You are on page 1of 11

Acta Diabetologica

https://doi.org/10.1007/s00592-021-01736-4

ORIGINAL ARTICLE

Klotho protects against diabetic kidney disease via AMPK‑


and ERK‑mediated autophagy
Meng Xue1 · Feng Yang2 · Ying Le1 · Yanlin Yang2 · Bingsen Wang1 · Yijie Jia2 · Zongji Zheng2 · Yaoming Xue2

Received: 9 March 2021 / Accepted: 6 May 2021


© Springer-Verlag Italia S.r.l., part of Springer Nature 2021

Abstract
Background Diabetic kidney disease (DKD) is a serious complication of diabetes mellitus and results in serious public
health problems. Although a great number of studies have been performed to elucidate the mechanisms of this disease, these
mechanisms remain largely unknown.
Methods Cell and animal models were first constructed using human renal proximal tubule cells stimulated by high glucose
(HG) and mice induced by streptozotocin (STZ). After Klotho overexpression, Klotho expression was assessed by RT-PCR
and western blot, immunofluorescence; autophagy and AMPK/ERK proteins were confirmed using western blot or immu-
nohistochemical assay; the autophagosomes were observed by transmission electron microscope; the pathological structure,
fibrosis, polysaccharides and glycogen of kidney were evaluated by H&E staining, Masson staining and PAS staining.
Results We first confirmed that Klotho expression and autophagic activity were reduced in DM mice and HG-induced
human renal proximal tubule cells. Besides, overexpression of Klotho could significantly enhance autophagy and AMPK
and ERK1/2 activities in vivo and in vitro, which also could be abolished by selective AMPK inhibitor and ERK activator.
Moreover, we proved that Klotho could inhibit hyperglycemia-induced renal tubular damage.
Conclusion In summary, our results proved that Klotho improved renal tubular cell autophagy via the AMPK and ERK path-
ways and played a role in renal protection. These findings provide new insight into the mechanism of Klotho and autophagy
in DKD.

Keywords Diabetic kidney disease · Klotho · Autophagy · AMPK · ERK

Introduction pathophysiology of DKD occurrence and development is


largely unknown, standard therapies can slow DKD progres-
Diabetic kidney disease (DKD) is a microvascular compli- sion but cannot arrest or reverse it. Therefore, studies to
cation that develops in approximately 30–40% of patients elucidate the pathogenesis of DKD are crucial in developing
with type 1 or 2 diabetes mellitus (DM) [1]. With increas- new targeted treatments for the disease.
ing prevalence and a lack of effective therapies, DKD has Autophagy is an evolutionarily conserved cellular self-
become the leading cause of end-stage renal disease and a consumption process and a crucial recycling system that
serious public health problem worldwide [2, 3]. Since the degrades aggregated proteins and dysfunctional organelles.
Cellular activity occurs at baseline levels in various cells to
Managed by Giuseppe Pugliese. preserve cytoplasmic homeostasis and plays an important
role in numerous physiological activities, including apopto-
* Yaoming Xue sis, necrosis, nutrient metabolism, development, immunity
yaomingxue@126.com and aging [4]. On the other hand, autophagy dysregulation
1
Department of Endocrinology and Metabolism, The Second contributes to the pathogenesis of a variety of diseases, such
Clinical Medical College, Shenzhen People’s HospitalJinan as cancers, infectious and inflammatory diseases, metabolic
UniversityThe First Affiliated Hospital, Southern University diseases, cardiovascular diseases and kidney diseases [5]. In
of Science and Technology), Shenzhen 518020, China the context of both type 1 and type 2 diabetes, autophagy is
2
Department of Endocrinology and Metabolism, Nanfang attenuated in tubular cells, which is associated with hyper-
Hospital, Southern Medical University, Guangzhou 510515, trophy [6]. In studies with streptozotocin (STZ)-induced
China

