Abstract
Renal interstitial fibrosis is believed to play a key role in the development of diabetic nephropathy (DN), and advanced glycation end-products (AGE) may contribute importantly to this. Recent reports have shown that nitric oxide (NO) is closely linked to the renal interstitial fibrosis of DN. In this study, the mechanisms by which NO and its downstream signals mediate the AGE-induced proliferative response in normal rat kidney fibroblasts (NRK-49F) are examined. AGE decreased NO production, cyclic guanosine 5′monophosphate (cGMP) synthesis, and cGMP-dependent protein kinase (PKG) activation time- and dose-dependently. These effects were not observed when cells were treated with nonglycated BSA. NO and inducible nitric oxide synthase (iNOS) stimulated by NO donors S-nitroso-N-acetylpenicillamine (SNAP)/sodium nitroprusside (SNP) and PKG activator 8-para-chlorophenylthio-cGMP (8-pCPT-cGMP) prevented both AGE-induced proliferation and Janus kinase 2 (JAK2)-signal transducers and activators of transcription 5 (STAT5) activation but not p42/p44 mitogen-activated protein kinase (MAPK) activation. The ability of NO-PKG to inhibit AGE-induced cell cycle progression was verified by the observation that SNAP, SNP, and 8-pCPT-cGMP inhibited both cyclin D1 and cdk4 activation. Furthermore, induction of NO-PKG significantly increased p21Waf1/Cip1 expression in AGE-treated NRK-49F cells. The data suggest that the NO-PKG pathway inhibits AGE-induced proliferation by suppressing activation of JAK2-STAT5 and cyclin D1/cdk4 and induction of p21Waf1/Cip1.
Diabetic nephropathy (DN) is characterized by an early period of renal growth with glomerular and tubular cell hypertrophy, but this is followed by progressive glomerulosclerosis and tubulointerstitial fibrosis (1, 2). Accumulating studies emphasized the crucial role of renal interstitial fibrosis as a mediator of development of DN (3–5). Renal interstitial fibrosis is characterized by aberrant growth of the renal fibroblasts and increased expression of interstitial extracellular matrix components (5–7). Moreover, renal fibroblasts can contribute to the increased expression of the basement membrane component collagen in renal interstitial fibrosis observed during DN (3, 8, 9). However, the molecular mechanisms underlying this have not been fully elucidated.
Renal accumulation of advanced glycation end products (AGE) has been closely linked to the progression of DN (10–12). Our previous studies have showed that the Janus kinase 2 (JAK2)-signal transducers and activators of transcription (STAT)-1/3/5 pathways were necessary for AGE-induced cellular mitogenesis and cell cycle progression in normal rat kidney fibroblasts (NRK-49F) (13, 14). Recent studies demonstrate that AGE activate the receptor for AGE (RAGE)–extracellular signal-regulated kinase (ERK)-1/2 pathway to mediate the early tubular epithelial-myofibroblast transdifferentiation (TEMT) process (15). AGE-RAGE may induce the expression of TGF-β and other cytokines that are proposed to mediate the transdifferentiation of epithelial cells to form myofibroblasts (16).
Nitric oxide (NO) is a key mediator in renal physiology and pathology (17, 18). Alterations of intrarenal NO synthesis play an important role in the pathogenesis and progression of diabetic complications (19). In DN, NO may exert destructive effects (peroxynitrate-mediated tissue injury, hyperfiltration) as well as exhibit certain protective properties (reduced TGF-β expression and extracellular matrix expansion) (20–22). NO inhibits cell proliferation via inhibition of polyamine synthesis and cell uptake and may well act as a “brake” on the proliferative response after cytokine exposure (23). In rat glomerular mesangial cells, MAPK inhibition and an antiproliferative effect could be induced by an increase in the cellular concentration of NO (24), whereas reduced NO synthesis by glomerular endothelial cells and increased proliferation of glomerular mesangial cells is associated with glomerular remodeling that leads to accelerate glomerulosclerosis (25). Nevertheless, the effect of NO on renal interstitial fibrosis and how NO mediates AGE-induced biologic responses remain elusive.
