Advanced glycation end products (AGEs) are involved in the progression of diabetic nephropathy. AGEs filtered by glomeruli or delivered from the circulation are endocytosed and degraded in the lysosomes of kidney proximal tubular epithelial cells (PTECs). Autophagy is a highly conserved degradation system that regulates intracellular homeostasis by engulfing cytoplasmic components. We have recently demonstrated that autophagic degradation of damaged lysosomes is indispensable for cellular homeostasis in some settings. In this study, we tested the hypothesis that autophagy could contribute to the degradation of AGEs in the diabetic kidney by modulating lysosomal biogenesis. Both a high-glucose and exogenous AGE overload gradually blunted autophagic flux in the cultured PTECs. AGE overload upregulated lysosomal biogenesis and function in vitro, which was inhibited in autophagy-deficient PTECs because of the impaired nuclear translocation of transcription factor EB. Consistently, streptozotocin-treated, PTEC-specific, autophagy-deficient mice failed to upregulate lysosomal biogenesis and exhibited the accumulation of AGEs in the glomeruli and renal vasculature as well as in the PTECs, along with worsened inflammation and fibrosis. These results indicate that autophagy contributes to the degradation of AGEs by the upregulation of lysosomal biogenesis and function in diabetic nephropathy. Strategies aimed at promoting lysosomal function hold promise for treating diabetic nephropathy.

Diabetic nephropathy is the most common cause of end-stage kidney disease worldwide (1). Although various factors contribute to the pathogenesis of diabetic complications, advanced glycation end products (AGEs), which are formed by the irreversible attachment of reducing sugars onto amino groups of proteins in the hyperglycemic state, are a common factor (25). Numerous studies have indicated that AGEs cause structural and functional alternations in various organs, including the kidneys (6). The formation of AGEs can cause generalized cellular dysfunction by inducing cross-linking of extracellular matrices such as collagen (7,8). In addition to the increased production in the hyperglycemic state, decreased clearance by the kidneys is responsible for the accumulation of AGEs in patients with diabetic nephropathy (9). AGEs filtered by glomeruli or delivered from the circulation are endocytosed and then, together with endogenous AGEs produced by the glycation of intracellular proteins, degraded in the lysosomes of the proximal tubular epithelial cells (PTECs) (10,11).

Macroautophagy, hereafter autophagy, is a highly conserved degradation system that functions to regulate intracellular homeostasis (12,13). Cytoplasmic components, such as organelles and proteins, are sequestered into a double-membrane cytosolic vesicle called an autophagosome. Then, the autophagosome fuses with a lysosome, resulting in the degradation of the sequestered materials. It has been demonstrated that autophagy deficiency leads to the accumulation of protein aggregates, resulting in tissue injury in various organs including the central nervous system, liver, and kidneys (1416). We have recently reported that autophagy plays a protective role not only in acute kidney injury, such as ischemic reperfusion injury and cisplatin nephropathy, but also in chronic disorders such as cyclosporine nephropathy and metabolic acidosis (1720). Recently, a novel relationship between autophagy and lysosomes has emerged: autophagy sequesters the damaged lysosome to control lysosomal biogenesis in acute hyperuricemic nephropathy, which is termed lysophagy (21,22).

With regard to the role of autophagy in diabetic nephropathy, it has been reported that autophagy in podocytes plays a protective role by maintaining lysosomal homeostasis (23). In addition, it has been demonstrated that AGE overload disrupts the autophagic pathway because of lysosomal membrane permeabilization (LMP) (24). However, it remains unclear whether autophagy in the proximal tubules can exert a protective role against diabetic nephropathy by the maintenance of lysosomes, where a large number of AGEs are endocytosed. Thus, we hypothesized that autophagy contributes to the degradation of AGEs in the diabetic kidney by modulating lysosomal biogenesis. To address this hypothesis, we investigated whether endogenous and exogenous AGE overload could affect autophagic activity and lysosomal biogenesis in vivo and in vitro. In addition, we explored the consequences of autophagy deficiency on kidney function and morphology in a streptozotocin (STZ)–induced murine diabetic nephropathy model, focusing especially on inflammasome activation and kidney fibrosis.

Experimental Animal Model

Atg5F/F:kidney androgen-regulated protein (KAP) mice have been described previously (18). Briefly, we crossed KAP-Cre transgenic mice, which express Cre recombinase in the kidney proximal tubules, with mice bearing an Atg5flox allele. We have demonstrated efficient knockout of the ATG5 protein in Atg5F/F:KAP mice (84.2% reduction in kidney cortex homogenates of 8-week-old mice) (17). All experiments were performed using 8-week-old male mice. Mice were allowed access to water and mouse chow ad libitum. All animal experiments were approved by the institutional committees of the Animal Research Committee of Osaka University and the Japanese Animal Protection and Management Law (no. 25). Diabetes was induced by intraperitoneal injection of STZ (50 mg/kg) dissolved in a sodium citrate buffer (pH 4.5) for 4 consecutive days. As a control, vehicle-injected (sodium citrate buffer) mice were followed concurrently. These mice were sacrificed 4 months after STZ or vehicle injection. Spot urine samples were collected before sacrifice.

Antibodies and Reagents

We used the following antibodies and reagents: antibodies for carboxymethyllysine (CML; Transgenic, Kobe, Japan); collagen type I and BSA (Abcam, Cambridge, U.K.); α-smooth muscle actin (α-SMA), β-actin, transcription factor EB (TFEB), and epidermal growth factor receptor (EGFR; Sigma-Aldrich, St. Louis, MO); F4/80 (Bio-Rad, Hercules, CA); lysosomal-associated membrane protein (Lamp) 1 (BD Biosciences, Franklin Lakes, NJ); LC3, interleukin (IL)-1β, and phosphorylated (p-)S6K1 (Cell Signaling Technology, Beverly, MA); biotinylated secondary antibodies (Vector Laboratories, Burlingame, CA); horseradish peroxidase–conjugated secondary antibodies (DakoCytomation, Glostrup, Denmark); Alexa 488– and 555–conjugated secondary antibodies (Molecular Probes, Eugene, OR); and bafilomycin, STZ, EGF, l-leucyl-l-leucine methyl ester (LLOMe), and wortmannin (Sigma-Aldrich).

Biochemical Measurements

Plasma glucose, blood urea nitrogen, serum creatinine, total cholesterol, and triglycerides were measured using the Glu-Test-Wako, BUN-Test-Wako, Cholesterol E-test Wako, Triglyceride E-test Wako (Wako Pure Chemical Industries, Osaka, Japan), and CRE-EN Kainos test (Kainos, Tokyo, Japan), respectively. Urinary albumin concentration was determined using the MICROFLUORAL Microalbumin test (Progen, Heidelburg, Germany). Plasma and urinary CML were measured using the Circulex anti-CML mouse autoantibody ELISA kit (CircuLex, Nagano, Japan). Urinary neutrophil gelatinase–associated lipocalin (NGAL) was measured using Mouse Lipocalin-2/NGAL Quantikine ELISA kit (R&D Systems, Minneapolis, MN).