13
Vol.:(0123456789)
Acta Diabetologica

diabetic mice, Wistar fatty rats with type 2 diabetes and by the Institutional Animal Care and Use Committee of the
kidney biopsies from type 2 diabetic patients, consistent Laboratory Animal Center, Nanfang Hospital, Southern
autophagy impairment was observed [5]. Medical University, Guangzhou, China (certificate number:
It is well known that autophagy is regulated by signaling SYXK2015-0056). One week before the experiment, all
pathways that sense nutrients [7, 8]. It has been suggested mice were housed in a specific pathogen-free environment
that dysregulation of these nutrient sensing pathways is with controlled temperature (20–26 °C), humidity (30%-
associated with the pathogenesis of DKD [9]. As a mas- 70%) and light (12-h light/dark cycle).
ter regulator of cellular metabolism that integrates diverse
pathways associated with energy and metabolism, the evo- Animal groups
lutionarily conserved heterotrimeric protein complex AMP-
activated protein kinase (AMPK) is activated under condi- The mice were randomized into two treatment groups: con-
tions of nutrient/energy depletion [10]. Therefore, alterations trol and high-fat diet (HFD)/STZ. The mice had free access
in the AMPK signaling pathway under diabetic conditions to water and were continuously fed either a normal chow
may inhibit autophagy, resulting in DKD and subsequently diet or an HFD containing 60% fat. After 4 weeks of an HFD
exacerbating renal injury [11]. As a critical member of the to induce obesity and a prediabetic status, HFD/STZ mice
mitogen-activated protein kinase (MAPK) family, extracel- were intraperitoneally administered 35 mg/kg STZ. Concur-
lular signal-regulated kinase (ERK) is well recognized for rently, control mice received the sodium citrate vehicle. One
its regulatory role in cell proliferation, survival and differ- week after STZ treatment, blood glucose >16.7 mmol/L was
entiation [12]. Recently, ERK signaling pathways have been defined as successful modeling. Then, each treatment group
shown to play important roles in energy homeostasis, such was randomly subdivided into 3 groups (n = 6/group): the
as regulating glycolysis and adipogenesis. Moreover, there 1-week, 6-week and 18-week groups. The mice were eutha-
is increasing evidence that the ERK pathway is an important nized at 1, 3 and 18 weeks after STZ or vehicle administra-
contributor to type 2 diabetes and the subsequent complica- tion. pCMV and pCMV-Klotho plasmids were purchased
tion of diabetic cardiomyopathy [13]. from Qiagen. Klotho overexpression was induced by inject-
Klotho was originally discovered as an antiaging factor ing the pCMV-Klotho plasmid via the tail vein in the HFD/
[6] and is primarily expressed in kidneys, particularly in STZ group, and pCMV was injected as a control. After an
renal distal tubular cells [14]. This protein acts in an endo- additional 18 weeks of HFD feeding, the mice were eutha-
crine/paracrine manner and plays multiple roles in oxidation nized, and the kidneys were harvested. During the experi-
suppression, lifespan extension, insulin sensitivity regula- ment, the health and certain behaviors of the mice were
tion and stem cell preservation [15]. Klotho expression has monitored daily, and a 20% loss of body weight was con-
been shown to be significantly decreased in renal tubules of sidered to be the humane end point for euthanasia. Then,
both humans and animals with DKD [12]. Additionally, a the mice were put into the euthanasia chamber containing
decrease in plasma Klotho was suggested to be indicative ­CO2 at a rate of 20% chamber volume/min. When the mice
of DKD progression in type 2 diabetic patients [16]. These lost consciousness and stopped breathing, the CO2 flow
findings have led to increasing interest in the role of Klotho continued for 1 min. The mice were then killed by cervical
in DKD. dislocation [17].
In the present study, we investigated the role of Klotho
in DKD and further explored the underlying mechanism. Cell culture and transfection
For the first time, we noted a close relationship between
Klotho and autophagy in diabetic nephropathy. Specifically, Human renal proximal tubule cells (Neuromics, Cat. No.
an increase in Klotho expression promoted autophagy via SC00A1-PTEC) were obtained from an agent (Biopike,
AMPK activation and ERK pathway inhibition. Our find- Beijing, China) and maintained in DMEM/F12 (Gibco, cat.
ings reveal new insight into the mechanism of Klotho and no. 11330–032) with 10% fetal bovine serum (FBS; Gibco).
autophagy in DKD. Human renal proximal tubule cells were seeded in 12-well
plates (5 × ­104 cells/well). Following overnight incubation,
the cells were exposed to normal (5 mM) or high (30 mM)
Materials and methods concentrations of glucose for 24 h. Some of the cells were
harvested and stored at -80 °C until analysis.
Animal experiment
Real‑time PCR (RT‑PCR)
Male C57BL/6 J mice aged 7 weeks were purchased from
the Guangdong Medical Laboratory Animal Center. All Total RNA was extracted from human renal proximal tubule
experimental animal procedures in this study were approved cells using TRIzol reagent (Invitrogen, CA, USA), and the

