YAMAMOTO 2016

You might also like

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

BASIC RESEARCH www.jasn.

org

High-Fat Diet-Induced Lysosomal Dysfunction and


Impaired Autophagic Flux Contribute to Lipotoxicity in
the Kidney
Takeshi Yamamoto,* Yoshitsugu Takabatake,* Atsushi Takahashi,* Tomonori Kimura,*
Tomoko Namba,* Jun Matsuda,* Satoshi Minami,* Jun-ya Kaimori,† Isao Matsui,*
Taiji Matsusaka,‡ Fumio Niimura,§ Motoko Yanagita,| and Yoshitaka Isaka*
Departments of *Nephrology and †Advanced Technology for Transplantation, Osaka University Graduate School of
Medicine, Suita, Osaka, Japan; ‡Institute of Medical Sciences and Department of Molecular Life Sciences and
§
Department of Pediatrics, Tokai University School of Medicine, Isehara, Kanagawa, Japan; and |Department of
Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan

ABSTRACT
Excessive fat intake contributes to the progression of metabolic diseases via cellular injury and
inflammation, a process termed lipotoxicity. Here, we investigated the role of lysosomal dysfunction and
impaired autophagic flux in the pathogenesis of lipotoxicity in the kidney. In mice, a high-fat diet (HFD)
resulted in an accumulation of phospholipids in enlarged lysosomes within kidney proximal tubular cells
(PTCs). In isolated PTCs treated with palmitic acid, autophagic degradation activity progressively stagnated
in association with impaired lysosomal acidification and excessive lipid accumulation. Pulse-chase experiments
revealed that the accumulated lipids originated from cellular membranes. In mice with induced PTC-specific
ablation of autophagy, PTCs of HFD-mice exhibited greater accumulation of ubiquitin-positive protein ag-
gregates normally removed by autophagy than did PTCs of mice fed a normal diet. Furthermore, HFD-mice
had no capacity to augment autophagic activity upon another pathologic stress. Autophagy ablation also
exaggerated HFD-induced mitochondrial dysfunction and inflammasome activation. Moreover, renal
ischemia-reperfusion induced greater injury in HFD-mice than in mice fed a normal diet, and ablation
of autophagy further exacerbated this effect. Finally, we detected similarly enhanced phospholipid accu-
mulation in enlarged lysosomes and impaired autophagic flux in the kidneys of obese patients compared
with nonobese patients. These findings provide key insights regarding the pathophysiology of lipotoxicity
in the kidney and clues to a novel treatment for obesity-related kidney diseases.

J Am Soc Nephrol 28: ccc–ccc, 2016. doi: 10.1681/ASN.2016070731

In the last several decades, the increased prevalence The mitochondrial b-oxidation of free fatty
of obesity has been considered a serious health and acids (FFAs) is a major source of renal ATP produc-
economic burden worldwide.1 A growing body of ev- tion, particularly in proximal tubular cells (PTCs),
idence suggests that excess lipid intake induces cell in-
Received July 8, 2016. Accepted November 7, 2016.
jury and organ dysfunction (termed lipotoxicity).2,3
Lipotoxicity has been intensively studied in organs T.Y. and Y.T. contributed equally to this work.
that have evolved specialized lipid handling functions, Published online ahead of print. Publication date available at
such as adipose tissue and liver. Although Moorhead www.jasn.org.
proposed the hypothesis that lipid abnormalities con-
Correspondence: Dr. Yoshitsugu Takabatake, Department of
tribute to the progression of kidney injury in 1982,4 Nephrology, Osaka University Graduate School of Medicine, Box
and increasing evidence has supported the lipid neph- D11, 2-2 Yamada-oka, Suita, Osaka 585-0871, Japan. Email:
takaba@kid.med.osaka-u.ac.jp
rotoxicity hypothesis, studies on lipid metabolism in
the kidney are scarce. Copyright © 2016 by the American Society of Nephrology

J Am Soc Nephrol 28: ccc–ccc, 2016 ISSN : 1046-6673/2805-ccc 1


BASIC RESEARCH www.jasn.org

which have a high energy demand.5–7 PTCs take up circulating led to Oil Red O-positive lipid droplet formation, especially in
FFAs dissociated from albumin through specific membrane nonobese mice (Supplemental Figure 1A). In contrast, almost
proteins, such as fatty acid (FA) translocase (CD36) and all vacuoles in obese mice were positively stained with Nile red,
FA-binding protein.8 In addition, PTCs retrieve albumin- which detects all neutral lipids, and with toluidine blue stain-
bound FFAs from the glomerular filtrate by receptor-mediated ing, which detects phospholipids (Supplemental Figure 1, B
albumin endocytosis. and C). Electron microscopy revealed that the vacuoles con-
Autophagy is one of the major cellular degradation path- tain multilamellar, onion skin-like structures indicative of
ways, along with the ubiquitin–proteasome system. 9–11 phospholipids in agreement with a previous report (Supple-
Recently, a close relationship between autophagic activity mental Figure 1, D–K).26 In addition, the margins of the
and lipid metabolism has been recognized. Firstly, lipid drop- vacuoles were immunostained with lysosomal-associated
lets are sequestered by autophagosomes and eventually broken membrane protein 1 (LAMP1) (Supplemental Figure 1L).
down by lysosomes through a process termed lipophagy.12–14
Secondly, several saturated and unsaturated FFAs appear to HFD/PA Induces Lysosomal Dysfunction with
modulate autophagic activity. For example, palmitic acid Impairment in Acidification
(PA) activates autophagy by downregulation of the mechanis- Inspired by the above observations, we postulated that
tic target of rapamycin (mTOR) signal15 or by the phosphor- HFD affects lysosomal machinery and/or autophagic degrada-
ylation of the double-stranded RNA-dependent protein kinase tion in the PTCs. Eight-week-old green fluorescent protein-
c-Jun N-terminal kinases 1 pathway.16 Moreover, PA causes conjugated microtubule-associated protein 1 light chain 3
the disruption of inhibitory signal transducer and activator of (GFP-MAP1LC3) transgenic mice, in which GFP-positive
transcription 3–RNA-dependent protein kinase interactions, which puncta reflect autophagosomes,27 were fed a ND or a HFD
facilitates autophagy induction.17 In contrast, a high-fat diet (HFD) for 2 months. We found lysosomal vacuoles containing GFP-
downregulates autophagy by reducing autophagosome/lysosome positive phospholipid accumulation, which should not be ob-
fusion,18 as well as by decreasing the number and the acidity served in acidic environment (Figure 1B).28 We then treated
of lysosomes.19 The conflicting results may be attributed to cultured PTCs stably expressing GFP-MAP1LC3 with
differences in cell types, observation period, FFAs concentra- PA-bound BSA. Colocalization of GFP and LysoTracker
tion and, more likely, difficulty in monitoring autophagic flux Red was significantly increased in PA-treated PTCs compared
in vivo. with BSA-treated controls, suggesting impairment in lyso-
We have previously demonstrated that enhanced autopha- somal acidification (Figure 1C). Furthermore, we utilized
gic activity in the kidney proximal tubules plays a protective the LysoSensor Yellow/Blue reagent, a pH-sensitive probe
role in several pathologic settings.20–24 More recently, we have that emits predominantly yellow fluorescence in acidic organ-
established methods for monitoring autophagic activity in elles and blue fluorescence in less acidic organelles. Treatment
vivo.25 Given that autophagic activity can be downregulated of PTCs with PA significantly increased the blue/yellow ratio in
by lipid overload, cellular functions may be jeopardized in the acidic vesicular organelles, reflecting an increase in pH
pathologic settings in which properly enhanced autophagy (Figure 1D). We also found a time-dependent decrease in cel-
should exert a compensatory or protective role. On the lular ATP levels in PTCs treated with PA (Figure 1E). Electron
basis of this background information, we investigated (1) microscopy analysis revealed a massive vacuolization contain-
the pathophysiology of lipotoxicity in the PTCs with a focus ing undigested proteins and organelles, and degenerated mito-
on lysosomes and mitochondria, (2) lipid overload-mediated chondria in the PTCs treated with PA (Figure 1, F–M).
alterations in autophagic activity in vivo and in vitro, and (3)
the effects of autophagy deficiency on kidney morphology and PA Treatment Mobilizes Phospholipids from Cellular
function during lipid overload. Membranes to Lysosomes via Autophagy
To investigate the origin of phospholipids and the role of auto-
phagy in phospholipid accumulation, we performed an FA
RESULTS pulse-chase assay using phosphatidylcholine fluorescence-
tagged at its FA tail (FL HPC) (Figure 2A). Cultured PTCs
HFD Induces Phospholipid Accumulation in the PTC were initially labeled overnight with trace amounts of FL
Lysosomes HPC, which is known to integrate into various cellular mem-
Eight-week-old mice were fed a normal diet (ND) or a HFD for branes, including the endoplasmic reticulum, mitochondria,
2 months (hereafter, referred to as nonobese and obese mice, and Golgi apparatus. Cultured PTCs chased with BSA treat-
respectively). Cytosolic vacuolar formation was induced in the ment preserved the FL HPC signal in cellular membrane
PTCs of the cortex by HFD, but not by ND (Figure 1A). structures (Figure 2B). In contrast, the FL HPC signal on
Twenty-four hours of starvation increased vacuole size in membranes was decreased in PA-treated PTCs. Colocaliza-
the PTC of obese mice, but not in nonobese mice. There tion of FL HPC and LysoTracker Red was significantly in-
were very few Oil Red O-positive vacuoles in the PTCs of creased and enlarged in PA-treated PTCs compared with
both obese and nonobese mice, whereas 24 hours of starvation BSA-treated controls, indicative of PA-induced redistribution

