You are on page 1of 15

BASIC RESEARCH www.jasn.

org

Impairment of PPARa and the Fatty Acid Oxidation


Pathway Aggravates Renal Fibrosis during Aging
Ki Wung Chung,1,2 Eun Kyeong Lee,1,2,3 Mi Kyung Lee,4 Goo Taeg Oh,5 Byung Pal Yu,6
and Hae Young Chung1,2
1
Molecular Inflammation Research Center for Aging Intervention and 2Department of Pharmacy, College of Pharmacy,
Pusan National University, Busan, Republic of Korea; 3Korea Institute of Toxicology, Yuseong-gu, Daejeon, Republic
of Korea; 4Department of Pathology, Ilsin Christian Hospital, Busan, Republic of Korea; 5Department of Life Sciences,
Ewha Womans University, Seoul, Republic of Korea; and 6Department of Physiology, The University of Texas Health
Science Center at San Antonio, San Antonio, Texas

ABSTRACT
Defects in the renal fatty acid oxidation (FAO) pathway have been implicated in the development of renal fibrosis.
Although, compared with young kidneys, aged kidneys show significantly increased fibrosis with impaired kidney
function, the mechanisms underlying the effects of aging on renal fibrosis have not been investigated. In this study,
we investigated peroxisome proliferator–activated receptor a (PPARa) and the FAO pathway as regulators of age-
associated renal fibrosis. The expression of PPARa and the FAO pathway–associated proteins significantly de-
creased with the accumulation of lipids in the renal tubular epithelial region during aging in rats. In particular,
decreased PPARa protein expression associated with increased expression of PPARa-targeting microRNAs. Among
the microRNAs with increased expression during aging, miR-21 efficiently decreased PPARa expression and im-
paired FAO when ectopically expressed in renal epithelial cells. In cells pretreated with oleic acid to induce lipid
stress, miR-21 treatment further enhanced lipid accumulation. Furthermore, treatment with miR-21 significantly
exacerbated the TGF-b–induced fibroblast phenotype of epithelial cells. We verified the physiologic importance
of our findings in a calorie restriction model. Calorie restriction rescued the impaired FAO pathway during aging and
slowed fibrosis development. Finally, compared with kidneys of aged littermate controls, kidneys of aged PPARa2/2
mice showed exaggerated lipid accumulation, with decreased activity of the FAO pathway and a severe fibrosis
phenotype. Our results suggest that impaired renal PPARa signaling during aging aggravates renal fibrosis devel-
opment, and targeting PPARa is useful for preventing age-associated CKD.

J Am Soc Nephrol 29: 1223–1237, 2018. doi: https://doi.org/10.1681/ASN.2017070802

Significance Statement
Age-related changes in kidney function, structure,
and their relationships have been extensively recog- Defective renal lipid metabolism has been implicated in
nized recently.1 The effects of aging on the kidneys the development of kidney disease. Defects in the fatty
are the most dramatic among any other organs acid oxidation (FAO) pathway may play a crucial role
during renal fibrosis progression. This paper describes
the important roles of PPARa and the FAO pathway
during age-related renal fibrosis. The studies demon-
Received July 26, 2017. Accepted January 3, 2018. strate that PPARa and the FAO pathway are signifi-
cantly decreased with severe interstitial fibrosis during
Published online ahead of print. Publication date available at kidney aging. The decreased PPARa was associated
www.jasn.org. with increased miR-21 during renal aging, which
Correspondence: Prof. Hae Young Chung, Molecular In-
showed the inhibitory effect on PPARa in the renal
flammation Research Center for Aging Intervention (MRCA) and epithelial cells. The importance of PPARa and the FAO
Department of Pharmacy, College of Pharmacy, Pusan National pathway was further confirmed in antiaging calorie re-
University, 2, Busandaehak-ro, 63beon-gil, Geumjeong-Gu, Bu- striction and PPARa knockout aging mouse models.
san 46241, Republic of Korea. Email: hyjung@pusan.ac.kr Collectively, targeting PPARa may be useful for pre-
venting age-associated renal fibrosis and CKD.
Copyright © 2018 by the American Society of Nephrology

J Am Soc Nephrol 29: 1223–1237, 2018 ISSN : 1046-6673/2904-1223 1223


BASIC RESEARCH www.jasn.org

because of diverse factors that can accelerate the changes.1 as a major energy source, the role of PPARa in the develop-
Epidemiologic studies have suggested that age-related renal ment of renal diseases has been extensively investigated re-
changes may be associated mainly with systemic hypertension, cently.15,19–21 It has been suggested that defects in PPARa
diabetes, dyslipidemia, and other environmental causes such and the FAO pathway play important roles during renal in-
as smoking.2,3 With aging, most subjects show a progressive terstitial fibrosis development. Defective FAO in tubule ep-
functional decline that includes decreases in GFR and renal ithelial cells has been associated with ATP depletion, cell
blood flow as well as increases in urinary excretion of proteins death, dedifferentiation, and lipid accumulation, which are
such as albumin.4,5 The prevalence of CKD is significantly the phenotypes resembling fibrosis.15 Furthermore, others
higher in older people, with more than one-third of those have suggested that miR-21-mediated downregulation of
aged 70 or older exhibiting moderate or severe CKD.6 Inter- PPARa contributes to fibrogenesis and epithelial injury
estingly, these functional changes occur in concert with struc- in a mouse fibrosis model.22
tural changes, which gives more accurate information on the In this study, we hypothesized as to whether age-associated
status of renal disease.7 Morphologic changes of aging kidneys renal fibrosis is associated with defective FAO in the renal
include loss of renal mass (primarily in the cortical region), tubule epithelium. We found that levels of PPARa and FAO-
development of glomerular sclerosis, tubular atrophy, and associated enzymes were reduced in aged rat kidneys with
tubulointerstitial fibrosis.7 significantly increased lipid accumulation and interstitial fi-
Fibrosis is characterized by a loss of parenchymal tissue with brosis. Reduced PPARa was associated with increases of
the accumulation of fibrillary collagens produced by activated miRNAs that regulate PPARa translation. Further animal
myofibroblasts. Although glomerular lesions are significantly studies using antiaging calorie restriction (CR) and aged
associated with renal functional decline and particular renal PPARa2/2 mouse models demonstrated the role of PPARa
diseases, renal fibrosis is always accompanied in all forms of in the regulation of age-associated renal fibrosis.
CKD.8 In renal tubulointerstitial fibrosis, tubular epithelial
cells play an important role.9 Loss of epithelial integrity causes
cell cycle arrest and dedifferentiation, leading to increased ex- RESULTS
pression of mesenchymal markers.10 During renal aging, it has
been well established that structural changes occur with glo- Aging Increases Renal Fibrosis, Functional Changes,
merulosclerosis, interstitial fibrosis, and tubular atrophy.11 and Epithelial Damage
Interestingly, a recent study has shown that age-related First, we verified renal fibrosis, functional changes, and
changes in the renal structure occur earlier than functional epithelial damages that accompany aging. To investigate
changes.12 The authors found a strong relationship between age-related fibrosis in our aging rat model, four different-
age and nephrosclerosis in healthy adults without functional aged kidneys (6, 12, 18, and 24 months) were examined.
changes in the kidney, thus implicating the importance of The expression levels of extracellular matrix (ECM) genes in-
structural changes. cluding Acta2, Fn, Col1a, Col3a1, Col4a, and Col11a1 were all
Alterations in lipid metabolism have been implicated in significantly increased at 24 months of aging (Figure 1A). We
various metabolic diseases.13 Although the kidney is not clas- next checked markers for mesenchymal and epithelial cells.
sified as a metabolic organ, renal tubular cells have high basal Interestingly, only markers of mesenchymal cells (Vim and
levels of baseline energy consumption and they prefer fatty Fsp) were increased during aging, whereas epithelial cell
acid oxidation (FAO) as a major energy source.14,15 The high markers (Krt8 and Cdh1) were not affected by aging (Supple-
energy yield of FAO (106 ATP equivalents per fatty acid [FA]), mental Figure 1A). The ECM protein levels were further in-
which is produced in the mitochondria and peroxisomal com- vestigated. The ECM proteins started to increase at 18 months
partments, makes it possible to support the high energy con- of age, and the extent was dramatically increased at 24 months
sumption of cells. Recently, defects in the FAO pathway have (Figure 1B). A histologic analysis also confirmed age-associ-
received substantial attention in the context of acute and ated ECM accumulation and fibrosis. Immunohistochemistry
chronic kidney diseases.15,16 It has been suggested that a de- (IHC) staining of collagen I was dramatically increased in the
fective FAO pathway induces abnormal lipid accumulation of interstitial region of aged kidneys (Figure 1C). However, the
triglycerides (TG), resulting in lipotoxicity that contributes to expression of collagen III did not show any differences be-
the development of kidney diseases. tween young and aged kidneys (Supplemental Figure 1B). The
Peroxisome proliferator–activated receptor a (PPARa) is accumulation of ECM proteins also showed an increase when
the key transcriptional factor that regulates intracellular lip- detected by Sirius red and Masson’s trichrome staining (Fig-
ids through direct transcriptional control of genes involved ure 1D). Fibrotic-positive regions were significantly increased
in peroxisomal and mitochondrial FAO pathways, FA up- at 24 months of aging (Figure 1E). We next measured serum
takes, and TG catabolism. 17,18 Accumulating evidence urea and creatinine levels to assess age-related kidney func-
supports a link between PPARa and metabolic diseases in- tion decline. Serum urea levels significantly increased at 24
cluding diabetes, obesity, dyslipidemia, and fatty liver.18 Be- months of aging (Figure 1F); however, creatinine levels did
cause the kidney is a metabolically active tissue that uses FAs not change significantly during aging (Supplemental Figure 1C).