13
Acta Diabetologica

quality was examined by a NanoDrop ND-1000 spectropho- temperature for 1 h. After being incubated with secondary
tometer. Then, cDNA was generated using 1 μg of RNA antibodies (Beyotime) at room temperature for 30 min, the
with a reverse transcription kit (Takara, Dalian, China). RT- sections were exposed to DAB and observed under a light
PCR was performed with a Roche 480 Thermal Cycler using microscope (× 200, Olympus Corporation, Tokyo, Japan).
a SYBR Green qPCR kit (Takara). The following primer The positive cell rate and degree of immunohistochemical
sequences were used: human Klotho: forward 5’-GAA​ staining were assessed by two pathologists. The positive
AAA​TGG​CTT​CCC​TCC​TT-3’ and reverse 5’-ACA​ACT​ cells are brown-yellow in color or granular and brown. The
CCC​CAA​GCA​AAG​TC; murine Klotho forward 5’-TTG​ score was calculated based on the percentage of positive
GGT​CAC​TGG​GTC​AAT​CTC-3’ and reverse 5’-CCG​GCA​ cells and the staining intensity. A score of 0–3 was consid-
CGA​TAG​GTC​ATG​TT-3’; human β-actin: forward 5’-GCA​ ered negative, and a score of > 3 was considered positive.
AGT​GCT​TCT​AGG​CGG​AC-3’ and reverse 5’-AAG​AAA​
GGG​TGT​AAA​ACG​CAGC-3’; and murine β-actin: forward Transmission electron microscope (TEM)
5’-GGC​TGT​ATT​CCC​CTC​CAT​CG-3’ and reverse 5’-CCA​
GTT​GGT​AAC​AAT​GCC​ATGT-3’. The relative mRNA lev- The ultrastructure of human renal proximal tubule cells
els were calculated by the ­2−ΔΔCt method. was observed with TEM to assess autophagy. After treat-
ment, the cells were harvested and fixed in 0.1 M cacodylate
Western blotting buffer containing 2.5% glutaraldehyde (Sigma) and post-
fixed in 1% osmium tetroxide. After treatment with 0.5%
Using RIPA lysis buffer (Beyotime, Nantong, China), the tannic acid and 1% sodium sulfate, the cells were cleared in
collected cells were lysed, and the kidneys were homog- 2-hydroxypropyl methacrylate, embedded in Ultracut (Leica,
enized for protein extraction. After determining the protein Wetzlar, Germany), sliced into sections with a thickness of
concentrations using a BCA assay (Beyotime), the proteins 60 nm and stained with uranyl acetate and lead citrate. The
were denatured at 95 °C. Equal amounts of protein were ultrathin sections were then examined using a JEM-1230
loaded into 10% SDS-PAGE gels, electrophoresed, trans- TEM (JEOL, Tokyo, Japan).
ferred to PVDF membranes, blocked with 5% nonfat milk
and then incubated with primary antibodies against Klotho, Immunofluorescence analysis
GAPDH, LC3B, AMPK, p-AMPK, ERK1/2 and p-ERK1/2
(Abcam, Cambridge, UK) at 4 °C overnight. After being After treatment, the cells were fixed in 4% PFA for 15 min
washed with 0.2% TBST, the membranes were incubated at room temperature and blocked with 5% BSA for 30 min
with peroxidase-conjugated secondary antibody at room at room temperature. The cells were incubated with anti-
temperature for 1 h. The images were acquired using an Klotho antibodies at 4 °C overnight. Subsequently, the cells
Odyssey infrared imaging system. Gel-Pro Analyzer soft- were incubated with secondary antibodies for 1 h. The cells
ware was used for densitometric analysis. The immunore- were counterstained with DAPI (5 μg/ml; Sigma-Aldrich)
activity was visualized with enhanced chemiluminescence for 5 min and observed under a laser confocal microscope
and calculated with ImageJ software (NIH). (Leica, Germany).

Histology and immunohistochemical staining Statistical analysis

The collected kidneys were fixed in 10% paraformaldehyde All results are presented as the mean ± SD. A two-tailed Stu-
(PFA). After being dehydrated with increasing concentra- dent’s t test was used for comparisons between two inde-
tions of ethanol and hyalinized with xylene, the tissues were pendent groups. One-way ANOVA was used to compare
embedded in paraffin and cut into serial sections at a thick- three or more independent groups. P values less than 0.05
ness of 5 μm. Then, the sections were dewaxed in xylene were considered significant.
and rehydrated in graded ethanol solutions. Some sections
were stained with hematoxylin and eosin (H&E) for histo-
pathological examination. Some sections underwent Mas- Results
son’s trichrome and periodic acid-Schiff (PAS) staining for
morphological assessments. Some sections were subjected Expression of Klotho in the kidneys of DM mice
to immunohistochemistry (IHC). Briefly, the sections were
heated in a citrate solution for antigen retrieval and blocked To confirm the role of Klotho in the pathogenesis of DKD,
with 2% bovine serum albumin (BSA) at room temperature we first evaluated Klotho expression in kidney samples from
for 30 min. Then, the sections were incubated with primary DM mice. As shown in Fig. 1, Klotho expression was sig-
antibodies against LC3B (Abcam, Cambridge, UK) at room nificantly decreased at the transcriptional and translational