2 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org BASIC RESEARCH

Figure 1. HFD contributes to phospholipid accumulation in the dysfunctional lysosomes of PTCs. (A) PAS-stained kidney cortical regions
of nonobese or obese mice that were either fed or subjected to 24 hours of starvation (n=6–9 in each group). Magnified images are
shown in the insets (original magnification, 31000). (B) Kidney sections of nonobese and obese GFP-MAP1LC3 transgenic mice (n=6–9
in each group) were immunostained for LAMP1, a marker of lysosome, or LRP2/MEGALIN, a marker of proximal tubules (red), and
counterstained with DAPI (blue). (C) PTCs isolated from wild-type mice stably expressing GFP-MAP1LC3 were stained with LysoTracker

J Am Soc Nephrol 28: ccc–ccc, 2016 Autophagy and Lipotoxicity 3


BASIC RESEARCH www.jasn.org

of phospholipids from cellular membranes into lysosomes autophagic activity in obese mice and low basal autophagy in
(Figure 2B). The active involvement of autophagy in FL nonobese mice. Next, autophagic flux was assessed in response
HPC transfer from cellular membranes was shown by immu- to 24 hours of starvation. Chloroquine administration signif-
nostaining for MAP1LC3 and LAMP1 in autophagy-deficient icantly increased GFP-positive puncta in starved nonobese
and -competent cultured PTCs (Figure 2, C and D). Under PA mice, whereas the increase was blunted in obese mice (Figure
treatment, FL HPC was occasionally found in autophago- 3, C and D). Western blot analysis (MAP1LC3, SQSTM1/p62,
somes in addition to lysosomes (Figure 2C). This FL HPC/ and ubiquitinated proteins) using whole kidney lysates
autophagosome/lysosome colocalization was inhibited by roughly confirmed this fluorescence study (Supplemental Fig-
autophagy deficiency (Figure 2D). ure 2, B and C). Overall, these results demonstrate that the
PTCs in nonobese mice exhibit low basal autophagic activity
HFD-Loaded Kidney Is More Reliant on Autophagy for and high starvation-induced autophagic flux, whereas those in
the Degradation of Increased Substrates obese mice exhibit high basal autophagic flux and stagnated
Next, HFD-mediated changes in demand and activity of basal autophagic flux by starvation.
autophagy were evaluated in the recently established tamoxifen-
inducible PTC-specific autophagy-deficient mice (Atg5F/F;N-myc PA Treatment Stimulates Autophagic Activity and
downstream regulated gene 1 [NDRG1]).25,29 Eight-week-old Subsequently Induces Downstream Suppression of
mice (Atg5F/F;NDRG1 and Atg5F/F control mice) were fed a Autophagic Flux
ND or HFD for 2 months. Three weeks before euthanasia, We next investigated the effects of PA overload on autophagic
ND-fed nonobese control and HFD-induced obese mice re- activity in cultured PTCs. BSA treatment promoted the con-
ceived tamoxifen to induce genetic ablation of Atg5. The accu- version of MAP1LC3-I to MAP1LC3-II, which was augmented
mulation of SQSTM1/p62 and ubiquitin represents the in the presence of bafilomycin A1 at all indicated times (Sup-
amount of substrate requiring degradation that accumulated plemental Figure 3A). Consistently, BSA treatment increased
over the 3-week period, because SQSTM1/p62 is the ubiquitin- the degradation of SQSTM1/p62 protein in a time-dependent
and MAP1LC3-binding protein that regulates the formation of manner, as shown by the reduction of SQSTM1/p62 turnover
protein aggregates and is removed by autophagy, thus index (defined as the proportion of the levels of SQSTM1/p62
serving as an index of autophagic degradation.30,31 Auto- of the indicated sample to the levels of SQSTM1/p62 of the
phagy deficiency was confirmed (Supplemental Figure 2). untreated controls at the baseline), indicating BSA-induced
PTCs of obese Atg5F/F;NDRG1 mice treated with tamoxifen upregulation of autophagic flux, as decreased SQSTM1/p62
exhibited mild cytosolic swelling compared with those protein levels are associated with autophagy activation (Sup-
of nonobese Atg5F/F;NDRG1 mice (Figure 3A). Moreover, plemental Figure 3B). In contrast, PA treatment strongly in-
3-week ablation of autophagy triggered increased accumula- creased the conversion of MAP1LC3-I to MAP1LC3-II with no
tion of SQSTM1/p62- and ubiquitin-positive protein aggre- additional increase in MAP1LC3-II observed in the presence
gates in obese Atg5F/F;NDRG1 mice, indicating that obese of bafilomycin A1 (Supplemental Figure 3A). Consistently, PA
mice utilize autophagy for the degradation of increasing treatment significantly increased the degradation of SQSTM1/
substrates (Figure 3B). p62 protein at early stages (3 hours), and thereafter, a significant
rebound at a late stage was observed (12 hours) (Supplemental
HFD Alters Autophagic Flux in Response to Starvation Figure 3B). The PA-induced rebound of the SQSTM1/p62 turn-
To assess HFD-mediated alterations in autophagic flux, GFP- over index was presumably because of the downstream suppres-
MAP1LC3 transgenic mice were administered chloroquine sion of autophagic flux, as SQSTM1/p62 mRNA expression was
6 hours before euthanasia. GFP-positive puncta were rarely unchanged (Supplemental Figure 3C).
observed in the fed, chloroquine-free nonobese mice,
whereas a substantial number of GFP-positive puncta were Mitophagy Could Protect PTCs from PA-Induced
observed in the LRP2/MEGALIN-positive PTCs of the fed, Mitochondrial Injury
chloroquine-free obese mice (Figure 3, C and D). Chloroquine To determine the role of autophagy during lipid overload, we
administration significantly increased the number of GFP- exposed autophagy-deficient and -competent cells to PA or
positive puncta in obese mice, whereas the number of puncta BSA. The mitochondrial membrane potential, assessed by
remained unchanged in nonobese mice, suggesting high basal tetramethylrhodamine ethyl ester (TMRE), was significantly

Red, and counterstained with DAPI (blue) after treatment with either 0.25% BSA or 0.25 mM PA for 12 hours (n=5). (D) PTCs were stained with
the pH indicator LysoSensor Yellow/Blue after treatment with either 0.25% BSA or 0.25 mM PA for 12 hours (n=5). The blue/yellow ratio
is presented (right). (E) Cellular ATP levels after treatment with either 0.25% BSA or 0.25 mM PA for 12 hours. (n=5). (F–M) Electron
micrographs of PTCs treated with 0.25% BSA (F–I) or with 0.25 mM PA (J–M) (n=2). (M’ and I’) Outer mitochondrial membranes are traced
(solid lines). Bars: 50 mm (A), 10 mm (B, C, and D), 5 mm (F and J), and 500 nm (G–I and K–M). Data are provided as mean6SEM. Statistically
significant differences (*P,0.05) are indicated. All images are representative of multiple experiments. M, mitochondria.

4 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org BASIC RESEARCH

Figure 2. PA treatment mobilizes phospholipids from cellular membranes via autophagy in the PTCs. (A) Schematic illustration of
fluorescence FA pulse-chase assay. (A and B) FL-HPC loaded PTCs isolated from wild-type mice were chased after treatment with either
0.25% BSA or 0.25 mM PA for 6 hours (n=5), and the subcellular localization of FL-HPC was determined by staining with LysoTracker
Red. To determine phospholipid accumulations in lysosomes, the number and size of merged dots were measured. (C and D) FL-HPC

