ABSTRACT
Experimental evidence demonstrated that macroautophagy/autophagy exerts a crucial role in maintain renal cellular homeostasis and represents a protective mechanism against renal injuries. Interestingly, it has been demonstrated that in the human proximal tubular renal cell line, HK-2, the MTOR inhibitor rapamycin enhanced autophagy and mitigated the apoptosis damage induced by urinary protein overload. However, the underlying molecular mechanism has not yet been elucidated. In our study we demonstrated, for the first time, that in HK-2 cells, the exposure to low doses of rapamycin transactivated the NGFR promoter, leading to autophagic activation. Indeed, we observed that in HK-2 cells silenced for the NGFR gene, the rapamycin-induced autophagic process was prevented, as the upregulation of the proautophagic markers, BECN1, as well as LC3-II, and the autophagic vacuoles evaluated by transmission electron microscopy, were not found. Concomitantly, using a series of deletion constructs of the NGFR promoter we found that the EGR1 transcription factor was responsible for the rapamycin-mediated transactivation of the NGFR promoter. Finally, our results provided evidence that the cotreatment with rapamycin plus albumin further enhanced autophagy via NGFR activation, reducing the proapoptotic events promoted by albumin alone. This effect was prevented in HK-2 cells silenced for the NGFR gene or pretreated with the MTOR activator, MHY1485. Taken together, our results describe a novel molecular mechanism by which rapamycin-induced autophagy, mitigates the tubular renal damage caused by proteinuria, suggesting that the use of low doses of rapamycin could represent a new therapeutic strategy to counteract the tubule-interstitial injury observed in patients affected by proteinuric nephropathies, avoiding the side effects of high doses of rapamycin.
KEYWORDS: autophagy, EGR1, MTOR, NGFR, proteinuric damage, rapamycin
Abbreviations
- BAF
bafilomycin A1
- BECN1
Beclin 1
- CQ
chloroquine
- EGR1
early growth response 1
- GAPDH
glyceraldehyde-3-phosphate dehydrogenase
- HDAC1
histone deacetylase 1
- LMNB
lamin B
- MAP1LC3B/LC3B
microtubule associated protein 1 light chain 3 beta
- MAPK1/ERK2
mitogen-activated protein kinase 1
- MAPK3/ERK1
mitogen-activated protein kinase 3
- MAPK/JNK
mitogen-activated protein kinase
- MHY1485/MH
4,6-Di-4-morpholinyl-N-(4-nitrophenyl)-1,3,5-triazin-2-amine
- MTOR
mechanistic target of rapamycin kinase
- MTORC1
mechanistic target of rapamycin complex 1
- MTORC2
mechanistic target of rapamycin complex 2
- NGFR
nerve growth factor receptor
- NF1
neurofibromin 1
- NR3C1/GR
nuclear receptor subfamily 3 group C member 1
- PARP1
poly(ADP-ribose) polymerase 1
- PD98059
2-(2-Amino-3-methoxyphenyl)-4H-1-benzopyran-4-one
- POLR2A
RNA polymerase II subunit A
- R
rapamycin
- SP1
Sp1 transcription factor
- SQSTM1/p62
sequestosome 1
- TFAP2A/AP2α
transcription factor AP-2 alpha
Introduction
Macroautophagy/autophagy is an adaptive intracellular process in which portions of cytoplasm are sequestered into double-membrane compartments, termed phagophores, that mature into autophagosomes, and then delivered to lysosomes for bulk degradation into their primary components to maintain cellular homeostasis and energy production. In mammalian systems baseline autophagy occurs under normal conditions, but it can be further stimulated by starvation or by various pathologic conditions, including ischemic, toxic, immunological, and oxidative insults [1]. Growing evidence suggests that basal constitutive autophagy is required for the homeostasis, viability and physiological functions of renal cells [2]. Interestingly, experimental models of acute and chronic kidney diseases demonstrate that, particularly in proximal tubular cells, autophagy plays an important cytoprotective effect against nephrotoxic injuries [3–5]. On the other hand, accumulating evidence underlines the emerging role of dysregulated autophagy in many kidney diseases [4–6].
Among various pathways known to regulate autophagy in mammalian cells, the best understood is the one regulated by MTOR (mechanistic target of rapamycin kinase) [1]. MTOR exists in 2 complexes, MTORC1, inhibited by rapamycin, regulates a wide variety of intracellular signals, protein synthesis, cellular energy metabolism, cellular stress, lipid and mitochondrial biogenesis, while MTORC2, insensitive to rapamycin, regulates survival and cytoskeletal organization [7,8].
Recently, Liu et al. have demonstrated, by an in vitro and in vivo study, that in proximal tubular renal cells the further induction of autophagy by rapamycin treatment, strongly mitigates the apoptotic damage caused by exposure to urinary protein overload, suggesting that activating autophagy flux in proximal tubular renal cells could represent a new therapeutic strategy in the management of nephropathies with proteinuria [5].
In this study we aimed to investigate the molecular mechanism by which rapamycin-induced autophagy protects proximal tubular renal cells against proteinuria injury, focusing our attention on NGFR (nerve growth factor receptor), a transmembrane glycoprotein belonging to the tumor necrosis receptor superfamily, significantly involved in the activation of proapoptotic events in neuronal and non-neuronal cells and in proximal tubular renal cells [9]. However, in literature it has been reported that in cerebellar Purkinje neurons deprived of trophic factors, NGFR promotes Purkinje neuron autophagy and death [10]. Therefore, as we have demonstrated that the signaling of NGFR is involved in the pathophysiology of many renal diseases [9,11,12], we explored the potential role of NGFR in modulating autophagy in proximal tubular renal cells in basal conditions as well as upon exposure to high doses of human albumin.
Results
Rapamycin transcriptionally upregulates NGFR expression in HK-2 cells
As first step of our in vitro study we tested the effects of rapamycin exposure on NGFR levels. HK-2 cells were plated in culture medium and treated with increasing concentrations of rapamycin (from 0.1 to 18 ng/ml). mRNA (Figure 1A) and protein (Figure 1B) expression levels of NGFR were evaluated, after 24 h and 48 h of treatment, by real time PCR and western blotting analysis, respectively. As showed in Figure 1, the treatment with rapamycin significantly induced NGFR mRNA and protein levels at all investigated doses. The transcriptional activation of NGFR was confirmed by transfection assay, using a luciferase reporter plasmid containing the wild-type NGFR promoter region (from −900 to +100 base pairs). After 24 h, transfected cells were treated for 18 h as reported and then luciferase activity was measured. Results showed a significant rapamycin-induced transactivation of the NGFR promoter, starting from the lower doses (Figure 1C). These data provided evidence, for the first time, that in HK-2 cells, the rapamycin exposure, upregulated neurotrophin receptor NGFR expression in a transcriptional dependent-manner.
Figure 1.
Rapamycin induces NGFR activation. HK-2 cells were untreated (-) or treated with increasing doses of rapamycin (R ng/ml) as indicated. (A) NGFR mRNA content, evaluated by real time RT-PCR after 24 h of exposure to treatment. Each sample was normalized to its GAPDH mRNA content. *P<0.05 compared with untreated cells (-); (B) Equal amounts of total cellular extract treated for 48 h were analyzed for NGFR levels by western blotting. The immunoblots show a single representative of 3 separate experiments. GAPDH was used as a loading control. Numbers over the blots represent the average fold change vs untreated cells (-). (C) HK-2 cells, transiently transfected with the wild-type NGFR promoter, were untreated (-) or treated for 18 h with increasing doses of R and then luciferase activity was measured. Luciferase activity of untreated cells was set as one-fold induction, upon which treatments were calculated. *P<0.05 compared to untreated cells (-). Bars represent the means ± SD of 3 different experiments each performed in triplicate.