1224 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 1223–1237, 2018
www.jasn.org BASIC RESEARCH

Figure 1. Aged kidneys show increased renal fibrosis, function decline, and epithelial damages. Four different-aged kidneys (6, 12, 18,
and 24 months, n=8) were examined to examine the effect of aging on renal changes. (A) Gene levels of ECM proteins were measured
by qPCR. *P,0.05 versus 6 months. (B) Protein levels of ECM proteins were measured by western blotting. Actin was used as the
loading control. (C) Collagen I expression in young and aged kidney was visualized by IHC. Scale bar=50 mm. (D) Masson’s trichrome
(MT) and Sirius red (SR) staining were performed to visualize fibrosis in kidney. Scale bar=100 mm. (E) Quantification of SR-stained
fibrosis in kidneys. *P,0.05 versus 6 months. (F) Urea levels were measured by biochemical method in serum to measure kidney
function. *P,0.05 versus 6 months. (G) KIM-1 levels were detected in serum by ELISA to measure renal epithelial damages. *P,0.05
versus 6 months.

Renal periodic acid–Schiff staining and evaluation were further Aging Increases Renal Lipid Accumulation
implemented in aged kidney. Aged rat showed significantly To investigate the relationship between increased fibrosis and
increased renal damages in both glomerular and tubule regions changes in lipid metabolism during aging, lipid metabolism–
(Supplemental Figure 1, D–F). To evaluate renal epithelial associated changes were further evaluated. Levels of extracted
damage, we measured kidney injury molecule-1 (KIM-1) levels TG were significantly increased at 24 months of aging (Figure
in the serum and kidney. Serum and kidney KIM-1 levels were 2A). Accumulated TG levels were further visualized via oil red
also significantly increased during aging (Figure 1G, Supple- O–staining analysis. As expected, renal lipid accumulation was
mental Figure 1, G and H). Taken together, we concluded that observed in 24-month-old kidneys (Figure 2B). Oil red
aging significantly increases renal fibrosis and damage with O–positive regions were mostly located in renal tubular epithe-
impaired renal function. lial cells (Figure 2B). We further detected an overall increase of

J Am Soc Nephrol 29: 1223–1237, 2018 PPARa and Age-Associated Renal Fibrosis 1225
BASIC RESEARCH www.jasn.org

Figure 2. Aging increases lipid accumulation with alteration of various lipid metabolism–associated transcriptional factors. Four dif-
ferent-aged kidneys (6, 12, 18, and 24 months, n=8) were examined to see the effect of aging on renal changes. (A) TGs were
quantified in aging kidney model. *P,0.05 versus 6 months. (B) Kidneys (6 months and 24 months) were stained with oil red O to

1226 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 1223–1237, 2018
www.jasn.org BASIC RESEARCH