The cyclic guanosine 5′ monophosphate (cGMP)-dependent protein kinases (PKG) are emerging as important components of mainstream signal transduction pathways (23, 26). NO-induced cGMP formation by stimulation of soluble guanylate cyclase is generally accepted as being the most widespread mechanism underlying PKG activation. PKG phosphorylates select tyrosine and serine/threonine residues on its substrate proteins, leading to subsequent activation/inactivation signals in the cell (27, 28). Accumulating evidence suggests that NO modulates gene expression. This involves activation or inhibition of different transcription factors such as AP-1, c-Myc, NF-κB, and Sp1 (29–32). Inhibition of DNA synthesis and proliferation by cGMP analogs is strictly dependent on PKG, and PKG activation inhibits the Ras-MAP kinase pathway (33). However, the relationship between the NO-PKG and JAK-STAT pathway remains poorly understood.
Therefore, in this study we investigated the effect of AGE on NO production and the antiproliferative mechanisms of NO-PKG in the AGE-induced proliferation in NRK-49F cells. To better understand the mechanisms underlying these actions, we further assessed the role of NO/PKG activation in cell cycle progression in the AGE-stimulated JAK-STAT signal cascade.
Materials and Methods
Reagents
FBS, DMEM, antibiotics, molecular weight standards, trypsin-EDTA, trypan blue stain, and all medium additives were obtained from Life Technologies, Inc. (Glasgow, UK). Anti-JAK2, -STAT5, -iNOS, -p42/p44 MAPK, -cyclin D1, -cdk4, and p21Waf1/Cip1 antibodies were obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). Antiphospho-p42/p44 MAPK antibody was purchased from New England Biolabs (Beverly, MA). Antiphospho-JAK2 and antiphospho-STAT5 antibodies were obtained from Upstate Biotechnology Inc. (Lake Placid, NY). Protein A/G-coupled agarose beads, SNAP, SNP, 8-para-chlorophenylthio-cGMP (8-pCPT-cGMP), KT5823, and anti-PKG-I antibody were purchased from Calbiochem (La Jolla, CA). Horseradish peroxidase (HRP)-conjugated goat anti-rabbit or anti-mouse secondary antibody, [γ-32P]ATP, and the enhanced chemiluminescence (ECL) kit were obtained from Amersham Corp. (Piscataway, NJ). N,N′-methylenebisacrylamide, acrylamide, SDS, ammonium persulfate, Temed, and Tween 20 were purchased from Bio-Rad Laboratories (Richmond, CA). cGMP enzymatic immunoassay (EIA) set was purchased from Amersham Pharmacia Biotech (Buckinghamshire, UK). Bovine serum albumin, anti–β-actin antibody, DMSO, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) colorimetric assay kit, and all other chemicals were obtained from Sigma Chemical Company (St. Louis, MO).
Culture Conditions
NRK-49F cells were obtained from the American Type Culture Collection (Manassas, VA). Cells were grown in culture flasks and maintained in DMEM (5.5 mM D-glucose) supplemented with 100 IU/ml penicillin, 100 μg/ml streptomycin, and 5% FBS in a humidified 5% CO2 incubator at 37°C. In some experiments, cells were exposed to serum-free (0.1% FBS) DMEM supplemented with the specific PKG inhibitor, KT5823, for 16 h before timed exposure to FBS and AGE. KT5823 was dissolved in DMSO. Cell viability was assessed by the trypan blue exclusion test and was routinely >92%. For cell number analysis, cells (1.5 × 105 cells/well) were cultured in 6-well culture plates (Nunclon) and grown in the added test agents. Cells were harvested and counted with a hemocytometer. Cells between passages 10 and 25 were used in all experiments. Each experimental data point represents the mean of duplicate wells from three independent experiments.