13
Acta Diabetologica

Fig. 1  Klotho expression was decreased in the kidneys of DM mice. ware was used to quantify the relative protein expression. The data
(a) Levels of Klotho mRNA and (b) Klotho protein were assessed in are the means ± SEM. n ≥ 3 per group. *p < 0.05, **p < 0.01 and
healthy mice and DM mice treated for different times. ImageJ soft- ***p < 0.001 compared to the control group

levels in DM mice compared to control mice. Furthermore, conversion in the samples from mice after 1, 6 or 18 weeks
the protein levels decreased in a time-dependent manner of treatment. As shown in Fig. 2a, the LC3-II/LC3-I ratio
(Fig. 1b). These data indicated that the changes in Klotho was significantly higher in DM mice after 1 week of
expression might be associated with the development of treatment than in control mice. In contrast, the LC3-II/
DKD. LC3-I ratio was significantly decreased after both 6 and
18 weeks of STZ treatment. The samples from the mice
Autophagic activity in the kidneys of DM mice after 18 weeks of treatment were additionally analyzed
by IHC. As expected, we observed a consistent decrease
LC3 conversion is a well-accepted biomarker of in LC3 in the renal cortex in DM mice (Fig. 2b). These
autophagic activity. In the present study, we determined results indicated that autophagy declined in DM mice after
the levels of LC3 by western blotting and IHC. Western long-term treatment with STZ.
blotting was used to examine the LC3 expression and

Fig. 2  LC3 conversion was increased in the kidneys of DM mice. (a) LC3 in the kidney cortex of healthy and DM mice after 18 weeks.
Western blot analysis of LC3-II/I expression in the kidney cortex at Original magnification: × 400, scale bar = 100 μm. The data are the
different treatment times. ImageJ software was used to quantify the means ± SEM; n ≥ 3 per group; *p < 0.05; and **p < 0.01 compared to
relative protein expression. (b) Immunohistochemical staining of the control group

13
Acta Diabetologica

Klotho expression in human renal proximal tubule proximal tubule cells compared to vector-transfected human
cells after high‑glucose stimulation renal proximal tubule cells (Fig. 4b). Moreover, the p-AMPK
level was increased, but p-ERK expression was decreased in
Due to their high sensitivity to diabetes-associated metabolic human renal proximal tubule cells treated with high glucose
factors, renal tubules are important in the pathogenesis and after transfection with pcDNA3.1-Klotho compared to those
development of DKD [18]. Human renal proximal tubule of human cells treated with high glucose after transfection
cells were used in this study to investigate tubular expression with pcDNA3.1-Vecto (Fig. 4c). These data indicated that
of Klotho after glucose stimulation. As shown in Fig. 3a, under high-glucose conditions, decreased expression of
the mRNA levels of Klotho were significantly decreased in Klotho could contribute to the suppression of autophagic
human renal proximal tubule cells exposed to high glucose activity in human renal proximal tubule cells.
for 48 h. Consistently, western blotting and immunofluo-
rescence staining showed a decrease in the protein level of
Klotho (Fig. 3b–c). These data suggested a potential cor- Role of Klotho in autophagy and AMPK and ERK1/2
relation between aberrant Klotho levels and tubular changes. activation in diabetic mouse kidneys

Effects of Klotho on autophagy and AMPK To further confirm the role of Klotho in autophagy and
and ERK1/2 activation in high‑glucose‑treated AMPK and ERK1/2 activation, we examined the kidneys of
human renal proximal tubule cells diabetic mice. After intravenously injecting pCMV-Klotho
into the mice, the renal levels of LC3-II/LC3-I were signifi-
To investigate the role of Klotho in tubular autophagy, cantly increased (Fig. 5a). Consistently, immunohistochemi-
Klotho was overexpressed in human renal proximal tubule cal analysis showed an increase in LC3 in the kidneys of
cells by pcDNA3.1 Klotho transfection. The protein levels of these mice (Fig. 5b). These results indicated that the level
LC3-1 and II were examined, and we observed that Klotho of autophagy was significantly increased after Klotho over-
overexpression significantly enhanced LC3-II conversion expression. Additionally, western blot analysis showed that
in cells after 48 and 72 h compared to that of control cells AMPK phosphorylation was increased, whereas ERK phos-
(Fig. 4a). Additionally, we examined autophagy by TEM, phorylation was decreased in pCMV-Klotho-treated mice,
and the results indicated that the number of autophagosomes indicating that AMPK was activated, but ERK was inhibited
was notably increased in Klotho-overexpressing human renal (Fig. 5c).