vacuoles in renal tubules during aging (Figure 2, C and D). lipids accumulated (Figure 3D). Furthermore, the expression
These data indicated that lipid accumulation was increased of PPARa and ACOX1 detected by IHC was also decreased
during renal aging, and the accumulation was especially in- during aging (Figure 3D). Taken together, we found that the
creased in the tubular epithelial region. levels of PPARa and its target proteins, CPT1a and ACOX1,
In the regulation of lipid metabolism, various transcrip- were significantly decreased during aging.
tional factors play important roles.23 We first checked the Because there were discrepancies between gene and pro-
mRNA levels of these transcription factors in the four dif- tein levels of PPARa during aging, we next checked the
ferent-aged kidneys; however, there were no significant upstream signaling of PPARa. However, the levels of
changes during aging (Figure 2E). Because it is generally AMP-activated protein kinase (AMPK) and activated
accepted that the localization of transcription factors is im- (phosphorylated) AMPK were both increased during aging
portant, we next checked the nuclear protein expression of (Figure 3E). The protein levels of PGC1a, which is an im-
these transcriptional factors. Surprisingly, unlike mRNA lev- portant coactivator of PPARa, were not changed during
els, there were remarkable changes in protein expression of aging (Figure 3E). We further checked various miRNAs
the transcription factors in the nucleus. Nuclear expression that can silence PPARa translation by epigenetic mecha-
of PPARa and Farnesoid X receptor (FXR) were dramatically nisms. Interestingly, among the seven miRNAs that are
decreased in 24-month-old rat kidneys, whereas SREBP1 known to suppress PPARa translation, levels of miR-21,
and ChREBP were rather increased during aging (Figure 2, miR-34a, and miR-155 were significantly increased during
F and G). Taken together, these data implicated that dysre- aging (Figure 3F). In summary, decreased PPARa expres-
gulated lipid metabolism during aging might be associated sion during aging may be affected by miRNA that regulates
with changes in nuclear expression of transcriptional factors the translation of PPARa epigenetically.
that regulate lipid metabolism.
PPARa Translation and FAO Are Suppressed by miR-21
Aging Decreases PPARa and FAO Pathway in Renal in Renal Tubular Epithelial Cells
Tubule Epithelial Region We next performed in vitro experiments to examine whether
Recent studies have suggested that defective FAO by impaired increased miRNA levels during aging suppress PPARa ex-
PPARa in renal tubular epithelial cells plays a pivotal role in pression and affect FAO in renal tubular epithelial cells.
kidney fibrosis development.15 On the basis of our results and NRK52E renal tubular epithelial cells were used to examine
those of previous studies, we hypothesized that impairments the effects of miRNAs on PPARa and the FAO pathway.
in PPARa and the FAO pathway may be important in the Among the three miRNAs (miR-21, miR-34a, and miR-
development of age-associated renal fibrosis. First, we 155) that were increased during aging, only miR-21 effi-
checked whether the decreased nuclear expression of PPARa ciently decreased PPARa protein expression in NRK52E cells
was due to defects in translocation. However, cytosolic frac- without changes in PPARa gene levels (Figure 4A, Supple-
tions of aged kidney also showed decreased PPARa expression mental Figure 3A). Furthermore, miR-21 also decreased
(Figure 3A). We next checked the expression levels of well PPRE binding activity as deduced from a luciferase assay
known PPARa gene targets by quantitative PCR (qPCR). (Figure 4B). PPARa target gene levels were further analyzed
Most of the PPARa target genes (Cpt1a, Acox1, Lcad, Mcad, under miR-21 treatment conditions. Most, but not all, of the
Acad10, and Hadh) were significantly decreased at 24 months PPARa target genes were downregulated by an miR-21 treat-
of aging (Figure 3B). However, other lipid metabolism–asso- ment (Figure 4C). Decreased CPT1a and ACOX1 levels were
ciated genes that play important roles in FA synthesis and TG further confirmed by measuring their protein levels (Figure
accumulation did not show significant changes (Supplemen- 4D). We next measured oxygen consumption rate (OCR)
tal Figure 2, A and B). We further checked carnitine palmi- under the miR-21 treatment conditions. miR-21 treatment
toyltransferase 1a (CPT1a) and acyl-coenzyme A oxidase 1 decreased OCR levels both under the normal and oleic acid
(ACOX1) protein levels. Aged rat kidneys showed signifi- treatment conditions (Figure 4E). Furthermore, consistent
cantly decreased CPT1a and ACOX1 protein levels (Figure with decreased OCR, miR-21 treatment reduced ATP pro-
3C). The decreased levels of PPARa and its target protein, duction in cells (Supplemental Figure 3B). However, treat-
ACOX1, were further verified by IHC. PPARa and ACOX1 ment with miR-21 resulted in marginal changes in the total
primarily localized to the tubular epithelial regions where the lipid content of the cells (Figure 4, F and G). Next, we

visualize lipid accumulation. Scale bar=50 mm. (C) Representative H&E staining shows increased vacuoles in renal tubules during aging. (D)
Number of vacuoles was quantified by counting vacuoles per tubule. *P,0.05 versus 6 months. (E) Gene levels of transcription factors
associated with lipid metabolism were measured by qPCR. *P,0.05 versus 6 months. (F) Nuclear protein expressions of transcription
factors associated with lipid metabolism were measured by western blotting. TFIIB was used as the loading control. (G) Western blot
results from 4 independent experiments were quantified by densitometry. *P,0.05 versus 6 months.

J Am Soc Nephrol 29: 1223–1237, 2018 PPARa and Age-Associated Renal Fibrosis 1227
BASIC RESEARCH www.jasn.org

Figure 3. Aging decreases PPARa and FAO signaling pathways in the renal tubule epithelial region. Four different-aged
kidneys (6, 12, 18, and 24 months, n=8) were examined to see the effect of aging on renal changes. (A) Nuclear and cytosolic
PPARa levels were measured in the rat kidney. TFIIB was used as the loading control for the nuclear fraction, and actin was
used as the loading control for the cytosolic fraction. (B) Gene levels of PPARa target proteins were measured by qPCR.
*P,0.05 versus 6 months. (C) Protein expression of CPT1a and ACOX1 was measured by western blotting. Actin was used as
the loading control. (D) PPARa and ACOX1 expression in young and aged kidneys was visualized by IHC. Scale bar=50 mm.
(E) Protein expression levels of signals upstream of PPARa (AMPK, p-AMPK, and PGC1a) were measured by western blotting.
Actin was used as a loading control. (F) The miRNA levels that are known to regulate PPARa expression were detected by
qPCR. *P,0.05 versus 6 months.

pretreated cells with 200 mM of oleic acid to induce lipid further checked FAO-related gene changes in the same con-
stress in the epithelial cells. Treatment with oleic acid signif- ditions. Although oleic acid treatment regulated FAO-related
icantly increased intracellular lipid content when measured gene expressions differently, miR-21 constantly reduced
quantitatively or visualized by oil red O staining (Figure 4, those gene expressions regardless of oleic acid treatment
F and G). The miR-21 treatment further increased the accu- (Supplemental Figure 3C). Furthermore, oleic acid with
mulation of intracellular lipids (Figure 4, F and G). We miR-21 treatment induced a decrease in cell viability,

1228 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 1223–1237, 2018
www.jasn.org BASIC RESEARCH

Figure 4. miR-21 suppresses PPARa translation and FAO in NRK52E cells. Suppression of PPARa translation and FAO by miR-21
in renal tubular epithelial cells. NRK52E renal epithelial cells were used to investigate the role of miRNAs in PPARa and FAO
signaling. (A) NRK52E cells were transfected with selected miRNAs (miR-21, miR-34a, and miR-155) and protein expression of
PPAR was measured by western blotting. Actin was used as the loading control. (B) NRK52E cells were transfected with selected
miRNAs (miR-21, miR-34a, and miR-155) and PPAR activities were measured by the PPRE luciferase activity. (C) Gene levels of
PPARa target proteins were measured by qPCR after miR-21 transfection in NRK52E cells. *P,0.05 versus nontreated control.
(D) Protein expression of CPT1a and ACOX1 was measured by western blotting in miR-21-treated NRK52E cells. Actin was used
as the loading control. (E) OCR was measured under normal and miR-21-treated conditions. Oleic acid (200 mM) and oligomycin
(1 mM) were treated at designated times. (F) Cellular TG levels were extracted and quantified in NRK52E cells treated with 200
mM oleic acid and/or miR-21. #P,0.05 versus nontreated control. *P,0.05 versus oleic acid treatment. (G) Cellular TG was
visualized by oil red O staining in NRK52E cells treated with 200 mM oleic acid and/or miR-21. Scale bar=50 mm.