J Am Soc Nephrol 28: ccc–ccc, 2016 Autophagy and Lipotoxicity 5


BASIC RESEARCH www.jasn.org

reduced in both cell lines with PA treatment compared with mice (Figure 5A). This remarkable finding confirms the in
those with BSA; however, the reduction was more prominent vitro data that PA-induced autophagy is involved in shuttling
in autophagy-deficient PTCs (Figure 4A). Furthermore, PA FL HPC from cellular membranes into lysosomes where phos-
significantly increased mitochondrial reactive oxygen species pholipids accumulate (Figure 2, A–D). In the kidneys of obese
(ROS), assessed by MitoSOX Red staining, in autophagy- Atg5F/F control littermates, massive lysosomal accumulations
deficient PTCs but not in autophagy-competent PTCs (Figure containing multilamellar, onion skin-like structures indicative
4B). PA significantly decreased mitochondrial DNA/nuclear of phospholipids were confirmed (Figure 5, B and C), whereas
DNA (mtDNA/nDNA) ratios, which was more pronounced mitophagy was occasionally observed (Figure 5D) and there
in autophagy-deficient PTCs than autophagy-competent was no remarkable morphologic change in the mitochondria
PTCs (Figure 4C). Consistent with these observations, PA (Figure 5E). In contrast, we found massive aggregation of
decreased cell survival of both PTCs, which was prominent fragmented and swollen mitochondria with derangement of
in autophagy-deficient PTCs (Supplemental Figure 4A). PA the cristae in obese Atg5F/F;KAP mice, although no remark-
significantly decreased mRNA of mitochondrial oxidative able morphologic change in lysosomes was observed (Figure
phosphorylation gene in both autophagy-competent and 5, F–I). COX and succinate dehydrogenase (SDH) staining
-deficient PTCs and increased mRNAs of FA b-oxidation were performed to evaluate HFD-induced alterations in mito-
genes in autophagy-competent PTCs but not in autophagy- chondrial respiration activity. PTCs of nonobese Atg5F/F;KAP
deficient PTCs (Supplemental Figure 4B). These data suggest mice showed less intense COX and SDH staining than those of
that PA places substantial stress on mitochondria, which is nonobese controls (Supplemental Figure 5, A and B). Tubular
alleviated by autophagy. COX and SDH staining was somewhat decreased in obese Atg5F/F
We next examined whether PA induces autophagy to selec- control littermates compared with nonobese Atg5F/F control
tively eliminate impaired mitochondria (mitophagy). Staining littermates; however, a more prominent decline in COX and
mitochondria with MitoTracker Red FM in GFP-MAP1LC3– SDH staining was observed in obese Atg5F/F;KAP mice. We
transfected PTCs demonstrated that more autophagosomes found increased staining for oxidative stress markers, including
were colocalized with mitochondria in PTCs treated with PA 4-hydroxy-2-nonenal (HNE), dityrosine, and N-carboxymethyl
than BSA (Figure 4D). Furthermore, to investigate whether lysine (CML), in obese Atg5F/F;KAP kidneys (Supplemental
ubiquitinated mitochondria were recognized as a target of Figure 5C). Nonobese Atg5F/F;KAP mice exhibited patchy
autophagic degradation, we stained GFP-MAP1LC3 and macrophage infiltration into the outer stripe of the outer
mStrawberry-ubiquitin double-transfected PTCs with medulla, which was significantly exaggerated by HFD (Fig-
MitoTracker Deep Red FM. More autophagosomes were ure 5J). This macrophage infiltration was associated with
localized with mitochondria and ubiquitin when treated change in the levels of mRNA expression of inflammatory
with PA than with BSA (Figure 4E). The phosphatase and tensin cytokines TNFa, chemokine CC motif ligand 2 (CCL2), and
homolog-induced putative kinase 1 (PINK1) and Parkin RBR E3 CCL3, as well as inflammasome-related molecules nod-like
ubiquitin protein ligase (PARK2/Parkin) pathway is critical in receptor, pyrin containing 3 (NLRP3); apoptosis-associated
controlling mitophagy.32 Western blot analysis revealed that PA speck-like protein containing; and caspase-1 (Figure 5,
increased the expression levels of PINK1 and PARK2/Parkin K and L). Western blot analysis (NLRP3, caspase-1, IL-1b,
but decreased level of cytochrome c oxidase subunit IV and IL-18) confirmed HFD-induced inflammasome activa-
(COX4), which confirmed PA-mediated mitophagy induction tion (Figure 5M). Obese Atg5F/F control mice showed in-
and mitochondrial depletion (Figure 5M). Together, although creased expression of IL-1b in F4/80-positive macrophages,
causal relationship between autophagy deficiency and mito- which was significantly exaggerated by autophagy deficiency
chondrial dysfunction remains to be examined, mitophagy (Figure 5, M and N). Additionally, to investigate the long-
could protect proximal tubules from PA-induced mitochon- term consequences of HFD-induced lipotoxicity, we ob-
drial damage. served Atg5F/F;KAP mice fed a HFD for up to 10 months.
Obese Atg5F/F;KAP mice exhibited severe brush border loss,
Autophagy Deficiency Aggravates HFD-Induced tubular dilation, cast formation, enhanced macrophage in-
Mitochondrial Dysfunction, Inflammation, and Fibrosis filtration, and interstitial fibrosis (Supplemental Figure 6,
We next examined the role of autophagy during lipid overload A–C). Collectively, autophagy protects kidney PTCs from
using PTC-specific Atg5-deficient mice (Atg5F/F;kidney an- HFD-induced reduction in mitochondrial function, sup-
drogen regulated protein [KAP] mice). Cytosolic vacuolar for- presses macrophage infiltration and inflammasome activa-
mation was almost completely resolved in obese Atg5F/F;KAP tion, and slows HFD-induced renal fibrosis.

loaded autophagy-competent and -deficient PTCs were immunostained for MAP1LC3 (red) and LAMP1 (white), and counterstained with
DAPI (blue) after treatment with either 0.25% BSA or 0.25 mM PA for 6 hours (n=3). Magnified images are shown in the insets (original
magnification, 37200). Bars: 10 mm. Data are provided as mean6SEM. Statistically significant differences (*P,0.05) are indicated. All
images are representative of multiple experiments.

6 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org BASIC RESEARCH

Figure 3. HFD induces autophagy dependency and alteration in autophagic flux in the PTCs. (A and B) The HFD-loaded kidney is more reliant on
autophagy for the degradation of increased substrates (n=4–5 in each group). (A) PAS-stained kidney cortical regions of vehicle- or tamoxifen-treated
nonobese and obese Atg5F/F and Atg5F/F;NDRG1 mice. (A, i and ii) Magnified images from tamoxifen-treated nonobese and obese Atg5F/F;
NDRG1 mice are presented (original magnification, 31000). (B) Kidney cortical regions of nonobese and obese Atg5F/F and Atg5F/F;NDRG1 mice
were immunostained for SQSTM1/p62 and ubiquitin after treatment with vehicle or tamoxifen 3 weeks before euthanasia. Magnified images
(original magnification, 31600) from tamoxifen-treated Atg5F/F;NDRG1 mice are presented. The number of SQSTM1/p62- or ubiquitin-positive

J Am Soc Nephrol 28: ccc–ccc, 2016 Autophagy and Lipotoxicity 7


BASIC RESEARCH www.jasn.org

HFD Enhances the Vulnerability toward Ischemia- overload stimulates autophagy; (2) elevated autophagy
Reperfusion Injury in Autophagy-Deficient Mice plays a crucial role in counteracting lipotoxicity through mi-
We exposed mice to ischemia-reperfusion (I/R) injury to deter- tochondrial quality control; (3) long-term lipid overload and
mine whether HFD-induced chronic lipotoxicity leads to vulner- subsequent autophagic activation places a burden on the ly-
ability to ischemic and/or oxidative stresses. More severely injured sosomal system, resulting in phospholipid accumulation and
tubules with massive tubular sediments and vacuolation inadequate acidification; (4) lysosomal dysfunction, in turn,
were observed in I/R-injured obese Atg5F/F mice compared with stagnates autophagic flux, which can lead to vulnerability to I/R
I/R-injured nonobese Atg5F/F mice (Figure 6A). The number of injury; and (5) some of these findings are recapitulated in pa-
apoptotic, terminal deoxynucleotidyl transferase–mediated tients with obesity-related nephropathy. We are convinced that
digoxigenin-deoxyuridine nick-end labeling (TUNEL)-positive our findings are novel and clinically important to elucidate a
tubular cells markedly increased in obese Atg5F/F mice after I/R molecular mechanism of lipid overload-mediated kidney injury.
injury (Figure 6B). These injuries were significantly exacerbated Accumulating evidence has suggested that several saturated
by autophagy deficiency. Renal function data, as assessed by plasma and unsaturated FAs appear to either activate or inactivate
urea nitrogen and creatinine, verified the morphologic results, autophagy.15–19 The significant variation in the literature
although not statistically significant presumably because mice could be attributed to differences in the cells used in the ex-
were subjected to unilateral (not bilateral) I/R injury (Figure 6C). periments, observation periods, concentration of FFAs, and
most importantly, the methods used to monitor autophagic
Phospholipid Accumulation in the Enlarged Lysosomes activity. In this study, we explored the effect of lipid overload
and Impaired Autophagic Flux in the Kidneys of Obese on autophagic activity in vitro and in vivo by using methods
Patients developed in our previous report25 and demonstrated com-
Kidney biopsy samples taken from obese and nonobese pa- plex alterations in autophagic activity, with several important
tients with various CKDs (n=5, respectively) were analyzed findings. A recent report has revealed that mTOR, a potent sup-
(Figure 7, A and B). Clinical characteristics of these patients pressor of autophagy,33 was activated in the proximal tubules of
are shown in Supplemental Table 1. All patients showed overt obese mice.34 It is also widely known that high levels of FFA lead
proteinuria. PTCs of all obese patients exhibited cytosolic vac- to constant activation of mTOR signaling, a process related to the
uolar formation (Figure 7A). The margins of the vacuoles were development of diseases such as diabetes and obesity.35 Indeed,
positive for LAMP1. SQSTM1/p62 accumulation was ob- in our study, mTOR complex 1 activation, as determined by
served in the PTCs from all these obese patients. In contrast, positive staining for phosphorylated ribosomal protein S6
the LAMP1-positive vacuoles or SQSTM1/p62-positive aggre- (Ser235/236), was observed in part of the PTCs and distal neph-
gates were rarely observed in the nonobese patients. The vac- ron segments of obese GFP-MAP1LC3 transgenic mice (Sup-
uoles observed in the PTCs of patient 2 were positively stained plemental Figure 7). This obesity-mediated mTOR complex
with toluidine blue (Figure 7B). Electron microscopy from 1 activation can certainly suppress canonical (nonselective)
patient 4 (Figure 7, C–M) revealed that the vacuoles contain autophagy induction; however, it should be noted that auto-
multilamellar, onion skin-like structures (Figure 7, F–K). A phagy, especially noncanonical (selective) autophagy, can be in-
massive vacuolization containing undigested proteins and or- duced by a variety of stresses such as ROS, endoplasmic reticulum
ganelles (Figure 7L), and aberrant and degenerated mitochon- stress, and hypoxia independently of mTOR activation.36 In
drial morphology (Figure 7M) were also observed in the PTCs. agreement with this notion, recent reports have demonstrated
These data suggest that phospholipid accumulation in the en- that PA induces autophagy independent of mTOR activity.37,38
larged lysosomes and impaired autophagic flux were recapit- Then, what triggers lipid overload-induced autophagy in-
ulated in the kidneys of obese patients. dependent of mTOR activation? What is degraded by lipid
overload-induced autophagy? We found that mitochondria
are a putative substrate. Indeed, other groups have previously
DISCUSSION reported the close association of mitochondria and lipid excess.
High levels of FFAs evoke mitochondrial DNA (mtDNA) dam-
In this study, we found a close association between autophagy age and impair mitochondrial dynamics and morphology in
and lipotoxicity in the kidney proximal tubule, as (1) lipid INS1 cells.39,40 FFAs modulate mitochondrial ROS production