Rapamycin induces autophagy via NGFR in HK-2 cells
First, we tested the effects of chronic exposure (48 h) to increasing concentrations of rapamycin (from 0.1 to 18 ng/ml) on HK-2 cells viability, by a luminescent cell viability assay. We observed that rapamycin at doses from 0.1 to 1 ng/ml did not elicit growth inhibitory effects. On the contrary, a slight reduction of HK-2 cell viability was observed at the doses of 7 ng/ml and 18 ng/ml respect to untreated cells (Figure 2A left panel). Next, in order to clarify if the reduction of vitality was associated with apoptotic events, in HK-2 treated with 7 ng/ml of rapamycin for 48 h we evaluated the PARP1 cleavage by western blotting and the fragmentation of DNA by a DNA laddering assay. We observed that rapamycin treatment did not induce change in PARP1 cleavage or in the inter-nucleosome fragmentation profile of genomic DNA (Figure 2A central and right panel), which is a diagnostic hallmark of cells undergoing apoptosis, suggesting that the growth inhibition (at doses of 7 ng/ml) did not occur through the apoptotic process, but may involve the autophagic pathway. Analysis of protein content confirmed the activation of autophagy at all doses investigated and an enhanced autophagic flux at the last 2 concentrations as shown by the SQSTM1/p62 decrease (Figure 2B left panel). To better confirm the obtained results, we performed the same experiment also in podocytes treated with rapamycin and/or bafilomycin A1 (BAF; 25 nM). As expected, the activated autophagic flux, confirmed by upregulation of BECN1/Beclin 1 and LC3-II, (the autophagosome-associated and phosphatidylethanolamine [PE]-conjugated form of LC3-I), as well as by a decrease in SQSTM1, was reversed in the presence of the inhibitor BAF (Figure 2B right panel). Because the most common signaling by which rapamycin exerts its biological effects is mediated by MTOR inhibition, we evaluated the involvement of this pathway using the MTOR activator MHY1485 that potently inhibits autophagy by suppression of fusion between autophagosomes and lysosomes. Therefore, HK-2 cells were cotreated in complete medium with increasing doses of rapamycin with or without 2 µM MHY1485 and then, protein levels of MTOR, NGFR, SQSTM1, BECN1, as well as LC3B-I and LC3B-II were evaluated by western blotting analysis. Results reported in Figure 2C showed that all doses of rapamycin inhibited MTOR, concomitant with an upregulation of both NGFR and BECN1, and induced conversion of LC3-I to LC3-II (Figure 2C left panel). These effects were reversed in cells treated with rapamycin plus MHY1485, suggesting that the proautophagic action of rapamycin occurred through inhibition of MTOR signaling (Figure 2C right panel). In order to confirm the activated autophagic flux in HK-2 cells, the same experiment was performed in the presence of the autophagic inhibitor chloroquine (25 μM). Results showed similar effect like MHY1485 except for MTOR that persisted in the inhibited form and NGFR levels that were mitigated but not completely reversed after chloroquine exposure (Figure 2D). To clarify the involvement of NGFR in autophagy activation, HK-2 cells were transfected with NGFR RNAi for 48 h and then treated for 6 h with increasing doses of rapamycin. Results reported in Figure 2F, showed that in cells silenced for NGFR (Figure 2E), the mRNA (Figure 2F upper panel) and protein (Figure 2F bottom panel) induction of the proautophagic markers BECN1, as well as LC3-II was reversed, highlighting the crucial role of NGFR in mediating rapamycin-induced autophagy.
Figure 2.
Rapamycin triggers autophagy via NGFR. (A left panel) luminescent cell viability assay of HK-2 treated for 48 h with increasing doses of rapamycin (R ng/ml) as indicated. Luciferase activity of untreated cells was set as one-fold induction, upon which treatments were calculated. *P<0.05 compared untreated cells (-). Bars represent the means ± SD of 3 different experiments each performed in triplicate. (A central panel) Western blotting analysis of PARP1 cleavage normalized on GAPDH content in HK-2 treated with R (7 ng/ml) for 48 h. The immunoblots show a single representative of 3 separate experiments. Numbers over the blots represent the average fold change vs untreated cells (-). (A right panel) DNA laddering in HK-2 treated with R (7 ng/ml) for 48 h. (B) protein expression levels of BECN1, LC3B-I and LC3B-II, SQSTM1 and NGFR in HK-2 cells exposed to R for 6 h (left panel) and in podocytes exposed to R alone or pretreated for 2 h with 25 nM BAF before rapamycin addition (right panel). Numbers on top of the blots represent the average fold change vs untreated cells normalized for internal loading. Particularly, western blotting analysis of LC3-II was normalized respect to GAPDH. (C) Immunoblotting analysis of phosphorylated and total MTOR, NGFR, BECN1, as well as LC3B-I and LC3-II, from total extracts of HK-2 cells treated for 6 h with increasing doses of R or/and the MTOR activator MHY1485 2 μM (MH). GAPDH was used as loading control. Numbers on top of the blots represent the average fold change vs untreated cells (-) normalized for internal loading. Particularly, western blotting analysis of LC3-II was normalized respect to GAPDH. (D) Immunoblotting analysis of phosphorylated and total MTOR, NGFR, SQSTM1, BECN1, as well as LC3B-I and LC3-II from total extracts of HK2 cells untreated (-) or treated for 6 h with increasing doses of R or/and the autophagy inhibitor Chloroquine 25 μM (CQ) added 2 h before starting treatments. GAPDH was used as loading control. Numbers on top of the blots represent the average fold change vs untreated cells (-) normalized for internal loading. Particularly, western blotting analysis of LC3-II was normalized with respect to GAPDH. (E) Western Blotting of NGFR in HK-2 cells transfected for 48 h with siRNA targeting the human NGFR mRNA sequence or with a control siRNA. GAPDH was used as loading control. Numbers on top of the blots represent the average fold change vs untreated cells (-) normalized for internal loading. (F) Total mRNA and proteins from HK-2 transfected with scrambled siRNA and NGFR siRNA and treated as indicated. Equal amounts of extracts were analyzed for BECN1, as well as LC3B-I and LC3-II mRNA and protein levels by Real-time PCR and immunoblotting analysis. GAPDH was used as loading control. Bars represent the means ± SD of 3 separate experiments, each performed in triplicate *P<0.05 vs untreated cells (-); $ P<0.05 vs untreated cells (-). Numbers on top of the blots represent the average fold change vs untreated cells normalized for GAPDH. Particularly, western blotting analysis of LC3-II was normalized with respect to GAPDH.
Rapamycin transactivates the NGFR promoter via the EGR1 transcription factor
Because we observed a significant role of NGFR in mediating rapamycin-induced autophagy, we aimed to study the consensus sites contained in its promoter (Figure 3A) that could be involved in autophagy activation.
Figure 3.
Rapamycin enhances NGFR promoter activation via the EGR1 consensus site. (A) Schematic representation of the WT human NGFR and its deletion constructs used in this study. (B) HK-2 cells were transfected for 24 h with WT NGFR promoter (NGFR -900+100) and its deletion constructs (NGFR -164+100, NGFR -315+100, NGFR -41+100), treated for 18 h with R (7 ng/ml) and then luciferase activity was measured. Luciferase activity of untreated cells was set as one-fold induction, upon which treatments were calculated. *P<0.05 compared with untreated cells (-). Bars represent the means ± SD of 3 different experiments each performed in triplicate. (C) cytosolic and nuclear contents of EGR1, NR3C1, SP1, TFAP2A and NF1 in HK-2 cells treated for 6 h with R (7 ng/ml). (D) cytosol/nucleus translocation of EGR1 in HK-2 transfected with scrambled and NGFR si RNAi and then treated as indicated. (E) cytosol to nucleus translocation of EGR1 in HK-2 treated with R and/or MH for 6 h. GAPDH and LMNB were used as loading control. Numbers on top of the blots represent the average fold change vs untreated cells normalized for internal loading.
To identify the transcription factor responsible for NGFR promoter transactivation induced by rapamycin exposure, HK-2 cells were transiently transfected for 24 h with a luciferase reporter plasmid containing the wild-type NGFR promoter region (from −900 to +100 base pairs), and with its deletion constructs (NGFR −164 + 100, NGFR −315 +100, NGFR −41 +100), each one lacking multiple consensus sites, and then treated for 18 h with 7 ng/ml of rapamycin.
Luciferase activity measurements showed that the transactivation of the NGFR promoter induced by rapamycin was still maintained using all deletion constructs (Figure 3B), suggesting that the region between −41 +100 could include consensus sites binding a transcription factor involved in the transactivation of the NGFR promoter, leading, in turn, to the activation of the autophagic process.
Promoter analysis for transcription factor binding sites, performed using Alibaba software, identified within the NGFR −41 +100 construct, multiple consensus sites for several transcription factors, as EGR1, NR3C1/GR, SP1, TFAP2A/AP2α, NF1, that act as modulators of genetic transcription, regulating various cellular processes such as cell survival, differentiation, apoptosis, autophagy and so on. Thus, we explored, in HK-2 cells treated with 7 ng/ml of rapamycin for 6 h, the nuclear translocation of above mentioned transcription factors. Results obtained by western blotting analysis reported in Figure 3C, showed that rapamycin exposure induced the nuclear translocation only of EGR1. Concomitantly, we observed that this effect was mediated by NGFR, as EGR1 nuclear translocation was reversed in cells silenced for NGFR (Figure 3D). To better confirm the involvement of MTOR inhibition pathway in activation of rapamycin-induced autophagy, in the same experimental condition, we explored the EGR1 cytosol-nucleus shuttle in HK-2 cells treated with rapamycin plus MHY1485. As reported in Figure 3E, the persistent activation of MTOR triggered by MHY1485 blocked the EGR1 transcription factor in the cytoplasm compartment. Taken together these results underlined that rapamycin induced autophagy through MTOR inhibition and activating EGR1 nuclear translocation that, in turn, transactivated NGFR promoter.
