Tubular damage is a major cause of renal failure, and both ischemic and toxic tubular injuries are very frequent causes of acute and chronic renal failure.1 The course of any renal tubular injury typically starts with loss of specialized function (e.g., proximal tubular cells lose the brush border and their ability to reabsorb low-molecular-weight proteins) and ends with cell death through necrosis, apoptosis, or necroptosis.2 Repair occurs through proliferation of the surviving epithelial cells and their redifferentiation into specialized tubular cells.3
Most patients with transplanted kidneys are treated with calcineurin inhibitors, which cause vascular and tubular damage.4 These drugs, mainly cyclosporin A (CsA) and tacrolimus, can initiate tubular lesions,5,6 but the exact mechanism is currently unknown. Calcineurin is a serine phosphatase involved in the dephosphorylation of various proteins and, most importantly, a family of transcription factors—the nuclear factors of activated T cells (NFATs). There are five of these; NFATs 1–4 are regulated by calcineurin, and NFAT5 is regulated by osmotic stress.7 Calcineurin, by dephosphorylating NFAT, allows it to enter the nucleus and begin its transcriptional activity. NFAT target genes involved in the inflammatory response have been extensively studied in lymphocytes, where NFAT acts as a transcriptional activator but also as a repressor for some genes.8In vivo, CsA induces renal vasoconstriction and tubular lesions.9 The spontaneous development of a renal phenotype reminiscent of cyclosporin toxicity in mice deficient for calcineurin A α indicates that the decrease in calcineurin activity is sufficient to trigger renal damage.10,11 Genetic deficiency of either cyclophilin or NFATc1 (i.e., the two substrates known to be inhibited by cyclosporin) is not sufficient to initiate tubular damage, but may increase it by impeding the tubular repair process.12–14
The mechanisms by which cyclosporin alters tubular function or repair are still poorly understood. Its vasoconstrictive properties on afferent arterioles might be involved in vivo, by causing subintrant ischemia downstream.4 A direct effect within the epithelium is also plausible since, like many xenobiotics, calcineurin inhibitors are metabolized by tubular epithelial cells. Endoplasmic reticulum (ER) stress is one hallmark of in vitro cyclosporin toxicity.15–17 It is a complex phenomenon with antagonistic consequences.18 The term refers both to the accumulation of misfolded proteins in the ER and to the molecular events triggered in response to this accumulation. The accumulation of misfolded proteins recruits the chaperone protein BiP, thus in turn activating double-stranded RNA activated protein kinase-like ER kinase (PERK), a kinase that phosphorylates the transcription factor EIF2α. EIF2α phosphorylation is the common end signal of kinases activated by amino-acid limitation (general control nonrepressible 2 [GCN2]), viral infection (double-strand RNA-activated kinase), heme limitation (heme-regulated inhibitor kinase) and protein misfolding (PERK).19 The biologic role of EIF2α phosphorylation is to decrease protein translation in general, except for that of adaptive proteins, including ATF4, a master transcription factor. ATF4 induces the expression of adaptive genes. If sustained, however, ATF4-induced upregulation of protein synthesis eventually causes ATP depletion and cell death.20 It is thus essential to control the ER stress pathway. Cyclosporin toxicity is known to be associated with impaired energy status and can be alleviated by ATP.21 Together, these data strengthen the hypothesis that ER stress plays a role in cyclosporin-induced renal injury.
Our group has extensively studied the phenotypic changes observed in tubular epithelial cells from human kidney recipients exposed to cyclosporin and has repeatedly shown, in particular, that immunohistochemical staining of two mesenchymal markers, vimentin and β-catenin, in tubular epithelial cells detects cyclosporin tubular toxicity at an early, reversible stage.5,22 We have also shown that the expression of these mesenchymal markers in tubular epithelial cells of kidney grafts in patients treated with cyclosporin is associated with initial ischemic injury and is predictive of further development of kidney fibrosis.23,24 To further investigate the mechanisms of CsA-induced tubular injury, we characterized the in vivo tubular transcriptomic changes that occur at an early stage of cyclosporin toxicity. Our findings point to a new response gene encoding nuclear protein, transcriptional regulator 1 (Nupr1), which has a central role in the ER stress pathway. We show here that Nupr1 is induced in several animal models of acute tubular damage and confers resistance to renal tubular damage in these animals presenting with cyclosporin toxicity.
Results
Cyclosporin Induces Tubular Transcriptomic Changes That Are Related to Protein Synthesis and Stress
We used laser microdissection coupled to an mRNA microarray to perform an in vivo comparative transcriptomic analysis of rat renal tubules exposed to either vehicle or nephrotoxic doses of cyclosporin (Figure 1, dataset one). Using a 1.5-fold threshold (with P<0.05 by t test), we found that 179 genes were downregulated and 286 genes were upregulated by cyclosporin (dataset one). The gene expression pattern confirmed that this transcriptomic analysis was limited to the tubular epithelium, since the expression of endothelial (Cd31) and podocyte-specific (Nephrin) genes was very low (three and two, respectively, on a logarithmic scale ranging from two to 14). Markers specific to the proximal tubule (Npt2a and Lrp2) were very strongly expressed (12 on the same logarithmic scale), whereas Aqp2, a marker of collecting ducts, was expressed only moderately (seven on the logarithmic scale). None of these cell-specific markers differed between the cyclosporin- and vehicle-treated groups. In contrast, we found that known markers of tubular damage—Havcr1 (also known as Kim1) and osteopontin (Opn)—were induced in renal tubules exposed to cyclosporin (three to seven for Kim1 and five to nine for Opn on the logarithmic scale). Figure 1 presents these expression data.
