytokines interleukin-1b (IL-1b) (D) and IL-18 (E) in mice 21. Li X, Du N, Zhang Q, et al. MicroRNA-30d regulates cardiomyocyte
pyroptosis by directly targeting foxo3a in diabetic cardiomyopathy. Cell
groups 8 hours or 24 hours after reperfusion. The experimental
Death Dis. 2014;23:e1479.
groups consist of phosphate-buffered saline–treated mice with sham 22. Wu H, Huang T, Ying L, et al. MiR-155 is involved in renal ischemia-
operation (sham, n ¼ 5), phosphate-buffered saline–treated mice reperfusion injury via direct targeting of FoxO3a and regulating renal
with ischemia reperfusion (IR) injury (n ¼ 5), and IR treated with tubular cell pyroptosis. Cell Physiol Biochem. 2016;40:1692–1705.
b-OHB (IRþb-OHB) (n ¼ 5) for 8 hours or 24 hours after reperfusion. 23. Robin E, Guzy RD, Loor G, et al. Oxidant stress during simulated ischemia
##P < 0.01 versus sham, #P < 0.05 versus sham, **P < 0.01 versus IR, primes cardiomyocytes for cell death during reperfusion. J Biol Chem.
*P < 0.05 versus IR, $$P < 0.01 versus IR (8 hours after reperfusion), 2007;282:19133–19143.
24. Shimazu T, Hirschey MD, Newman J, et al. Suppression of oxidative stress
$P < 0.05 versus IR (8 hours after reperfusion), {{P < 0.01 versus by b-hydroxybutyrate, an endogenous histone deacetylase inhibitor.
IRþb-OHB (8 hours after reperfusion). Bar represents mean  SEM. Science. 2013;339:211–214.
Supplementary material is linked to the online version of the paper at 25. Rahman M, Muhammad S, Khan MA, et al. The b-hydroxybutyrate
www.kidney-international.org. receptor HCA2 activates a neuroprotective subset of macrophages. Nat
Commun. 2014;5:3944.
26. Mitchell JR, Verweij M, Brand K, et al. Short-term dietary restriction and
REFERENCES fasting precondition against ischemia reperfusion injury in mice. Aging
1. Veech RL, Chance B, Kashiwaya Y, et al. Ketone bodies, potential Cell. 2010;9:40–53.
therapeutic uses. IUBMB Life. 2001;51:241–247. 27. van Ginhoven TM, Mitchell JR, Verweij M, et al. The use of preoperative
2. Cahill GF Jr, Herrera MG, Morgan AP, et al. Hormone-fuel nutritional interventions to protect against hepatic ischemia-reperfusion
interrelationships during fasting. J Clin Invest. 1966;45:1751–1769. injury. Liver Transpl. 2009;15:1183–1191.
3. Robinson AM, Williamson DH. Physiological roles of ketone bodies as 28. Verweij M, van Ginhoven TM, Mitchell JR, et al. Preoperative
substrates and signals in mammalian tissues. Physiol Rev. 1980;60: fasting protects mice against hepatic ischemia/reperfusion injury:
143–187. mechanisms and effects on liver regeneration. Liver Transpl.
4. Cahill GF Jr. Fuel metabolism in starvation. Annu Rev Nutr. 2006;26:1–22. 2011;17:695–704.
5. Laffel L. Ketone bodies: a review of physiology, pathophysiology and 29. Youm YH, Nguyen KY, Grant RW, et al. The ketone metabolite
application of monitoring to diabetes. Diabetes Metab Res Rev. 1999;15: b-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory
412–426. disease. Nat Med. 2015;21:263–269.
6. Rojas-Morales P, Tapia E, Pedraza-Chaverri J. b-hydroxybutyrate: a 30. Fulop M, Murthy V, Michilli A, et al. Serum beta-hydroxybutyrate
signaling metabolite in starvation response? Cell Signal. 2016;28: measurement in patients with uncontrolled diabetes mellitus. Arch Intern
917–923. Med. 1999;159:381–384.
7. Katayama M, Hiraide A, Sugimoto H, et al. Effect of ketone bodies on 31. Hu J, Wang Y, Geng X, et al. Metabolic acidosis as a risk factor for the
hyperglycemia and lactic acidemia in hemorrhagic stress. J Parenter development of acute kidney injury and hospital mortality. Exp Ther Med.
Enteral Nutr. 1994;18:442–446. 2017;13:2362–2374.