Preparation of AGE
AGE-BSA was prepared according to standard procedures: 20% (wt/vol) fatty acid-free BSA was incubated at 37°C under sterile phosphate-buffered saline (PBS), pH 7.2, supplemented with 250 mM glucose-6-phosphate, 100 μg/ml gentamycin, and 10 μg/ml ceftriaxone for 8 wk. Unincorporated glucose-6-phosphate and antibiotics were then removed by dialysis against PBS. Control nonglycated BSA was incubated in the same conditions with the exception of the absence of glucose-6-phosphate. They were then sterile-filtered through 0.2-μm nylon filters. Preparations were tested for endotoxin using the Endospecy ES-20S system; no endotoxin was detectable. The AGE content of the preparations was determined spectrofluorometrically with excitation set at 390 nm and emission set at 450 nm, and expressed as the percentage of relative fluorescence compared with nonincubated aliquots of the same batch of BSA in glucose-6-phosphate solution, stored frozen immediately after preparation.
NO Analysis
NO levels were measured with the Griess method. In brief, 1.2 × 103 cells were plated in each well of a 96-well plate in DMEM medium with 5% FBS. After being passed through 50-kD ultrafilters, 20 μl of the medium was diluted with 120 μl assay buffer and mixed with 5 μl cofactor and 5 μl nitrate reductase (NO colorimetric assay kit, Calbiochem). After the medium had been kept at room temperature for 2 h to convert nitrate to nitrite, total nitrite was measured at 540 nm absorbance by reaction with Griess reagent (sulfanilamide and naphthalene–ethylene diamine dihydrochloride). Amounts of nitrite in the medium were estimated by a standard curve obtained from enzymatic conversion of NaNO3 to nitrite.
cGMP and Protein Determination
Cells were grown in 24-well plates, 1.2 × 104 cells/well. Sixteen hours before stimulation, culture media were replaced by serum-free DMEM supplemented with the specific inhibitors or other drugs. Basal cGMP level was checked in the wells to which a corresponding volume of incubation medium without activators was added. Reaction was terminated by adding 10% (vol/vol) ice-cold lysis buffer A from cGMP EIA kit. After 30 min of extraction on ice, the supernatant was collected to the Eppendorf tubes and stored at −20°C until cGMP assay. cGMP was determined using the acetylation method in a cGMP immunoassay system as described in the manual provided by the manufacturer. For protein measurement, the cells were rinsed twice with PBS and solubilized in 500 μl solution of SDS in 0.01 N NaOH. The 50-μl aliquots were used for Bio-Rad protein assay. Measurements were performed in triplicate.
Preparation of Nuclear Extracts
Nuclear extracts were prepared as in our previous study (13). Briefly, cells were maintained in 5% FBS medium for 1 d. After fasting (0.1% FBS) for 48 h, fresh DMEM (5% FBS) with different concentrations of AGE or agents were added. After stimulation, cells were harvested and vigorously vortexed. Then cell lysates were centrifuged and nuclear pellets were resuspended in nuclear extraction buffer. Nuclear proteins were measured for protein by using a Bio-Rad protein assay kit. The extracts were stored at −70°C for further use.
Immunoprecipitation and Western Blotting
For protein analysis, 1.5 × 107 serum-deprived cells were treated with agents or AGE as described above. Total cell lysates were harvested, resolved by 10% SDS-polyacrylamide gel electrophoresis (SDS-PAGE), and then transferred to Protran membranes (0.45 μm; Schieicher & Schuell, Keene, NH). The membranes were blocked in blocking solution and subsequently probed with primary antibody (1 μg/ml). The membrane was incubated in 4000× diluted HRP-conjugated goat anti-rabbit or anti-mouse secondary antibody. The protein bands were detected using the ECL system, and the percentage of phosphorylated form of protein was determined using a scanning densitometer.
For p42/p44 MAPK and JAK/STAT activation assays, cell extracts were immunoprecipitated with p42/p44 MAPK and JAK/STAT monoclonal antibodies and with protein A/G-agarose beads. Then the samples were resolved by SDS-PAGE, and transferred to Protran membranes. The membranes were probed with anti-phospho-p42/p44 MAPK (0.75 μg/ml), antiphospho-JAK2 (0.75 μg/ml), anti-phospho-STAT5 (0.75 μg/ml), anti-p42/p44 MAPK (1:1000), anti-JAK2 (1:1000), and anti-STAT5 (1:1000) antibodies. The anti-phospho-STAT5 polyclonal antibody detects the tyrosine-phosphorylated forms of both STAT5a and STAT5b. Immunoreactive proteins were detected with the ECL system as described above.