Fig. 3  Klotho expression in


human renal proximal tubule
cells after high-glucose stimula-
tion. (a) Levels of Klotho
mRNA and (b) Klotho protein
were assessed in human renal
proximal tubule cells treated
with low glucose and high glu-
cose. (c) Morphological obser-
vations of Klotho expression
by confocal microscopy. Scale
bar: 20 μM. The data are the
means ± SEM. n ≥ 3 per group.
**p < 0.01 and ***p < 0.001
compared to the control group

13
Acta Diabetologica

Fig. 4  Klotho overexpression increased autophagy via the AMPK/ with damaged organelles and other cytoplasmic components. Origi-
ERK pathway in human renal proximal tubule cells stimulated with nal magnification: × 13,500. (c) p-AMPK, AMPK, p-ERK and ERK
high glucose. LC3 protein expression in human renal proximal tubule protein expression in human renal proximal tubule cells cultured in
cells transfected with the vector or Klotho and treated with high high-glucose conditions for 72 h after transfection with the vector or
glucose for 24 h, 48 h and 72 h. (b) Changes in autophagosomes in Klotho. The data are the means ± SEM. n ≥ 3 per group. *p < 0.05 and
human renal proximal tubule cells cultured in high glucose for 72 h **p < 0.01 compared to the control group
after transfection with Klotho. Red arrows indicate autophagosomes

Role of Klotho in renal injury in diabetic mice a thicker glomerular basement membrane and the accumu-
lation of extracellular matrix or mesangial matrix (Fig. 6).
DM mice were injected with pCMV-Klotho, and we col- Masson’s trichome staining was used to examine fibrosis.
lected kidneys to evaluate the association between pathologi- DM mice injected with pCMV-Klotho exhibited significant
cal changes in the kidney and Klotho. H&E staining showed decreases in fibers with only small collagen fibers in the
that DM mice with Klotho overexpression exhibited the kidney cortex. PAS staining showed significantly decreased
densest tissue arrangement without obvious damage, such as expression of polysaccharides and glycogen in the kidneys

13
Acta Diabetologica

Fig. 5  Klotho increased renal autophagy via AMPK activation and p-AMPK, AMPK, p-ERK and ERK protein expression in the kid-
ERK1/2 pathway inhibition in diabetic mice. (a) Western blot show- ney cortex after injection of the vector or Klotho. The data are the
ing LC3-I/II protein expression. (b) Immunohistochemical staining means ± SEM; n ≥ 3 per group; **p < 0.01 and ***p < 0.001 com-
of LC3 in the kidney cortex after intravenous injection of the vec- pared to the vector group
tor or Klotho. Original magnification: × 400, scale bar = 100 μm. (c)

of DM mice injected with pCMV-Klotho. These results indi- comparable between cells with or without Klotho over-
cated that Klotho overexpression suppressed renal tubular expression. However, AMPK phosphorylation was sig-
damage in diabetic mice. nificantly increased, while ERK1/2 phosphorylation was
inhibited. After curcumin and compound C treatment, the
Role of AMPK and ERK1/2 in the effect of Klotho changes in the phosphorylation of AMPK and ERK1/2
on human renal proximal tubule cell autophagy were restored. LC3-I and LC3-II expression was exam-
after high‑glucose stimulation ined and showed that the increased conversion of LC3 was
abolished by curcumin or compound C. These results indi-
To examine whether AMPK and ERK1/2 were involved cated that in tubule cells exposed to high glucose, Klotho
in Klotho-mediated regulation of autophagy in human regulated AMPK and ERK1/2 phosphorylation, which in
renal proximal tubule cells, we first determined the lev- turn played a critical role in Klotho-mediated regulation
els of these factors in the cells transfected with Klotho. of autophagy in these cells.
As shown in Fig. 7, the AMPK and ERK1/2 levels were

13
Acta Diabetologica

Fig. 6  Klotho protected against DKD in mice. Photomicrographs deposition (Masson’s trichome staining) and glycogen or polysaccha-
of kidney sections from the vector and Klotho groups stained with ride (periodic acid-Schiff (PAS) staining) expression. (Original mag-
hematoxylin and eosin (H&E), showing tubulointerstitial collagen nification: × 400)

Fig. 7  AMPK and ERK1/2 regulated autophagy in human renal expression of AMPK, p-AMPK, ERK and p-ERK in cells was inves-
proximal tubule cells simulated with high glucose. (a) After transfec- tigated. (b, c and d) ImageJ software was used to quantify the relative
tion with Klotho and culture under high-glucose conditions, human protein expression. The data are the means ± SEM. n ≥ 3 per group.
renal proximal tubule cells were further treated without or with **p < 0.01: compared to the control group. &&p < 0.01: compared to
compound C (AMPK inhibitor) or curcumin (ERK inhibitor). The the Klotho group