Figure 1.: In vivo renal tubular transcriptome analysis reveals transcriptomic patterns associated with cyclosporine toxicity. (A) Principal component analysis of the gene expression results. The six samples circled in red passed the quality check for further analysis. (B) Volcano plot with −log P value (t test) along the y-axis and log fold-change along the x-axis. Nupr1 is circled in red. (C) mRNA expression by microarray (on a logarithmic scale ranging from two to 14) of representative markers for various segments of the nephron (megalin and Npt2a for the proximal renal tubule, Aqp2 for the collecting duct, Nephrin for the glomeruli, and Cd31 for the endothelium) and known markers of renal tubular injury (Kim1 and Opn). CsA-treated animals (CsA, n=3) were compared with the control group (CTL, n=3). Results were considered not significant if the P value (t test) >0.05. (D) Summary of the logistic regression pathway analysis on KEGG pathways. The five pathways were enriched in the differential analysis between the CsA and the CTL group. Genes from the first three pathways were upregulated by cyclosporin, and genes from the last two pathways were downregulated by it. KEGG, Kyoto Encyclopedia of Genes and Genomes.
The pathway analysis using the Pathway Analysis using Logistic Regression (LRpath) identified significant changes in the expression of genes involved in five Kyoto Encyclopedia of Genes and Genomes pathways: ribosome, protein processing in the ER, protein export, peroxisome, and terpenoid synthesis (Figure 1, Supplemental Table 1). Notably, searching the Transcription Factor Binding Sites database (Biobase; Qiagen, Waltham, MA) for transcription factors associated with the observed transcriptomic changes did not identify NFAT or any other transcription factor. The first three pathways (ribosome, protein processing in the ER, and protein export), all involved in protein synthesis, showed overexpression in the CsA group, whereas the latter two (peroxisome and terpenoid synthesis) were underexpressed. This transcriptional signature of cyclosporin in vivo in the tubular epithelium is strongly reminiscent of the ATF4-dependent gene expression induced by tunicamycin (an ER stress inducer) in vitro.20 Accordingly, we observed that nephrotoxic doses of cyclosporin consistently enhanced the level of ATF4 in renal tubular cells, as shown by immunohistochemical analysis of kidney sections from CsA-treated mice. Moreover, ATF4 was induced by CsA in vitro in cultured mouse tubular epithelial cells, as shown by Western blot (Figure 2). Interestingly, Nupr1, an ATF4-responsive gene,25,26 was the gene most highly overexpressed (86 times) under cyclosporin in this microarray analysis (Supplemental Table 1). To our knowledge, the role of Nupr1 in acute tubular damage has not previously been studied in vivo.
Figure 2.: Cyclosporine induces ATF4 in the kidney. ATF4 protein expression after CsA treatment assessed by immunohistochemical analysis (A) and by Western blot (B) in renal tubules of mice, and at different time points (C). (A) Tubules injured by CsA 100 mg/kg per day, for 14 days (CsA) overexpressed ATF4, compared with control kidneys (CTL). Arrows indicate strongly stained nuclei. (B) Photograph of the full Western blots for ATF4 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), with the protein ladder on the left (ATF4 and GAPDH corresponding to 50 and 40 kDa, respectively), showing induction of ATF4 after 24 hours exposure to 5 µg/ml of CsA in a primary cell culture of mouse proximal tubular cells (representative of three independent experiments). Extinction of ATF4 staining after transfection with an anti-ATF4 small interfering RNA attests to the specificity of the anti-ATF4 antibody. (C) Time course of ATF4 protein (n=2) with Nupr1, Atf4, Chop, and Bip mRNA (n=4), after 1, 2, 3, 6, and 24 hours of CsA (1 µg/ml) exposure, compared with vehicle. Scr, scrambled (non specific) RNA; siRNA, small interfering RNA.