Rapamycin enhanches autophagy in HK-2 cells exposed to albumin overload
In order to clarify the biological role of activated autophagy induced by rapamycin in our experimental model, HK-2 cells were exposed for 6 h to rapamycin with or without 8 mg/ml of human albumin to mime in vitro, the proteinuria. Firstly, we observed, by western blotting analysis, that the upregulation of NGFR was more evident in cells cotreated with albumin plus rapamycin with respect to cells treated with rapamycin alone (Figure 4A). Our results showed that the combined treatment with rapamycin plus albumin significantly upregulated the autophagic marker BECN1 and enhanced the conversion of LC3-I to its PE-conjugated form LC3-II. In our experimental condition, performed for 6 h, albumin alone did not promote autophagy (Figure 4B left panel), according to that reported by Liu et al [13], that detected it at longer time (8 h). Interestingly, these effects were reversed in cells in which the expression of NGFR was silenced (Figure 4B right panel), highlighting that NGFR, activated by rapamycin, triggered the autophagy process, protecting proximal tubular renal cells from proteinuria damage. Next, to further examine autophagic vacuoles we performed transmission electron microscopy (TEM). As reported in Figure 4C, numerous autophagic structures (black arrows) with partially degraded material have been recognized in rapamycin-treated cells (Figure 4C-ii upper panel). Rapamycin-induced autophagy was not accompanied by destructive cytoplasmic changes, while silencing of NGFR drastically blocked rapamycin-induced autophagy (Figure 4C-iv upper panel). In albumin-treated cells, small lipid droplets (LDs) accumulated and became dispersed within the cytoplasm (Fig 4C-i bottom panel). Induction of autophagy has been demonstrated upon treatment of the cells with albumin plus rapamycin (Figure 4C-ii bottom panel). Autophagic vacuoles (black arrows) contained dense, amorphous materials and membranous myelin-like whorls. Interestingly, silencing of NGFR inhibited autophagy although did not attenuate lipid accumulation (Figure 4C-iii e -iv bottom panel).
Figure 4.
Rapamycin-induced autophagy protects HK-2 cells from albumin damage. (A) Protein levels of NGFR in HK-2 treated with R alone or with albumin (A) (8 mg/ml) for 6 h. Numbers on top of the blots represent the average fold change vs untreated cells normalized for internal loading. (B) Protein levels of BECN1, as well as LC3B-I and LC3B-II in HK-2 cells transfected with scrambled and NGFR siRNA for 48 h and then treated as indicated. Numbers above the blots represent the average fold change vs untreated cells normalized for internal loading. (C upper panel) Representative TEM images showing the effect of NGFR silencing on rapamycin-induced autophagy. (i) Control cells; (ii) Numerous autophagic structures (black arrows) with partially degraded material can be recognized in rapamycin-treated cells. Rapamycin-induced autophagy is not accompanied by destructive cytoplasmic changes; (iii) Control cells treated with NGFR siRNA; (iv) Silencing of NGFR drastically blocks rapamycin-induced autophagy in cultured renal cells. Scale bars: 2 µm (i, iii, iv), 5 µm (ii). (C bottom panel) Ultrastructural analysis showing the effect of NGFR silencing on A and R cotreated cells. In albumin-treated cells, small lipid droplets (LDs) accumulate and become dispersed within the cytoplasm. Characteristically, LDs appear as round to oval shaped structures of moderate electron density; (ii) Induction of autophagy was demonstrated upon treatment of the cells with albumin plus rapamycin. Autophagic vacuoles (black arrows) contain dense, amorphous materials and membranous myelin-like whorls; (iii) siRNA and albumin-treated cells; cells treated with NGFR siRNA and albumin; (iv) In cells cotreated with A plus R, silencing of NGFR inhibits autophagy but does not attenuate lipid accumulation. Scale bars: 5 µm (i, iii, iv), 2 µm (ii). (D) Phosphorylated and total levels of MAPK1/3 in HK-2 cells treated with R alone or in combination with A for 4 h (left panel) and in HK-2 cells pretreated for one h with 10 μM PD98059 and then treated as indicated above (right panel). Numbers on top of the blots represent the average fold change vs untreated cells normalized for GAPDH. (E) EGR1 cytosol-nucleus translocation in HK-2 cells treated with R alone or in combination with A for 4 h (left panel) and in HK-2 cells pretreated for one h with 10 μM PD98059 and then treated as indicated above (right panel). GAPDH and LMNB were used as internal loading controls. Numbers on top of the blots represent the average fold change vs untreated cells normalized to internal loading controls. (F) Cell viability assay of HK-2 cells treated for 6 h as indicated. Cell proliferation is expressed as fold change with respect to untreated cells (-). *P<0.05.
Next, we explored which NGFR downstream pathways was involved in the autophagic process rapamycin-induced. Our results, obtained by of western blotting, revealed that the enhanced phosphorylation of MAPK1/ERK2-MAPK3/ERK1 induced by rapamycin, was strengthened in the presence of albumin overload, concomitantly with a significant EGR1 translocation in the nuclear compartment (Figure 4D and Figure 4E left panel). These effects were reversed in cells pretreated for 30 min with PD98059 (10 μM), a specific inhibitors of MAPK pathway (Figure 4D and Figure 4E right panel). Conversely, the pathway of MAPK/JNK was not activated in the same experimental condition (data not shown). Data obtained were in agreement with a luminescent cell viability assay (Figure 4F) that showed a reduction of cell death in HK-2 cells cotreated for 6 h with rapamycin plus albumin respect to cells treated with albumin alone, suggesting that NGFR via MAPK1/3 phosphorylates EGR1 that migrates to the nucleus, exerting its proautophagic effects.
The recruitment of EGR1 at the EGR1 site on the NGFR promoter region regulates rapamycin-induced autophagy in HK-2 cells
Results reported in Figure 3 suggested the crucial role of EGR1 transcription factor on activation of autophagy via NGFR. These data prompted us to investigate the molecular mechanism by which rapamycin induced nuclear EGR1 translocation, transactivating the NGFR promoter. Therefore, HK-2 cells were transfected with WT (NGFR −900 +100) and its shorter deletion (NGFR −41+100) constructs and then treated with rapamycin and albumin alone or in combination for 18 h. Luciferase activity of both constructs showed that the rapamycin-induced transactivation of NGFR was potentiated in cells cotreated as reported above, while no significant effect was observed upon albumin alone (Figure 5A).
Figure 5.
Rapamycin exposure enhances EGR1 recruitment to the NGFR promoter. (A) HK-2 cells were transiently transfected for 24 h with NGFR -900+100 and NGFR −41 +100 constructs and then subjected to treatments with R and A alone or in combination for 18 h. Luciferase activity was measured. Results represent the mean ± s.d. of 3 different experiments each performed in triplicate. *P < 0.05 compared with untreated cells (-); **P < 0.05 compared with cells treated with R. (B) HK-2 cells were treated with R and A alone or in combination for 6 h, then crosslinked with formaldehyde and lysed. The precleared chromatin was immunoprecipitated with anti-EGR1, anti-POLR2A and anti-HDAC1 antibodies. A 5 µl volume of extracted DNA was analyzed by real-time PCR using specific primers to amplify the NGFR promoter sequence, including the EGR1 site. Similar results were obtained in 3 independent experiments. *P < 0.01 compared with vehicle (-). **P < 0.05 compared with cells treated with R. (C) Promoter activity in HK-2 cells transfected for 24 h with the shorter construct (NGFR −41+100) and then treated with R, A alone or in combination, with or without mithramycin A for 18 h. Luciferase activity was measured. Results represent the mean ± s.d. of 3 different experiments each performed in triplicate. *P < 0.05 compared with untreated cells (-), $ P < 0.05 compared with cells treated with R. **P < 0.05 compared with cells treated with R, $$ P < 0.05 compared with cells treated with R plus A. (D) real time PCR from HK-2 cells that were treated for 6 h with R, A and mithramycin A alone or in combination and then subjected to ChIP assay as in B. The precleared chromatin was immunoprecipitated with anti-EGR1 and anti-POLR2A, antibodies. Similar results were obtained in 3 independent experiments. *P < 0.05 compared with untreated cells (-), $ P < 0.05 compared with cells treated with R. **P < 0.05 compared with cells treated with R, $$ P < 0.05 compared with cells treated with R plus A. (E) Site-directed mutagenesis of the EGR1 site in the NGFR promoter of HK-2 cells transfected with NGFR −41 +100 or NGFR −41 +100 (NGFREGR1∆) constructs as described in A. The luciferase activities were normalized to the internal control and values of untreated cells (-) were set as one-fold induction upon which the activity induced by treatment was calculated. The values represent the mean ± SD of 3 separate experiments. *P < 0.05 compared with untreated cells (-), $ P < 0.05 compared with cells treated with R. **P < 0.05 compared with cells treated with R, $$ P < 0.05 compared with cells treated with R plus A. (F) protein expression of LC3B-I and LC3B-II in HK-2 treated as indicated in C. GAPDH was used as internal loading. Numbers on top of the blots represent the average fold change vs untreated cells (-) normalized for internal loading. Particularly, western blotting analysis of LC3-II was normalized with respect to GAPDH.