dots was counted in at least ten high-power fields (original magnification, 3400). (C and D) GFP-positive puncta formation was assessed in
the proximal tubules of nonobese or obese GFP-MAP1LC3 transgenic mice that were either fed or subjected to 24 hours of starvation, with
or without chloroquine administration 6 hours before euthanasia (n=6–9 in each group). (C) Kidney sections were immunostained for LRP2/
MEGALIN, a marker of proximal tubules (red), and counterstained with DAPI (blue). (D) The number of GFP-positive puncta per proximal
tubule under each condition was counted in at least ten high-power fields (original magnification, 3600) (each high-power field contained
10–15 proximal tubules). Bars: 50 mm (A) and 10 mm (B and C). Data are provided as mean6SEM. Statistically significant differences
(*P,0.05) are indicated. All images are representative of multiple experiments. F/F, Atg5F/F mice; F/F:NDRG1, Atg5F/F;NDRG1 mice.

8 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org BASIC RESEARCH

Figure 4. PA-induced mitophagy protects PTCs from mitochondrial injury. (A and B) The mitochondrial membrane potential (A) and the
mitochondrial ROS (B) of autophagy-competent and -deficient PTCs treated with either 0.25% BSA or 0.25 mM PA for 12 hours were
assessed by TMRE and MitoSOX Red stainings, respectively (n=4–6). Quantitative data of relative intensity were also provided. (C) The
mtDNA copy number of autophagy-competent and -deficient kidney PTCs was quantified after treatment with either 0.25% BSA or
0.25 mM PA for 12 hours. (A–C) Values are normalized by the signal intensity of BSA-treated autophagy-competent PTCs. (D and E)
PTCs stably expressing GFP-MAP1LC3 (D) or GFP-MAP1LC3 and mStrawberry-ubiquitin (E) were stained with MitoTracker Red FM (D)

J Am Soc Nephrol 28: ccc–ccc, 2016 Autophagy and Lipotoxicity 9


BASIC RESEARCH www.jasn.org

by several mechanisms, such as interaction with components of previously observed that, in the aged kidney, basal autophagic
the respiratory chain, thereby inhibiting the electron transport.41 activity is enhanced to cope with increasing “garbage” with age,
Our observation that PA significantly increased mitochon- but upregulation of autophagic flux in response to additional
drial ROS and decreased mitochondrial membrane potential stress is blunted partly through lysosomal dysfunction (e.g.,
in autophagy-deficient PTCs, both of which could trigger lipofustin).25 In other words, aging and lipid overload have a
mitophagy,42 suggests that autophagy could be triggered to common mechanistic platform, lysosomal dysfunction, and
target dysfunctional mitochondria during lipid overload. In- consequent impaired autophagic flux. The interesting obser-
deed, PA-induced mitophagy has been recently reported in vation that may strengthen the close mechanistic relationship
human aortic endothelial cells.43 In keeping with suppressed mi- between aging and lipid overload are that caloric restriction
tochondrial bioenergetics, we found a time-dependent decrease potentially promotes longevity in many mammalian mod-
in cellular ATP levels in PTCs treated with PA, which may explain els,47 whereas high-fat and/or -carbohydrate diets accelerates
the impairment of lysosomal acidification, as H+ pump activity is aging.48 Furthermore, ischemic injury in the elderly and pa-
highly sensitive to decreases in ATP concentration.44 Collectively, tients with hyperlipidemia is a major risk factor for ESRD.49,50
our findings demonstrate that, during lipid overload, PINK1- In this point, therapeutic effort to restore lysosomal function
PARK2/Parkin–mediated mitophagy continues to protect and autophagic flux will pave the way to improving the clinical
proximal tubules from increasing mitochondrial stress; however, outcome of kidney injury related to aging or HFD.
persistent mitophagy inevitably places a burden on the lyso- The NLRP3 inflammasome senses obesity-associated dan-
somal system (as manifested by impaired acidification and phos- ger signals, such as intracellular ceramide-induced caspase-1
pholipids accumulation), leading to impaired autophagic flux. cleavage, in macrophages and adipose tissue and contributes to
It has been previously reported that HFD-mice have prox- obesity-induced inflammation and insulin resistance.51 In an-
imal tubule injury with enlarged LAMP1-positive lysosomes other report, PA stimulation results in AMP-activated protein
containing phospholipids as multilaminar inclusions.26 In kinase inhibition, leading to defective autophagy in myeloid
addition, a significant increase in the levels of specific glomer- cells, which activate inflammasomes.52 In addition, inflamma-
ular and tubular lipid species, including phospholipids, has some activation in hematopoietic cells impairs insulin signal-
been recently reported in diabetes.45 Our study confirmed ing in several target tissues to reduce glucose tolerance and
these findings, not only in HFD-loaded mouse kidney, but insulin sensitivity.52 In this study, we extend these previous
also in the clinical setting (obesity-related nephropathy). Al- observations to the kidney and have demonstrated that HFD-
though lipotoxicity in organs that have evolved specialized induced impaired autophagic flux evokes macrophage infiltra-
lipid handling functions has been generally attributed to FFAs tion and inflammasome activation, followed by renal fibrosis. It
and/or triglyceride accumulation, in our study, phospholipid remains to be elucidated whether lipid overload-induced inflam-
accumulation has emerged as being central to lipotoxicity in masome activation could lead to reduced glucose tolerance and
the kidney. The origin of phospholipids appears to be the insulin sensitivity.
mitochondrial membrane. Phospholipids play an important In conclusion, we report that HFD-induced elevation in
role in cellular homeostasis by engagement in various cellular autophagy plays a crucial role in counteracting lipotoxicity
signaling pathways.46 However, the mechanism underlying the in the kidney through mitochondrial quality control.
accumulation of undigested phospholipids in the lysosome Furthermore, a reduced capacity for upregulation of autopha-
during lipid overload and consequent alterations in cellular gic flux is associated with HFD-induced vulnerability to I/R
function remain to be elucidated. injury. Strategies aimed at modulating autophagy hold promise
We have previously demonstrated that autophagy deficiency for treating obesity-related kidney disease.
exacerbates kidney injury in many mouse models. In this study,
we demonstrated that impairment in autophagic activity is
observable in patients as well as genetically engineered mice. CONCISE METHODS
Although largely overlooked and not fully understood relative
to the process of phagophore formation, the mechanism Mice
through which autophagosomes fuse with lysosomes is a crit- GFP-MAP1LC3 transgenic mice and Atg5F/F;NDRG1 and Atg5F/F;KAP
ical aspect of macroautophagy. Similar to this study, we have mice on a C57BL/6 background have been described previously.20,25,27

or MitoTracker Deep Red FM (E) after treatment with either 0.25% BSA or 0.25 mM PA for 6 hours (n=5). (D) Yellow dots in merged images
represent colocalization of mitochondria and autophagosomes. Colocalization was assessed by Pearson correlation. (E) White dots in
merged images represent colocalization of ubiquitinated mitochondria and autophagosomes. (F) Western blot analysis (PINK1, PARK2/Parkin,
and COX4) (n=4). Actin, b (ACTB) was used as loading control. The mean value of untreated controls at the baseline was adjusted to “1” as a
reference. Bars: 10 mm. Data are provided as mean6SEM. Statistically significant differences: *P,0.05 versus autophagy-competent cells of
corresponding treatment; #P,0.05 versus BSA-treated control cells (A–C); *P,0.05 (D and E). Atg5(+), autophagy-competent PTC; Atg5(2),
autophagy-deficient PTC. All images are representative of multiple experiments.

10 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org BASIC RESEARCH

Figure 5. Autophagy deficiency aggravates HFD-induced mitochondrial dysfunction and inflammation, leading to renal fibrosis. (A) PAS-stained
kidney cortexes of nonobese and obese Atg5F/F;KAP mice and Atg5F/F control littermates (n=10–14 in each group). Magnified images are shown
in the insets (original magnification, 31000). (B–I) Electron micrographs of obese Atg5F/F (B–E) and Atg5F/F;KAP mice (F–I) (n=3 in each group).
Arrow indicates the lysosome and asterisk indicates the nucleus. (J–N) Immunostaining for F4/80, a macrophage marker (J), mRNA expression
levels of inflammatory cytokine- (K) and inflammasome- (L) related genes, Western blot analysis of NLRP3, caspase-1, IL-1b, and IL-18 (M), and
immunostaining for F4/80 (green) and IL-1b (red) (N) in the kidney outer medulla of Atg5F/F;KAP and Atg5F/F control mice fed ND or HFD for
2 months. n=7–9 (J) and 6–7 (K–N) in each group. Bars: 50 mm (A and J), 10 mm (B, F and N), 5 mm (F and J), and 500 nm (C–E and G–I). Data are
expressed as the fold change relative to the mean value of nonobese Atg5F/F mice. Values represent the mean6SEM. Statistically significant
differences: *P,0.05 versus treatment-matched Atg5F/F control littermates; #P,0.05 versus nonobese mice. (N) Sections were counterstained
with DAPI (blue). All images are representative of multiple experiments. F/F, Atg5F/F mice; F/F;KAP, Atg5F/F;KAP mice; M, mitochondria.