Nupr1 Confers Resistance to Epithelial Injury
To gain insight into the role of Nupr1 in CsA nephrotoxicity, we translated our experimental model to mice. C57bl6/J wild-type (WT) mice showed evidence of epithelial toxicity with renal failure after 2 weeks of cyclosporin at doses between 50 and 100 mg/kg per day subcutaneously. A dose of 100 mg/kg per day induced AKI, weight loss, tubular damage, apoptosis, and proliferation together with dose-dependent Nupr1 induction by CsA and ER stress (Supplemental Figures 1–3). We used Nupr1−/− mice to investigate the role of this gene in cyclosporin-induced renal injury. Although these Nupr1-deficient mice showed no spontaneous renal phenotype, the renal tubular lesions they developed when challenged with cyclosporin were substantially worse than those observed in WT mice. They had more tubular vacuolization and dilation than their WT littermates (median semiquantitative score 2 interquartile range [0.88–2.5] versus 0.5 [0.5–1]; P=0.01 for vacuolization and 1.75 [0.5–2] versus 0 [0–0.5]; P=0.003 for dilation, Figure 3, A, B, G, and F). Accordingly, Kim-1 mRNA expression was significantly higher in the cortex of Nupr1−/− compared with WT mice; a trend toward higher plasma urea levels in the Nupr1−/− mice was also observed, but did not reach statistical significance (Supplemental Figure 4).
Figure 3.: Nupr1 alleviates cyclosporine nephrotoxicity. (A, C, and E) WT mice treated by cyclosporin 100 mg/kg per day for 14 days. (B, D, and F) Nupr1 −/− animals treated by cyclosporin 100 mg/kg per day for 14 days. (A and B) Masson Trichrome. Nupr1 deficiency exacerbated histologic tubular lesions caused by cyclosporin: tubular vacuolization and dilation. (C and D) Ki67 immunohistochemistry. Nupr1 deficiency was associated with increased tubular cell proliferation. (E and F) TUNEL assay. Nupr1 deficiency was associated with increased apoptosis. Quantification for tubular dilation (G), tubular vacuolization (H) in WT (n=12) and knockout (n=10) mice, Ki67 (I) and TUNEL (J) in WT (n=6) and knockout (n=6) mice. KO, knockout; TUNEL, terminal deoxynucleotidyl transferase–mediated digoxigenin-deoxyuridine nick-end.
The Mechanism by Which Nupr1 Protects the Renal Epithelium Involves Cell Survival but Not Decreased Cyclosporin Exposure, Vascular Reactivity, or Enhanced Tubular Repair
Measurements of serum cyclosporin trough levels showed that the groups did not differ for cyclosporin exposure (Supplemental Figure 5A). Knockout mice did not differ from the WT mice in their expression of the Il27 gene, a close neighbor of the Nupr1 gene on the mouse genome and a potential regulator of cyclosporin-induced inflammation (Supplemental Figure 5B).27 Measurements of aortic BP and renal blood flow in Nupr1−/− and WT mice after 5 days of cyclosporin (100 mg/kg per day subcutaneously) or 5 days of vehicle showed no hemodynamic effect of Nupr1 (Supplemental Figure 5, C and D).
After cyclosporin treatment, apoptosis, assessed by terminal deoxynucleotidyl transferase–mediated digoxigenin-deoxyuridine nick-end labeling, was more frequent in renal tubular cells from Nupr1−/− (18.5 [9.8–33.3] nuclei/field) than in WT mice (4 [1.75–6.5]; P=0.01) (Figure 3, E, F, and J). This observation suggests that Nupr1 protects tubular cells from death. Tubular proliferation assessed by Ki67 staining was again more frequent in the renal tubular cells of the Nupr1−/− (89.5 [55.5–117.3] nuclei/field) than WT animals (43.5 [33.3–56.5]; P=0.03) (Figure 3, C, D, and I). Both tubular apoptosis and proliferative repair increased in the absence of Nupr1.
Nupr1 Limits Protein Synthesis, Proliferation, and Cell Death, and Preserves the Energy Status of Epithelial Proximal Tubular Cells during In Vitro Cyclosporin Toxicity
We used primary cultures of proximal epithelial tubular cells from Nupr1−/− and WT mice to investigate the effect of Nupr1 on apoptosis under CsA treatment. In WT cells, we measured ATF4 protein, Nupr1, Atf4, Chop, and Bip mRNA expression before and after 1, 2, 3, 6, and 24 hours of cyclosporin exposure. All these parameters were induced already after 1 hour, with ATF4 protein induction being the strongest at this early time point, except Bip which was induced only after 3 hours (Figure 2C). Nupr1−/− cells had a higher level of ATF4 expression after CsA treatment than WT cells (Supplemental Figure 6). Using a pulse of homopropargylglycine (a methionine analogue) to evaluate protein synthesis by immunofluorescence and flow cytometry, we showed that cyclosporin decreased protein synthesis in WT cells but did not do so in Nupr1−/− cells (Figure 4).
Figure 4.: Assessment of protein synthesis using a pulse of L-homopropargylglycine (methionine analogue) revealed by Alexa Fluor 488 shows Nupr1-dependent repression of protein synthesis under cyclosporine A. (A) Protein synthesis evaluation by immunofluorescence (in green) in WT and Nupr1 −/− proximal tubular cells incubated with 5 µg/ml CsA or vehicle for 24 hours. Main images: magnification ×100, insets: magnification ×600. (B) Protein synthesis evaluation by flow cytometry showing the percentage of cells in cell-cycle phase G1 undergoing protein synthesis, and the rate of protein synthesis (median fluorescence intensity) in these cells for WT and Nupr1 −/− cells treated for 24 hours with 5 µg/ml cyclosporin or vehicle. CHX, cycloheximide; CTL, control.