The functional role of the EGR1 motif, located in the NGFR promoter, in mediating autophagy was investigated by chromatin immunoprecipitation assay (ChIP). After 6 h of treatment the protein-chromatin complexes were immunoprecipitated using antibodies against EGR1 and the POLR2A subunit of RNA Polymerase II. Results indicated that EGR1 was constitutively bound on the EGR1 site of the NGFR promoter and that its recruitment was increased after rapamycin exposure and to a higher extent, in the presence of rapamycin plus albumin. The same result was obtained using an antibody against POLR2A, highlighting that the cotreatment increased transcription of the NGFR promoter that, in turn, activated the autophagy process (Figure 5B). In accordance with these data, the potentiated transcription of POLR2A was substantiated by re-ChIP assay that showed a reduced recruitment of the chromatin silencer HDAC1 on the EGR1 motif in HK-2 cells treated with rapamycin alone or with albumin (Figure 5B). The crucial role of EGR1 on NGFR promoter transactivation was better demonstrated in HK-2 cells transfected with the construct NGFR −41 +100 and treated with 100 nM mithramycin A, a GC-rich inhibitor that displaces EGR1 binding to DNA. As reported in Figure 5C, exposure to mithramycin A reduced the transactivation of NGFR induced by rapamycin alone or with albumin, concomitantly with a decrease of EGR1 and POLR2A recruitment on the EGR1 site (Figure 5D).
Finally, the crucial role of EGR1 was better assessed by site-directed mutagenesis assay on the EGR1 site. We found that the mutated EGR1 motif abrogated the transactivation of the NGFR −41+100 construct, induced by rapamycin alone or with albumin (Figure 5E upper panel). In vitro data of Chen et al. have provided the first evidence that in chronic obstructive pulmonary disease, the activation of the E2F4 and EGR1 transcription factors by HDAC inhibition induces the expression of LC3B [14]. Therefore, we evaluated the protein levels of LC3B-I and LC3B-II in HK-2 cells treated as indicated above. Results reported in Figure 5F showed that the rapamycin-induced upregulation of LC3-II was enhanced in the presence of albumin overload and that LC3B-II levels were decreased in HK-2 cells treated with mithramycin A.
The chronic exposure to rapamycin protects HK-2 cells from apoptosis via MTOR inhibition
To explore whether the chronic exposure to rapamycin could activate apoptosis, HK-2 cells were treated with rapamycin with or without albumin for 48 h and then PARP1 cleavage was evaluated by western blotting. As evident in Figure 6A, the exposure to rapamycin reduced the apoptosis induced by human albumin alone, concomitant with a persistent autophagy activation. This data fit well with cell viability results (Figure 6B) that showed that the chronic activation of MTOR signaling, mimed by MHY1485 exposure, abrogated the protective effects induced by rapamycin in HK-2 cells exposed to overload of human albumin. These results confirmed the importance of MTOR inhibition in protecting HK-2 cells through autophagy activation.
Figure 6.
Autophagy protects tubular renal cells from albumin. (A) western blotting analysis of PARP1, BECN1, as well as LC3B-I and LC3B-II modulation normalized to its GAPDH content in HK-2 cells treated with R and A, alone or in combination, for 48 h. Numbers on top of the blots represent the average fold change vs untreated cells normalized for internal loading. Particularly, western blotting analysis of LC3-II was normalized with respect to GAPDH. (B) Cell viability assay of HK-2 cells treated with R, A, alone or in combination, with or without MH for 48 h. Cell viability is expressed as fold change with respect to untreated cells (-). Results are representative of 3 different experiments each performed in triplicate. *P<0.05 compared untreated cells (-), **P<0.05 compared with cells treated with R. $ P<0.05 compared with cells treated with R, $$ P<0.05 compared with cells treated with R plus A.
Discussion
In the current study we demonstrated, for the first time, that in HK-2 proximal tubular renal cells, rapamycin exposure induced autophagy through the transactivation of the NGFR promoter, protecting cells from the damage caused by albumin exposure. In addition, we elucidated the underlying molecular mechanism by which rapamycin transactivated the NGFR promoter.
NGFR modulates cell-fate decisions through its highly ramified signaling pathways. Its biological effects are cell-type dependent and include survival, apoptosis, migration, differentiation, (pro)-neurotrophin binding, interacting transmembrane coreceptor expression, intracellular adaptor molecule availability and post-translational modifications [15]. However, although the proapoptotic role of NGFR is well known [16–22], little is known about NGFR as a potential mediator of autophagy. Indeed, to date, only one study reports that in cerebellar Purkinje neurons deprived of trophic factors, NGFR triggers autophagy, leading to Purkinje neuron death [10]. However, nothing is known about the role of NGFR in mediating renal autophagy.
Previously, we have demonstrated that in HK-2 cells, the cotreatment with nerve growth factor and the immunosuppressant cyclosporine A, causes a transcriptional activation of NGFR, leading to apoptosis [9]. In the current study, our results showed that in HK-2 cells the treatment with increasing doses of the immunosuppressant rapamycin, induced a transcriptional activation of NGFR that was not followed by apoptotic events, as we did not observe PARP1 cleavage or DNA fragmentation. Therefore, to explore whether the detected reduction of cellular viability induced by rapamycin could be due to autophagy activation, we evaluated the mRNA and protein expression of key autophagy regulators, that resulted in activation after rapamycin exposure, and was abrogated in HK-2 cells transfected with NGFR siRNA, suggesting that in our cellular model, rapamycin promoted autophagic flux via NGFR. To confirm our findings, we investigated the activation of rapamycin-induced autophagy, using immortalized human podocytes. Indeed, glomerular podocytes are highly specialized epithelial cells, with high basal autophagy, whose injury in glomerular diseases causes proteinuria. Data reported in literature demonstrate that in podocytes, increased autophagy represents a protective mechanism to counteract the renal damage [23]. As expected, our results showed that in podocytes treated with increasing concentrations of rapamycin and BAF, the autophagic flux persisted. The obtained results were similar to those observed in HK-2 cells, highlighting that MTOR inhibitors, through autophagy induction, could protect kidney from many injuries.
In addition, we confirmed that the inhibition of MTOR was crucial for NGFR-mediated autophagy activation, as treatment with the MTOR activator, MHY1485, reversed the upregulation of NGFR, as well as that of the autophagic markers reported above. Similar patterns of modulation were observed in HK-2 cells pretreated with Chloroquine, which inhibits autolysosomal degradation, by increasing lysosomal pH. These data suggest that autophagic flux inhibitors have similar effects to MHY1485, although the levels of NGFR, in the presence of chloroquine, are mitigated.
Surprisingly, in our in vitro model, in the presence of autophagy inhibitors, we did not observe a significant amount in LC3-II levels. This could be due to drawbacks in western blot method used for measuring endogenous LC3-II. We did not used additional methods to detect LC3-II levels, as other investigated markers (as BECN1 and SQSTM1), with or without autophagy inhibitors, confirmed the modulation of autophagic flux. In addition, we supposed that the difference in LC3-II content between the treated and untreated samples could be subtle and difficult to detect because of high basal autophagy. Finally, it is also important to note that when cells are treated with unsaturating concentrations of CQ or BAF alone in regular full culture medium, LC3 lipidation would be insufficiently blocked, leading to a lack of increase of LC3-II levels [24,25].
To overcome these issues, we evaluated the modulation of SQSTM1 levels; as expected, its downregulation was reversed in cells treated with autophagy inhibitors.
Starting from these data we explored the molecular mechanism by which rapamycin transactivated the NGFR promoter, leading to autophagy activation. The human NGFR promoter region contains multiple transcription regulatory elements, including NR3C1/GR, NF1, SP1, EGR1 and TFAP2A sites. Our functional experiments conducted using wild-type and deletion constructs of the NGFR promoter, showed that the promoter transactivation induced by rapamycin occurred through the EGR1 consensus site, that, in turn, bound the corresponding transcription factor, EGR1, whose nuclear translocation, mediated by NGFR and MTOR, was promoted by rapamycin exposure.