Preparation of Atg5-Deficient and -Competent PTECs and GFP–Galectin 3–Expressing Cell Lines

Atg5-deficient PTECs (Atg5 [−] PTECs) isolated from 3-week-old Atg5F/F:KAP mice and Atg5-revertant PTECs (Atg5 [+] PTECs) were described previously (17). Construction of GFP–galectin 3–expressing retrovirus vector and establishment of GFP–galectin 3 stably expressing Atg5 (+) and Atg5 (−) PTECs were described previously (21).

Preparation of AGE-BSA

According to previous reports, AGE-BSA and BSA were prepared by incubating endotoxin-free BSA (Sigma-Aldrich) with 0.5 mol/L d-glucose in 0.4 mol/L PBS or with PBS alone at 37°C for 8 weeks, respectively (25,26). Free glucose was removed through Amicon centrifugal filters with a 50-kDa molecular weight cutoff (Millipore, Temecula, CA). The concentrations of AGE-BSA and BSA were estimated by the Bradford method.

Cell Culture Conditions

Atg5 (−) and Atg5 (+) PTECs were cultured in DMEM (Sigma-Aldrich) containing 5% FBS. To evaluate the effect of high glucose on intracellular AGE formation, confluent Atg5 (−) and Atg5 (+) PTECs were incubated for up to 72 h in DMEM containing either 5.6 or 25 mmol/L glucose and 1% FBS. To examine the effect of extracellular AGEs (and BSA) administration, confluent Atg5 (−) and Atg5 (+) PTECs were incubated for 6 or 24 h in DMEM (containing 5.6 mmol/L glucose and 1% FBS) with vehicle (PBS) or 40 μmol/L BSA or AGE-BSA.

Western Blot Analysis

Western blot analysis was conducted as described previously (18). Band intensities were measured using ImageJ software (National Institutes of Health, Bethesda, MD).

EGFR Assay

Atg5 (−) and Atg5 (+) PTECs were precultured in DMEM without FBS for 5 h and incubated in DMEM containing 1% FBS with vehicle (PBS) or 40 μmol/L BSA or AGE-BSA for 24 h. After adding 100 ng/mL EGF (time point: 0 min), cells were cultured for the indicated times, and then Western blot analysis for EGFR was performed.

Quantitative RT-PCR

Quantitative RT-PCR was performed as described previously (27). Primers were as follows: nucleotide binding domain, leucine-rich repeats–containing family, pyrin domain-containing (NLRP) 3, 5′-ATGCTGCTTCGACATCTCCT-3′ and 5′-AACCAATGCGAGATCCTGAC-3′; apoptotic speck protein containing a caspase recruitment domain (ASC), 5′-ACAGAAGTGGACGGAGTGCT-3′ and 5′-CTCCAGGTCCATCACCAAGT-3′; caspase-1, 5′-CACAGCTCTGGAGATGGTGA-3′ and 5′-TCTTTCAAGCTTGGGCACTT-3′; MCP-1, 5′-CCCAATCAGTAGGCTGGAGA-3′ and 5′-TCTGGACCCATTCCTTCTTG-3′; NGAL, 5′-CTACAACCAGTTCGCCATGG-3′ and 5′-ACACTCACCACCCATTCAGT-3′; α-SMA, 5′-AGATCAAGATCATTGCCCCTCC-3′ and 5′-TTGTGTGCTAGAGGCAGAGC-3′; collagen type I, 5′-ACGCCATCAAGGTCTACTGC-3′ and 5′-ACTCGAACGGGAATCCATCG-3′; and 18S ribosomal RNA, 5′-AAACGGCTACCACATCCAAG-3′ and 5′-CCTCCAATGGATCCTCGTTA-3′. The relative amounts of RNA were depicted as ratios to 18S ribosomal RNA.

Immunohistochemistry, Immunocytochemistry, and the Evaluation of LMP

Immunohistochemical staining for CML, Lamp1, megalin, F4/80, IL-1β, α-SMA, and collagen type I was performed as described previously (18). Antigen retrieval was performed by autoclaving in 0.01 mmol/L citrate buffer (pH 6) for 10 min at 120°C (for CML, Lamp1, megalin, IL-1β, α-SMA, and collagen type I) or by treatment with proteinase K solution (20 μg/mL) in TE buffer (10 mmol/L Tris-HCl [pH 8] and 1 mmol/L EDTA) for 3 min at room temperature (for F4/80).

Immunocytochemistry for TFEB using Atg5 (+) and Atg5 (−) PTECs, which were exposed to PBS, BSA, or AGE-BSA, was performed as described previously (21).

To assess LMP, the number of GFP-galectin 3–positive dots were counted in Atg5 (+) and Atg5 (−) PTECs stably expressing GFP-galectin 3, which were exposed to PBS, BSA, or AGE-BSA (or LLOMe as a positive control). Cells were observed using an FV1000 confocal microscope (Olympus, Tokyo, Japan).

Statistics

All results are presented as means ± SE. Statistical analysis was conducted using JMP software (SAS institute, Cary, NC). Multiple group comparisons were performed using ANOVA with posttesting according to the Tukey-Kramer test. A P value <0.05 was considered statistically significant.

AGEs Accumulate in the Kidney Tubular Cells of Autophagy-Deficient Diabetic Mice

STZ (50 mg/kg) or a vehicle was intraperitoneally injected into control (Atg5F/F) and KAP-Cre/floxed Atg5 (Atg5F/F:KAP) mice, in which autophagy is deficient in the PTECs, at 8 weeks of age for 4 consecutive days. Four months after treatment, we found no significant difference in plasma glucose levels, kidney weight, blood pressure, or kidney function as assessed by blood urea nitrogen and serum creatinine between STZ-treated control and Atg5F/F:KAP mice (Table 1). Notably, urinary albumin excretion, as evaluated by the albumin-to-creatinine ratio, was not elevated in either of the STZ-treated mice (control and Atg5F/F:KAP), in agreement with the previous reports (28,29). Although periodic acid Schiff staining revealed no apparent morphological changes in STZ-treated control and Atg5F/F:KAP mice, immunohistochemical and Western blot analysis for CML demonstrated that more AGEs accumulated in the STZ-treated Atg5F/F:KAP mice than in the STZ-treated control mice (Fig. 1A–C). The amount of CML in urine significantly increased by STZ treatment in both control and Atg5F/F:KAP mice (Fig. 1D). Of interest, the plasma CML level significantly increased only in STZ-treated Atg5F/F:KAP mice (Fig. 1E). In addition, accumulation of CML in the glomerulus was prominent in STZ-treated Atg5F/F:KAP mice compared with STZ-treated control mice (Supplementary Fig. 1A). Moreover, the intimal thickness of intrarenal artery by CML accumulation was observed in STZ-treated Atg5F/F:KAP mice, whereas it was rarely observed in STZ-treated control mice (Supplementary Fig. 1B). These results indicate that autophagy in the proximal tubules suppresses the accumulation of AGEs in the whole kidney.