suggesting the role of FAO in regulating lipotoxicity under impaired FAO pathway during fibrosis signaling in renal ep-
the stress condition (Supplemental Figure 3D). These data ithelial cells. First, we measured miR-21 expression changes
suggest that miR-21 suppresses the translation of PPARa in on TGF-b-treated NRK52E cells. TGF-b significantly in-
renal epithelial cells, thus inhibiting FAO and inducing lipid duced miR-21 expression as previously described (Figure
accumulation. 5A). We next checked whether TGF-b treatment affected
lipid accumulation that was induced by oleic acid. TGF-b
The FAO Pathway Is Important during TGF-b-Induced treatment further increased oleic acid–induced lipid accu-
Fibrosis Signaling in Renal Tubular Epithelial Cells mulation (Figure 5, B and C). To further demonstrate the
It has been suggested that TGF-b-induced miR-21 plays an role of miR-21-mediated PPARa silencing and the FAO
important role during the mediation of fibrosis signaling in pathway on TGF-b, an miR-21 inhibitor (anti-miR-21 oligo)
renal epithelial cells.24 To this end, we further investigated was utilized. TGF-b treatment reduced PPARa expression in
the roles of miR-21-mediated PPARa silencing and an epithelial cells, and anti-miR-21 abrogated this reduction

J Am Soc Nephrol 29: 1223–1237, 2018 PPARa and Age-Associated Renal Fibrosis 1229
BASIC RESEARCH www.jasn.org

Figure 5. FAO signaling is important in TGF-b-induced fibrosis signaling in renal tubular epithelial cells. NRK52E renal epithelial cells
were transfected with miR-21 under TGF-b treatment conditions to investigate the role of FAO signaling in fibrosis. (A) The miR-21
levels were measured by qPCR under TGF-b treatment conditions. (B) Cellular TG was visualized by oil red O staining in NRK52E cells
treated with 200 mM oleic acid or/and TGF-b. Scale bar=50 mm. (C) Cellular TG levels were extracted and quantified in NRK52E cells
treated with 200 mM oleic acid or/and TGF-b. #P,0.05 versus nontreated control. *P,0.05 versus oleic acid treatment. (D) Protein
expression of PPARa, CPT1a, and ACOX1 was measured by western blotting in TGF-b and anti-miR-21–treated NRK52E cells. Actin
was used as the loading control. (E) Gene levels of PPARa target proteins were measured by qPCR after TGF-b and anti-miR-21
treatment in NRK52E cells. #P,0.05 versus nontreated control. *P,0.05 versus TGF-b treatment. (F) OCR was measured under
designated conditions. Oleic acid (200 mM) and oligomycin (1 mM) were treated at designated times. (G) Cellular TG was visualized by
oil red O staining in NRK52E cells treated with designated conditions. Scale bar=50 mm. (H) Cellular TG levels were extracted and
quantified in NRK52E cells treated with designated conditions. #P,0.05 versus nontreated control. *P,0.05 versus TGF-b and anti-
miR-21 treatment. (I) Changes in expression of ECM genes in NRK52E cells. Gene expression levels of ECM proteins were measured by
qPCR under oleic acid or/and TGF-b treatment conditions. #P,0.05 versus nontreated control. *P,0.05 nontreated control versus
TGF-b, oleic acid, and anti-miR-21 treatment.

(Figure 5D). PPARa target genes and protein expression increases TGF-b-induced fibrosis-related signaling in cells.
changes were also reduced by TGF-b treatment and blocked Although oleic acid treatment itself did not alter fibrosis-
by anti-miR-21 treatment (Figure 5, D and E). We next mea- related gene expression, oleic acid under the TGF-b treat-
sured the effect of anti-miR-21 on OCR under the TGF-b ment conditions significantly increased fibrosis-related
treatment condition. Treatment with anti-miR-21 blocked gene expression (Supplemental Figure 5). The effect of oleic
TGF-b-induced OCR level decrease (Figure 5F) and rescued acid was partially blocked by anti-miR-21 treatment
ATP levels (Supplemental Figure 4). In addition, anti-miR- (Figure 5I). Collectively, these data implicated the impor-
21 treatment significantly reduced oleic acid–induced lipid tance of the FAO pathway in TGF-b-induced fibrosis sig-
accumulation under the TGF-b treatment conditions naling in renal tubular epithelial cells, especially under
(Figure 5, G and H). We next checked whether lipid stress excessive lipid stresses.

1230 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 1223–1237, 2018
www.jasn.org BASIC RESEARCH

CR Restores PPARa Expression and Slows Renal PPARa2/2 mice showed significantly higher lipid accumula-
Fibrosis tion compared with littermates (Figure 8C). Interestingly,
CR is one of the most powerful antiaging regimens that aged PPARa2/2 mouse kidneys showed distinct histologic
can retard aging and age-related diseases.25 To investigate structures in the renal tubule region when analyzed by
whether CR can restore defective lipid metabolism during hematoxylin and eosin (H&E) staining (Figure 8D). Aged
aging, calorie-restricted aged rats were used. CR significantly PPARa2/2 mouse kidneys showed increased vacuoles in tu-
rescued aging-induced decreases in PPARa expression (Figure 6A). bules (Supplemental Figure 8, A and B). These distinct histo-
Furthermore, CPT1a and ACOX1 expression levels were also logic structures were found to be due to excessive lipid droplet
increased in response to CR (Figure 6A). The increased PPARa formation when the kidneys were subjected to oil red
and ACOX1 levels were confirmed by IHC analysis. Increased O staining (Figure 8D). These data suggested that PPARa de-
expressions of PPARa and ACOX1 were detected especially in ficiency significantly decreases FAO with high lipid accumu-
the epithelial region of CR kidneys (Figure 6B). Other PPARa lation in the tubule epithelial region during aging.
target genes that modulate FAO were also increased by CR We further investigated whether PPARa-deficient mice ex-
(Figure 6C). The increased expression of FAO regulators was hibit accelerated renal fibrosis during aging. Aged PPARa2/2
associated with decreased lipid accumulation in the CR kid- mice developed significantly more interstitial fibrosis than
neys (Figure 6, D and E). In contrast, other lipid metabolism– control littermates as detected by mRNA and protein expres-
associated genes were not affected by CR (Supplemental Fig- sion levels of ECM proteins (Figure 8, E and F). IHC staining
ure 6, A and B). We further detected lipid vacuoles in renal of collagen I, Sirius red, and Masson’s trichrome staining were
tubules during CR (Figure 6, F and G). CR significantly re- also higher in the interstitial region of aged PPARa2/2 kidneys
duced the number of vacuoles in tubules. PPARa-silencing (Figure 8G). Consistently, fibrotic scores were increased in
miRNAs were further investigated. CR resulted in significant aged PPARa2/2 kidneys when calculated by using Sirius
reductions in the levels of all three miRNAs (miR-21, miR- red–staining–positive area (Figure 8H). Serum urea levels
34a, miR-155) that showed increase during aging (Figure 6H). and histologic damages were also increased only in the aged
Together, these data suggested that CR restored PPARa ex- PPARa2/2 mice group (Figure 8I, Supplemental Figure 8, C–E).
pression through the regulation of miRNAs, implicating the Similarly, the epithelial injury marker KIM-1 was also expressed
importance of the FAO pathway in the regulation of renal at high levels in the aged PPARa2/2 mouse serum and kidneys
lipid accumulation. (Figure 8J, Supplemental Figure 8, F and G). Collectively, these
Because defective FAO is known to play a role in the devel- data indicated that PPARa deficiency not only increases renal
opment of fibrosis, we further examined the effects of CR on lipid accumulation but also accelerates renal interstitial fibro-
age-related fibrosis development. CR significantly reduced gene sis, suggesting the importance of PPARa on the development of
expression of several ECM components (Figure 7A). CR also age-associated renal fibrosis.
reduced protein expression of ECM components including
procollagen I, collagen I, and collagen IV (Figure 7B). IHC
staining of collagen I, Sirius red, and Masson’s trichrome stain- DISCUSSION
ing also showed decreased expression of ECM in the interstitial
region of the kidneys (Figure 7C). Consistently, fibrotic scores The role of lipid metabolism has been recently implicated in
were decreased in the CR group when the area positive for various renal diseases.26,27 The kidney is not a major metabolic
Sirius red staining was calculated (Figure 7D). Furthermore, organ that actively participates in the systemic regulation of
CR improved renal function with decreased epithelial damage lipid metabolism. However, recent evidence suggests that lipid
(Figure 7, E and F, Supplemental Figure 6, C–G). In summary, accumulation in nonadipose tissues can contribute to organ
CR significantly delayed aging-induced renal fibrosis with the injury.28 Clinical observations have implicated a potential as-
restoration of PPARa expression and FAO-associated proteins. sociation between renal lipid accumulation and CKD devel-
opment.29 Furthermore, there is an abundance of animal data
PPARa Deficiency Increases Renal Lipid Accumulation demonstrating an association between renal lipid accumula-
and Accelerates Renal Fibrosis tion and kidney dysfunction, including models of metabolic
To confirm the role of PPARa in aging-induced renal fibrosis, and nonmetabolic renal diseases.15,30 In addition, several pre-
we subjected PPARa2/2 mice and sex-matched control litter- vious reports have examined the effects of renal lipid metab-
mates to aging. We first compared changes in lipid metabo- olism on age-associated renal diseases.31,32 Aged animals show
lism. As expected, aged control mice showed decreased PPARa higher lipid accumulation with increased glomerulosclerosis
and FAO-associated gene expression (Figure 8, A and B). and tubulointerstitial fibrosis. However, although the occur-
Compared with aged control littermates, PPARa2/2 mice rence of renal lipid accumulation in aged kidneys was coinci-
showed reduced expression of FAO-associated proteins and dent in our aged model, the regions of lipid accumulation were
genes (Figure 8, A and B). In contrast, other lipid metabolism different compared with those reported by previous studies.
genes were not significantly changed in aged PPARa2/2 Our results clearly indicated that the site of lipid accumulation
mice except for Scd (Supplemental Figure 7, A and B). Aged was the renal tubular epithelial region.