We measured cell proliferation using three different techniques: a simple cell count 48 hours after seeding various numbers of cells, continuous monitoring of cell density with xCELLigence technology, and 5-bromo-2′-deoxyuridine incorporation (Figure 5, Figure 6, A and B, Supplemental Figure 7). All techniques showed that Nupr1−/− proximal tubular cells proliferated faster than WT cells. Treatment with CsA virtually abolished proliferation in WT cells, but only decreased proliferation in knockout cells (Figure 6, A and B). Although proliferation was maintained, assessment by xCELLigence of CsA-treated Nupr1−/− cells showed a decrease in cell density, suggesting cell death.
Figure 5.: Proliferation assessment of WT and Nupr1 −/− cells within 48 hours after seeding shows increased proliferation in Nupr1 −/− cells. (A) Number of cells counted by flow cytometry in a well 48 hours after seeding 500–20,000 of either knockout (KO) or WT proximal tubular cells/well (96-well plate). (B) Photograph of the cells 48 hours after seeding 5000 of KO or WT proximal tubular cells per well. (C) Real-time monitoring of cell proliferation for 48 hours after seeding 1000–7000 cells/well (96-wells plate) of either KO or WT proximal tubular cells, and monitoring of cell proliferation for 24 hours after seeding 5000 of KO or WT proximal tubular cells per well, followed by 24 hours incubation with 1 µg/ml cyclosporine or vehicle.
Figure 6.: Nupr1 decreases proliferation and cell death, and preserves cellular ATP levels. (A) Cell-cycle analysis using flow cytometry to assess DNA content by propidium iodide staining and DNA synthesis by BrdU incorporation, under normal conditions or under 1 µg/ml cyclosporin for 24 hours, according to Nupr1 status (n=3). (B) Rate of BrdU incorporation in BrdU-positive cells assessed by flow cytometry, under normal conditions or under 1 µg/ml cyclosporin for 24 hours, according to Nupr1 status (n=3). (C) Apoptosis quantified by flow cytometry (propidium iodide for necrosis and YO-PRO-1 iodide for apoptosis) in vitro under normal conditions or under 5 µg/ml cyclosporin for 24 hours, according to Nupr1 status (n=5). (D) ATP/ADP ratio in vitro under normal conditions (n=12) or under 1 µg/ml cyclosporin for 24 hours (n=6), according to Nupr1. BrdU, 5-bromo-2′-deoxyuridine; CTL, control; KO, knockout; MFI, mean fluorescence intensity.
Similar to CsA renal toxicity in vivo, 5 µg/ml CsA induced cell death, with more dead cells in Nupr1−/− cells compared with WT cells (Figure 6C).
We investigated the effect of Nupr1 on the energy status of proximal tubular epithelial cells by measuring ATP and ADP in Nupr1−/− and WT cells incubated for 24 hours with or without 1 µg/ml CsA (Figure 6D). ATP/ADP ratio was lower in Nupr1−/− mice compared with WT mice. Moreover, the ATP/ADP ratio decreased further still after cyclosporin was added. The effect of Nupr1 on the ATP/ADP ratio was explained mainly by a fall in ATP, although ADP increased slightly as well (Supplemental Figure 8).
Rescuing Nupr1 deficiency by transfecting a flag-Nupr1 transgene into Nupr1−/− proximal tubular cells did inhibit proliferation (less cells in S phase, and more cells in G2/M) in cells expressing the transgene, thus confirming the instrumental role of Nupr1 in proliferation and apoptosis (Figure 7, Supplemental Figure 9).
Figure 7.: Proliferation control is rescued in Nupr1 −/− cells by transfection of a flag-Nupr1 transgene (n=3). (A) Gating of single cells on DNA content using an area versus height dot-plot. (B) Transfection resulted in only approximately 1% of flag-expressing cells. (C) Transfection selective cell-cycle analysis, based on DNA content assessed by Hoechst staining.
Nupr1 Induction is Relevant to Other Models of Kidney Injury in Mice and in Human Kidney Transplantation
To determine if Nupr1 was specifically upregulated after CsA treatment, we measured Nupr1 mRNA in kidney cortices of mice in control conditions and after ischemia-reperfusion injury, unilateral ureteral obstruction, and angiotensin II-induced hypertension. As shown in Supplemental Figure 10, we found that Nupr1 was upregulated in each of these different experimental models. This finding suggests that Nupr1 may exert its protective effects in many forms of kidney injury.