Table 1

Physiological parameters of vehicle- and STZ-treated mice

Vehicle (n = 6)
STZ (n = 10)
F/FF/F:KAPF/FF/F:KAP
Body weight (g), day 0 23 ± 2 22 ± 3 23 ± 1 23 ± 1 
Body weight (g), 4 months 30 ± 3 30 ± 3 22 ± 3* 21 ± 1* 
Kidney weight (mg) 158 ± 14 167 ± 13 182 ± 13* 180 ± 25* 
Kidney weight/body weight 5.4 ± 0.8 5.6 ± 0.3 8.2 ± 1.2* 8.8 ± 2.0* 
Glucose (mg/dL) 237 ± 22 268 ± 16 438 ± 65* 430 ± 57* 
Total cholesterol (mg/dL) 66 ± 12 74 ± 13 72 ± 14 100 ± 17 
Triglycerides (mg/dL) 73 ± 12 93 ± 16 104 ± 13 116 ± 13 
Day 0     
 SBP (mmHg) 100 ± 2 94 ± 13 99 ± 3 97 ± 7 
 DBP (mmHg) 65 ± 7 57 ± 8 59 ± 9 57 ± 4 
4 months     
 SBP (mmHg) 97 ± 2 97 ± 9 109 ± 10 97 ± 12 
 DBP (mmHg) 66 ± 5 54 ± 6 57 ± 10 57 ± 11 
Blood urea nitrogen (mg/dL) 28 ± 1 27 ± 3 28 ± 5 29 ± 4 
Creatinine (mg/dL) 0.20 ± 0.06 0.20 ± 0.06 0.18 ± 0.07 0.24 ± 0.02 
Urinary albumin/urinary creatinine 0.22 ± 0.04 0.21 ± 0.01 0.19 ± 0.03 0.24 ± 0.04 
Vehicle (n = 6)
STZ (n = 10)
F/FF/F:KAPF/FF/F:KAP
Body weight (g), day 0 23 ± 2 22 ± 3 23 ± 1 23 ± 1 
Body weight (g), 4 months 30 ± 3 30 ± 3 22 ± 3* 21 ± 1* 
Kidney weight (mg) 158 ± 14 167 ± 13 182 ± 13* 180 ± 25* 
Kidney weight/body weight 5.4 ± 0.8 5.6 ± 0.3 8.2 ± 1.2* 8.8 ± 2.0* 
Glucose (mg/dL) 237 ± 22 268 ± 16 438 ± 65* 430 ± 57* 
Total cholesterol (mg/dL) 66 ± 12 74 ± 13 72 ± 14 100 ± 17 
Triglycerides (mg/dL) 73 ± 12 93 ± 16 104 ± 13 116 ± 13 
Day 0     
 SBP (mmHg) 100 ± 2 94 ± 13 99 ± 3 97 ± 7 
 DBP (mmHg) 65 ± 7 57 ± 8 59 ± 9 57 ± 4 
4 months     
 SBP (mmHg) 97 ± 2 97 ± 9 109 ± 10 97 ± 12 
 DBP (mmHg) 66 ± 5 54 ± 6 57 ± 10 57 ± 11 
Blood urea nitrogen (mg/dL) 28 ± 1 27 ± 3 28 ± 5 29 ± 4 
Creatinine (mg/dL) 0.20 ± 0.06 0.20 ± 0.06 0.18 ± 0.07 0.24 ± 0.02 
Urinary albumin/urinary creatinine 0.22 ± 0.04 0.21 ± 0.01 0.19 ± 0.03 0.24 ± 0.04 

Data are presented as mean ± SE. Physiological parameters obtained from the samples of vehicle- or STZ-treated Atg5F/F and Atg5F/F:KAP mice.

DBP, diastolic blood pressure; F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice; SBP, systolic blood pressure.

*P < 0.05 vs. vehicle-treated mice in each group.

Figure 1

Accumulation of AGEs in the kidney tubules of diabetic autophagy-deficient mice. Representative images of periodic acid Schiff staining (A), Western blot analysis (B), and immunohistochemical staining (C) for CML using the kidney of vehicle- or STZ-treated Atg5F/F:KAP or control (Atg5F/F) mice. B: Western blot analysis was performed using whole-kidney lysates. Densitometric analysis was performed. C: Quantitative analysis of at least 10 high-power fields (×400) was performed in each animal. CML levels in the urine (corrected by urinary creatinine concentration) (D) and in the plasma (E) were determined by ELISA from samples of vehicle- or STZ-treated Atg5F/F:KAP or control mice. The mean value of vehicle-treated control mice is adjusted to 1 as a reference (D). Data are means ± SE (n = 6, vehicle-treated mice; n = 10, STZ-treated mice). Scale bars: 50 μm (A and C). *P < 0.05. F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice.

Figure 1

Accumulation of AGEs in the kidney tubules of diabetic autophagy-deficient mice. Representative images of periodic acid Schiff staining (A), Western blot analysis (B), and immunohistochemical staining (C) for CML using the kidney of vehicle- or STZ-treated Atg5F/F:KAP or control (Atg5F/F) mice. B: Western blot analysis was performed using whole-kidney lysates. Densitometric analysis was performed. C: Quantitative analysis of at least 10 high-power fields (×400) was performed in each animal. CML levels in the urine (corrected by urinary creatinine concentration) (D) and in the plasma (E) were determined by ELISA from samples of vehicle- or STZ-treated Atg5F/F:KAP or control mice. The mean value of vehicle-treated control mice is adjusted to 1 as a reference (D). Data are means ± SE (n = 6, vehicle-treated mice; n = 10, STZ-treated mice). Scale bars: 50 μm (A and C). *P < 0.05. F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice.

Close modal

Endogenous or Exogenous AGE Overload Gradually Stagnates Autophagic Flux in the PTECs

To investigate whether endogenous AGE overload affects autophagic flux in the proximal tubular cells, autophagy-deficient PTECs isolated from Atg5F/F:KAP mice (Atg5 [−] PTECs) and Atg5-revertant cells (Atg5 [+] PTECs) were exposed to normal (5.6 mmol/L) or high (25 mmol/L) glucose medium for a prolonged period (72 h). It was demonstrated that more CML accumulated in the Atg5 (−) PTECs incubated in the high-glucose medium compared with the other conditions (Fig. 2A). Next, we incubated Atg5 (+) PTECs in normal or high-glucose states for up to 72 h to examine autophagic flux. LC3-II level was increased in the cells incubated in the high-glucose medium for 72 h, which was unchanged after bafilomycin treatment, whereas a substantial conversion of LC3-I to LC3-II by bafilomycin treatment was observed in other conditions (Fig. 2B). We excluded the possibility that the observation with regard to autophagic activity was not attributed to high osmolarity itself by the addition of mannitol to the normal glucose medium. Moreover, wortmannin, an inhibitor of autophagosome formation, suppressed the increase of LC3-II by bafilomycin in the high-glucose state (Supplementary Fig. 2A). These results suggest that the high-glucose state gradually blunted in the later step of autophagy.