Kidney International (2019) -, -–- 17


basic research T Tajima et al.: Effects of b-hydroxybutyrate on IRI

32. Bilici M, Tavil B, Dogru O, Davutoglu M, Bosnak M. Diabetic ketoasidosis 50. Nho RS, Hergert P. FoxO3a and disease progression. World J Biol Chem.
is associated with prothrombotic tendency in children. Pediatr Hematol 2014;5:346–354.
Oncol. 2011;28:418–424. 51. Lee BS, Woo DC, Woo CW, et al. Exogenous b-hydroxybutyrate treatment
33. Dong T, Liao D, Liu X, et al. Using small molecules to dissect non- and neuroprotection in a suckling rat model of hypoxic-ischemic
apoptotic programmed cell death: necroptosis, ferroptosis, and encephalopathy. Dev Neurosci. 2018;40:73–83.
pyroptosis. Chembiochem. 2015;16:2557–2561. 52. Qian J, Zhu W, Lu M, et al. D-b-hydroxybutyrate promotes functional
34. Croker BA, Silke J, Gerlic M. Fight or flight: regulation of emergency recovery and relieves pain hypersensitivity in mice with spinal cord
hematopoiesis by pyroptosis and necroptosis. Curr Opin Hematol. injury. Br J Pharmacol. 2017;174:1961–1971.
2015;22:293–301. 53. Alsarraf O, Fan J, Dahrouj M, et al. Acetylation: a lysine modification with
35. Yang Y, Jiang G, Zhang P, et al. Programmed cell death and its role in neuroprotective effects in ischemic retinal degeneration. Exp Eye Res.
inflammation. Mil Med Res. 2015;2:12. 2014;127:124–131.
36. Schoenlaub L, Cherla R, Zhang Y, et al. Coxiella burnetii avirulent nine 54. Chen M, Liu Q, Chen L, et al. Remifentanil postconditioning ameliorates
mile phase II induces caspase-1-dependent pyroptosis in murine histone H3 acetylation modification in H9c2 cardiomyoblasts after
peritoneal B1a B cells. Infect Immun. 2016;84:3638–3654. hypoxia/reoxygenation via attenuating endoplasmic reticulum stress.
37. Lorenz G, Darisipudi MN, Anders HJ. Canonical and non-canonical effects Apoptosis. 2017;22:662–671.
of the NLRP3 inflammasome in kidney inflammation and fibrosis. 55. Marumo T, Hishikawa K, Yoshikawa M, et al. Epigenetic regulation of
Nephrol Dial Transplant. 2014;29:41–48. BMP7 in the regenerative response to ischemia. J Am Soc Nephrol.
38. Eichholz K, Bru T, Tran TT, et al. Immune-complexed adenovirus induce 2008;19:1311–1320.
AIM2-mediated pyroptosis in human dendritic cells. PLoS Pathog. 56. Havasi A, Haegele JA, Gall JM, et al. Histone acetyl transferase (HAT)
2016;12:e1005871. HBO1 and JADE1 in epithelial cell regeneration. Am J Pathol. 2013;182:
39. Yang J, Zhao Y, Zhang P, et al. Hemorrhagic shock primes for lung 152–162.
vascular endothelial cell pyroptosis: role in pulmonary inflammation 57. Levine MH, Wang Z, Bhatti TR, et al. Class-specific histone/protein
following LPS. Cell Death Dis. 2016;7:e2363. deacetylase inhibition protects against renal ischemia reperfusion
40. Vande Walle L, Lamkanfi M. Pyroptosis. Curr Biol. 2016;26:R568–R572. injury and fibrosis formation. Am J Transplant. 2015;15:965–973.
41. Hutton HL, Ooi JD, Holdsworth SR, et al. The NLRP3 inflammasome in 58. Yamashita J, Kita S, Iwamoto T, et al. Attenuation of ischemia/
kidney disease and autoimmunity. Nephrology (Carlton). 2016;21:736– reperfusion-induced renal injury in mice deficient in Naþ/Ca2þ