We studied the expression of Nupr1 in cultured human renal epithelial cells. Increasing the cyclosporin concentration dose-dependently induced Nupr1 mRNA expression in human proximal tubular cells (Figure 8A). We also performed immunohistochemical analysis for NUPR1 on normal kidney (stable renal allografts) and on injured kidney (preimplantation biopsies from donors with acute tubular necrosis and allografts undergoing acute tubular necrosis or allograft dysfunction). Using multiple stainings for NUPR1, megalin, and nuclei, we found that NUPR1 expression was absent in nuclei from renal proximal tubules in normal kidneys. Conversely, in injured kidneys, NUPR1 was strongly expressed in many nuclei from renal proximal tubules. (Figure 8, B–F).
Figure 8.: NUPR1 is induced upon injury in human renal proximal tubular cells. (A) Analysis of Nupr1 expression (normalized on housekeeping gene Hprt) in human proximal tubular HK2 cells showed a dose-dependent induction of Nupr1 mRNA by cyclosporin. (B–E) Immunohistochemical analysis for NUPR1 in human renal allografts. (B) semi-quantitative assessment of NUPR1 expression in normal surveillance biopsies 3 months after transplantation (Nal M3), allograft dysfunction, acute tubular necrosis after renal transplantation (ATN), or acute tubular necrosis on native kidneys (pre-implantation biopsies from deceased donors, donor ATN). Two examples showing a normal biopsy 3 months after transplantation with no NUPR1 expression (C and E), and a biopsy with acute antibody-mediated rejection and chronic allograft dysfunction (D and F). (C and D) show NUPR1 expression, and (E–F) show the triple staining for nuclei (DAPI, blue), megalin (green), and NUPR1 (magenta). Arrows indicate NUPR1-positive nuclei in megalin-postive tubules. DAPI, 4′,6-diamidino-2phénylindole.
Discussion
Kidney resistance to injury varies greatly,28 as exemplified by cyclosporin toxicity; it is widely prescribed to patients in standardized conditions and results in variable toxicity, depending on the dose, on additional insults, and on the characteristics of the patient.4 Several genes involved in xenobiotic detoxification have been suggested as risk factors for calcineurin inhibitor nephrotoxicity.29 Nonetheless, cyclosporin nephrotoxicity remains poorly understood, especially its in vivo effect on renal tubular epithelium. Our investigation provides the first description of the genome-wide transcriptional effect of this drug there. Our primary hypothesis was that calcineurin inhibition would cause toxic effects by inactivating NFAT, a pleiotropic transcription factor. NFAT-induced genes (e.g.,Il2) are expected to be downregulated by cyclosporin through calcineurin inhibition.8 Other genes have been shown to be repressed by NFAT (e.g.,Cdk4) and might thus be enhanced under cyclosporin treatment.8,30–32 Interestingly, one of the genotypes at risk of cyclosporin toxicity is a low-activity variant of Cyp2c9,29 the gene most downregulated by cyclosporin in our data (Figure 1). However, the Cyp2c9 promoter contains no NFAT response element,33 and neither the NFAT nor the calcineurin pathways were modified by cyclosporin on a transcriptional level in our data. These seemingly contradictory results can be interpreted in two ways: (1) the calcineurin and NFAT pathways in the tubular epithelium (our data) are very different from these pathways in the lymphocytes (previously published data7,8,31), thus, bioinformatics tools like LRpath do not link our data to calcineurin and NFAT; and (2) the pattern of gene expression we observed is not directly caused by the inhibition of the calcineurin and NFAT pathways. Other substrates of calcineurin might be involved, or a secondary transcriptomic response to injury. This pattern is reminiscent of the genes induced by ATF4, a master transcription factor that orchestrates the expression of stress genes, including Nupr1 and other stress-related genes that were overexpressed in our analysis, like Asparagine synthetase and several aminoacyl-transfer RNA-synthetases. ATF4 induces a transcriptional program that upregulates the energy-expending mechanisms of protein synthesis potentially leading to cell death.20 Decreased ATP levels are instrumental in cell death, whether due to diminished ATP production (decreased fatty acid oxidation) or to enhanced ATP consumption (increased protein synthesis).20,34 Here, after administration of toxic doses of cyclosporin in vitro and in vivo, renal tubular cells expressed ATF4-dependent pathways associated with decreased expression of peroxisome genes and increased rates of protein synthesis, energy depletion, and cell death.
Nupr1, essentially unstudied in renal diseases, is a chromatin-binding protein induced by various epithelial stresses and has important properties: it is antiapoptotic and anti-inflammatory, and it regulates cell proliferation.35 Its expression is instrumental in endothelin-mediated mesangial hypertrophy.36 Because Nupr1 is an early stress response gene induced by ATF4 in the renal tubular epithelium and protects tubular cells from cell death,25 we hypothesized that it might protect against apoptosis by preserving the cell energy status.