Figure 2

Endogenous or exogenous AGE overload gradually stagnates autophagic flux in PTECs. A: Western blot analysis for CML using the lysates of Atg5 (+) or Atg5 (−) PTECs, which had been exposed to high-glucose (HG) medium (25 mmol/L) or normal-glucose (NG) medium (5.6 mmol/L) for 72 h. B: Western blot analysis for LC3 using the lysates of Atg5 (+) or Atg5 (−) PTECs, which had been exposed to high-glucose medium and normal-glucose medium (with or without mannitol) for 24 and 72 h. To assess autophagic flux, 100 nmol/L bafilomycin was administered to the medium 2 h before harvest. A and B: Representative images are presented. Densitometric analysis was performed. Western blot analysis for BSA and CML (C) and LC3 (D) was performed using the lysates of Atg5 (+) or Atg5 (−) PTECs, which had been exposed to vehicle (PBS), BSA, or AGE-BSA for 6 and 24 h. To assess autophagic flux, 100 nmol/L bafilomycin was administered to the medium 60 min before harvest. Representative images are presented. D (bottom panel): Densitometric analysis was performed. Data are means ± SE (n = 6 in each group). *P < 0.05. Mn, mannitol.

Figure 2

Endogenous or exogenous AGE overload gradually stagnates autophagic flux in PTECs. A: Western blot analysis for CML using the lysates of Atg5 (+) or Atg5 (−) PTECs, which had been exposed to high-glucose (HG) medium (25 mmol/L) or normal-glucose (NG) medium (5.6 mmol/L) for 72 h. B: Western blot analysis for LC3 using the lysates of Atg5 (+) or Atg5 (−) PTECs, which had been exposed to high-glucose medium and normal-glucose medium (with or without mannitol) for 24 and 72 h. To assess autophagic flux, 100 nmol/L bafilomycin was administered to the medium 2 h before harvest. A and B: Representative images are presented. Densitometric analysis was performed. Western blot analysis for BSA and CML (C) and LC3 (D) was performed using the lysates of Atg5 (+) or Atg5 (−) PTECs, which had been exposed to vehicle (PBS), BSA, or AGE-BSA for 6 and 24 h. To assess autophagic flux, 100 nmol/L bafilomycin was administered to the medium 60 min before harvest. Representative images are presented. D (bottom panel): Densitometric analysis was performed. Data are means ± SE (n = 6 in each group). *P < 0.05. Mn, mannitol.

Close modal

Next, to examine the role of autophagy when PTECs are exposed to exogenous AGEs, Atg5 (+) and Atg5 (−) PTECs were exposed to AGE-BSA or BSA for 24 h. We found more AGE-BSA accumulated in the AGE-BSA–treated Atg5 (−) PTECs than in the AGE-BSA–treated Atg5 (+) PTECs, whereas we observed less BSA in the BSA-treated Atg5 (−) PTECs compared with the BSA-treated Atg5 (+) PTECs (Fig. 2C). We also examined autophagic flux in AGE-BSA– or BSA-treated Atg5 (+) PTECs for up to 24 h. LC3-II level was not significantly increased in the cells treated with AGE-BSA for 24 h, which was unchanged after bafilomycin treatment similar to endogenous AGEs, whereas a substantial conversion of LC3-I to LC3-II by bafilomycin treatment was observed in other conditions (Fig. 2D). Moreover, wortmannin suppressed the increase of LC3-II by bafilomycin in AGE-BSA–treated Atg5 (+) PTECs (Supplementary Fig. 2B). These results suggest that exogenous AGEs also gradually disrupt the later step of autophagy.

Autophagy Upregulates Lysosomal Biogenesis and Function in Response to AGE Overload

The observation that high glucose and AGE overload led to the gradual stagnation of autophagic flux prompted us to investigate whether they could modulate lysosomal biogenesis and function. Atg5 (+) and Atg5 (−) PTECs were exposed to high glucose (or normal glucose) or AGE-BSA (or BSA). The protein levels of Lamp1 were comparable between normal and high-glucose conditions in both Atg5 (+) and Atg5 (−) PTECs, indicating that high glucose itself does not affect lysosomal biogenesis (Fig. 3A). In contrast, we observed increased protein levels of Lamp1 in BSA- and AGE-BSA–exposed Atg5 (+) PTECs, but not in BSA- or AGE-BSA–exposed Atg5 (−) PTECs, suggesting that autophagy upregulates lysosomal biogenesis upon protein overload (Fig. 3B). In line with these observations, the protein level of TFEB, a master regulator of lysosomal biogenesis, was higher in Atg5 (+) PTECs than in Atg5 (−) PTECs, and the difference was prominent after AGE overload (Fig. 3B). In addition, nuclear translocation of TFEB was impaired in Atg5 (−) PTECs irrespective of protein overload (Fig. 3C). To investigate the mechanism underlying autophagy-dependent TFEB activation, we explored the mammalian target of rapamycin (mTOR) pathway, which regulates the nuclear translocation of TFEB (30). Protein levels of p-S6K1 (downstream from the mTOR pathway) were comparably increased after 24 h of exposure to BSA or AGE-BSA in both Atg5 (+) and Atg5 (−) PTECs (Supplementary Fig. 3).

Figure 3

Autophagy upregulates lysosomal biogenesis and function in response to AGE overload. Representative images of Western blot analysis for Lamp1 (and TFEB) using the lysates of Atg5 (+) or Atg5 (−) PTECs cultured in high-glucose (HG) and normal-glucose (NG) medium (A) or in PBS-, BSA-, or AGE-BSA–containing medium (B) for the indicated times. Densitometric analysis was performed. The mean value of Atg5 (+) PTECs at the base point is adjusted to 1 as a reference. C: Representative images of immunocytochemistry for TFEB in Atg5 (+) or Atg5 (−) PTECs, which had been exposed to PBS, BSA, or AGE-BSA for 24 h. Quantification of the number of cells positive for nuclear TFEB was performed (right panel). Values are means of measurements from five fields containing at least 30 cells. D: EGFR degradation assay using the lysates of Atg5 (+) or Atg5 (−) PTECs, which had been exposed to PBS, BSA, or AGE-BSA for the indicated times. The mean value at baseline is adjusted to 1 as a reference. E: Representative fluorescent images of Atg5 (+) or Atg5 (−) PTECs stably expressing GFP-galectin 3. Images of LLOMe-treated Atg5 (+) PTECs are shown as a positive control for LMP. Atg5 (+), Atg5 (+) PTEC; Atg5 (−), Atg5 (−) PTEC. Data are means ± SE (n = 6 in each group). Scale bars: 10 μm (C and E). *P < 0.05. Mn, mannitol.