We used two different mice models, tamoxifen-inducible (Atg5F/F; mice were used for most of the experiments for evaluating the effects
NDRG1) and conditional (Atg5F/F;KAP) PTC-specific autophagy- of autophagy deficiency during lipid overload. Atg5F/F;KAP mice
deficient mice; Atg5F/F;NDRG1 mice were used only for the experi- were generated by crossing KAP-Cre transgenic mice, in which Cre
ment assessing HFD-mediated changes in demand and activity of recombinase is expressed under control of the KAP gene promoter,
basal autophagy because using corn oil to dissolve tamoxifen could and mice bearing an Atg5 flox allele (Atg5F/F mice). KAP-Cre/CAG-
modify HFD-mediated changes such as metabolic profiles, and Atg5F/F;KAP CAT-Z male mice, in which b-galactosidase expression reflects Cre

J Am Soc Nephrol 28: ccc–ccc, 2016 Autophagy and Lipotoxicity 11


BASIC RESEARCH www.jasn.org

Figure 6. HFD enhances the vulnerability against I/R injury to autophagy-deficient mice. (A and B) Representative images of PAS-staining (A)
and TUNEL staining (B) on kidney cortexes of nonobese and obese Atg5F/F;KAP mice and Atg5F/F control littermates 2 days after unilateral
I/R injury or sham operation. (A) The tubular injury score was shown. (B) The number of TUNEL-positive PTCs was calculated in at least ten
high-power fields (original magnification, ☓400). Bars: 500 mm. (C) Plasma urea nitrogen and creatinine were measured (n=6–8 in each
group). Values represent the mean6SEM. Statistically significant differences: *P,0.05 versus treatment-matched Atg5F/F control littermates;
#
P,0.05 versus nonobese mice; ‡P,0.05 versus sham-operated mice. F/F, Atg5F/F mice; F/F;KAP, Atg5F/F;KAP mice.

activity, exhibited LacZ-positive tubules in the cortex and the outer (Sigma-Aldrich; 50 mg/g body wt) was injected intraperitoneally and
medulla, whereas LacZ-positive tubules were absent in female mice were euthanized 6 hours after the injection.
mice.20 Therefore, only male mice were used in this study. Eight-
week-old mice were housed in box cages, maintained on a 12-hour Antibodies
light/dark cycle, and fed an ND (12.8% of kilocalories from fat: 5% We used the following antibodies: antibodies for LRP2/MEGALIN
fat, 23% protein, and 55% carbohydrate) or HFD (62.2% of kilocalories (a gift from T. Michigami, Department of Bone and Mineral Research,
from fat: 35% fat, 23% protein, and 25% carbohydrate) (Oriental Yeast, Osaka Medical Center and Research Institute for Maternal and Child
Tokyo, Japan) for 2 or 10 months. For the induction of Atg5 deletion in Health, Osaka, Japan), COL1A1 (Abcam, Inc., Cambridge, MA),
Atg5F/F;NDRG mice, we intraperitoneally injected tamoxifen (Sigma- ATG5 (MBL, Nagoya, Japan), SQSTM1 (MBL), ubiquitin (Cell Sig-
Aldrich, St. Louis, MO) (1 mg/10 g body wt) dissolved in corn oil naling Technology, Danvers, MA), MAP1LC3 (Cell Signaling Tech-
(Sigma-Aldrich) at a concentration of 10 mg/ml three times every other nology), IL-1b (Cell Signaling Technology), IL-18 (Abcam, Inc.),
day. In the experiment assessing the autophagic flux in vivo, chloroquine NLRP3 (Adipogen, Seoul, Korea), caspase-1 (Santa Cruz Biotechnology,

12 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org BASIC RESEARCH

Figure 7. Phospholipid accumulation and impaired autophagic flux in the kidneys of obese patients. (A) Representative images of kidney
specimens obtained from obese and nonobese patients with overt proteinuria (n=5, respectively). Kidney sections were immunostained for LAMP1
and SQSTM1/p62, and counterstained with hematoxylin. Magnified images are shown in the insets (original magnification, 31000). (B)
Representative images of toluidine blue staining from obese patient 2 (original magnification, 31000). (C–M) Representative images of
electron micrographs from obese patient 4. A lysosome with normal morphology is shown (D and E). Magnified images of lysosomes are
presented (original magnification, 33000). Bars: 50 mm (A and B), 5 mm (C, F, and I), and 500 nm (D, E, G, H, J–M). Arrows indicate the
lysosome and asterisks indicate the nucleus. BM, basement membrane; M, mitochondria; TL, tubular lumen.

J Am Soc Nephrol 28: ccc–ccc, 2016 Autophagy and Lipotoxicity 13


BASIC RESEARCH www.jasn.org

Santa Cruz, CA), PINK1 (Abcam, Inc.), PARK2/Parkin (Cell Signaling isopropylene for 1 minute, and washed with PBS three times. Tissue
Technology), COX4 (MBL), phosphorylated ribosomal protein sections were counterstained with hematoxylin.
S6 (Ser235/236; Cell Signaling Technology), ACTB (Sigma-Aldrich),
F4/80 (AbD Serotec, Oxford, UK), LAMP1 (BD Biosciences, San Jose, COX/SDH Staining
CA), 4HNE (Japan Institute for the Control of Aging, Fukuroi, Japan), COX/SDH staining were performed as previously described.23 Briefly,
dityrosine (Japan Institute for the Control of Aging), CML (Transgenic, we used fresh cryosections (5 mm thick) of kidney tissues. For COX
Kobe, Japan), biotinylated secondary antibodies (Vector Laboratories, staining, the sections were incubated in the solution composed of
Burlingame, CA), horseradish peroxidase-conjugated secondary anti- 2 mg/ml of catalase (Sigma-Aldrich), 1 mg/ml of cytochrome c
bodies (DAKO, Glostrup, Denmark), and Alexa Fluor 555- and Alexa (Sigma-Aldrich), and 0.5 mg/ml of diaminobenzidine (Wako) in
Fluor 488-conjugated secondary antibodies (Invitrogen, Carlsbad, CA). 0.1 M of phosphate buffer (pH 7.4) for 1 hour at 37°C. The SDH
staining was performed according to the modified method of Martin
Histologic Analysis et al.53 In brief, we prepared nitro blue tetrazolium stock composed of
Histologic analysis was performed as previously described, with mod- KCN 6.5 mg (Nacalai, Kyoto, Japan), EDTA 185 mg (Sigma-Aldrich),
ification.25 To examine the number of GFP-positive dots, postfixed and nitro blue tetrazolium 100 mg (Nacalai) in 100 ml of 0.1 M
frozen kidney was sectioned and immunostained for LRP2/MEGALIN phosphate buffer (pH 7.6) and 500 mM of sodium succinate solution
(PTC maker). The number of GFP-MAP1LC3 dots per proximal tubule (Nacalai) diluted in distilled water. Then the sections were incubated
were counted in at least ten high-power fields (3600) (each high-power in a solution mixed with 2 ml of nitro blue tetrazolium stock solution,
field contained 10–15 proximal tubules). The fluorescence images 0.2 ml of succinate solution, and 0.7 mg of phenazine methosulfate
were collected using confocal microscopy (FV1000-D; Olympus, Tokyo, (Nacalai) for 15 minutes at 37°C. The intensity of the positive staining
Japan). Immunohistochemical staining for SQSTM1/p62, ubiquitin, area was measured using the imaging software, ImageJ. For each
COL1A1/collagen type I, HNE, dityrosine, CML, and IL-1b was kidney, at least ten high-power fields (3400) were analyzed.
performed on paraffin-embedded sections after antigen retrieval
via autoclaving in 0.01 mM citrate buffer (pH 6.0) for 10 minutes Cell Culture
at 120°C and blocking with 1.5% BSA (Sigma-Aldrich) in PBS for Autophagy-deficient and autophagy-competent PTC lines were as
60 minutes. Immunohistochemical staining for F4/80 (or double stain- previously described.20 In brief, Atg5-deficient kidney PTCs (Atg5-
ing for F4/80 and IL-1b) was performed on paraffin-embedded sections negative PTCs) were isolated from 3-week-old Atg5F/F;KAP mice
after enzyme pretreatment using proteinase K solution (20 mg/ml) in TE and immortalized by using pEF321-T, an SV-40 large T antigen ex-
buffer (10 mM Tris-HCl [pH 8.0], 1 mM EDTA) for 3 minutes at room pression vector. As a control, we generated Atg5-positive PTCs by
temperature. For double staining for SQSTM1 (or ubiquitin) and LRP2/ stably transfecting pMRX-IRES-Atg5-bsr (Atg5-expressing retroviral
MEGALIN, each molecule was first visualized using a horseradish vector cassette) to Atg5-negative PTCs. We also isolated PTCs from
peroxidase-DAB compound (Nichirei, Chuo-ku, Tokyo, Japan), after wild-type mice and immortalized cell lines were established by trans-
detection of LRP2/MEGALIN using alkaline phosphatase and nitro blue formation using pEF321-T, an SV-40 large T antigen expression vector.
tetrazolium and 5-bromo-4-chloro-3-indolyl phosphate Stock Solution We transfected the pEGFP-MAP1LC3 vector (a gift from T. Yoshimori,
(Roche Diagnostics, Basel, Switzerland). The percentage of the Department of Genetics, Osaka University) to the wild-type PTC lines
immune-positive area for COL1A1/collagen type I (or F4/80) was and then constructed GFP-MAP1LC3 transgenic PTC lines. Cells were
calculated using a digital image-analyzing software, ImageJ (available at cultured in DMEM (Sigma-Aldrich) containing 5% FBS at 37°C under
http://rsbweb.nih.gov/ij/index.html; National Institutes of Health). a humidified atmosphere of 5% CO2 and 95% air. Lipid-containing
For each kidney, at least ten high-power fields (3400) were analyzed. media (PA-BSA) were prepared by conjugation of PA with essentially
For electron microscopy, kidney specimens were fixed with 2.5% FA-free, low endotoxin BSA (Sigma-Aldrich). Sodium palmitate
glutaraldehyde (Wako, Osaka, Japan) and observed using a Hitachi (Sigma-Aldrich) were dissolved in 50% ethanol and vigorously mixed
H-7650 transmission electron microscope (Hitachi, Tokyo, Japan). with BSA in PBS at a molar ratio of 6.6:1,54 filter-sterilized, and added to
Characterization of tubular vacuole accumulation was performed culture media at the final FA (PA) concentration of 0.25 mM. In
using the toluidine blue staining on 4% paraformaldehyde (PFA)-fixed this experimental condition, the BSA concentration was 2.5 mg/ml, or
paraffin-embedded sections and using the Nile Red staining (Wako) on 37.88 mM. This concentration of FFA was used because the molar ratio
4% PFA-fixed frozen sections. In all quantitative or semiquantitative of FFA to albumin could reach up to 8.59,55 and the albumin concen-
analyses of histologic staining, at least ten high-power fields were re- tration in the proximal tubular lumen could be as high as 2.9 mg/ml, or
viewed for each tissue by two nephrologists (T.Y. and A.T.) in a blinded 43 mM.56 Twenty-four hours after the seeding, the cells were treated
manner. with PA-BSA or 2.5 mg/ml BSA for 6–24 hours before harvest or anal-
ysis. According to previous studies, we used 2.5 mg/ml BSA treatment
Oil Red O Staining as a control for 0.25 mM PA treatment because it has been reported
Sections (5 mm thick) of frozen, 4% PFA-embedded tissue were dried that, at a higher concentration, FA-free BSA has several cytotoxic effects,
for 1 hour at room temperature and rinsed in PBS, and placed in 60% including ROS production, mitochondrial damage, and endoplasmic
isopropylene for 1 minute. They were stained in 60% Oil Red O so- reticulum stress,57,58 all of which could induce autophagy and apoptosis
lution (100% solution: 0.3 g of Oil Red O [Sigma-Aldrich] dissolved in the PTCs.59 To assess autophagic activity, PTCs were treated with
in 100 ml of isopropylene) for 20 minutes at 37°C, placed in 60% 200 nM of bafilomycin A1 (Wako) for 1 hour at 37°C before harvest.