J Am Soc Nephrol 29: 1223–1237, 2018 PPARa and Age-Associated Renal Fibrosis 1231
BASIC RESEARCH www.jasn.org

Figure 6. CR restores PPARa expression and FAO pathways with decreased lipid accumulation in the kidneys. Young (6-month), old
(24-month), and calorie-restricted old (24-month) rat kidneys were used to investigate the effects of CR on age-related renal fibrosis
(n=7–8). (A) Nuclear protein expression of PPARa and cytosolic protein expression of CPT1a and ACOX1 were measured by western
blotting. TFIIB and actin were used as the loading controls, respectively. (B) PPARa and ACOX1 expression in kidney samples were
visualized by IHC. Scale bar=50 mm. (C) Gene levels of PPARa target proteins were measured by qPCR. #P,0.05 versus young.
*P,0.05 versus old. (D) TGs were quantified in the kidneys. #P,0.05 versus young. *P,0.05 versus old. (E) Kidneys were stained with
oil red O to visualize lipid accumulation. Scale bar=50 mm. (F) Representative H&E staining shows increased vacuoles in renal tubules
during aging and CR. (G) Number of vacuoles was quantified by counting vacuoles per tubule. #P,0.05 versus young. *P,0.05 versus
old. (H) The miRNA levels that were changed during aging were detected by qPCR. #P,0.05 versus young. *P,0.05 versus old.

1232 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 1223–1237, 2018
www.jasn.org BASIC RESEARCH

Figure 7. CR prevents the development of age-related renal fibrosis. Young (6-month), old (24-month), and CR-old (24-month) rat
kidneys were used to investigate the effects of CR on age-related renal fibrosis (n=7–8). (A) Gene levels of ECM proteins were mea-
sured by qPCR. #P,0.05 versus young. *P,0.05 versus old. (B) Protein levels of ECM proteins were measured by western blotting.
Actin was used as the loading control. (C) Collagen I expression was visualized by IHC, and Masson’s trichrome (MT) and Sirius red (SR)
staining were performed to visualize fibrosis. Scale bar=50 mm. (D) Quantification of SR-stained fibrosis in kidneys. #P,0.05 versus
young. *P,0.05 versus old. (E) Urea levels were measured by biochemical methods in the serum to measure kidney function. #P,0.05
versus young. *P,0.05 versus old. (F) KIM-1 levels were detected in the serum via ELISA to measure renal epithelial damages. #P,0.05
versus young. *P,0.05 versus old.

Recently, roles of the impaired FAO pathway in the develop- accumulation was associated with an impaired FAO pathway.
ment of renal interstitial fibrosis have been investigated.15 The Critically, the protein levels of CPT1a, which is the rate-limiting
authors found that renal tubular epithelial cells depend critically enzyme of FAO, showed great reduction in the tubular epithelial
on FAO as their energy source, and depressed FAO was associ- region. However, we found that upstream regulators of PPARa,
ated with intracellular lipid accumulation. This observation was AMPK and PGC1a, which were shown to be significantly re-
consistent with previous reports demonstrating that mitochon- duced in the fibrosis model, were actually increased or un-
drial b-oxidation of FAs is a major source for renal energy pro- changed during aging. These differences led us to investigate
duction, which has a high energy demand with relatively low other mechanisms associated with decreased PPARa signaling
glycolytic capacity.33 They also suggested that impaired FAO was during aging.
owing to a decreased PPARa-PGC1a axis during renal fibrosis miRNAs are endogenously encoded, evolutionarily con-
development. We also found that age-associated renal lipid served small RNAs that regulate gene expression predominantly

J Am Soc Nephrol 29: 1223–1237, 2018 PPARa and Age-Associated Renal Fibrosis 1233
BASIC RESEARCH www.jasn.org

Figure 8. PPARa deficiency increases renal lipid accumulation and accelerates renal fibrosis. PPARa2/2 mice and their littermates were
subjected to aging to investigate the effect of PPARa on renal lipid metabolism and fibrosis changes during aging (n=5–7). (A) Gene