Assay of PKG Activity
Cells were treated with agents or AGE as described above. After rinsing the monolayer with PBS, cells were harvested and homogenized with cold buffer consisting of 20 mM sodium phosphate (pH 6.8), 2 mM EDTA, 15 mM 2-mercaptoethanol, 150 mM NaCl, 2 mM benzamidine, 0.1 mM phenylmethylsulfonyl fluoride, 10 μg/ml pepstatin A, and 10 μg/ml leupeptin. The suspension was centrifuged for 10 min at 14,000 × g to obtain cell extract. Aliquots of extract were analyzed for PKG activity and also for Western blotting with an affinity-purified polyclonal rabbit anti-PKG-I antibody (1:500). The anti-PKG-I polyclonal antibody detects the activation forms of both PKG-Iα and PKG-Iβ. PKG activity was assayed as described in Suhasini et al. (33) and Soh et al. (34) with a peptide substrate (RKISASEFDRPL) selective for PKG. The difference in the phosphorylation of substrate in the presence and absence of cGMP was taken as PKG activity.
Cdk4 Kinase Assay
For immunoprecipitation of cdk4, cells were washed twice with ice-cold PBS, harvested in cdk4 lysis buffer [50 mM HEPES (pH 7.5), 10% glycerol, 150 mM NaCl, 1 mM EDTA, 2.5 mM EGTA, 1 mM dithiothreitol, and 0.1% Tween 20, supplemented with the phosphatase and protease inhibitors 5 mM NaF, 0.1 mM sodium orthovanadate, 5 μg/ml leupeptin, 10 μg/ml aprotinin, 50 μg/ml phenylmethylsulfonyl fluoride, and 5 μg/ml pepstatin A], and lysed by repeated passages through a 25-gauge needle. Cellular debris was removed from soluble extracts by centrifugation at 16,000 × g for 10 min at 4°C. After normalization of protein content, lysates were precleared by incubation with protein A/G-agarose beads and preimmune rabbit serum for 30 min at 4°C. Endogenous cdk4-containing complexes were immunoprecipitated for 3 h at 4°C, using a rabbit polyclonal anti-human cdk4 antibody. Immunoprecipitates were washed twice with cdk4 lysis buffer and four times with glutathione S-transferase-RB (GST-RB) kinase buffer [50 mM HEPES (pH 7.5), 10 mM MgCl2, 1 mM dithiothreitol, 2.5 mM EGTA, 10 mM β-glycerophosphate, 0.1 mM orthovanadate, 1 mM NaF] and then resuspended in 50 μl of GST-RB kinase buffer.
Kinase activity associated with anti-cdk4 immunocomplexes was assayed in 50 μl of GST-RB kinase buffer containing 2 μg of GST-RB substrate, and in each case supplemented with 2 mM EGTA and 2 μCi of [γ-32P]ATP. Reactions were carried out for 30 min at room temperature; cold ATP (final concentration, 30 μM) was then added to each reaction mixture to reduce background signal. Reactions were stopped by addition of Laemmli sample buffer, and the reaction products were electrophoresed in 12% SDS-PAGE, whereupon the gels were dried, visualized by autoradiography, and quantitated with a scanning densitometer.
Synthesis of Oligodeoxynucleotides
The sequences of phosphorothioate double-stranded oligodeoxynucleotides (ODN) used in this study were synthesized using a DNA/RNA synthesizer (Applied Biosystems, Foster City, CA). An ODN containing the STAT5 binding, γ-IFN–activated sequence (GAS)-like element from the β-casein promoter (35), was used as a probe. The sequences of STAT5 ODN were 5′-TGCTTCTTGGAATT-3′, 3′-ACGAAGAACCTTAA-5′. The single-stranded ODN were annealed for 2 h as temperature descended from 80°C to 25°C.