Figure 3

Autophagy upregulates lysosomal biogenesis and function in response to AGE overload. Representative images of Western blot analysis for Lamp1 (and TFEB) using the lysates of Atg5 (+) or Atg5 (−) PTECs cultured in high-glucose (HG) and normal-glucose (NG) medium (A) or in PBS-, BSA-, or AGE-BSA–containing medium (B) for the indicated times. Densitometric analysis was performed. The mean value of Atg5 (+) PTECs at the base point is adjusted to 1 as a reference. C: Representative images of immunocytochemistry for TFEB in Atg5 (+) or Atg5 (−) PTECs, which had been exposed to PBS, BSA, or AGE-BSA for 24 h. Quantification of the number of cells positive for nuclear TFEB was performed (right panel). Values are means of measurements from five fields containing at least 30 cells. D: EGFR degradation assay using the lysates of Atg5 (+) or Atg5 (−) PTECs, which had been exposed to PBS, BSA, or AGE-BSA for the indicated times. The mean value at baseline is adjusted to 1 as a reference. E: Representative fluorescent images of Atg5 (+) or Atg5 (−) PTECs stably expressing GFP-galectin 3. Images of LLOMe-treated Atg5 (+) PTECs are shown as a positive control for LMP. Atg5 (+), Atg5 (+) PTEC; Atg5 (−), Atg5 (−) PTEC. Data are means ± SE (n = 6 in each group). Scale bars: 10 μm (C and E). *P < 0.05. Mn, mannitol.

Close modal

We next examined the effect of autophagy on lysosomal function using an EGFR degradation assay, in which the capacity of EGF-induced degradation of EGFR reflects lysosomal function. After 24 h of exposure to BSA or AGE-BSA, the degradation of EGFR protein was comparably promoted in Atg5 (+) PTECs, whereas the degradation was significantly delayed in Atg5 (−) PTECs (Fig. 3D). Collectively, autophagy upregulates lysosomal biogenesis and function in response to protein or AGE overload.

Previous reports that autophagy is induced to degrade the damaged lysosome after the uptake of mineral crystals such as silica and monosodium urate or after photodamage prompted us to investigate whether BSA or AGE overload could cause lysosomal damage (21,31). We explored galectin 3, which is localized to the endosomal compartment from cytoplasm in response to endosomal membrane rupture (32). The lysosomotropic compound LLOMe, which evokes lysosomal damage, caused the formation of GFP-positive dots in Atg5 (+) PTECs stably expressing GFP–galectin 3, whereas few GFP-positive dots were observed in both Atg5 (+) and Atg5 (−) PTECs during BSA or AGE-BSA exposure (Fig. 3E). These data indicate that LMP was not induced during BSA or AGE overload.

Autophagy Contributes to the Upregulation of Lysosomal Biogenesis in the Diabetic State In Vivo

To recapitulate the in vitro observation that autophagy contributes to the upregulation of lysosomal biogenesis in response to AGE overload in vivo, we next examined the effect of autophagy on lysosomal biogenesis in the proximal tubules of diabetic mice. The protein level of Lamp1 in the kidney of STZ-treated control mice was increased compared with that in the nondiabetic state (Fig. 4A). Intriguingly, this upregulation was not evident in the kidney of STZ-treated Atg5F/F:KAP mice (Fig. 4A). Consistently, immunohistochemical analysis indicated that the increase in Lamp1-positive lysosomes in the proximal tubules of STZ-treated control mice was not observed in those of STZ-treated Atg5F/F:KAP mice (Fig. 4B). These results suggest that autophagy contributes to the upregulation of lysosomal biogenesis in the diabetic state in vivo.

Figure 4

Autophagy contributes to the upregulation of lysosomal biogenesis in the diabetic state in vivo. Representative images of Western blot analysis for Lamp1 (A) and immunohistochemistry for Lamp1 and megalin (proximal tubule marker) (B) using the kidney of vehicle- or STZ-treated Atg5F/F:KAP or control mice. A and B (right): Densitometric analysis for Lamp1 (A) and quantification of Lamp1- positive intensity (B) are shown. The mean value of vehicle-treated control mice is adjusted to 1 as a reference. Data are means ± SE (n = 6, vehicle-treated mice; n = 10, STZ-treated mice). Scale bar: 50 μm. *P < 0.05. F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice.

Figure 4

Autophagy contributes to the upregulation of lysosomal biogenesis in the diabetic state in vivo. Representative images of Western blot analysis for Lamp1 (A) and immunohistochemistry for Lamp1 and megalin (proximal tubule marker) (B) using the kidney of vehicle- or STZ-treated Atg5F/F:KAP or control mice. A and B (right): Densitometric analysis for Lamp1 (A) and quantification of Lamp1- positive intensity (B) are shown. The mean value of vehicle-treated control mice is adjusted to 1 as a reference. Data are means ± SE (n = 6, vehicle-treated mice; n = 10, STZ-treated mice). Scale bar: 50 μm. *P < 0.05. F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice.

Close modal

Autophagy Inhibits Macrophage Infiltration and Inflammasome Activation in the Diabetic Kidney

Because it has been reported that AGE accumulation could evoke chronic inflammation, we examined the effect of autophagy on macrophage infiltration and inflammasome activation induced by the diabetic state (33,34). Immunohistochemistry for F4/80 showed more macrophage infiltration in the kidneys of STZ-treated Atg5F/F:KAP mice compared with those in nondiabetic mice (control and Atg5F/F:KAP) and STZ-treated control mice (Fig. 5A). Moreover, mRNA levels of NLRP3, ASC, and caspase-1 increased in the kidneys of STZ-treated Atg5F/F:KAP mice compared with STZ-treated control mice (Fig. 5B). In addition, the protein level of IL-1β, as assessed by Western blot analysis and immunohistochemistry, increased in the kidneys of STZ-treated control mice compared with vehicle-treated control mice, which was more pronounced in the Atg5F/F:KAP mice (Fig. 5C and D). These results indicate that exacerbation of macrophage infiltration and NLRP3-mediated inflammation induced by the diabetic state becomes apparent with autophagy deficiency.

Figure 5

Autophagy inhibits macrophage infiltration and inflammasome activation in the diabetic kidney. A: Representative images of immunohistochemical staining for F4/80 (left panel) of the kidney sections of vehicle- or STZ-treated Atg5F/F:KAP or control mice. Quantification of F4/80 positive area was performed (right panel). BD: mRNA levels of NLRP3, ASC, and caspase-1 obtained from quantitative RT-PCR (B) and representative image of Western blot analysis (C) and immunohistochemical analysis (D) for IL-1β using kidney samples from vehicle- or STZ-treated Atg5F/F:KAP or control mice. C (right panel): Densitometric analysis was performed. E: mRNA level of MCP-1 obtained from quantitative RT-PCR analysis using the lysates of Atg5 (+) or Atg5 (−) PTECs after PBS, BSA, or AGE-BSA exposure. B, C, and E: The mean value of vehicle-treated control mice (B and C) or of vehicle-treated Atg5 (+) PTECs (E) is adjusted to 1 as a reference. Data are means ± SE (n = 6, vehicle-treated mice; n = 10, STZ-treated mice; n = 6, in vitro assay). Scale bars: 200 μm (A) and 50 μm (D). *P < 0.05. F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice.