EGR1 is a transcription factor implicated in cell proliferation, apoptosis, signal transduction and in carcinogenesis. In vitro studies report that in hepatocellular carcinoma, EGR1 transcriptionally regulates hypoxia-induced autophagy by binding the MAP1LC3B promoter, resulting in resistance of cancer cells to chemotherapeutic agents [26]. A report, conducted in ovarian cancer cells, shows that the activation of EGR1 and MIR152 may be a useful therapeutic strategy to overcome cisplatin-resistance by preventing cytoprotective autophagy [27]. Zhi-Hua Chen et al., using an in vitro and in vivo models of chronic obstructive pulmonary disease, demonstrate a critical role of EGR1 in promoting autophagy and apoptosis in response to cigarette smoke exposure, suggesting that autophagy could represents a novel therapeutic target for the treatment of cigarette smoke-induced lung injury [14]. Our results suggested a potential involvement of EGR1 in tubular rapamycin-induced autophagy that, to date, has not been investigated.
Recently, Liu et al. reported that in proximal tubular renal cells, the rapamycin-promoted autophagic flux protected cells from the apoptotic damage induced by urinary protein exposure [13]. Although the specific mechanism has not been elucidated, many authors report that MTOR inhibitors can prevent or mitigate the tubule-interstitial injury observed in patients affected by primary and secondary nephropaties with excessive proteinuria [28]. The results of Bonegio et al. indicate that MTOR inhibitors may exert their beneficial effects in proteinuric nephropathy at doses considerably lower than those required for the prevention of transplant rejection [29]. On the other hand, Stallone et al. report that sirolimus-induced proteinuria, which is observed in renal transplant recipients during conversion from calcineurin inhibitors to sirolimus-based immunosuppression, might be avoided aiming at producing a lower blood level of the drug [30]. Taking into account these data, cotreating HK-2 cells with high doses of human albumin plus low doses of rapamycin (7 ng/ml) we observed a marked activation of autophagy that was reversed in the presence of NGFR RNAi. NGFR exerts many of its biological effects through the MAP3K and MAPK/JNK pathways [31], whose activation is also involved in the autophagic process [32]. Yoon et al. provide evidence that in endothelial cells, EGR1 activation regulates the angiogenic activity of colorectal cancer cell-derived extracellular vesicles, through the MAPK1/3 and MAPK/JNK pathways [33]. In our study we observed that in HK-2 cells the cotreatment with rapamycin plus albumin enhanced the phosphorylation of MAPK1/3, whose inhibition with the MAPK1/3 inhibitor PD98059, was accompanied by a decreased nuclear retention of EGR1, suggesting that the activation of NGFR downstream signaling was involved in the increased receptor promoter activity mediated by EGR1.
Furthermore, by in vivo studies, performed by using ChIP assay, we observed that in HK-2 cells transfected with the −41 +100 deletion construct, the cotreatment with rapamycin plus albumin promoted the interaction between EGR1 and NGFR, followed by a an increased transcriptional activity of polymerase II. As expected, the recruitment of the chromatin silencer HDAC1 was reduced in cells treated with rapamycin alone or plus albumin. The enhanced NGFR promoter activity (−41 +100) and the recruitment of EGR1 and POLR2A on the EGR1 site, induced by rapamycin with or without albumin were abrogated in the presence of inhibitor or mutated EGR1 motif. It's known that EGR1, binding on the promoter of its target genes, triggers autophagy [14,26]. In our experimental model, we observed enhanced levels of LC3-II in cells treated as reported above, while mithramycin A reversed this effect, suggesting that the enhanced recruitment of EGR1 on the NGFR promoter upregulates protein expression of LC3-II.
Taken together these results confirmed the role of NGFR in modulating the tubular autophagic pathway through the nuclear translocation of EGR1.
To date, several in vitro and in vivo studies are focused on the role of autophagy in the kidney, as it seems to exert protective effects in several kidney diseases [3,6,34]. Kimura et al. demonstrate that the renal function of ATG5-deficient mice exposed to acute ischemic injury worsens compared with control animals [4]. The same result is confirmed by in vitro studies performed in human and rat proximal tubular cells exposed to cisplatinum nephrotoxicity [35–37]. In order to confirm that activated autophagy persists over a long time to protect damaged proximal tubule, we chronically exposed HK-2 cells to rapamycin with or without human albumin. Results of western blotting didn't detect apoptotic events while BECN1 and LC3-II levels where enhanced. Concomitantly, we observed a significant reduction of cell viability in HK-2 cells pretreated with MHY1485 and then exposed to the cotreatment with rapamycin plus albumin, suggesting that the loss of autophagy reactivation caused cell death.
In conclusion, our data demonstrated for the first time that autophagy, through MTOR inhibition and transcriptional NGFR activation, protects proximal tubular cells against proteinuric damage. The challenge is to design more favorable therapeutic strategies to use low concentrations of MTOR inhibitors, reducing their dose-dependent side effects, in order to prevent at an early stage, tubular injury via autophagy activation.
Materials and methods
Cell culture and treatments
Human renal proximal tubular cells, HK-2 (ATCC, CRL-2190), were grown in Keratinocyte-SFM (Invitrogen, 17005–034) containing 5 ng/ml EGF (Invitrogen, 10450-013), bovine pituitary extract (0.5 mg/ml; Invitrogen, 13028-014) and 1 mg/ml penicillin/streptomycin (Invitrogen, 15140-122). Human immortalized podocytes, a gift from professor G. Camussi (Department of Internal Medicine and Center of Experimental Research and Medical Sciences (CERMS), University of Turin, Turin, Italy) [38], were grown in Dulbecco's modified Eagle' medium (Gibco, 41966-029) plus glutamax (Gibco, 25030-024) containing 10% fetal bovine serum (Invitrogen, 10500-064) and 1 mg/ml penicillin-streptomycin. All experiments were performed in complete medium because starvation conditions induce basal autophagy activation. The cells were exposed to the following treatments: rapamycin (from 0.1 to 18 ng/ml; Sigma Aldrich, R8781); albumin from human serum (8 mg/ml; Sigma Aldrich, A1653); chloroquine (25 µM; Sigma Aldrich, C6628), bafilomicyn A1/BAF (25 nM; Sigma Aldrich, B1793), MHY1485 (2 µM; Sigma Aldrich, SML0810); PD98059 (10 µM; Sigma Aldrich, P215).
Immunoblot analysis
Cells were treated in complete medium as indicated and then subjected to total protein extraction as described previously [8]. Nitrocellulose membrane were probed with: PARP1 (1:1000; Santa Cruz Biotechnology, sc-7150), NGFR (1:300; Santa Cruz Biotechnology, sc-8317), SQSTM1/p62 (1:1000; Santa Cruz Biotechnology, sc-28359), BECN1/Beclin 1 (1:1000; Santa Cruz Biotechnology, sc-48341), MAP1LC3B (1:200; Santa Cruz Biotechnology, sc-28266), pMTOR (Ser2448) (1:500; Cell Signaling Technology, sc-101738), total MTOR (1:500; Santa Cruz Biotechnology, sc-136269), phosphorylated and total MAPK (MAPK1/ERK2/p42 and MAPK3/ERK1/p44) (1:1000; Santa Cruz Biotechnology, sc-7383 and Cell Signaling Technology, 9102) and MAPK/JNK (1:500; pan-phospho-MAPK8/9/10[JNK1/2/3, respectively] Santa Cruz Biotechnology, sc-6254 and pan-nonphospho-MAPK8/9/10 [JNK1/2/3], Santa Cruz Biotechnology, sc-571) antibodies. As internal control, all membranes were subsequently stripped (0.2 M glycine, pH 2.6, for 30 min at room temperature) of the first antibody and reprobed with anti-GAPDH antibody (1:10000; Santa Cruz Biotechnology, sc-25778). The antigen-antibody complex was detected by incubation of the membranes for 1 h at room temperature with peroxidase-coupled goat anti-mouse (1:2000, IgG-HRP; Santa Cruz Biotechnology, sc-2005) or anti-rabbit (1:7000, IgG-HRP; Santa Cruz Biotechnology, sc-2004) and revealed using the enhanced chemiluminescence system (Santa Cruz Biotechnology, sc-2048). Blots were then exposed to film (Santa Cruz Biotechnology).