We found that Nupr1 was strongly expressed during cyclosporin-induced renal epithelial lesions in vivo and in vitro and that it exerted a protective role on cell survival and energy status. Nupr1 also contributed to inhibiting protein synthesis and slowing the proliferation of proximal tubular cells. Ischemia-reperfusion injury causes p53-dependent G2/M arrest of renal tubular cells and subsequent renal fibrosis.37Nupr1 is a positive regulator of p53.38 Because we found a consistent increase in Nupr1 levels in all the kidney injury models we tested (including ischemia-reperfusion injury), Nupr1 induction appears to be a generic response to any epithelial injury and is not cyclosporin-specific. Whether Nupr1 induction is also protective in these other models remains to be tested. The mechanism by which the ATF4-NUPR1 pathway might influence evolution toward cell death or survival is outlined in Figure 9. Overall, Nupr1 inhibits energetically costly processes (proliferation and protein synthesis) and preserves ATP cellular concentrations. ADP/ATP ratio is associated with apoptosis, and may be instrumental in this model. However, the molecular targets of Nupr1 that lead to cell protection are still unknown. Further studies should therefore: (1) investigate the precise mechanisms that regulate energy consumption, protein synthesis, and proliferation downstream of Nupr1; (2) test strategies aimed at producing a time-limited expression of Nupr1-related pathways in AKI; and (3) determine whether Nupr1 variants are involved in susceptibility to kidney injury in humans.
Figure 9.: Nupr1 acts as a pro-survival factor in the renal proximal tubule. Schematic representation of the relations between kidney injury, the ATF4-NUPR1 pathway, and epithelial fate.
Concise Methods
For a full description of the methods please see Supplemental Material.
Animals
We used a model of in vivo renal toxicity created by daily injections of cyclosporin for 2 weeks.39 J.L.I. created the Nupr1−/− mouse strain.38 Blood and kidneys were harvested under general anesthesia. For creatinine clearance measurements, animals were placed overnight in metabolic cages for urine collection, and blood was collected from the retro-orbital sinus under isoflurane anesthesia.
Morphologic Studies and Laser Microdissection
Ki67 and terminal deoxynucleotidyl transferase–mediated digoxigenin-deoxyuridine nick-end quantification were performed with the particle analysis function of ImageJ 1.45o software. For each sample, 200 cortical tubules were harvested by laser capture microdissection (Supplemental Figure 11).
RT-PCR and mRNA Microarray
Microdissected tubules were studied with the GeneChip Rat Genome 230 2.0 microarrays (Affymetrix, Santa Clara, CA). The pathway analysis was performed with LRpath web-based software.40 The quantitative RT-PCR expression results relative to the housekeeping gene were obtained by the ∆Cp method (see Supplemental Table 2 for primers sequences).
In Vitro Studies
Proximal epithelial cells were isolated from fresh kidneys of Nupr1−/− and WT mice.41,42
Statistical Analyses
Statistical analysis used JMP Pro 9.0.2 software. Data are presented by their median values and interquartile ranges, and a nonparametric Wilcoxon test was used to assess statistical significance, except where otherwise indicated. A P value <0.05 was defined as significant.
Study Approval
Animals were handled in a licensed facility (approval number B-75–20–01) in compliance with French legislation and Institut National de la Santé et de la Recherche Médicale standards. Biopsies from renal-transplanted patients receiving cyclosporin treatment were used for NUPR1 immunohistochemistry. The biopsies were indicated for state-of-the-art routine care, and all patients provided written consent for use of the leftover material for research purposes. The study performed on human biopsies was approved by our institutional review board (Comité de Protection des Personnes Ile de France V).
Disclosures
None.
P.G. designed and performed the experiments and drafted the manuscript. M.W., A.B., and C.M. participated in the in vivo experiments. D.L. and S.V. participated in the in vitro experiments. S.P. performed the hemodynamics study. N.B. participated in the in vivo studies and RT-qPCRs. Y.-C.X.-D., C.J., and D.B. participated in the histologic evaluation of the kidneys (mice and humans). A.H. participated in the interpretation of the results and reviewed the manuscript. J.I. created the Nupr1−/− mice and reviewed the manuscript. E.R. participated in the study design and in writing the manuscript.
Project funding: Assistance Publique–Hôpitaux de Paris, Agence de Biomédecine. Equipment funding: Domaine d'Intérêt Majeur Cardiovasculaire-Obésité-Diabète (CODDIM). Scholarships: Assistance Publique–Hôpitaux de Paris (to P.G. and A.B.), Société Francophone de Transplantation (to P.G.), Société de Néphrologie (to P.G.), Société Française d'Anesthésie-Réanimation (to A.B.), and Contrat d’Interface hospitalier Institut National de la Santé et de la Recherche Médicale - Assistance Publique—Hôpitaux de Paris (to A.H.)