Figure 5

Autophagy inhibits macrophage infiltration and inflammasome activation in the diabetic kidney. A: Representative images of immunohistochemical staining for F4/80 (left panel) of the kidney sections of vehicle- or STZ-treated Atg5F/F:KAP or control mice. Quantification of F4/80 positive area was performed (right panel). BD: mRNA levels of NLRP3, ASC, and caspase-1 obtained from quantitative RT-PCR (B) and representative image of Western blot analysis (C) and immunohistochemical analysis (D) for IL-1β using kidney samples from vehicle- or STZ-treated Atg5F/F:KAP or control mice. C (right panel): Densitometric analysis was performed. E: mRNA level of MCP-1 obtained from quantitative RT-PCR analysis using the lysates of Atg5 (+) or Atg5 (−) PTECs after PBS, BSA, or AGE-BSA exposure. B, C, and E: The mean value of vehicle-treated control mice (B and C) or of vehicle-treated Atg5 (+) PTECs (E) is adjusted to 1 as a reference. Data are means ± SE (n = 6, vehicle-treated mice; n = 10, STZ-treated mice; n = 6, in vitro assay). Scale bars: 200 μm (A) and 50 μm (D). *P < 0.05. F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice.

Close modal

To determine whether lysosomal overload of AGE-BSA in the proximal tubules directly induces macrophage infiltration and inflammation as observed in vivo, we evaluated MCP-1 expression in vitro. BSA or AGE-BSA exposure did not affect mRNA levels of MCP-1 in Atg5 (+) PTECs, whereas significantly increased expression of MCP-1 was detected in Atg5 (−) PTECs (Fig. 5E), suggesting that lysosomal overload induced by BSA or AGE-BSA exposure can directly induce inflammation in autophagy-deficient PTECs.

Autophagy Protects Kidneys From Injury and Fibrosis in the Diabetic State

We next investigated the effect of autophagy on kidney injury and fibrosis in the diabetic state. mRNA expression and urinary concentration of the tubular injury marker, NGAL, was increased in STZ-treated Atg5F/F:KAP mice compared with STZ-treated control mice (Fig. 6A). Because it has been reported that AGEs induce fibrotic change in the diabetic state, we evaluated the expression of α-SMA and collagen type I, hallmarks of fibrosis (25). Immunohistochemistry for α-SMA and collagen type I demonstrated that the kidneys of STZ-treated Atg5F/F:KAP mice exhibited increased areas of positive staining compared with STZ-treated control mice (Fig. 6B and C). mRNA expression of α-SMA and collagen type I were more significantly increased in the kidneys of STZ-treated Atg5F/F:KAP mice than those in STZ-treated control mice (Fig. 6D and E). These results indicate that autophagy protects the kidneys from injury and fibrosis in the diabetic state.

Figure 6

Autophagy protects the kidney from injury and fibrosis in the diabetic state. A: mRNA level in the kidney (left panel) and urinary concentration (corrected for urinary creatinine concentration) (right panel) of kidney injury marker NGAL using vehicle- or STZ-treated Atg5F/F:KAP or control mice. Representative images of immunohistochemical analysis (B and C) and mRNA levels (D and E) of α-SMA (B and D) and collagen type I (C and E) using the kidneys of vehicle- or STZ-treated Atg5F/F:KAP and control mice. The mean value of vehicle-treated control mice is adjusted to 1 as a reference. Data are means ± SE (n = 6, vehicle-treated mice; n = 10, STZ-treated mice). Scale bars: 100 μm. *P < 0.05. F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice.

Figure 6

Autophagy protects the kidney from injury and fibrosis in the diabetic state. A: mRNA level in the kidney (left panel) and urinary concentration (corrected for urinary creatinine concentration) (right panel) of kidney injury marker NGAL using vehicle- or STZ-treated Atg5F/F:KAP or control mice. Representative images of immunohistochemical analysis (B and C) and mRNA levels (D and E) of α-SMA (B and D) and collagen type I (C and E) using the kidneys of vehicle- or STZ-treated Atg5F/F:KAP and control mice. The mean value of vehicle-treated control mice is adjusted to 1 as a reference. Data are means ± SE (n = 6, vehicle-treated mice; n = 10, STZ-treated mice). Scale bars: 100 μm. *P < 0.05. F/F, Atg5F/F mice; F/F:KAP, Atg5F/F:KAP mice.

Close modal

In this study, we demonstrated that 1) lysosomal biogenesis and function in the kidney proximal tubules are upregulated in the diabetic state, at least partially, to cope with AGE overload; 2) both endogenous and exogenous AGEs gradually disrupt autophagic flux; and 3) autophagy in PTECs plays an essential role in the degradation of AGEs via lysosomal degradation.

Our study cannot completely exclude the possibility that high glucose itself stimulates lysosomal biogenesis in vivo, because we cannot segregate the effects of hyperglycemia and AGE overload on AGE accumulation in autophagy deficiency; however, our result that high glucose does not stimulate lysosomal biogenesis in vitro supports the possibility that lysosomal biogenesis and function are upregulated in response to AGE overload.

In this study, more AGEs accumulated in the proximal tubules of proximal tubule-specific, autophagy-deficient mice than in those of control mice in the diabetic state. One possible explanation is that the amount of intracellular proteins that could be a target of glycation increase because of the lack of canonical (bulk) autophagy in the diabetic state, which is supported by the in vitro study using a high-glucose medium. Alternatively, but not exclusively, accumulation of AGEs in the autophagy-deficient kidney is attributed to insufficient degradation in the lysosomes of the proximal tubules. The fact that plasma concentration and glomerular deposition of CML increased in proximal tubule-specific, autophagy-deficient diabetic mice indicates that autophagy in the PTECs copes with the increasing number of AGEs that exist in urine and plasma by promoting upregulation of the endocytosis-lysosomal system. How are lysosomal biogenesis and function upregulated by autophagy during AGE overload? We previously reported that ruptured lysosomes are directly sequestered by autophagy in the PTECs when exposed to monosodium urate crystals, termed lysophagy (21,35). In this study, LMP, as assessed by GFP–galectin 3–positive dots and electron microscopic analysis of the PTECs, was not observed, even during AGE overload (Fig. 3E and data not shown). Recently, it has been reported that AGE-BSA retards autophagic activity because of LMP in HK-2 cells (24). The precise mechanism to explain this discrepancy remains unknown, but it may be related to the experimental conditions such as the type of cells used or the concentration of AGE-BSA; however, our result that the nuclear translocation of TFEB was impaired in autophagy-deficient PTECs even without AGE overload could shed light on an essential role of autophagy in lysosomal biogenesis other than lysophagy. Because the levels of p-S6K1 protein were comparable between Atg5 (+) and Atg5 (−) PTECs, autophagy-dependent TFEB activation seems to be mTOR independent, at least in our experimental conditions.