Electrophoretic Mobility Shift Assay
Electrophoretic mobility shift assay (EMSA) was performed as in our previous study (13). Briefly, 32P-labeling of STAT5 ODN was carried out using T4-polynucleotide kinase (New England BioLabs) and [γ-32P]ATP (3000 Ci/mmol). Labeled DNA was separated from the unincorporated radioactivity. Binding reactions were carried out by adding 5 μg of nuclear protein to 20 μl of binding buffer and [γ-32P]ATP labeled STAT5 ODN probes. Where indicated, cold competitive oligonucleotides were included during the preincubation periods. Samples were incubated at room temperature for 25 min and fractionated by electrophoresis. After electrophoresis, gels were transferred to filter paper, dried, and exposed to x-ray Hyperfilm-MP at −70°C using an intensifying screen. The results were quantified by a scanning densitometer.
MTT Assay
MTT assays were performed to evaluate the proliferation of NRK-49F cells. Briefly, 100 μl of each cell type (1 × 105 cells/ml) were plated and incubated for 24 h in wells of a 96-well plate. Then various concentrations of each drug were added to the wells. After another 24 h incubation, 10 μl of sterile MTT dye were added, and the cells were incubated for 6 h at 37°C. Then 100 μl of acidic isopropanol (0.04 M HCl in isopropanol) were added and thoroughly mixed. Spectrometric absorbance at 595 nm (for formazan dye) was measured with the absorbance at 655 nm for reference.
Flow-Cytometric Analysis of the Cell Cycle
For flow-cytometric experiments to determine DNA content and the cell cycle profile, cells were plated at a density of 1 × 106 to 2 × 106 cells per T-25 flask. At various time points, cells were harvested and fixed with ice-cold 100% ethanol with vortexing at low speed, and cells were then stored at −20°C for overnight. After fixation, cells were centrifuged and washed once with PBS containing 1% bovine serum albumin. For staining with DNA dye, cells were resuspended in 0.5 to 1 ml of propidium iodide (PI) solution containing RNase and incubated at 37°C for 30 min, and then they were incubated overnight at 4°C. Cell cycle profiles were obtained with a FACScan flow cytometer (Becton Dickinson, San Jose, CA) and data were analyzed with ModFit software (Verity Software House, Inc., Topsham, ME) for cell cycle analysis.
Statistical Analyses
The results were expressed as the mean ± standard errors of the mean (SEM). Unpaired t tests were used for the comparison between two groups. One-way ANOVA followed by unpaired t test was used for the comparison among more than three groups. P < 0.05 considered statistically significant.
Results
Effects of AGE on NO Production, cGMP Synthesis, and PKG-I Activation in NRK-49F Cells
We previously reported that AGE induces NRK-49F cell proliferation via activation of the JAK-STAT pathway (13). However, the involvement of NO in the proliferative effects of AGE is unclear. Therefore, the goal of this study was to elucidate the mechanisms by which NO and its downstream signals mediate the AGE-induced proliferative response in NRK-49F cells. To investigate whether AGE could affect NO production in NRK-49F cells, we treated these cells with nonglycated BSA and AGE and endogenously analyzed NO production by the Griess method. We found that AGE dose-dependently decreased nitrite production at 8 and 12 h (Figure 1). However, nonglycated BSA (100 μg/ml) did not induce any changes at the same times. It is well established that the downstream target of the NO pathway is PKG, and that many of the biologic effects of NO are mediated through the important kinase PKG. We examined whether NO blockade prevented cGMP synthesis and PKG activation. We found that AGE (100 μg/ml) significantly decreased cGMP synthesis and PKG activity at 1, 1.5, and 2 h (Figure 2, A and B), while nonglycated BSA (100 μg/ml) did not affect control cGMP synthesis and PKG activity in NRK-49F cells. We also found that the synthesis of PKG-I was significantly reduced by AGE but not by nonglycated BSA at 0.5, 1, and 2 h (Figure 2C). The activation and protein synthesis of PKG in the absence of added test agents were similar to nonglycated BSA (100 μg/ml) (data not shown). These results indicate that the biologic effects of AGE in NRK-49F cells may be caused by inhibition of the NO-cGMP-PKG pathway.