References
1. Cerdá J, Lameire N, Eggers P, Pannu N, Uchino S, Wang H, Bagga A, Levin A: Epidemiology of acute kidney injury. Clin J Am Soc Nephrol 3: 881–886, 200818216347
2. Bonventre JV, Yang L: Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest 121: 4210–4221, 201122045571
3. Humphreys BD, Czerniak S, DiRocco DP, Hasnain W, Cheema R, Bonventre JV: Repair of injured proximal tubule does not involve specialized progenitors. Proc Natl Acad Sci U S A 108: 9226–9231, 201121576461
4. Naesens M, Kuypers DR, Sarwal M: Calcineurin inhibitor nephrotoxicity. Clin J Am Soc Nephrol 4: 481–508, 200919218475
5. Galichon P, Vittoz N, Xu-Dubois YC, Cornaire E, Vandermeersch S, Mesnard L, Hertig A, Rondeau E: Epithelial phenotypic changes detect cyclosporine in vivo nephrotoxicity at a reversible stage. Transplantation 92: 993–998, 201121909056
6. Ojo AO, Held PJ, Port FK, Wolfe RA, Leichtman AB, Young EW, Arndorfer J, Christensen L, Merion RM: Chronic renal failure after transplantation of a nonrenal organ. N Engl J Med 349: 931–940, 200312954741
7. Macian F: NFAT proteins: key regulators of T-cell development and function. Nat Rev Immunol 5: 472–484, 200515928679
8. Im SH, Rao A: Activation and deactivation of
gene expression by Ca2+/calcineurin-NFAT-mediated signaling. Mol Cells 18: 1–9, 200415359117
9. Kopp JB, Klotman PE: Cellular and molecular mechanisms of cyclosporin nephrotoxicity. J Am Soc Nephrol 1: 162–179, 19902104260
10. Gooch JL, Toro JJ, Guler RL, Barnes JL: Calcineurin A-alpha but not A-beta is required for normal kidney development and function. Am J Pathol 165: 1755–1765, 200415509543
11. Gooch JL, Roberts BR, Cobbs SL, Tumlin JA: Loss of the alpha-isoform of calcineurin is sufficient to induce nephrotoxicity and altered expression of transforming growth factor-beta. Transplantation 83: 439–447, 200717318077
12. Hong F, Lee J, Piao YJ, Jae YK, Kim YJ, Oh C, Seo JS, Yun YS, Yang CW, Ha J, Kim SS: Transgenic mice overexpressing cyclophilin A are resistant to cyclosporin A-induced nephrotoxicity via peptidyl-prolyl cis-trans isomerase activity. Biochem Biophys Res Commun 316: 1073–1080, 200415044094
13. Pallet N, Bouvier N, Bendjallabah A, Rabant M, Flinois JP, Hertig A, Legendre C, Beaune P, Thervet E, Anglicheau D: Cyclosporine-induced endoplasmic reticulum stress triggers tubular phenotypic changes and death. Am J Transplant 8: 2283–2296, 200818785955
14. Langworthy M, Zhou B, de Caestecker M, Moeckel G, Baldwin HS: NFATc1 identifies a population of proximal tubule cell progenitors. J Am Soc Nephrol 20: 311–321, 200919118153
15. Pallet N, Rabant M, Xu-Dubois YC, Lecorre D, Mucchielli MH, Imbeaud S, Agier N, Hertig A, Thervet E, Legendre C, Beaune P, Anglicheau D: Response of human renal tubular cells to cyclosporine and sirolimus: a toxicogenomic study. Toxicol Appl Pharmacol 229: 184–196, 200818331748
16. Slattery C, Campbell E, McMorrow T, Ryan MP: Cyclosporine A-induced renal fibrosis: a role for epithelial-mesenchymal transition. Am J Pathol 167: 395–407, 200516049326
17. Choi YM, Cho HY, Anwar MA, Kim HK, Kwon JW, Choi S: ATF3 attenuates cyclosporin A-induced nephrotoxicity by downregulating CHOP in HK-2 cells. Biochem Biophys Res Commun 448: 182–188, 201424768635
18. Pallet N, Bouvier N, Beaune P, Legendre C, Thervet E, Anglicheau D: [Endoplasmic reticulum stress in kidney diseases: a question of life and death?]. Nephrol Ther 5: 173–180, 200919071081
19. Kilberg MS, Shan J, Su N: ATF4-dependent transcription mediates signaling of amino acid limitation. Trends Endocrinol Metab 20: 436–443, 200919800252
20. Han J, Back SH, Hur J, Lin YH, Gildersleeve R, Shan J, Yuan CL, Krokowski D, Wang S, Hatzoglou M, Kilberg MS, Sartor MA, Kaufman RJ: ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nat Cell Biol 15: 481–490, 201323624402
21. Sumpio B, Baue AE, Chaudry IH: Treatment with verapamil and adenosine triphosphate-MgCl2 reduces
cyclosporine nephrotoxicity. Surgery 101: 315–322, 19872950608
22. Hazzan M, Hertig A, Buob D, Copin MC, Noël C, Rondeau E, Dubois-Xu YC: Epithelial-to-mesenchymal transition predicts
cyclosporine nephrotoxicity in renal transplant recipients. J Am Soc Nephrol 22: 1375–1381, 201121719789