14 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org BASIC RESEARCH

Assessment of Lysosomal Acidification Colocalization of autophagosomes and mitochondria was calculated


To assess autolysosome/lysosomal acidification, PTCs stably expressing using ImageJ and represented as Pearson correlation between fluo-
GFP-MAP1LC3 were stained with 50 nM LysoTracker Red DND-99 rescence of GFP and MitoTracker Red FM. We also constructed GFP-
(Invitrogen) for 30 minutes at 37°C. Colocalization of autophago- MAP1LC3 and mStrawberry-ubiquitin double-transfected PTCs to
somes and lysosome was calculated using ImageJ. The lysosomal assess the association between ubiquitin and mitophagy. GFP-
acidification was determined by staining with 1 mM of LysoSensor MAP1LC3 and mStrawberry-ubiquitin double-transfected PTCs
Yellow/Blue DND-160 (PDMPO) (Invitrogen) for 1 minute at 37°C. were stained with 25 nM of MitoTracker Deep Red FM (Invitrogen)
The fluorescence images were collected at the wavelength range from for 15 minutes at 37°C.
510 to 641 nm (yellow) and at the wavelength range from 404 to
456 nm (blue) with the Olympus FV1000-D (Olympus). The mean Relative Quantification of mtDNA Copy Numbers
fluorescence intensity in the fluorescence-positive area was mea- Relative quantification of mtDNA copy numbers were performed as
sured using ImageJ, for yellow and blue fluorescence, respectively. previously described.25 In brief, total DNA (mtDNA and nuclear
The blue/yellow ratio was calculated by division process in each DNA) was extracted from isolated proximal tubules with Wizard
section. The average blue/yellow ratio of a given sample was calcu- Genomic DNA Purification Kit according to the manufacturer’s in-
lated from five sections. Intensity was measured using at least three struction (Promega, Madison, WI). Real-time quantitative PCR of
independent experiments (.150 cells per experiment). the mtDNA-encoded gene mt-Co1 (GenBank accession no.
NC_001807) and the single-copy nuclear gene Ndufv1 (NADH de-
Fluorescence FA Pulse-Chase Assay hydrogenase [ubiquinone] flavoprotein 1) (GenBank accession no.
Fluorescence FA pulse-chase assay was performed as previously de- NM_133666) was carried out with an ABI PRISM 7900HT sequence
scribed, with slight modification. 60 PTCs were incubated with detection system (Applied Biosystems, Foster City, CA). Relative
DMEM with 5% FBS containing 2 mM b-BODIPY FL C12-HPC quantification of mtDNA copy numbers were depicted as the ratio
(FL HPC) (Invitrogen) for 16 hours. Cells were then washed three of the relative concentrations of mtDNA/nDNA. The sequences of
times with DMEM with 5% FBS, incubated for 1 hour to allow the primers for the MT-CO1 and Ndufv1 gene are as follows: mt-Co1-F,
fluorescent lipids to incorporate into cellular membranes, and then 59-tgctagccgcaggcattac-39; mt-Co1-R, 59-gggtgcccaaagaatcagaac-39;
chased for the indicated times under PA-BSA treatment or 0.25% Ndufv1-F, 59-cttccccactggcctcaag-39; Ndufv1-R, 59-ccaaaacccagtgatccagc-39).
BSA treatment. FL-HPC–loaded and chased PTCs were stained
with 50 nM LysoTracker Red DND-99 for 30 minutes at 37°C Cell Proliferation Assay
or stained with 25 nM of MitoTracker Red FM (Invitrogen) for MTS assays were performed using CellTiter 96 AQueous One Solution
15 minutes at 37°C immediately before imaging, or fixed with 4% Cell Proliferation Assay (Promega), according to the manufacturer’s
PFA for 15 minutes at room temperature for immunocytochemistry. instructions, in a 96-well plate. Each experiment was performed in
The number and size of phospholipid/lysosome-merged dots were triplicate and repeated at least three times. Cell viability in percentage
measured using ImageJ “analyze particles” function in threshold refers to the MTS value of the induced cells compared with the
images. value of the untreated control cells (cultured in DMEM containing
5% FBS).
Measurement of Mitochondrial Membrane Potential,
ROS Production, and ATP Quantitative RT-PCR and Western Blot Analysis
Cells cultured on 35 mm glass-bottomed dishes were subjected to the Quantitative RT-PCR and Western blot analyses were performed as
confocal analysis. The mitochondrial membrane potential was deter- previously described.61 The sequences of the primers used were as
mined by staining with 50 nM of TMRE (Invitrogen) for 30 minutes at follows: Cpt1-F, 59-accactggccgaatgtcaag-39; Cpt1-R, 59-agcgagtagcg-
37°C. To determine mitochondrial ROS production, cells were incu- catggtcat-39; Aox-F, 59-tcaacagcccaactgtgacttccatta-39; Aox-R, 59-
bated with 0.3 mM MitoSOX Red (Invitrogen) for 10 minutes. The tcaggtagccattatccatctcttca-39; Scad-F, 59-ttacctggcctactccatcg-39;
fluorescence images for TMRE or MitoSOX Red were collected using Scad-R, 59-tgatccactgttgcttctgc-39; Mcad-F, 59-taatcggtgaaggag-
the Olympus FV1000-D (Olympus), and the mean fluorescence in- caggttt-39; Mcad-R, 59-ggcatacttcgtggcttcgt-39; Lcad-F, 59-gcatcaa-
tensity in the fluorescence-positive area was measured using ImageJ. catcgcagagaaa-39; Lcad-R, 59-acgcttgctcttcccaagta-39; Cox5b-F,
The average mean fluorescence intensity of a given sample was 59-aggcagcttcaggcaccaag-39; Cox5b-R, 59-ggtggggcaccagcttgtaa-39;
calculated from five sections. Intensity was measured using at Tnfa-F, 59-atggcctccctctcatcagt-39; Tnfa-F -R, 59-cttggtggtttgctac-
least three independent experiments (.150 cells per experiment). gacg-39; Ccl2-F, 59-cccaatgagtaggctggaga-39; Ccl2-R, 59-tctggacc-
ATP was measured using ATP Colorimetric/Fluorometric Assay cattccttcttg-39; Ccl3-F, 59-tgcccttgctgttcttctct-39; Ccl3-R,
Kit (BioVision, Inc., Milpitas, CA) according to manufacturer’s 59-gatgaattggcgtggaatct-39; Nlrp3-F, 59-atgctgcttcgacatctcct-
instruction. 39; Nlrp3-R, 59-aaccaatgcgagatcctgac-39; Asc-F, 59-acagaagtg-
gacggagtgct-39; Asc-R, 59-ctccaggtccatcaccaagt-39; caspase-1-F,
Assessment of Mitophagy 59-cacagctctggagatggtga-39; caspase-1-R, 59-tctttcaagcttgggcactt-39;
Assessment of mitophagy were performed as previously described.23 Sqstm1-F, 59-ccccagagtcgaagtagctg-39; Sqstm1-R, 59-agtgagaa-
To prove mitophagy, PTCs stably expressing GFP-MAP1LC3 were gaggctggtgga-39; Gapdh-F, 59-aactttggcattgtggaagg-39; Gapdh-R, 59-
stained with 25 nM of MitoTracker Red FM for 15 minutes at 37°C. acacattgggggtaggaaca-39.