Time course and dose-dependent effects of advanced glycation end-products (AGE) on nitric oxide (NO) production in normal rat kidney fibroblasts (NRK-49F) cells. Serum-deprived NRK-49F cells (1.2 × 103 cells/well) were treated with serum-free (SF) medium, 5% FBS (control), AGE (50, 100, and 200 μg/ml) or BSA (100 μg/ml) for 0, 4, 8, and 12 h. NO production was determined by monitoring nitrite production by Griess assay as described in Materials and Methods. AGE (but not BSA) significantly decreased nitrite production at 8 and 12 h. Results were expressed as the mean ± SEM (n = 6). *P < 0.01 versus control.
Inhibition of cyclic guanosine 5′ monophosphate (cGMP) synthesis and cGMP-dependent protein kinase (PKG) activity by AGE in NRK-49F cells. Serum-deprived NRK-49F cells were treated with SF medium, BSA (100 μg/ml), or AGE (100 μg/ml) for 0, 0.5, 1, 1.5, and 2 h, and then assayed for cGMP synthesis (A) and PKG activity (B) as described in Materials and Methods. *P < 0.01 versus control. (C) Serum-deprived cells were treated with BSA (100 μg/ml) or AGE (100 μg/ml) for 0, 0.5, 1, and 2 h, and then total cell lysates were subjected to Western blot analysis for PKG-I. Results were expressed as the mean ± SEM (n = 4). *P < 0.01 versus BSA.
Effects of NO Donors, PKG Inhibitor, and PKG Activator on AGE-Inhibited NO-PKG Pathway
To investigate whether exogenously induced NO/PKG could affect AGE-inhibited NO-PKG pathway in NRK-49F cells, we treated these cells with NO donors, PKG inhibitor, and PKG activator and examined iNOS synthesis, nitrite production, and PKG activation in the presence of BSA (100 μg/ml) and AGE (100 μg/ml). Figure 3 illustrates that AGE significantly reduced iNOS synthesis and nitrite production at 8 h as compared with the control and BSA. Basal (control) nitrite production was not affected by BSA treatment. The NO donors SNAP (5 μM), SNP (5 μM), and the PKG activator 8-pCPT-cGMP (5 μM) markedly induced iNOS synthesis and nitrite production in the presence of AGE at 8 h. However, the inhibitor of PKG, KT5823, slightly enhanced AGE-inhibited nitric oxide production in NRK-49F cells. We also examined whether the NRK-49F cells express other NOS isoenzymes and found that the amount of endothelial NOS or neuronal NOS in the cells is very small (data not shown). Furthermore, the effects of NO donors, PKG inhibitor, and PKG activator on AGE-inhibited cGMP synthesis and PKG activation were also tested in NRK-49F cells and similar results were obtained (Figure 4). These data suggest that AGE can inhibit iNOS synthesis and NO-PKG pathway and NO donors or PKG activator can reverse these AGE-induced effects.
Effects of NO donors, PKG inhibitor, and PKG activator on AGE-blocked inducible NO synthase (iNOS) synthesis and NO production. Serum-deprived NRK-49F cells were treated with S-nitroso-N-acetylpenicillamine (SNAP) (5 μM), sodium nitroprusside (SNP) (5 μM), KT5823 (1 μM), 8-para-chlorophenylthio-cGMP (8-pCPT-cGMP) (5 μM), and DMSO for 8 h in the presence of BSA or AGE, and then assayed for iNOS synthesis by Western blotting (A). Cells treated with the same conditions (1.2 × 103 cells/well) were assayed for nitrite production by Griess assay (B). SF and Control represent cells grown in the presence of 0.1% FBS and 5% FBS, respectively. DMSO was the internal control used to dissolve SNAP, SNP, KT5823, and 8-pCPT-cGMP. Results were expressed as the mean ± SEM (n = 6). *P < 0.01 versus AGE.