23. Hertig A, Anglicheau D, Verine J, Pallet N, Touzot M, Ancel PY, Mesnard L, Brousse N, Baugey E, Glotz D, Legendre C, Rondeau E, Xu-Dubois YC: Early epithelial phenotypic changes predict graft fibrosis. J Am Soc Nephrol 19: 1584–1591, 200818434568
24. Hertig A, Verine J, Mougenot B, Jouanneau C, Ouali N, Sebe P, Glotz D, Ancel PY, Rondeau E, Xu-Dubois YC: Risk factors for early epithelial to mesenchymal transition in renal grafts. Am J Transplant 6: 2937–2946, 200617061992
25. Averous J, Lambert-Langlais S, Cherasse Y, Carraro V, Parry L, B’chir W, Jousse C, Maurin AC, Bruhat A, Fafournoux P: Amino acid deprivation regulates the stress-inducible gene p8 via the GCN2/ATF4 pathway. Biochem Biophys Res Commun 413: 24–29, 201121867687
26. Jin HO, Seo SK, Woo SH, Choe TB, Hong SI, Kim JI, Park IC: Nuclear protein 1 induced by ATF4 in response to various stressors acts as a positive regulator on the transcriptional activation of ATF4. IUBMB Life 61: 1153–1158, 200919946894
27. Wang C, Tian Y, Lei B, Xiao X, Ye Z, Li F, Kijlstra A, Yang P: Decreased IL-27 expression in association with an increased Th17 response in Vogt-Koyanagi-Harada disease. Invest Ophthalmol Vis Sci 53: 4668–4675, 201222669715
28. Sharma A, Mucino MJ, Ronco C: Renal functional reserve and renal recovery after acute kidney injury. Nephron Clin Pract 127: 94–100, 201425343829
29. Jacobson PA, Schladt D, Israni A, Oetting WS, Lin YC, Leduc R, Guan W, Lamba V, Matas AJ; DeKAF Investigators: Genetic and clinical determinants of early, acute calcineurin inhibitor-related nephrotoxicity: results from a kidney transplant consortium. Transplantation 93: 624–631, 201222334041
30. Baksh S, Widlund HR, Frazer-Abel AA, Du J, Fosmire S, Fisher DE, DeCaprio JA, Modiano JF, Burakoff SJ: NFATc2-mediated repression of cyclin-dependent kinase 4 expression. Mol Cell 10: 1071–1081, 200212453415
31. Feske S, Giltnane J, Dolmetsch R, Staudt LM, Rao A: Gene regulation mediated by calcium signals in T lymphocytes. Nat Immunol 2: 316–324, 200111276202
32. Kim HB, Kong M, Kim TM, Suh YH, Kim WH, Lim JH, Song JH, Jung MH: NFATc4 and ATF3 negatively regulate adiponectin
gene expression in 3T3-L1 adipocytes. Diabetes 55: 1342–1352, 200616644691
33. Gerbal-Chaloin S, Daujat M, Pascussi JM, Pichard-Garcia L, Vilarem MJ, Maurel P: Transcriptional regulation of CYP2C9 gene. Role of glucocorticoid receptor and constitutive androstane receptor. J Biol Chem 277: 209–217, 200211679585
34. Kang HM, Ahn SH, Choi P, Ko YA, Han SH, Chinga F, Park AS, Tao J, Sharma K, Pullman J, Bottinger EP, Goldberg IJ, Susztak K: Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development. Nat Med 21: 37–46, 201525419705
35. Goruppi S, Iovanna JL: Stress-inducible protein p8 is involved in several physiological and pathological processes. J Biol Chem 285: 1577–1581, 201019926786
36. Goruppi S, Bonventre JV, Kyriakis JM: Signaling pathways and late-onset gene induction associated with renal mesangial cell hypertrophy. EMBO J 21: 5427–5436, 200212374743
37. Yang L, Besschetnova TY, Brooks CR, Shah JV, Bonventre JV. Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after injury.
Nat Med 16: 535-543, 1p following 143, 2010
38. Vasseur S, Hoffmeister A, Garcia-Montero A, Mallo GV, Feil R, Kühbandner S, Dagorn JC, Iovanna JL: p8-deficient fibroblasts grow more rapidly and are more resistant to adriamycin-induced apoptosis. Oncogene 21: 1685–1694, 200211896600
39. Elzinga LW, Rosen S, Bennett WM: Dissociation of glomerular filtration rate from tubulointerstitial fibrosis in experimental chronic cyclosporine nephropathy: role of sodium intake. J Am Soc Nephrol 4: 214–221, 19938400085
40. Sartor MA, Leikauf GD, Medvedovic M: LRpath: a logistic regression approach for identifying enriched biological groups in
gene expression data. Bioinformatics 25: 211–217, 200919038984
41. Legouis D, Bataille A, Hertig A, Vandermeersch S, Simon N, Rondeau E, Galichon P: Ex vivo analysis of renal proximal tubular cells. BMC Cell Biol 16: 12, 201525881040
42. Bens M, Vallet V, Cluzeaud F, Pascual-Letallec L, Kahn A, Rafestin-Oblin ME, Rossier BC, Vandewalle A: Corticosteroid-dependent sodium transport in a novel immortalized mouse collecting duct principal cell line. J Am Soc Nephrol 10: 923–934, 199910232677