Discussion [20]. Therefore, it is important to understand the potential


pathological mechanism of DKD for the development of
With the recent increase in diabetes cases, DKD has novel therapeutic strategies.
become an extreme threat to human life [19]. Despite An animal model of DM is the main method for study-
dramatic developments in renoprotective drugs, DKD ing the mechanism of disease and drug therapy. Studies
treatment remains a formidable challenge for clinicians have shown that HFD can lead to insulin resistance and DM
induction, and low-dose STZ treatment can cause moderate

13
Acta Diabetologica

impairments in β-cell function and incomplete insulin expression could improve autophagic clearance and attenu-
secretion, which leads to impaired glucose metabolism. It ate cell injury, ultimately serving as a protective factor [29,
has been reported that HFD combined with low-dose STZ 30]. In DKD, Klotho is aberrantly expressed and was sug-
induces insulin resistance and elevates blood lipids in mice. gested to participate in this disease [31]. A clinical study
The pathogenesis and progression of DM in mice induced by also explored the potential association of Klotho with DKD
an HFD and STZ were similar to those of human DM. This and noted that Klotho was negatively associated with DKD
animal model can simulate the disease process in human DM [32]. In STZ-induced diabetic mice, a study showed that
patients and has been used in research to study the mecha- Klotho deficiency exacerbated early DKD [33]. Another
nism of DM and develop drugs to treat DM. Therefore, the study also indicated that enhanced serum Klotho levels
DM mouse model was induced by an HFD combined with might be a mechanism by which atrasentan inhibited renal
low-dose STZ in this study. Previous studies also revealed tubular injury in DKD [34]. Moreover, it was suggested that
that the level of Klotho was significantly decreased in DKD autophagy is the convergent mechanism of Klotho pathways
patient serum; thus, Klotho was identified as a potential bio- and is central to DKD [35]. In this study, we observed a
marker of DKD [21, 22]. Our results also confirmed that protective effect of Klotho on renal tubular cells and against
Klotho was frequently diminished in the kidney cortex in DKD. Furthermore, our data are the first to demonstrate that
DM mice, and high glucose also suppressed the expression the induction of autophagy by Klotho critically contributes
of Klotho in human renal proximal tubule cells. to renoprotection.
DKD is a complex disease that is associated with ultras- Multiple pathways are involved in the biological function
tructural changes in all compartments in the kidney. These of Klotho. Using a selective inhibitor or activator, we noted
changes are characterized by basement membrane thicken- that Klotho markedly induced the activation of AMPK and
ing, glomerular and tubular hypertrophy, mesangial expan- the inactivation of ERK, ultimately regulating autophagic
sion, glomerulosclerosis and tubulointerstitial fibrosis [23]. activity and DKD. In some types of tumor cells, Klotho
Although glomerular changes have attracted the most atten- overexpression or treatment with soluble protein consist-
tion, renal tubules, an energy-consuming structure suscep- ently increased AMPK phosphorylation [36]. In the aortas
tible to various metabolic and hemodynamic fluctuations, of Klotho-expressing mice, AMPK activity was also reduced
is damaged before the occurrence of glomerulopathy, sug- [37]. In an additional study with db/db mice, Klotho ame-
gesting that tubular injury critically contributes to the patho- liorated DKD via AMPK [38]. The role of Klotho in regu-
genesis and development of DKD [24, 25]. In the present lating ERK signaling has been reported to vary according
study, we used tubular cells to investigate the underlying to the pathways. When ERK phosphorylation was induced
mechanism by which high glucose induces DKD. Our data by TGFβ1, AngII and phosphate, Klotho had no inhibitory
showed that a critical contributor to high glucose-induced effects. However, Klotho inhibited ERK activity via FGF2
DKD might be a decrease in autophagy, which is consistent signaling [39]. These results raised the question of which
with previous studies [26]. With the highest mitochondrial pathway Klotho regulates ERK phosphorylation through in
content in the kidney, renal tubular cells are particularly sen- DKD.
sitive to mitochondrial dysfunction, which has been well In summary, our present study demonstrated the protec-
recognized as a pivotal factor that initiates DKD and is a tive role of Klotho in DKD. In addition, we uncovered a
therapeutic target. By degrading damaged organelles, includ- novel mechanism associated with the protective effect of
ing mitochondria, autophagy is essential for homeostasis Klotho against DKD and found that the control of Klotho-
and cell survival [27]. Defective autophagy may ultimately mediated autophagy might be a therapeutic strategy for
contribute to the accumulation of cellular damage, resulting DKD. Therefore, we suggest that the increase in Klotho is a
in the development of metabolic or age-related kidney dis- promising strategy for DKD treatment. However, there are
eases [5]. In addition to autophagy, reactive oxygen species limitations to current research. For example, models of both
(ROS) and apoptosis are also triggered by hyperglycemia type 1 and type 2 diabetes should be established to explore
and lead to DKD [28]. Autophagy is upstream of these two the function and mechanism of Klotho in DKD, and the
pathways and has been demonstrated to play an important phosphorylation of Klotho should also be further investi-
role in regulating their activities [5]. These findings further gated in DKD.
suggest that inducing autophagy is a promising strategy for
treating DKD. Acknowledgements This study was supported by the National Natural
Science Foundation of China (No. 81870570; No. 81900378).
The core autophagy machinery is regulated upstream
by a complex signaling network. As an antiaging protein, Author contributions MX, FY, YL, YY, BW and YJ performed the
Klotho has been reported to be associated with autophagy experiments and analyzed the data. MX and FY wrote the manuscript.
and a variety of diseases. For example, in Alzheimer’s dis- ZZ and YX revised the manuscript. All authors read and approved the
ease and acute kidney injury, the upregulation of Klotho final manuscript.