Effects of PKG inhibitor, NO donors, and PKG activator on AGE-inhibited cGMP and PKG-I synthesis and PKG activity. Total cell lysates from NRK-49F cells treated with KT5823 (1 μM), SNAP (5 μM), SNP (5 μM), and 8-pCPT-cGMP (5 μM) in the presence of AGE (100 μg/ml) for 2 h were subjected to analysis for synthesis of cGMP (A) and PKG-I (B). Serum-deprived NRK-49F cells were treated with the same test agents as above for 2 h, and then assayed for PKG activity as described in Materials and Methods (C). The concentration of BSA (control) was 100 μg/ml. These are representative experiments, each performed at least four times. *P < 0.01 versus AGE.
NO Donors and PKG Activator Block AGE-Induced Proliferation and JAK2-STAT5 Activation, But Not p42/p44 MAPK Activation
To determine whether the NO-PKG pathway played a role in AGE-induced mitogenesis, NO donors and PKG activator were used to pretreat NRK-49F cells in proliferation and JAK2-STAT5 activities. Both NO donors and PKG activator interfered with AGE-induced mitogenesis at 3 d (Table 1). This finding suggests that the antiproliferative effect of NO on AGE-induced proliferation is associated with PKG activation. Previously, we have shown that AGE induced tyrosine phosphorylation of JAK2 (but not JAK1, JAK3 or TYK2) and STAT5 at 15 min (14). Therefore, we also tested the effects of SNAP, SNP, and 8-pCPT-cGMP on AGE-induced JAK2-STAT5 activation at the same time. By using antibodies specific for JAK2 or phospho-JAK2, Western blots revealed that NO donors and PKG activator markedly reduced phospho-JAK2 without affecting JAK2 protein levels in AGE-treated NRK-49F cells (Figure 5, A and B). Interestingly, the PKG inhibitor KT5823 moderately enhanced the effect of AGE on phosphorylation of JAK2. Additionally, the experiments on phosphorylation of STAT5 showed similar results when cells were treated with AGE. To test whether the inhibition seen at the phosphorylation of STAT5 were also observed on protein-DNA binding activities, EMSA was performed. As shown in Figure 5C, the binding protein complexes were characterized by incubation with a 20-fold molar excess of unlabeled consensus ODN (Figure 5C, lane Comp.). Consistent with the results from the phosphorylation assays, treatment with SNAP, SNP, and 8-pCPT-cGMP inhibited AGE-enhanced STAT5 protein-DNA binding activities. On the other hand, recent studies have indicated that activation of the MAPK pathway is associated with AGE-increased cell proliferation (36, 37). Therefore, we also examined whether NO-PKG regulates p42/p44 MAPK activity in NRK-49F cells treated with AGE. We measured p42/p44 MAPK activity after exposure of cultured NRK-49F cells to FBS (0.1%), FBS (5%), BSA (100 μg/ml), and AGE (100 μg/ml). We found that AGE (100 μg/ml) markedly increased p42/p44 MAPK phosphorylation at 30 min in comparison with FBS (5%) and BSA (100 μg/ml) (data not shown). However, we found that p42/p44 MAPK phosphorylation was unaffected by NO donors or PKG activator when cells were treated with AGE (100 μg/ml) for 30 min (Figure 6). These observations demonstrate that JAK2-STAT5 but not MAPK may play roles as important signal mediators in the antiproliferative effects of the NO-PKG pathway in AGE-treated NRK-49F cells.
Effects of PKG inhibitor, NO donors, and PKG activator on AGE-induced phosphorylation of Janus kinase 2 (JAK2) and signal transducers and activators of transcription 5 (STAT5) and STAT5 protein-DNA binding activity. (A) Total cell lysates from NRK-49F cells treated with KT5823 (1 μM), SNAP (5 μM), SNP (5 μM), and 8-pCPT-cGMP (5 μM) in the presence of AGE (100 μg/ml) for 15 min were immunoprecipitated (IP) with anti-JAK2 and anti-STAT5. Immune complexes were separated by a polyacrylamide gel and immunoblotted (IB) with anti-phospho-JAK2 (P-JAK2) and anti-phospho-STAT5 (P-STAT5) antibodies (upper panels) or antibodies corresponding to the above antibodies (lower panels). (B) Laser densitometry of the gels shown in (A) and two additional tyrosine phosphorylation experiments. *P < 0.01 versus AGE. (C) Effects of KT5823 (1 μM), SNAP (5 μM), SNP (5 μM), and 8-pCPT-cGMP (5 μM) on AGE-induced STAT5 protein-DNA binding activities in NRK-49F cells. Serum-deprived cells were treated with the same above test agents for 15 min. Nuclear extracts were prepared and assayed by electrophoretic mobility shift assay (EMSA), as described in Materials and Methods. It was obvious that 20-fold molar excess of unlabeled STAT5 probe (Comp.) markedly abolished the DNA-protein complexes. Effects of BSA (100 μg/ml) on JAK2 and STAT5 activation are used as control. This is a representative experiment independently performed four times.