For nuclear and cytoplasmic extracts cells were harvested in cold PBS (Sigma-Aldrich, P4417) and then 300 μl of cytosolic buffer (50 mM HEPES, pH 7.5 [Sigma Aldrich, H3375, 150 mM NaCl [Sigma Aldrich, S7653] 1% Triton X-100 [Sigma Aldrich, T8787], 1.5 mM MgCl2 [Sigma Aldrich, M8266], 1 mM EGTA, pH 7.5 [Sigma Aldrich, E3889, 10% glycerol [Sigma Aldrich, G5516], protease inhibitors [Sigma-Aldrich, P8340]) were added. Following centrifugation (14000 g, 4°C, 10 min), the supernatant was referred to as cytoplasmic fraction. The pellet containing nuclei was resuspended in high salt buffer (20 mM HEPES pH 7.9, 25% [v:v] glycerol, 420 mM NaCl, 1.5 mM MgCl2, 0.2 mM EDTA [Sigma Aldrich, E5134], protease inhibitors). Extraction of nuclear proteins was achieved by vortexing this solution thoroughly, incubating overnight with agitation and subsequent centrifugation (14000 g, 4°C, 10 min). The collected supernatant represents the nuclear fraction. The purity of the cytoplasmic and nuclear fractions was confirmed by immunoblotting respectively with an anti-GAPDH antibody (Santa Cruz Biotechnology, sc-25778) and anti-LMNB/Lamin B (1:3000; Santa Cruz Biotechnology, sc-6217). Protein concentration was determined by Bradford Protein Assay (Bio-Rad, 5000006) and then equal amounts (40 μg) of cytosolic and nuclear proteins were prepared, resolved by 8% SDS-PAGE and probed with antibodies directed against: EGR1 (1:300; Santa Cruz Biotechnology, sc-110); NR3C1/GR (1:1000; Santa Cruz Biotechnology, sc-8992); SP1 (1:1000; Santa Cruz Biotechnology, sc-59); TFAP2A/AP2α (1:500; Santa Cruz Biotechnology, sc-12726); NF1 (1:500; Santa Cruz Biotechnology, sc-870). The bands of interest were quantified using the ImageJ densitometry scanning program.
Real-time RT-PCR assays
HK-2 cells were grown in 6-well plates to 70% to 80% confluence and exposed to treatments as indicated. Total RNA was isolated from cells with TRIZOL reagent (Invitrogen, 15596026) according to the manufacturer's protocol. The purity and integrity of the RNA was confirmed both spectroscopically and electrophoretically. Analysis of gene expression was performed using real-time reverse transcription PCR.
cDNA was synthesized from 2 μg of total RNA with random hexamer primers using the High capacity cDNA Archive Kit (Applied Biosystems, 4368813. Real-time PCR was performed in an iCycler iQ Detection System (Bio-Rad, 170–8740, Hercules, CA, USA) using SYBR Green Universal PCR Master Mix (Applied Biosystems, 4309155) with 0.1 mmol/l of each primer in a total volume of a 30-μl reaction mixture following the manufacturer's recommendations. Negative control containing water instead of first-strand cDNA was used. Each sample was normalized on its GAPDH mRNA content.
Primers used for the amplification were:
NGFR: forward 5’-CGTATTCCGACGAGGCCAACC-3’ and reverse 5’-CCACAAGGCCCACAACCACAGC-3’
BECN1: forward 5’-GGCCAATAAGATGGGTCTGA-3’ and reverse 5’-GCTTTTGTCCACTGCTCCTC-3’
MAP1LC3B: forward 5’-GATGTCCGACTTATTCGAGAGC-3’ and reverse 5’-TTGAGCTGTAAGCGCCTTCTA-3’
EGR1: forward 5’-CTTCAACCCTCAGGCGGACA-3’ and reverse 5’-GGAAAAGCGGCCAGTATAGGT-3’
(GAPDH): forward 5’-CCCACTCCTCCACCTTTGAC-3’ and reverse 5’-CATACCAGGAAATGAGCTTGACAA-3’
The relative gene expression levels were normalized as previously described [12].
DNA laddering
HK-2 cells were plated in 10-cm dishes to 70% confluence and exposed to 7 ng/ml rapamycin for 48 h. Cells were pelleted at 125 × g for 5 min, washed in PBS and resuspended in 0.5 ml of extraction buffer (50 mmol/L Tris-HCl, pH 8, 10 mmol/L EDTA, 0.5% SDS) for 20 min in rotation at 4°C. DNA was extracted with a phenol-chloroform protocol. The aqueous phase was used to precipitate nucleic acids with 0.1 volumes of 3 M sodium acetate and 2.5 volumes cold ethanol overnight at −20°C. The DNA pellet was resuspended in 15 µl of H2O treated with RNase A (Invitrogen, 12091039) for 30 min at 37°C. The absorbance of the DNA solution at 260 and 280 nm was determined by spectrophotometry (Eppendorf BioPhotometer® D30, 6133000001, Hamburg Germany). The extracted DNA was subjected to electrophoresis on 1.8% agarose gel.
Transfection assay
HK-2 cells were transfected using Lipofectamine 2000 (Invitrogen, 11668019), according to the procedure outlined in the product manual, in complete medium without penicillin/streptomycin with 100 nM of validated stealth NGFR targeted for the human NGFR mRNA sequence RNAi (Invitrogen, ID: 1299001), or with a negative control RNAi (Invitrogen, 452001) that does not match with any currently known human mRNA (scrambled siRNA). Efficiency of knockdown for each gene and the negative control was determined by western blot analyses (data not shown). After 48 h with the respective siRNA complexes the cells were treated as indicated and experiments were performed.
The WT NGFR promoter (NGFR −900 +100) and its deletion constructs (NGFR −164+100, NGFR −315+100, NGFR −41 +100) were a gift from E. Valli (University of Bologna, Department of Biology, Bologna, Italy) [39].
Functional studies were performed in HK-2 cells transfected for 24 h with WT and deletion constructs (0.5 µg) containing the NGFR promoter region, concomitant with 5 ng of pRL-CMV (Promega, E2261), which expresses Renilla luciferase enzymatically distinguishable from firefly luciferase by the strong cytomegalovirus enhancer/promoter, using the Fugen 6 reagent (Promega, E2692). Then HK-2 cells were treated as reported for 18 h. The firefly luciferase reporter is measured first by adding Luciferase Assay Reagent II (LAR II) to generate a stabilized luminescent signal. After quantifying the firefly luminescence, this reaction is quenched, and the Renilla luciferase reaction is simultaneously initiated by adding Stop & Glo Reagent to the same tube. The Stop & Glo Reagent also produces a stabilized signal from the Renilla luciferase, which decays slowly over the course of the measurement. The integrated format of the DLR™ Assay provides rapid quantification of both reporters either in transfected cells or in cell-free transcription and translation reactions. The firefly luciferase values of each sample were normalized by Renilla luciferase activity used as internal control.
Site-directed mutagenesis
The NGFR promoter plasmid containing the EGR1-responsive element-mutated site was created by site-directed mutagenesis using a mutagenesis kit (Thermofisher, F541), according to the manufacturer's method using as template the shorter construct (−41 +100). Primers utilized are the following: (mutations are shown as lowercase letters): Fw: 5’-CGCTGCCGCGGGAGttagtCGATGGGGCAGGTGC-3’ and Rv: 5’-GCACCTGCCCCATCGactaaCTCCCGCGGCAGCG-3’. The mutations were confirmed by DNA sequencing.
Next, the mutated construct (mutated NGFREGR1 promoter) was transfected in HK-2 cells as reported above.
Cell viability assay
The CellTiter-Glo® Luminescent Cell Viability Assay (Promega, G7571) is a homogeneous method to determine the number of viable cells in culture based on quantification of ATP, which signals the presence of metabolically active cells. The amount of ATP is directly proportional to the number of cells present in culture. Briefly, HK-2 cells (20,000 cells per well) were grown in 96-well plates and exposed to treatments as indicated in complete culture medium. A volume of CellTiter-Glo® Reagent equal to the volume of cell culture medium was added to each well, cells were placed in a shaker for 2 min to induce lysis and incubated at room temperature for 10 min to stabilize the luminescent signal. The luminescence was read by a luminometer (Lumat LB 9507 Tube Luminometer, 2334, Berthold Detection Systems GmbH Pforzheim Germany) and results were reported as fold respect to untreated cells.
Transmission electron microscopy
For transmission electron microscopy (TEM), samples were fixed in 3% glutaraldehyde prepared in 0.1 M phosphate buffer for 2 h at 4°C, postfixed in 3% osmium tetroxide, dehydrated in graded acetone, and embedded in Araldite (Fluka, 10951). Ultrathin sections were prepared using a diamond knife, collected on copper grids (EMS, G 300 Cu) and then examined with a Jeol JEM 1400 Plus electron microscope (Peabody, Massachusetts, USA) at 80 kV.