Similar to AGE-BSA, BSA activated the endocytosis-lysosomal system in autophagy-competent PTECs, as is evidenced by the Lamp1 protein level and EGFR assay, although BSA did not retard autophagic activity. In autophagy-deficient PTECs, unlike AGE-BSA, BSA did not accumulate despite the lack of upregulation of lysosomal function. This is presumably because BSA may be more easily degraded than AGE-BSA, which is irreversibly glycated. Another possibility is that an endocytosis pathway is disturbed in autophagy-deficient PTECs because of the nonautophagic effect of Atg5, as is suggested by our observation (Fig. 2C). In either case, BSA-induced upregulation of lysosomal biogenesis and function via autophagy could be critical for proteinuric glomerular diseases including advanced diabetic nephropathy.

Another prominent finding of this study is that tubular injury, inflammation, and interstitial fibrosis are exacerbated in the diabetic proximal tubule–specific, autophagy-deficient kidney. Considering that the blood glucose levels were comparable, it is possible that AGE accumulation because of autophagy deficiency leads to such changes. In fact, many reports have shown that AGEs induce extracellular matrix expansion and epithelial mesenchymal transition and inflammation (26,36). In our study, the PTECs produced more MCP-1 after AGE-BSA exposure in the autophagy-deficient PTECs than in autophagy-competent PTECs. In addition, mRNA levels of NLRP3, ASC, and caspase-1 and the protein level of IL-1β increased, especially in the diabetic proximal tubule–specific, autophagy-deficient kidney. These results indicate that autophagy suppresses kidney inflammation and fibrosis by upregulation of lysosomal degradation of AGEs.

Considering that AGEs blunt autophagic flux in cultured PTECs, it is easy to infer that AGEs should accumulate even in autophagy-competent cells treated with AGEs, but actually, that has not been the case. Previous reports have suggested that AGEs are directly endocytosed to lysosomes for degradation (10,11). Although AGEs place an inordinate level of stress on the lysosomal systems (which will blunt the autophagic flux), lysosomal function is preserved compared with the autophagy-deficient kidney, which will allow the degradation of AGEs. Although we cannot easily assess autophagic flux in vivo, immunostaining for SQSTM1/p62, the substrate of autophagy, demonstrated that the numbers of SQSTM1/p62-positive dots were comparable between STZ- and vehicle-treated control mice, whereas they significantly increased in STZ-treated Atg5F/F:KAP mice compared with vehicle-treated Atg5F/F:KAP mice, indicating that autophagic flux is not completely inhibited in diabetic control mice (Supplementary Fig. 4).

In addition to hyperglycemia, STZ administration induces various physiological changes such as lipid metabolism, which may affect autophagic activity. However, most physiological changes including total cholesterol and triglycerides were comparable between STZ-treated control and Atg5F/F:KAP mice. Therefore, it is possible that autophagy deficiency per se leads to lysosomal dysfunction and accumulation of AGEs.

Although several agents that reduce or inhibit AGE production have been experimentally or clinically evaluated, most research efforts and trials have failed to clarify clinical efficacy in diabetic nephropathy (3). Our findings suggest that upregulation of autophagic activity and thereby enhancement of upregulation of lysosomal function for AGE degradation could be a promising therapeutic strategy against diabetic nephropathy. The proposed mechanisms by which autophagy inhibits accumulation of AGEs in diabetic nephropathy are depicted schematically (Fig. 7).

Figure 7

Schematic illustration of possible mechanisms by which autophagy protects PTECs from the accumulation of AGEs in diabetic nephropathy. Autophagy inhibits the accumulation of endogenous AGEs by degrading intracellular proteins and exogenous AGEs by upregulating lysosomal functions, resulting in the alleviation of inflammation and fibrotic change in diabetic nephropathy.

Figure 7

Schematic illustration of possible mechanisms by which autophagy protects PTECs from the accumulation of AGEs in diabetic nephropathy. Autophagy inhibits the accumulation of endogenous AGEs by degrading intracellular proteins and exogenous AGEs by upregulating lysosomal functions, resulting in the alleviation of inflammation and fibrotic change in diabetic nephropathy.

Close modal

In conclusion, autophagy in the proximal tubules of the kidney suppresses the accumulation of AGEs by stimulating the upregulation of lysosomal biogenesis and function and alleviates inflammation and fibrosis in the diabetic state.

Acknowledgments. The authors thank Noboru Mizushima (University of Tokyo, Tokyo, Japan) for donation of the Atg5F/F mice, Toshimi Michigami (Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan) for megalin antibody, and Naoko Horimoto and Megumi Kameda (Osaka University, Osaka, Japan) for technical and secretarial assistance.