13
Acta Diabetologica

Availability of data and materials The datasets used during the present 16. Kim SS, Song SH, Kim IJ et al (2016) Decreased plasma alpha-
study are available from the corresponding author. Klotho predict progression of nephropathy with type 2 diabetic
patients. J Diab Complicat 30:887–892
17. Creamer-Hente MA, Lao FK, Dragos ZP et al (2018) Sex- and
Declarations Strain-related differences in the stress response of Mice to ­CO2
Euthanasia. J Am Assoc Lab Anim Sci 57:513–519
Conflict of interest The authors declare that they have no conflict of 18. Thomas MC, Burns WC, Cooper ME (2005) Tubular changes
interests. in early diabetic nephropathy. Adv Chronic Kidney Dis
12:177–186
Patient consent for publication Not applicable. 19. Cheng MF, Chen LJ, Cheng JT (2010) Decrease of Klotho in the
kidney of streptozotocin-induced diabetic rats. J Biomed Biotech-
Human and Animal Rights All procedures performed in studies involv- nol 2010:513853
ing animal participants were in accordance with the ethical standards 20. Jha V, Garcia-Garcia G, Iseki K et al (2013) Chronic kidney dis-
of the institutional or national research commitee and with the 1964 ease: global dimension and perspectives. Lancet 382:260–272
Heisinki declaration and its later amendments or comparable ethical 21. Lv M, Chen Z, Hu G et al (2015) Therapeutic strategies of dia-
standards. This article does not contain any human experiments. betic nephropathy: recent progress and future perspectives. Drug
Discov Today 20:332–346
Informed consent All animal studies and procedures were approved 22. Hu MC, Moe OW (2012) Klotho as a potential biomarker and
by the Institutional Animal Care and Use Committee of the Labora- therapy for acute kidney injury. Nat Rev Nephrol 8:423–429
tory Animal Center, Nanfang Hospital, Southern Medical University, 23. Magri CJ, Fava S (2009) The role of tubular injury in diabetic
Guangzhou, China (certificate number: SYXK2015-0056). nephropathy. Eur J Intern Med 20:551–555
24. Zhang YH, Zhang YQ, Guo CC et al (2020) Prostaglandin E1
attenuates high glucose-induced apoptosis in proximal renal tubu-
lar cells by inhibiting the JNK/Bim pathway. Acta Pharmacol Sin
41:561–571
References 25. Guo C, Li Y, Zhang R, Zhang Yet al (2018) Protective effect
of salidroside against diabetic kidney disease through inhibiting
1. Thomas MC, Brownlee M, Susztak K et al (2015) Diabetic kidney BIM-mediated apoptosis of proximal renal tubular cells in rats.
disease. Nat Rev Dis Primers 1:15018 Front Pharmacol 9:1433
2. Alsaad KO, Herzenberg AM (2007) Distinguishing diabetic 26. Bhatia D, Choi ME (2019) The emerging role of mitophagy in
nephropathy from other causes of glomerulosclerosis: an update. kidney diseases. J Life Sci (Westlake Village) 1:13–22
J Clin Pathol 60:18–26 27. Lionaki E, Markaki M, Palikaras K et al (2015) Mitochondria,
3. Zhang L, Long J, Jiang W et al (2016) Trends in chronic kidney autophagy and age-associated neurodegenerative diseases:
disease in China. N Engl J Med 375:905–906 New insights into a complex interplay. Biochim Biophys Acta
4. Chaabane W, User SD, El-Gazzah M et al (2013) Autophagy, 1847:1412–1423
apoptosis, mitoptosis and necrosis: interdependence between 28. Forbes JM, Thorburn DR (2018) Mitochondrial dysfunction in
those pathways and effects on cancer. Arch Immunol Ther Exp diabetic kidney disease. Nat Rev Nephrol 14:291–312
(Warsz) 61:43–58 29. Zeng CY, Yang TT, Zhou HJ et al (2019) Lentiviral vector-medi-