Chromatin immunoprecipitation (ChIP) assay
HK-2 cells were treated for 6 h as indicated and cross-linked in PBS containing 1X formaldehyde for 7 min at 37°C. Then cells were pelletted and resuspendend in a lysis buffer (1% SDS, 10 mmol/L EDTA, 50 mmol/L Tris-HCl, pH 8.1), and left on ice for 10 min. The HK-2 cells were sonicated 4 times for 10 sec at 30% of maximal power (Vibra Cell 500 W; Sonics and Materials, Inc., NEWTOWN, Connecticut, USA) and collected by centrifugation at 4°C for 10 min at 11,000 × g. Sample (30 µl) was used as input (non immunoprecipitated). The samples were diluted in immunoprecipitation buffer (0.01% SDS, 1.1% Triton X-100, 1.2 mmol/L EDTA, 16.7 mmol/L Tris-HCl, pH 8.1, 16.7 mmol/L NaCl), precleared with 60 µl of protein A/G Plus (Santa Cruz Biotechnology, SC-2003) for 1 h at 4°C.
Precleared chromatin was immunoprecipitated with anti-EGR1 or anti-POLR2A antibodies (Santa Cruz Biotechnology, sc-899) overnight at 4°C and reimmunoprecipitated with antibody against HDAC1 (Santa Cruz Biotechnology, sc-8410). The next day 60 µl of protein A/G Plus was added, and precipitation was further continued for 2 h at 4°C. Immunocomplexes DNA-proteins were washed sequentially for 5 min with the following buffers: Wash A (0.1% SDS, 1% Triton X-100, 2 mmol/L EDTA, 20 mmol/L Tris-HCl, pH 8.1, 150 mmol/L NaCl); Wash B (0.1% SDS, 1% Triton X-100, 2 mmol/L EDTA, 20 mmol/L Tris-HCl, pH 8.1, 500 mmol/L NaCl]; and Wash C [0.25 M/L LiCl, 1% NP-40 [Sigma Aldrich, I3021], 1% sodium deoxycholate [Sigma Aldrich, 30970], 1 mmol/L EDTA, 10 mmol/L Tris-HCl, pH 8.1), and then twice with 10 mmol/L Tris, pH 8.0, 1 mmol/L EDTA. Samples were eluted with elution buffer (1% SDS, 0.1 M/L NaHCO3). 5 M NaCl was added to the ChIP samples overnight at 65°C followed by digestion with proteinase K (0.5 mg/ml; Bioline, BIO-37037) at 45°C for 1 h. DNA was extracted using phenolchloroform-isoamyl alcohol. Two microliters of 10 mg/ml yeast tRNA (Sigma Aldrich, R4018) were added to each sample, DNA was precipitated with 95% ethanol for 24 h at −20°C, washed with 70% ethanol and resuspended in 20 µl of 10 mmol/L Tris, pH 8.0, 1 mmol/L EDTA buffer. Purified DNA (0.5 µl) was used as a template for real-time PCR detection using primers flanking the EGR1 site sequence present in the human NGFR promoter: Fw: 5’-GGACCGAGCTGGAAGTCG-3’ and Rv: 5’-GCTAACACTCACCCCCAGAA- 3’.
Results were normalized in comparison to total input DNA.
Statistical analysis
All experiments were performed in, at least, triplicate per treatment and repeated in 3 independent experiments. Real Time RT-PCR and Luciferase activity results are presented as fold induction over basal condition. Optical densities were measured using the ImageJ 1.47T software and their results are presented as fold induction respect to control. All results are presented as mean ± SD of data from 3 combined experiments. Data were analyzed by the Student t test using the GraphPad Prism 4 software program. P < 0.05 was considered as statistically significant.
Acknowledgements
We thank Dr. Emanuele Valli, Department of Biology, University of Bologna, Italy, for the wild-type NGFR promoter and its deletion constructs and Prof. Giovanni Camussi, University of Turin, Italy, who kindly gifted us with the immortalized human podocytes.
Disclosure of potential conflicts of interest
No potential conflict of interest needed to be reported by the authors.
Reference
- [1].De Rechter S, Decuypere JP, Ivanova E, et al. Autophagy in renal diseases. Ped Nephrol (Berlin, Germany). 2016;31(5):737–752. doi: 10.1007/s00467-015-3134-2 PubMed PMID: 26141928; eng. [DOI] [PubMed] [Google Scholar]
- [2].Takabatake Y, Kimura T, Takahashi A, et al. Autophagy and the kidney: health and disease. Nephrol Dial Transplant. 2014;29(9):1639–1647. doi: 10.1093/ndt/gft535 PubMed PMID: 24520117; eng. [DOI] [PubMed] [Google Scholar]
- [3].Jiang M, Liu K, Luo J, et al. Autophagy is a renoprotective mechanism during in vitro hypoxia and in vivo ischemia-reperfusion injury. Am J Pathol. 2010;176(3):1181–1192. doi: 10.2353/ajpath.2010.090594 PubMed PMID: 20075199; PubMed Central PMCID: PMCPmc2832141. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].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(5):902–913. doi: 10.1681/asn.2010070705 PubMed PMID: 21493778; PubMed Central PMCID: PMCPmc3083312. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Liu S, Hartleben B, Kretz O, et al. Autophagy plays a critical role in kidney tubule maintenance, aging and ischemia-reperfusion injury. Autophagy. 2012;8(5):826–837. doi: 10.4161/auto.19419 PubMed PMID: 22617445; eng. [DOI] [PubMed] [Google Scholar]
- [6].Takahashi A, Kimura T, Takabatake Y, et al. Autophagy guards against cisplatin-induced acute kidney injury. Am J Pathol. 2012;180(2):517–525. doi: 10.1016/j.ajpath.2011.11.001 PubMed PMID: 22265049; eng. [DOI] [PubMed] [Google Scholar]
- [7].Grahammer F, Ramakrishnan SK, Rinschen MM, et al. mTOR Regulates endocytosis and nutrient transport in proximal tubular cells. J Am Soc Nephrol. 2017;28(1):230–241. doi: 10.1681/asn.2015111224 PubMed PMID: 27297946; PubMed Central PMCID: PMCPmc5198276. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Raught B, Gingras AC, Sonenberg N. The target of rapamycin (TOR) proteins. Proc Nat Acad Sci U S A. 2001;98(13):7037–7044. doi: 10.1073/pnas.121145898 PubMed PMID: 11416184; PubMed Central PMCID: PMCPmc34619. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Vizza D, Perri A, Lofaro D, et al. Exposure to nerve growth factor worsens nephrotoxic effect induced by Cyclosporine A in HK-2 cells. PloS one. 2013;8(11):e80113. doi: 10.1371/journal.pone.0080113 PubMed PMID: 24244623; PubMed Central PMCID: PMCPmc3820545. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Florez-McClure ML, Linseman DA, Chu CT, et al. The p75 neurotrophin receptor can induce autophagy and death of cerebellar Purkinje neurons. J Neurosci. 2004;24(19):4498–4509. doi: 10.1523/jneurosci.5744-03.2004 PubMed PMID: 15140920; PubMed Central PMCID: PMCPmc1876689. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Antonucci MT, Bonofiglio R, Papalia T, et al. Nerve growth factor and its monocyte receptors are affected in kidney disease. Nephron Clin Prac. 2009;111(1):c21–c28. doi: 10.1159/000178819 PubMed PMID: 19169037; eng. [DOI] [PubMed] [Google Scholar]
- [12].Vizza D, Perri A, Toteda G, et al. Nerve growth factor exposure promotes tubular epithelial-mesenchymal transition via TGF-beta1 signaling activation. Growth factors (Chur, Switzerland). 2015;33(3):169–180. doi: 10.3109/08977194.2015.1054989 PubMed PMID: 26066770; eng. [DOI] [PubMed] [Google Scholar]
- [13].Liu WJ, Luo MN, Tan J, et al. Autophagy activation reduces renal tubular injury induced by urinary proteins. Autophagy. 2014;10(2):243–256. doi: 10.4161/auto.27004 PubMed PMID: 24345797; PubMed Central PMCID: PMCPmc5396082. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Chen ZH, Kim HP, Sciurba FC, et al. Egr-1 regulates autophagy in cigarette smoke-induced chronic obstructive pulmonary disease. PloS one. 2008;3(10):e3316. doi: 10.1371/journal.pone.0003316 PubMed PMID: 18830406; PubMed Central PMCID: PMCPmc2552992. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Tomellini E, Lagadec C, Polakowska R, et al. Role of p75 neurotrophin receptor in stem cell biology: more than just a marker. Cell Mol Life Sci. 2014;71(13):2467–2481. doi: 10.1007/s00018-014-1564-9 PubMed PMID: 24481864; eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Asai K, Tamakawa S, Yamamoto M, et al. Activated hepatic stellate cells overexpress p75NTR after partial hepatectomy and undergo apoptosis on nerve growth factor stimulation. Liver Int. 2006;26(5):595–603. doi: 10.1111/j.1478-3231.2006.01267.x PubMed PMID: 16762005; eng. [DOI] [PubMed] [Google Scholar]