J Am Soc Nephrol 28: ccc–ccc, 2016 Autophagy and Lipotoxicity 15


BASIC RESEARCH www.jasn.org

Induction of Kidney I/R Injury [to Y.T.], and 24659416 [to Y.I.]), Takeda Medical Research Foun-
Kidney ischemia was induced as previously described.20 In brief, the dation (to Y.T.), and Merck Sharp & Dohme (to Y.I.).
animals were anesthetized and were kept on a homeothermic table.
For unilateral clamping, back incisions were made to expose the kid-
ney pedicles to induce 35 minutes of kidney ischemia. The clamps DISCLOSURES
were released for reperfusion. Control animals were subjected to None.
sham operation without renal pedicle clamping. Kidney tubular
cells of the cortex were examined by two nephrologists (T.Y. and
A.T.) in a blind manner, and were scored from 0 to 10 according REFERENCES
to the percentage of damaged tubules. Tubular damage was defined
as cell necrosis, which was evaluated in this study by loss of brush 1. Withrow D, Alter DA: The economic burden of obesity worldwide: A
border, tubular dilation, and cast formation. At least ten high-power systematic review of the direct costs of obesity. Obes Rev 12: 131–141,
fields (3400) were reviewed for each periodic acid–Schiff-stained 2011
2. Unger RH, Clark GO, Scherer PE, Orci L: Lipid homeostasis, lipotoxicity
slide. Plasma urea nitrogen and creatinine were measured using and the metabolic syndrome. Biochim Biophys Acta 1801: 209–214,
the BUN-Test-Wako (Wako), the CRE-EN Kainos (Kainos, Tokyo, 2010
Japan), respectively. 3. Muoio DM: Intramuscular triacylglycerol and insulin resistance: Guilty
as charged or wrongly accused? Biochim Biophys Acta 1801: 281–288,
2010
TUNEL Staining 4. Moorhead JF, Chan MK, El-Nahas M, Varghese Z: Lipid nephrotoxicity
Apoptotic cells were detected using the TUNEL assay with an in situ in chronic progressive glomerular and tubulo-interstitial disease. Lancet 2:
apoptosis detection kit (Takara, Kyoto, Japan). Tissue sections were 1309–1311, 1982
counterstained with Methyl Green Solution (Wako) and examined 5. Gullans SR, Brazy PC, Mandel LJ, Dennis VW: Stimulation of phosphate
under a microscope. transport in the proximal tubule by metabolic substrates. Am J Physiol
247: F582–F587, 1984
6. Balaban RS, Mandel LJ: Metabolic substrate utilization by rabbit
Kidney Biopsy Specimens proximal tubule. An NADH fluorescence study. Am J Physiol 254:
Human kidney biopsy specimens were obtained from patients with F407–F416, 1988
overt proteinuria. All patients provided written informed consent. 7. Uchida S, Endou H: Substrate specificity to maintain cellular ATP along
the mouse nephron. Am J Physiol 255: F977–F983, 1988
8. Stremmel W, Pohl L, Ring A, Herrmann T: A new concept of cellular
Statistical Analyses uptake and intracellular trafficking of long-chain fatty acids. Lipids 36:
All results are presented as means6SEM. Statistical analyses were 981–989, 2001
conducted using JMP software (SAS Institute Inc., Cary, NC). Multiple- 9. Levine B, Kroemer G: Autophagy in the pathogenesis of disease. Cell
group comparisons were performed using analysis of variance with 132: 27–42, 2008
post-testing using the Tukey–Kramer test. The difference between 10. Choi AMK, Ryter SW, Levine B: Autophagy in human health and dis-
two experimental values was assessed by paired t test when appro- ease. N Engl J Med 368: 651–662, 2013
11. Mizushima N, Komatsu M: Autophagy: Renovation of cells and tissues.
priate. Statistical significance was defined as P,0.05. Cell 147: 728–741, 2011
12. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, Tanaka K,
Study Approval Cuervo AM, Czaja MJ: Autophagy regulates lipid metabolism. Nature
All animal experiments were approved by the institutional committee 458: 1131–1135, 2009
of the Animal Research Committee of Osaka University and the 13. Singh R, Cuervo AM: Lipophagy: Connecting autophagy and lipid
metabolism. Int J Cell Biol 2012: 282041, 2012
Japanese Animal Protection and Management Law (No.25). In the 14. Settembre C, Ballabio A: Lysosome: Regulator of lipid degradation
case of human studies, written informed consent was received from pathways. Trends Cell Biol 24: 743–750, 2014
participants before inclusion in the study. 15. Choi S-E, Lee S-M, Lee Y-J, Li L-J, Lee S-J, Lee J-H, Kim Y, Jun H-S, Lee
K-W, Kang Y: Protective role of autophagy in palmitate-induced INS-1
beta-cell death. Endocrinology 150: 126–134, 2009
16. Komiya K, Uchida T, Ueno T, Koike M, Abe H, Hirose T, Kawamori R,
ACKNOWLEDGMENTS Uchiyama Y, Kominami E, Fujitani Y, Watada H: Free fatty acids stim-
ulate autophagy in pancreatic b-cells via JNK pathway. Biochem Bio-
T.Y., A.T., Y.T., and Y.I. were involved in study design and writing phys Res Commun 401: 561–567, 2010
17. Shen S, Niso-Santano M, Adjemian S, Takehara T, Malik SA, Minoux H,
manuscript. The other authors and T.Y. performed experiments and
Souquere S, Mariño G, Lachkar S, Senovilla L, Galluzzi L, Kepp O,
analyzed data. All authors contributed to the discussions. We thank Pierron G, Maiuri MC, Hikita H, Kroemer R, Kroemer G: Cytoplasmic
N. Mizushima, University of Tokyo, for Atg5F/F and GFP-MAP1LC3 STAT3 represses autophagy by inhibiting PKR activity. Mol Cell 48:
mice; T. Michigami, Osaka Medical Center and Research Institute, 667–680, 2012
for LRP2/MEGALIN antibody; and N. Horimoto and for technical 18. Koga H, Kaushik S, Cuervo AM: Altered lipid content inhibits auto-
phagic vesicular fusion. FASEB J 24: 3052–3065, 2010
assistance.
19. Las G, Serada SB, Wikstrom JD, Twig G, Shirihai OS: Fatty acids suppress
This work was supported by a Grant-in-Aid for Scientific Re- autophagic turnover in b-cells. J Biol Chem 286: 42534–42544, 2011
search from the Ministry of Education, Culture, Sports, Science and 20. Kimura T, Takabatake Y, Takahashi A, Kaimori JY, Matsui I, Namba T,
Technology in Japan (15H06371 [to T.Y.], 24591196 and 15K09260 Kitamura H, Niimura F, Matsusaka T, Soga T, Rakugi H, Isaka Y:

16 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org BASIC RESEARCH

Autophagy protects the proximal tubule from degeneration and acute 37. Jia S-N, Lin C, Chen D-F, Li A-Q, Dai L, Zhang L, Zhao L-L, Yang J-S,
ischemic injury. J Am Soc Nephrol 22: 902–913, 2011 Yang F, Yang W-J: The transcription factor p8 regulates autophagy in
21. Takahashi A, Kimura T, Takabatake Y, Namba T, Kaimori J, Kitamura H, response to palmitic acid stress via a mammalian target of rapamycin
Matsui I, Niimura F, Matsusaka T, Fujita N, Yoshimori T, Isaka Y, Rakugi (mTOR)-independent signaling pathway. J Biol Chem 291: 4462–4472,
H: Autophagy guards against cisplatin-induced acute kidney injury. Am 2016
J Pathol 180: 517–525, 2012 38. Tan SH, Shui G, Zhou J, Li JJ, Bay B-H, Wenk MR, Shen H-M: Induction
22. Kimura T, Takahashi A, Takabatake Y, Namba T, Yamamoto T, Kaimori of autophagy by palmitic acid via protein kinase C-mediated signaling
J-Y, Matsui I, Kitamura H, Niimura F, Matsusaka T, Soga T, Rakugi H, pathway independent of mTOR (mammalian target of rapamycin).
Isaka Y: Autophagy protects kidney proximal tubule epithelial cells J Biol Chem 287: 14364–14376, 2012
from mitochondrial metabolic stress. Autophagy 9: 1876–1886, 39. Grishko V, Rachek L, Musiyenko S, Ledoux SP, Wilson GL: Involvement
2013 of mtDNA damage in free fatty acid-induced apoptosis. Free Radic Biol
23. Namba T, Takabatake Y, Kimura T, Takahashi A, Yamamoto T, Matsuda Med 38: 755–762, 2005
J, Kitamura H, Niimura F, Matsusaka T, Iwatani H, Matsui I, Kaimori J, 40. Molina AJA, Wikstrom JD, Stiles L, Las G, Mohamed H, Elorza A, Walzer
Kioka H, Isaka Y, Rakugi H: Autophagic clearance of mitochondria in the G, Twig G, Katz S, Corkey BE, Shirihai OS: Mitochondrial networking
kidney copes with metabolic acidosis. J Am Soc Nephrol 25: 2254– protects beta-cells from nutrient-induced apoptosis. Diabetes 58:
2266, 2014 2303–2315, 2009
24. Takabatake Y, Kimura T, Takahashi A, Isaka Y: Autophagy and the 41. Schönfeld P, Wojtczak L: Fatty acids as modulators of the cellular pro-
kidney: Health and disease. Nephrol Dial Transplant 29: 1639–1647, duction of reactive oxygen species. Free Radic Biol Med 45: 231–241,
2014 2008
25. Yamamoto T, Takabatake Y, Kimura T, Takahashi A, Namba T, 42. Zhu J, Wang KZQ, Chu CT: After the banquet: Mitochondrial bio-
Matsuda J, Minami S, Kaimori J-Y, Matsui I, Kitamura H, Matsusaka genesis, mitophagy, and cell survival. Autophagy 9: 1663–1676, 2013
T, Niimura F, Yanagita M, Isaka Y, Rakugi H: Time-dependent dys- 43. Wu W, Xu H, Wang Z, Mao Y, Yuan L, Luo W, Cui Z, Cui T, Wang XL,
regulation of autophagy: Implications in aging and mitochondrial Shen YH: PINK1-parkin-mediated mitophagy protects mitochondrial
homeostasis in the kidney proximal tubule. Autophagy 12: 801– integrity and prevents metabolic stress-induced endothelial injury.
813, 2016 PLoS One 10: e0132499, 2015
26. Declèves A-E, Zolkipli Z, Satriano J, Wang L, Nakayama T, Rogac 44. Yamashiro DJ, Fluss SR, Maxfield FR: Acidification of endocytic
M, Le TP, Nortier JL, Farquhar MG, Naviaux RK, Sharma K: Reg- vesicles by an ATP-dependent proton pump. J Cell Biol 97: 929–934,
ulation of lipid accumulation by AMP-activated kinase [corrected] 1983
in high fat diet-induced kidney injury. Kidney Int 85: 611–623, 45. Grove KJ, Voziyan PA, Spraggins JM, Wang S, Paueksakon P, Harris RC,
2014 Hudson BG, Caprioli RM: Diabetic nephropathy induces alterations in
27. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y: In vivo the glomerular and tubule lipid profiles. J Lipid Res 55: 1375–1385,
analysis of autophagy in response to nutrient starvation using trans- 2014
genic mice expressing a fluorescent autophagosome marker. Mol Biol 46. Baxter AA, Hulett MD, Poon IK: The phospholipid code: A key com-
Cell 15: 1101–1111, 2004 ponent of dying cell recognition, tumor progression and host-microbe
28. Kimura S, Noda T, Yoshimori T: Dissection of the autophagosome interactions. Cell Death Differ 22: 1893–1905, 2015
maturation process by a novel reporter protein, tandem fluorescent- 47. Omodei D, Fontana L: Calorie restriction and prevention of age-associated
tagged LC3. Autophagy 3: 452–460, 2007 chronic disease. FEBS Lett 585: 1537–1542, 2011
29. Endo T, Nakamura J, Sato Y, Asada M, Yamada R, Takase M, Takaori K, 48. Honma T, Shinohara N, Ito J, Kijima R, Sugawara S, Arai T, Tsuduki T,
Oguchi A, Iguchi T, Higashi AY, Ohbayashi T, Nakamura T, Muso E, Ikeda I: High-fat diet intake accelerates aging, increases expression of
Kimura T, Yanagita M: Exploring the origin and limitations of kidney Hsd11b1, and promotes lipid accumulation in liver of SAMP10 mouse.
regeneration. J Pathol 236: 251–263, 2015 Biogerontology 13: 93–103, 2012
30. Bjørkøy G, Lamark T, Brech A, Outzen H, Perander M, Overvatn A, 49. Kusaka J, Koga H, Hagiwara S, Hasegawa A, Kudo K, Noguchi T: Age-
Stenmark H, Johansen T: p62/SQSTM1 forms protein aggregates dependent responses to renal ischemia-reperfusion injury. J Surg Res
degraded by autophagy and has a protective effect on huntingtin- 172: 153–158, 2012
induced cell death. J Cell Biol 171: 603–614, 2005 50. Koshiba K, Kumano K, Watanabe T, Takashima Y, Cynshi O: [Effects
31. Komatsu M, Waguri S, Koike M, Sou Y-S, Ueno T, Hara T, Mizushima of high fat diet and a novel antioxidant (BO653) on ischemia re-
N, Iwata J, Ezaki J, Murata S, Hamazaki J, Nishito Y, Iemura S, perfusion injury of rat kidney]. Nihon Jinzo Gakkai Shi 39: 455–463,
Natsume T, Yanagawa T, Uwayama J, Warabi E, Yoshida H, Ishii T, 1997
Kobayashi A, Yamamoto M, Yue Z, Uchiyama Y, Kominami E, 51. Vandanmagsar B, Youm Y-H, Ravussin A, Galgani JE, Stadler K, Mynatt
Tanaka K: Homeostatic levels of p62 control cytoplasmic inclusion RL, Ravussin E, Stephens JM, Dixit VD: The NLRP3 inflammasome in-
body formation in autophagy-deficient mice. Cell 131: 1149–1163, stigates obesity-induced inflammation and insulin resistance. Nat Med
2007 17: 179–188, 2011
32. Springer W, Kahle PJ: Regulation of PINK1-Parkin-mediated mitophagy. 52. Wen H, Gris D, Lei Y, Jha S, Zhang L, Huang MT-H, Brickey WJ,
Autophagy 7: 266–278, 2011 Ting JP-Y: Fatty acid-induced NLRP3-ASC inflammasome activa-
33. Galluzzi L, Pietrocola F, Levine B, Kroemer G: Metabolic control of tion interferes with insulin signaling. Nat Immunol 12: 408–415,
autophagy. Cell 159: 1263–1276, 2014 2011
34. Yamahara K, Kume S, Koya D, Tanaka Y, Morita Y, Chin-Kanasaki M, 53. Martin TP, Vailas AC, Durivage JB, Edgerton VR, Castleman KR:
Araki H, Isshiki K, Araki S, Haneda M, Matsusaka T, Kashiwagi A, Quantitative histochemical determination of muscle enzymes:
Maegawa H, Uzu T: Obesity-mediated autophagy insufficiency ex- Biochemical verification. J Histochem Cytochem 33: 1053–1059,
acerbates proteinuria-induced tubulointerstitial lesions. J Am Soc 1985
Nephrol 24: 1769–1781, 2013 54. Brasaemle DL, Barber T, Kimmel AR, Londos C: Post-translational
35. Muoio DM, Newgard CB: Mechanisms of disease:Molecular and met- regulation of perilipin expression. Stabilization by stored intracellular
abolic mechanisms of insulin resistance and beta-cell failure in type 2 neutral lipids. J Biol Chem 272: 9378–9387, 1997
diabetes. Nat Rev Mol Cell Biol 9: 193–205, 2008 55. Ghiggeri GM, Ginevri F, Candiano G, Oleggini R, Perfumo F, Queirolo
36. Kroemer G, Mariño G, Levine B: Autophagy and the integrated stress C, Gusmano R: Characterization of cationic albumin in minimal change
response. Mol Cell 40: 280–293, 2010 nephropathy. Kidney Int 32: 547–553, 1987

J Am Soc Nephrol 28: ccc–ccc, 2016 Autophagy and Lipotoxicity 17


BASIC RESEARCH www.jasn.org

56. Lewy JE, Pesce A: Micropuncture study of albumin transfer in amino- 60. Rambold AS, Cohen S, Lippincott-Schwartz J: Fatty acid traffick-
nucleoside nephrosis in the rat. Pediatr Res 7: 553–559, 1973 ing in starved cells: Regulation by lipid droplet lipolysis, auto-
57. Erkan E, Devarajan P, Schwartz GJ: Mitochondria are the major targets phagy, and mitochondrial fusion dynamics. Dev Cell 32: 678–692,
in albumin-induced apoptosis in proximal tubule cells. J Am Soc 2015
Nephrol 18: 1199–1208, 2007 61. Matsui I, Ito T, Kurihara H, Imai E, Ogihara T, Hori M: Snail, a tran-
58. Ohse T, Inagi R, Tanaka T, Ota T, Miyata T, Kojima I, Ingelfinger JR, scriptional regulator, represses nephrin expression in glomerular epi-
Ogawa S, Fujita T, Nangaku M: Albumin induces endoplasmic re- thelial cells of nephrotic rats. Lab Invest 87: 273–283, 2007
ticulum stress and apoptosis in renal proximal tubular cells. Kidney Int
70: 1447–1455, 2006
59. Liu WJ, Luo M-N, Tan J, Chen W, Huang LZ, Yang C, Pan Q, Li B, Liu HF:
Autophagy activation reduces renal tubular injury induced by urinary This article contains supplemental material online at http://jasn.asnjournals.
proteins. Autophagy 10: 243–256, 2014 org/lookup/suppl/doi:10.1681/ASN.2016070731/-/DCSupplemental.

18 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016

You might also like