5. Yang D, Livingston MJ, Liu Z et al (2018) Autophagy in dia- ated overexpression of Klotho in the brain improves Alzheimer’s
betic kidney disease: regulation, pathological role and therapeutic disease-like pathology and cognitive deficits in mice. Neurobiol
potential. Cell Mol Life Sci 75:669–688 Aging 78:18–28
6. Barbosa Júnior Ade A, Zhou H, Hültenschmidt D et al (1992) 30. Chen K, Sun Z (2019) Autophagy plays a critical role in Klotho
Inhibition of cellular autophagy in proximal tubular cells of the gene deficiency-induced arterial stiffening and hypertension. J
kidney in streptozotocin-diabetic and uninephrectomized rats. Mol Med (Berl) 97:1615–1625
Virchows Archiv B, Cell Pathol Includ Molec Pathol 61:359–366 31. Yang YL, Xue M, Jia YJ et al (2020) Long noncoding RNA
7. He C, Klionsky DJ (2009) Regulation mechanisms and signaling NEAT1 is involved in the protective effect of Klotho on renal
pathways of autophagy. Annu Rev Genet 43:67–93 tubular epithelial cells in diabetic kidney disease through the
8. Kroemer G, Marino G, Levine B (2010) Autophagy and the inte- ERK1/2 signaling pathway. Exp Mol Med 52:266–280
grated stress response. Mol Cell 40:280–293 32. Navarro-Gonzalez JF, Sanchez-Nino MD, Donate-Correa J et al
9. Kume S, Koya D, Uzu T et al (2014) Role of nutrient-sensing sig- (2018) Effects of pentoxifylline on soluble klotho concentrations
nals in the pathogenesis of diabetic nephropathy. Biomed ResInt and renal tubular cell expression in diabetic kidney disease. Dia-
2014:315494 betes Care 41:1817–1820
10. Steinberg GR, Kemp BE (2009) AMPK in Health and Disease. 33. Lin Y, Kuro-o M, Sun Z (2013) Genetic deficiency of anti-aging
Physiol Rev 89:1025–1078 gene klotho exacerbates early nephropathy in STZ-induced dia-
11. Kume S, Thomas MC, Koya D (2012) Nutrient sensing, betes in male mice. Endocrinology 154:3855–3863
autophagy, and diabetic nephropathy. Diabetes 61:23–29 34. Kang WL, Xu GS (2016) Atrasentan increased the expression of
12. Guo YJ, Pan WW, Liu SB et al (2020) ERK/MAPK signalling klotho by mediating miR-199b-5p and prevented renal tubular
pathway and tumorigenesis. Exp Ther Med 19:1997–2007 injury in diabetic nephropathy. Sci Rep 6:19979
13. Xu Z, Sun J, Tong Q et al (2016) The Role of ERK1/2 in the 35. Guo J, Zheng HJ, Zhang W et al (2020) Accelerated Kidney Aging
Development of Diabetic Cardiomyopathy. Int J Mol Sci 17:2001 in Diabetes Mellitus. Oxid Med Cell Longev 2020:1234059
14. Kuro-o M (2009) Klotho and aging. Biochim Biophys Acta 36. Shmulevich R, Rubinstein T, Scherf T et al (2015) The tumor sup-
1790:1049–1058 pressor klotho: a master regulator of metabolism in breast cancer.
15. Wang Q, Ren D, Li Y et al (2019) Klotho attenuates diabetic Cancer Research 75:1184–1184
nephropathy in db/db mice and ameliorates high glucose-induced 37. Cheng XY, Li YY, Huang C et al (2017) AMP-activated protein
injury of human renal glomerular endothelial cells. Cell Cycle kinase reduces inflammatory responses and cellular senescence
18:696–707 in pulmonary emphysema. Oncotarget 8:22513–22523

13
Acta Diabetologica

38. Lee J, Tsogbadrakh B, Yang S et al (2020) Klotho ameliorates Publisher’s Note Springer Nature remains neutral with regard to
diabetic nephropathy via LKB1-AMPK-PGC1 alpha-mediated jurisdictional claims in published maps and institutional affiliations.
renal mitochondrial protection. Biochem Biophys Res Commun
534
39. Mencke R, Olauson H, Hillebrands JL (2017) Effects of Klotho on
fibrosis and cancer: a renal focus on mechanisms and therapeutic
strategies. Adv Drug Deliv Rev 121:85–100

13

You might also like