- [17].Caporali A, Pani E, Horrevoets AJ, et al. Neurotrophin p75 receptor (p75NTR) promotes endothelial cell apoptosis and inhibits angiogenesis: implications for diabetes-induced impaired neovascularization in ischemic limb muscles. Circ Res. 2008;103(2):e15–e26. doi: 10.1161/circresaha.108.177386 PubMed PMID: 18566344; PubMed Central PMCID: PMCPmc2640941. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Imbschweiler I, Seehusen F, Peck CT, et al. Increased p75 neurotrophin receptor expression in the canine distemper virus model of multiple sclerosis identifies aldynoglial Schwann cells that emerge in response to axonal damage. Glia. 2012;60(3):358–371. doi: 10.1002/glia.22270 PubMed PMID: 22072443; eng. [DOI] [PubMed] [Google Scholar]
- [19].Lowry KS, Murray SS, McLean CA, et al. A potential role for the p75 low-affinity neurotrophin receptor in spinal motor neuron degeneration in murine and human amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2001;2(3):127–134. PubMed PMID: 11771768; eng. doi: 10.1080/146608201753275463. [DOI] [PubMed] [Google Scholar]
- [20].Mufson EJ, Ma SY, Dills J, et al. Loss of basal forebrain P75(NTR) immunoreactivity in subjects with mild cognitive impairment and Alzheimer's disease. J Comp Neurol. 2002;443(2):136–153. PubMed PMID: 11793352; eng. doi: 10.1002/cne.10122. [DOI] [PubMed] [Google Scholar]
- [21].Truzzi F, Marconi A, Atzei P, et al. p75 neurotrophin receptor mediates apoptosis in transit-amplifying cells and its overexpression restores cell death in psoriatic keratinocytes. Cell Death Differ. 2011;18(6):948–958. doi: 10.1038/cdd.2010.162 PubMed PMID: 21151024; PubMed Central PMCID: PMCPmc3131939. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Zhu Z, Friess H, Shi X, et al. Up-regulation of p75 neurotrophin receptor (p75NTR) is associated with apoptosis in chronic pancreatitis. Digest Dis Sci. 2003;48(4):717–725. PubMed PMID: 12741461; eng. doi: 10.1023/A:1022880524395. [DOI] [PubMed] [Google Scholar]
- [23].Huang SS, Ding DF, Chen S, et al. Resveratrol protects podocytes against apoptosis via stimulation of autophagy in a mouse model of diabetic nephropathy. Sci Rep. 2017;7:45692. doi: 10.1038/srep45692 PubMed PMID: 28374806; PubMed Central PMCID: PMCPmc5379482. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Ni HM, Bockus A, Wozniak AL, et al. Dissecting the dynamic turnover of GFP-LC3 in the autolysosome. Autophagy. 2011;7(2):188–204. PubMed PMID: 21107021; PubMed Central PMCID: PMCPmc3039769. eng. doi: 10.4161/auto.7.2.14181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research. Cell. 2010;140(3):313–326. doi: 10.1016/j.cell.2010.01.028 PubMed PMID: 20144757; PubMed Central PMCID: PMCPmc2852113. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Peng WX, Xiong EM, Ge L, et al. Egr-1 promotes hypoxia-induced autophagy to enhance chemo-resistance of hepatocellular carcinoma cells. Exp Cell Res. 2016;340(1):62–70. doi: 10.1016/j.yexcr.2015.12.006 PubMed PMID: 26708617; eng. [DOI] [PubMed] [Google Scholar]
- [27].He J, Yu JJ, Xu Q, et al. Downregulation of ATG14 by EGR1-MIR152 sensitizes ovarian cancer cells to cisplatin-induced apoptosis by inhibiting cyto-protective autophagy. Autophagy. 2015;11(2):373–384. doi: 10.1080/15548627.2015.1009781 PubMed PMID: 25650716; PubMed Central PMCID: PMCPmc4502709. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Rangan GK, Coombes JD. Renoprotective effects of sirolimus in non-immune initiated focal segmental glomerulosclerosis. Nephrol Dial Transplant. 2007;22(8):2175–2182. doi: 10.1093/ndt/gfm191 PubMed PMID: 17550925; eng. [DOI] [PubMed] [Google Scholar]
- [29].Bonegio RG, Fuhro R, Wang Z, et al. Rapamycin ameliorates proteinuria-associated tubulointerstitial inflammation and fibrosis in experimental membranous nephropathy. J Am Soc Nephrol. 2005;16(7):2063–2072. doi: 10.1681/asn.2004030180 PubMed PMID: 15917339; eng. [DOI] [PubMed] [Google Scholar]
- [30].Stallone G, Infante B, Pontrelli P, et al. Sirolimus and proteinuria in renal transplant patients: evidence for a dose-dependent effect on slit diaphragm-associated proteins. Transplantation. 2011;91(9):997–1004. doi: 10.1097/TP.0b013e318211d342 PubMed PMID: 21364499; eng. [DOI] [PubMed] [Google Scholar]
- [31].Bhakar AL, Howell JL, Paul CE, et al. Apoptosis induced by p75NTR overexpression requires Jun kinase-dependent phosphorylation of Bad. J Neurosci. 2003;23(36):11373–11381. PubMed PMID: 14673001; eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Li T, Xu XH, Tang ZH, et al. Platycodin D induces apoptosis and triggers ERK- and JNK-mediated autophagy in human hepatocellular carcinoma BEL-7402 cells. Acta Pharmacologica Sinica. 2015;36(12):1503–1513. doi: 10.1038/aps.2015.99 PubMed PMID: 26592509; PubMed Central PMCID: PMCPmc4816242. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Yoon YJ, Kim DK, Yoon CM, et al. Egr-1 activation by cancer-derived extracellular vesicles promotes endothelial cell migration via ERK1/2 and JNK signaling pathways. PloS one. 2014;9(12):e115170. doi: 10.1371/journal.pone.0115170 PubMed PMID: 25502753; PubMed Central PMCID: PMCPmc4264882. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Zhang YL, Zhang J, Cui L-Y, et al. Autophagy activation attenuates renal ischemia-reperfusion injury in rats. Exp Bio Med (Maywood, NJ). 2015;240(12):1590–1598. doi: 10.1177/1535370215581306 PubMed PMID: 25898836; PubMed Central PMCID: PMCPmc4935330. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Inoue K, Kuwana H, Shimamura Y, et al. Cisplatin-induced macroautophagy occurs prior to apoptosis in proximal tubules in vivo. Clin Exp Nephrol. 2010;14(2):112–122. doi: 10.1007/s10157-009-0254-7 PubMed PMID: 20013139; eng. [DOI] [PubMed] [Google Scholar]
- [36].Periyasamy-Thandavan S, Jiang M, Wei Q, et al. Autophagy is cytoprotective during cisplatin injury of renal proximal tubular cells. Kidney Int. 2008;74(5):631–640. doi: 10.1038/ki.2008.214 PubMed PMID: 18509315; eng. [DOI] [PubMed] [Google Scholar]
- [37].Yang C, Kaushal V, Shah SV, et al. Autophagy is associated with apoptosis in cisplatin injury to renal tubular epithelial cells. Am J Physiol Renal Physiol. 2008;294(4):F777–F787. doi: 10.1152/ajprenal.00590.2007 PubMed PMID: 18256309; eng. [DOI] [PubMed] [Google Scholar]
- [38].Bruno S, Bussolati B, Grange C, et al. Isolation and characterization of resident mesenchymal stem cells in human glomeruli. Stem Cells Dev. 2009;18(6):867–880. doi: 10.1089/scd.2008.0320 PubMed PMID: 19579288; eng. [DOI] [PubMed] [Google Scholar]
- [39].Iraci N, Diolait D, Papa A, et al. A SP1/MIZ1/MYCN repression complex recruits HDAC1 at the TRKA and p75NTR promoters and affects neuroblastoma malignancy by inhibiting the cell response to NGF. Cancer Res. 2011;71(2):404–412. doi: 10.1158/0008-5472.can-10-2627 PubMed PMID: 21123453; eng. [DOI] [PubMed] [Google Scholar]