1234 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 1223–1237, 2018
www.jasn.org BASIC RESEARCH

by facilitating the degradation and translation inhibition of tar- It has been suggested that sirtuin 1 (SIRT1) is a main reg-
get mRNAs, thus having a powerful role in regulating various ulator that connects calorie intake and life span.38,39 Interest-
biologic processes.34 Additionally, miRNAs have also been im- ingly, SIRT1 has been shown to directly or indirectly regulate
plicated in regulating various kidney diseases, including renal PPARa expression and activity, and is also associated with re-
carcinoma, diabetic nephropathy, and AKI.35 We found a dis- nal fibrosis, aging, and other age-related diseases.40,41 Further-
crepancy between the gene and protein levels of PPARa, which more, SIRT1 is also regulated by miRNAs through epigenetic
suggests that there may be epigenetic mechanisms regulating regulation.42 Although we did not check the SIRT1 levels in
PPARa translation. Among the miRNAs that are known to our animal models, previous studies have shown decreased
regulate PPARa, miR-21 showed great efficiency in regulating SIRT1 expression and activity during renal aging.43 Because
PPARa expression in renal tubular epithelial cells. Interest- SIRT1 mediates various beneficial roles during CR, a possible
ingly, miR-212/2 mice have been reported to exhibit far less role of SIRT1 in age-related renal fibrosis and its association
interstitial fibrosis in response to kidney injury, and the lipid with PPARa and CR should be further considered.
metabolism pathway regulated by PPARa has been implicated Increasing evidence has also demonstrated an association
in this model.22 In addition, transfection of an miR-21 shRNA between PPARa and aging.44,45 The decreased expression or
plasmid halted the progression of renal fibrosis in established activity of PPARa has been detected during aging in several
obstructive nephropathy.24 We demonstrated the importance of tissues including kidney, heart, and spleen.46–48 More direct
the miR-21-PPARa axis in age-related renal fibrosis. Likewise, evidence on the relationship between PPARa and aging comes
miR-21-mediated PPARa silencing had significant effects in the from observations of PPARa2/2 mice. Accelerated physiolog-
regulation of lipid metabolism and TGF-b response in renal ically aged phenotypes were first reported in PPARa2/2 mice,
tubular epithelial cells. Given the previous reported data and along with decreased longevity and increased age-dependent
our results, miR-21 has been shown to play a role in the regula- lesions.49–51 In the liver, the roles of PPARa during aging have
tion of PPARa and lipid metabolism as an important regulator also been shown. PPARa2/2 mice and hepatocyte-specific
of renal fibrosis. PPARa2/2 mice showed significant alterations of systemic
Through the regulation of various biologic processes rang- lipid metabolism that lead to early hepatic steatosis during
ing from metabolic changes to epigenetic processes, CR is aging.52 Although it is evident that PPARa deficiency acceler-
known to regulate aging and age-related diseases in diverse ates renal aging with defects in systemic lipid metabolism, the
species.25 We hypothesized that changes in renal lipid metab- phenotypes of aged PPARa2/2 mice regarding renal lipid
olism are important in inducing age-associated renal fibrosis; metabolism have not been previously reported. We found
therefore, the effects of CR on renal lipid metabolism and that renal lipid metabolism was severely impaired in aged
fibrosis were further investigated. We found that CR signifi- PPARa2/2 mice with increased lipid accumulation. Impaired
cantly reduced renal lipid amounts with increased PPARa ex- lipid metabolism in the kidney was associated with early onset
pression and FAO-associated genes. CR also reduced miRNAs of renal fibrosis in aged PPARa2/2 mice. However, we cannot
that regulate PPARa silencing during aging. These changes in exclude other possibilities that can induce an early onset of the
lipid metabolism were associated with decreased renal fibrosis fibrosis phenotype in aged PPARa2/2 mice, because we used
during aging. The effects of CR on age-associated fibrosis have whole-body PPARa2/2 mice. Performing aging studies in kid-
been reported in other studies. Although the suggested mech- ney epithelial–specific PPARa2/2 mice will be important.
anisms vary between the model and tissues used, it is evident To conclude, these studies identify PPARa and the FAO
that CR can alleviate age-related kidney, heart, liver, and aorta pathway as important regulators in kidney lipid metabolism
fibrosis.32,36,37 As a strong metabolic modifier, CR can allevi- that amplifies age-associated renal fibrosis (Supplemental
ate renal fibrosis by regulating abnormal lipid metabolism Figure 9). The post-transcriptional regulator of PPARa,
during aging. which is miR-21, critically decreased FAO pathways in renal

levels of PPARa target proteins were measured by qPCR. #P,0.05 versus 7-month wild type (WT). *P,0.05 versus 20-month WT. (B)
Nuclear protein expression of PPARa and cytosolic protein expression of CPT1a and ACOX1 were measured by western blotting. TFIIB
and actin were used as the loading controls, respectively. (C) TGs were quantified in the kidneys. #P,0.05 versus 7-month WT. *P,0.05
versus 20-month WT. (D) Kidneys were stained with H&E to visualize structural differences, and oil red O staining was used to visualize lipid
accumulation. Scale bar in H&E staining=100 mm. Scale bar in oil red O staining=50 mm. (E) Gene levels of ECM proteins were measured
by qPCR. #P,0.05 versus 7-month WT. *P,0.05 versus 20-month WT. (F) Protein levels of ECM proteins were measured by western
blotting. Actin was used as the loading control. (G) Collagen I expression was visualized by IHC, and Masson’s trichrome (MT) and Sirius red
(SR) staining were performed to visualize fibrosis. Scale bar=100 mm. (H) Quantification of SR-stained fibrosis in the kidneys. #P,0.05
versus 7-month WT. *P,0.05 versus 20-month WT. (I) Urea levels were measured by biochemical methods in the serum to measure kidney
function. #P,0.05 versus 7-month WT. *P,0.05 versus 20-month WT. (J) KIM-1 levels were detected in the serum by ELISA to measure
renal epithelial damages. #P,0.05 versus 7-month WT. *P,0.05 versus 20-month WT. ORO, oil red O; Ppara, Peroxisome proliferator-
activated receptor alpha.

J Am Soc Nephrol 29: 1223–1237, 2018 PPARa and Age-Associated Renal Fibrosis 1235
BASIC RESEARCH www.jasn.org

epithelial cells with increased fibrotic responses. Antiaging ACKNOWLEDGMENTS


CR rescued age-associated PPARa and the FAO pathway
with slowed renal fibrosis. PPARa2/2 mice showed a severely The authors thank the Korean Aging Tissue Bank for providing re-
disrupted renal FAO pathway with high accumulation of lip- search materials. The authors also thank G.T.O. (Ewha Womans
ids, and showed earlier onset of renal fibrosis, demonstrating University) for providing PPARa2/2 mice.
the importance of PPARa in the development of age-related This work was supported by the National Research Foundation of
renal fibrosis. Future studies may explore the effects of im- Korea (NRF) funded by the Korean government (MSIP: Minister of
paired PPARa and FAO pathways on human aged kidney Science, ICT, and Future Planning) (grant Nos. 2009-0083538,
samples. It will also be necessary to assess the pharmacologic 2013M3A9B6076431 and 2015R1A2A2A01004137), and the Bio &
interventions (e.g., PPARa agonist, miR-21 antagonist) for Medical Technology Development Program of the NRF funded by
the treatment of age-associated renal fibrosis. If so, this could the MSIP (grant No. 2015M3A9B8029074).
provide novel therapeutics to intervene in renal fibrosis and K.W.C. and H.Y.C. developed the concept and designed the ex-
functional decline during aging. periments. K.W.C., E.K.L., and M.K.L. performed the experiments.
K.W.C., B.P.Y., and H.Y.C. prepared the manuscript. All of the au-
thors analyzed the results and edited the manuscript.
CONCISE METHODS