Funding. This work was supported by the Japan Foundation for Applied Enzymology (to A.T.) and the Manpei Suzuki Diabetes Foundation and Uehara Memorial Foundation (to T.K.).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. A.T., Y.T., and T.K. designed the study. A.T. conducted all experiments and drafted the manuscript. Y.T. edited and revised the manuscript. I.Mae. gave some helpful advice about in vitro experiments. T.K., T.N., T.Ya., J.M., S.M., J.-y.K., and I.Mat. contributed to discussion and reviewed the manuscript. T.M. and F.N. provided mice and commented on the manuscript. T.Yo. and Y.I. supervised this study. Y.T. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Cooper
ME
.
Pathogenesis, prevention, and treatment of diabetic nephropathy
.
Lancet
1998
;
352
:
213
219
[PubMed]
2.
Tan
AL
,
Forbes
JM
,
Cooper
ME
.
AGE, RAGE, and ROS in diabetic nephropathy
.
Semin Nephrol
2007
;
27
:
130
143
[PubMed]
3.
Goh
SY
,
Cooper
ME
.
Clinical review: the role of advanced glycation end products in progression and complications of diabetes
.
J Clin Endocrinol Metab
2008
;
93
:
1143
1152
[PubMed]
4.
Kanwar
YS
,
Sun
L
,
Xie
P
,
Liu
FY
,
Chen
S
.
A glimpse of various pathogenetic mechanisms of diabetic nephropathy
.
Annu Rev Pathol
2011
;
6
:
395
423
[PubMed]
5.
Beisswenger
PJ
,
Howell
SK
,
Russell
GB
,
Miller
ME
,
Rich
SS
,
Mauer
M
.
Early progression of diabetic nephropathy correlates with methylglyoxal-derived advanced glycation end products
.
Diabetes Care
2013
;
36
:
3234
3239
[PubMed]
6.
Ahmed
N
.
Advanced glycation endproducts--role in pathology of diabetic complications
.
Diabetes Res Clin Pract
2005
;
67
:
3
21
[PubMed]
7.
Forbes
JM
,
Cooper
ME
,
Oldfield
MD
,
Thomas
MC
.
Role of advanced glycation end products in diabetic nephropathy
.
J Am Soc Nephrol
2003
;
14
(
Suppl. 3
):
S254
S258
[PubMed]
8.
Bohlender
JM
,
Franke
S
,
Stein
G
,
Wolf
G
.
Advanced glycation end products and the kidney
.
Am J Physiol Renal Physiol
2005
;
289
:
F645
F659
[PubMed]
9.
Miyata
T
,
Ueda
Y
,
Horie
K
, et al
.
Renal catabolism of advanced glycation end products: the fate of pentosidine
.
Kidney Int
1998
;
53
:
416
422
[PubMed]
10.
Gugliucci
A
,
Bendayan
M
.
Renal fate of circulating advanced glycated end products (AGE): evidence for reabsorption and catabolism of AGE-peptides by renal proximal tubular cells
.
Diabetologia
1996
;
39
:
149
160
[PubMed]
11.
Saito
A
,
Takeda
T
,
Sato
K
, et al
.
Significance of proximal tubular metabolism of advanced glycation end products in kidney diseases
.
Ann N Y Acad Sci
2005
;
1043
:
637
643
[PubMed]
12.
Levine
B
,
Kroemer
G
.
Autophagy in the pathogenesis of disease
.
Cell
2008
;
132
:
27
42
[PubMed]
13.
Mizushima
N
,
Levine
B
,
Cuervo
AM
,
Klionsky
DJ
.
Autophagy fights disease through cellular self-digestion
.
Nature
2008
;
451
:
1069
1075
[PubMed]
14.
Komatsu
M
,
Waguri
S
,
Chiba
T
, et al
.
Loss of autophagy in the central nervous system causes neurodegeneration in mice
.
Nature
2006
;
441
:
880
884
[PubMed]
15.
Komatsu
M
,
Waguri
S
,
Koike
M
, et al
.
Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice
.
Cell
2007
;
131
:
1149
1163
[PubMed]
16.
Takabatake
Y
,
Kimura
T
,
Takahashi
A
,
Isaka
Y
.
Autophagy and the kidney: health and disease
.
Nephrol Dial Transplant
2014
;
29
:
1639
1647
[PubMed]
17.
Kimura
T
,
Takabatake
Y
,
Takahashi
A
, et al
.
Autophagy protects the proximal tubule from degeneration and acute ischemic injury
.
J Am Soc Nephrol
2011
;
22
:
902
913
[PubMed]
18.
Takahashi
A
,
Kimura
T
,
Takabatake
Y
, et al
.
Autophagy guards against cisplatin-induced acute kidney injury
.
Am J Pathol
2012
;
180
:
517
525
[PubMed]
19.
Kimura
T
,
Takahashi
A
,
Takabatake
Y
, et al
.
Autophagy protects kidney proximal tubule epithelial cells from mitochondrial metabolic stress
.
Autophagy
2013
;
9
:
1876
1886
[PubMed]
20.
Namba
T
,
Takabatake
Y
,
Kimura
T
, et al
.
Autophagic clearance of mitochondria in the kidney copes with metabolic acidosis
.
J Am Soc Nephrol
2014
;
25
:
2254
2266
[PubMed]
21.
Maejima
I
,
Takahashi
A
,
Omori
H
, et al
.
Autophagy sequesters damaged lysosomes to control lysosomal biogenesis and kidney injury
.
EMBO J
2013
;
32
:
2336
2347
[PubMed]
22.
Hasegawa
J
,
Maejima
I
,
Iwamoto
R
,
Yoshimori
T
.
Selective autophagy: lysophagy
.
Methods
2015
;
75
:
128
132
[PubMed]
23.
Tagawa
A
,
Yasuda
M
,
Kume
S
, et al
.
Impaired podocyte autophagy exacerbates proteinuria in diabetic nephropathy
.
Diabetes
2016
;
65
:
755
767
[PubMed]
24.
Liu
WJ
,
Shen
TT
,
Chen
RH
, et al
.
Autophagy-lysosome pathway in renal tubular epithelial cells is disrupted by advanced glycation end products in diabetic nephropathy
.
J Biol Chem
2015
;
290
:
20499
20510
[PubMed]
25.
Oldfield
MD
,
Bach
LA
,
Forbes
JM
, et al
.
Advanced glycation end products cause epithelial-myofibroblast transdifferentiation via the receptor for advanced glycation end products (RAGE)
.
J Clin Invest
2001
;
108
:
1853
1863
[PubMed]
26.
Burns
WC
,
Twigg
SM
,
Forbes
JM
, et al
.
Connective tissue growth factor plays an important role in advanced glycation end product-induced tubular epithelial-to-mesenchymal transition: Implications for diabetic renal disease
.
J Am Soc Nephrol
2006
;
17
:
2484
2494
[PubMed]
27.
Kusunoki
Y
,
Matsui
I
,
Hamano
T
, et al
.
Excess 25-hydroxyvitamin D3 exacerbates tubulointerstitial injury in mice by modulating macrophage phenotype
.
Kidney Int
2015
;
88
:
1013
1029
[PubMed]
28.
Fujita
H
,
Haseyama
T
,
Kayo
T
, et al
.
Increased expression of glutathione S-transferase in renal proximal tubules in the early stages of diabetes: a study of type-2 diabetes in the Akita mouse model
.
Exp Nephrol
2001
;
9
:
380
386
[PubMed]
29.
Gurley
SB
,
Clare
SE
,
Snow
KP
,
Hu
A
,
Meyer
TW
,
Coffman
TM
.
Impact of genetic background on nephropathy in diabetic mice
.
Am J Physiol Renal Physiol
2006
;
290
:
F214
F222
[PubMed]
30.
Settembre
C
,
Zoncu
R
,
Medina
DL
, et al
.
A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB
.
EMBO J
2012
;
31
:
1095
1108
[PubMed]
31.
Hung
YH
,
Chen
LM
,
Yang
JY
,
Yang
WY
.
Spatiotemporally controlled induction of autophagy-mediated lysosome turnover
.
Nat Commun
2013
;
4
:
2111
[PubMed]
32.
Paz
I
,
Sachse
M
,
Dupont
N
, et al
.
Galectin-3, a marker for vacuole lysis by invasive pathogens
.
Cell Microbiol
2010
;
12
:
530
544
[PubMed]
33.
Vlassara
H
,
Palace
MR
.
Diabetes and advanced glycation endproducts
.
J Intern Med
2002
;
251
:
87
101
[PubMed]
34.
Tahara
N
,
Yamagishi
S
,
Takeuchi
M
, et al
.
Positive association between serum level of glyceraldehyde-derived advanced glycation end products and vascular inflammation evaluated by [(18)F]fluorodeoxyglucose positron emission tomography
.
Diabetes Care
2012
;
35
:
2618
2625
[PubMed]
35.
Shen
HM
,
Mizushima
N
.
At the end of the autophagic road: an emerging understanding of lysosomal functions in autophagy
.
Trends Biochem Sci
2014
;
39
:
61
71
[PubMed]
36.
Vallon
V
.
The proximal tubule in the pathophysiology of the diabetic kidney
.
Am J Physiol Regul Integr Comp Physiol
2011
;
300
:
R1009
R1022
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data