744. exchanger. J Pharmacol Exp Ther. 2003;304:284–293.
42. Lin CF, Kuo YT, Chen TY, et al. Quercetin-rich guava (Psidium guajava) 59. Chang YK, Choi DE, Na KR, et al. Erythropoietin attenuates renal injury in
juice in combination with trehalose reduces autophagy, apoptosis and an experimental model of rat unilateral ureteral obstruction via anti-
pyroptosis formation in the kidney and pancreas of type II diabetic rats. inflammatory and anti-apoptotic effects. J Urol. 2009;181:1434–1443.
Molecules. 2016;21:334. 60. Chang YK, Choi H, Jeong JY, et al. Dapagliflozin, SGLT2 inhibitor, attenuates
43. Yang CC, Yao CA, Yang JC, et al. Sialic acid rescues repurified renal ischemia-reperfusion injury. PLoS One. 2016;11:e0158810.
lipopolysaccharide-induced acute renal failure via inhibiting TLR4/PKC/ 61. Choi DE, Jeong JY, Lim BJ, et al. Pretreatment of sildenafil attenuates
gp91-mediated endoplasmic reticulum stress, apoptosis, autophagy, and ischemia-reperfusion renal injury in rats. Am J Physiol Renal Physiol.
pyroptosis signaling. Toxicol Sci. 2014;141:155–165. 2009;297:F362–F370.
44. Chung SD, Lai TY, Chien CT, et al. Activating Nrf-2 signaling depresses 62. Lee KW, Jeong JY, Lim BJ, et al. Sildenafil attenuates renal injury in an
unilateral ureteral obstruction-evoked mitochondrial stress-related experimental model of rat cisplatin-induced nephrotoxicity. Toxicology.
autophagy, apoptosis and pyroptosis in kidney. PLoS One. 2012;7: 2009;257:137–143.
e47299. 63. Choi HI, Kim HJ, Park JS, et al. PGC-1a attenuates hydrogen peroxide-induced
45. Wu H, Craft ML, Wang P, et al. IL-18 contributes to renal damage after apoptotic cell death by upregulating Nrf-2 via GSK3b inactivation mediated
ischemia-reperfusion. J Am Soc Nephrol. 2008;19:2331–2341. by activated p38 in HK-2 Cells. Sci Rep. 2017;7:4319.
46. Linkermann A, Bräsen JH, Himmerkus N, et al. Rip1 (receptor-interacting 64. Andreucci M, Fuiano G, Presta P, et al. Downregulation of cell survival
protein kinase 1) mediates necroptosis and contributes to renal signalling pathways and increased cell damage in hydrogen peroxide-
ischemia/reperfusion injury. Kidney Int. 2012;81:751–761. treated human renal proximal tubular cells by alpha-erythropoietin. Cell
47. Mulay SR, Linkermann A, Anders HJ. Necroinflammation in kidney Prolif. 2009;42:554–561.
disease. J Am Soc Nephrol. 2016;27:27–39. 65. Kong G, Huang Z, Ji W, et al. The ketone metabolite b-hydroxybutyrate
48. Sarhan M, Land WG, Tonnus W, et al. Origin and consequences of attenuates oxidative stress in spinal cord injury by suppression of class I
necroinflammation. Physiol Rev. 2018;98:727–780. histone deacetylases. J Neurotrauma. 2017;34:2645–2655.
49. Yatim N, Jusforgues-Saklani H, Orozco S, et al. RIPK1 and NF-kB signaling 66. Applied Biosystems. Guide to Performing Relative Quantitation of Gene
in dying cells determines cross-priming of CD8⁺ T cells. Science. 2015;350: Expression Using Real-Time Quantitative PCR. Foster City, CA: Applied
328–334. Biosystems; 2008.

18 Kidney International (2019) -, -–-

You might also like