Animals DISCLOSURES
To investigate the changes in lipid metabolism and fibrosis during None.
aging, 6-, 12-, 18-, and 24-month-old SD rats (n=8) were used. To
compare the effects of CR on lipid metabolism and fibrosis
changes, young (6 months), aged (24 months), and CR (60% of REFERENCES
ad libitum for 1 month) aged (24 months) rats (n=7–8) were used.
To evaluate the effects of PPARa deficiency on age-associated re- 1. Weinstein JR, Anderson S: The aging kidney: Physiological changes.
nal fibrosis, age-matched (3, 7, and 20 months) PPARa2/2 mice Adv Chronic Kidney Dis 17: 302–307, 2010
and their wild-type littermates (n=5–7) were used. Animal studies 2. Fox CS, Larson MG, Leip EP, Culleton B, Wilson PW, Levy D: Predictors
of new-onset kidney disease in a community-based population. JAMA
were designed by the Aging Tissue Bank, approved by the Insti-
291: 844–850, 2004
tutional Animal Care Committee of Pusan National University, 3. de Boer IH, Katz R, Fried LF, Ix JH, Luchsinger J, Sarnak MJ, et al.:
and performed in accordance with the guidelines for animal ex- Obesity and change in estimated GFR among older adults. Am J Kidney
perimentation issued by Pusan National University. Rats and mice Dis 54: 1043–1051, 2009
were maintained under controlled environmental conditions 4. Coresh J, Selvin E, Stevens LA, Manzi J, Kusek JW, Eggers P, et al.:
Prevalence of chronic kidney disease in the United States. JAMA 298:
under a 12-hour light/dark cycle, and allowed ad libitum access
2038–2047, 2007
to water and a standard laboratory diet except for the CR study. 5. Davies DF, Shock NW: Age changes in glomerular filtration rate, ef-
PPARa2 /2 mice were genotyped by PCR as previously de- fective renal plasma flow, and tubular excretory capacity in adult males.
scribed. 53 For all animal experiments, serum was collected for J Clin Invest 29: 496–507, 1950
biochemical analysis. Kidneys were collected and either frozen 6. Stevens LA, Viswanathan G, Weiner DE: Chronic kidney disease and
end-stage renal disease in the elderly population: Current prevalence,
immediately in liquid nitrogen for qPCR, western blotting, and
future projections, and clinical significance. Adv Chronic Kidney Dis
biochemical tests, or fixed in neutral-buffered formalin for his- 17: 293–301, 2010
tochemical examination. For long-term storage, tissue samples 7. Denic A, Glassock RJ, Rule AD: Structural and functional changes with
were moved to a 280°C deep freezer located at the Aging Tissue the aging kidney. Adv Chronic Kidney Dis 23: 19–28, 2016
Bank in Pusan National University. 8. Duffield JS: Cellular and molecular mechanisms in kidney fibrosis. J Clin
Invest 124: 2299–2306, 2014
9. Farris AB, Colvin RB: Renal interstitial fibrosis: Mechanisms and evalu-
Histologic Analysis ation. Curr Opin Nephrol Hypertens 21: 289–300, 2012
Kidneys were fixed in 10% neutral formalin and paraffin-embedded 10. Allison SJ: Fibrosis: Targeting EMT to reverse renal fibrosis. Nat Rev
sections were stained with H&E. Sirius red staining and Masson’s Nephrol 11: 565, 2015
trichrome staining were performed as described previously to deter- 11. Sangaralingham SJ, Heublein DM, Grande JP, Cataliotti A, Rule AD,
McKie PM, et al.: Urinary C-type natriuretic peptide excretion: A po-
mine the degree of aging-associated fibrosis.54
tential novel biomarker for renal fibrosis during aging. Am J Physiol
Renal Physiol 301: F943–F952, 2011
Statistical Analyses 12. Rule AD, Amer H, Cornell LD, Taler SJ, Cosio FG, Kremers WK, et al.:
The t test was used to analyze differences between two groups, and The association between age and nephrosclerosis on renal biopsy
ANOVA was used to analyze intergroup differences. P values of ,0.05 among healthy adults. Ann Intern Med 152: 561–567, 2010
13. Kaur J: A comprehensive review on metabolic syndrome. Cardiol Res
were considered statistically significant. The analysis was performed
Pract 2014: 943162, 2014
using GraphPad Prism 5 (GraphPad software). 14. Meyer C, Nadkarni V, Stumvoll M, Gerich J: Human kidney free fatty
Detailed protocols for the other experiments are provided in the acid and glucose uptake: Evidence for a renal glucose-fatty acid cycle.
Supplemental Material. Am J Physiol 273: E650–E654, 1997

1236 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 1223–1237, 2018
www.jasn.org BASIC RESEARCH

15. Kang HM, Ahn SH, Choi P, Ko YA, Han SH, Chinga F, et al.: Defective 37. Ahmet I, Tae HJ, de Cabo R, Lakatta EG, Talan MI: Effects of calorie
fatty acid oxidation in renal tubular epithelial cells has a key role in restriction on cardioprotection and cardiovascular health. J Mol Cell
kidney fibrosis development. Nat Med 21: 37–46, 2015 Cardiol 51: 263–271, 2011
16. Simon N, Hertig A: Alteration of fatty acid oxidation in tubular epithe- 38. Hou L, Wang D, Chen D, Liu Y, Zhang Y, Cheng H, et al.: A systems
lial cells: From acute kidney injury to renal fibrogenesis. Front Med approach to reverse engineer lifespan extension by dietary restriction.
(Lausanne) 2: 52, 2015 Cell Metab 23: 529–540, 2016
17. Lefebvre P, Chinetti G, Fruchart JC, Staels B: Sorting out the roles of 39. Kitada M, Koya D: SIRT1 in Type 2 diabetes: Mechanisms and thera-
PPAR alpha in energy metabolism and vascular homeostasis. J Clin peutic potential. Diabetes Metab J 37: 315–325, 2013
Invest 116: 571–580, 2006 40. Huang XZ, Wen D, Zhang M, Xie Q, Ma L, Guan Y, et al.: Sirt1 activation
18. Wang YX: PPARs: Diverse regulators in energy metabolism and meta- ameliorates renal fibrosis by inhibiting the TGF-b/Smad3 pathway.
bolic diseases. Cell Res 20: 124–137, 2010 J Cell Biochem 115: 996–1005, 2014
19. Li S, Mariappan N, Megyesi J, Shank B, Kannan K, Theus S, et al.: 41. Purushotham A, Schug TT, Xu Q, Surapureddi S, Guo X, Li X: Hepatocyte-
Proximal tubule PPARa attenuates renal fibrosis and inflammation specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic
caused by unilateral ureteral obstruction. Am J Physiol Renal Physiol steatosis and inflammation. Cell Metab 9: 327–338, 2009
305: F618–F627, 2013 42. Yamakuchi M: MicroRNA regulation of SIRT1. Front Physiol 3: 68, 2012
20. Mori Y, Hirano T, Nagashima M, Shiraishi Y, Fukui T, Adachi M: De- 43. Yuan Y, Cruzat VF, Newsholme P, Cheng J, Chen Y, Lu Y: Regulation of
creased peroxisome proliferator-activated receptor alpha gene ex- SIRT1 in aging: Roles in mitochondrial function and biogenesis. Mech
pression is associated with dyslipidemia in a rat model of chronic renal Ageing Dev 155: 10–21, 2016
failure. Metabolism 56: 1714–1718, 2007 44. Chung KW, Yu BP, Chung HY: Anti-aging action of PPARs: Potential
21. Han SH, Malaga-Dieguez L, Chinga F, Kang HM, Tao J, Reidy K, et al.: therapeutic targets. In: Anti-aging Drugs: From Basic Research to
Deletion of Lkb1 in renal tubular epithelial cells leads to CKD by altering Clinical Practice, edited by Vaiserman AM, United Kingdom, Royal
metabolism. J Am Soc Nephrol 27: 439–453, 2016 Society of Chemistry, 2017, pp 393–415
22. Chau BN, Xin C, Hartner J, Ren S, Castano AP, Linn G, et al.: MicroRNA- 45. Chung JH, Seo AY, Chung SW, Kim MK, Leeuwenburgh C, Yu BP, et al.:
21 promotes fibrosis of the kidney by silencing metabolic pathways. Sci Molecular mechanism of PPAR in the regulation of age-related in-
Transl Med 4: 121ra18, 2012 flammation. Ageing Res Rev 7: 126–136, 2008
23. Desvergne B, Michalik L, Wahli W: Transcriptional regulation of me- 46. Sung B, Park S, Yu BP, Chung HY: Modulation of PPAR in aging, in-
tabolism. Physiol Rev 86: 465–514, 2006 flammation, and calorie restriction. J Gerontol A Biol Sci Med Sci 59:
24. Zhong X, Chung AC, Chen HY, Meng XM, Lan HY: Smad3-mediated 997–1006, 2004
upregulation of miR-21 promotes renal fibrosis. J Am Soc Nephrol 22: 47. Iemitsu M, Miyauchi T, Maeda S, Tanabe T, Takanashi M, Irukayama-
1668–1681, 2011 Tomobe Y, et al.: Aging-induced decrease in the PPAR-alpha level in
25. Chung KW, Kim DH, Park MH, Choi YJ, Kim ND, Lee J, et al.: Recent hearts is improved by exercise training. Am J Physiol Heart Circ Physiol
advances in calorie restriction research on aging. Exp Gerontol 48: 283: H1750–H1760, 2002
1049–1053, 2013 48. Sanguino E, Roglans N, Alegret M, Sánchez RM, Vázquez-Carrera M,
26. Trevisan R, Dodesini AR, Lepore G: Lipids and renal disease. J Am Soc Laguna JC: Atorvastatin reverses age-related reduction in rat hepatic
Nephrol 17[Suppl 2]: S145–S147, 2006 PPARalpha and HNF-4. Br J Pharmacol 145: 853–861, 2005
27. Bobulescu IA: Renal lipid metabolism and lipotoxicity. Curr Opin 49. Poynter ME, Daynes RA: Peroxisome proliferator-activated receptor
Nephrol Hypertens 19: 393–402, 2010 alpha activation modulates cellular redox status, represses nuclear
28. van Herpen NA, Schrauwen-Hinderling VB: Lipid accumulation in non- factor-kappaB signaling, and reduces inflammatory cytokine pro-
adipose tissue and lipotoxicity. Physiol Behav 94: 231–241, 2008 duction in aging. J Biol Chem 273: 32833–32841, 1998
29. Herman-Edelstein M, Scherzer P, Tobar A, Levi M, Gafter U: Altered 50. Howroyd P, Swanson C, Dunn C, Cattley RC, Corton JC: Decreased
renal lipid metabolism and renal lipid accumulation in human diabetic longevity and enhancement of age-dependent lesions in mice lacking
nephropathy. J Lipid Res 55: 561–572, 2014 the nuclear receptor peroxisome proliferator-activated receptor alpha
30. Kume S, Uzu T, Araki S, Sugimoto T, Isshiki K, Chin-Kanasaki M, et al.: (PPARalpha). Toxicol Pathol 32: 591–599, 2004
Role of altered renal lipid metabolism in the development of renal injury 51. Atherton HJ, Gulston MK, Bailey NJ, Cheng KK, Zhang W, Clarke K,
induced by a high-fat diet. J Am Soc Nephrol 18: 2715–2723, 2007 et al.: Metabolomics of the interaction between PPAR-alpha and age in
31. Jiang T, Liebman SE, Lucia MS, Li J, Levi M: Role of altered renal lipid the PPAR-alpha-null mouse. Mol Syst Biol 5: 259, 2009
metabolism and the sterol regulatory element binding proteins in the 52. Montagner A, Polizzi A, Fouché E, Ducheix S, Lippi Y, Lasserre F, et al.:
pathogenesis of age-related renal disease. Kidney Int 68: 2608–2620, 2005 Liver PPARa is crucial for whole-body fatty acid homeostasis and is
32. Jiang T, Liebman SE, Lucia MS, Phillips CL, Levi M: Calorie restriction protective against NAFLD. Gut 65: 1202–1214, 2016
modulates renal expression of sterol regulatory element binding pro- 53. Lee SS, Pineau T, Drago J, Lee EJ, Owens JW, Kroetz DL, et al.:
teins, lipid accumulation, and age-related renal disease. J Am Soc Targeted disruption of the alpha isoform of the peroxisome pro-
Nephrol 16: 2385–2394, 2005 liferator-activated receptor gene in mice results in abolishment of
33. Balaban RS, Mandel LJ: Metabolic substrate utilization by rabbit the pleiotropic effects of peroxisome proliferators. Mol Cell Biol 15:
proximal tubule. An NADH fluorescence study. Am J Physiol 254: 3012–3022, 1995
F407–F416, 1988 54. Bajwa A, Huang L, Kurmaeva E, Ye H, Dondeti KR, Chroscicki P, et al.:
34. Mendell JT, Olson EN: MicroRNAs in stress signaling and human dis- Sphingosine kinase 2 deficiency attenuates kidney fibrosis via IFN-g.
ease. Cell 148: 1172–1187, 2012 J Am Soc Nephrol 28: 1145–1161, 2017
35. Trionfini P, Benigni A, Remuzzi G: MicroRNAs in kidney physiology and
disease. Nat Rev Nephrol 11: 23–33, 2015
36. Horrillo D, Gallardo N, Lauzurica N, Barrus MT, San Frutos MG, Andres
A, et al.: Development of liver fibrosis during aging: Effects of caloric This article contains supplemental material online at http://jasn.asnjournals.
restriction. J Biol Regul Homeost Agents 27: 377–388, 2013 org/lookup/suppl/doi:10.1681/ASN.2017070802/-/DCSupplemental.

J Am Soc Nephrol 29: 1223–1237, 2018 PPARa and Age-Associated Renal Fibrosis 1237

You might also like