Effects of PKG inhibitor, NO donors, and PKG activator on AGE-induced phosphorylation of p42/p44 mitogen activated protein kinase (MAPK). Total cell lysates from NRK-49F cells treated with KT5823 (1 μM), SNAP (5 μM), SNP (5 μM), and 8-pCPT-cGMP (5 μM) in the presence of AGE (100 μg/ml) for 30 min were immunoprecipitated (IP) with anti-p42/p44 MAPK. Immune complexes were separated by a polyacrylamide gel and immunoblotted (IB) with anti-phospho-p42/p44 MAPK antibody (upper panel) or antibodies corresponding to the above antibodies (lower panel). This is a representative experiment independently performed four times.
Effects of PKG inhibitor, NO donors, and PKG activator on AGE-induced cellular mitogenesis in NRK-49F cells
NO Donors and PKG Activator Block the Effects of AGE on Cell Cycle Progression, Cyclin D1/cdk4 Activation, and p21Waf1/Cip1 Expression
To gain further insight into the mechanism exerted by NO-PKG, we next wished to determine whether activation of the NO-PKG pathway is responsible for inhibition of AGE-mediated cell cycle progression in NRK-49F cells. AGE-incubated cells were treated for the indicated times with KT5823, SNAP, SNP, and 8-pCPT-cGMP, and then examined by flow-cytometric analysis. We found that a significant proportion of the cells entered S phase after the start of AGE treatment, whereas control cells largely remained arrested in the G1 phase (Table 2). Clearly, SNAP, SNP, and 8-pCPT-cGMP caused potent G1 arrest in AGE-treated NRK-49F cells. No significant difference in percentage of S phase was observed between AGE and KT5823-treated cells. These results demonstrate that AGE can induce S phase entry, and activation of the NO-PKG pathway may also have the ability to induce cell cycle arrest from AGE-treated cells. To investigate the possible role of NO-PKG in cell cycle progression, we examined the effects of SNAP, SNP, and 8-pCPT-cGMP on AGE-regulated expression of cell cycle regulatory molecules and cdk4 kinase activation. SNAP, SNP, and 8-pCPT-cGMP can prevent synthesis of cyclin D1 but not cdk4 (Figure 7A). However, cdk4 kinase analysis reveals that both NO donors and PKG activators reduced cdk4 kinase activation in AGE-treated cells (Figure 7B). Furthermore, NO donors and PKG activator significantly reversed AGE-inhibited expression of p21Waf1/Cip1 (Figure 7A). These results indicated that suppression of cyclin D1/cdk4 activation and induction of p21Waf1/Cip1 are the underlying mechanisms by NO donors and PKG activator to promote inhibition of cell cycle progression.
Effects of PKG inhibitor, NO donors, and PKG activator on AGE-regulated expression of cell cycle regulatory molecules and cdk4 kinase activation. (A) Total cell lysates from NRK-49F cells treated with KT5823 (1 μM), SNAP (5 μM), SNP (5 μM), and 8-pCPT-cGMP (5 μM) in the presence of AGE (100 μg/ml) for 4 h were subjected to Western blot analysis for cyclin D1, cdk4, and p21Waf1/Cip1. (B) Serum-deprived NRK-49F cells were treated with the same above test agents for 4 h, and then assayed for cdk4 kinase activation was described in Materials and Methods. The concentration of BSA (control) was 100 μg/ml. These are representative experiments, each performed at least four times. *P < 0.01 versus AGE.
Effects of PKG inhibitor, NO donors, and PKG activator on AGE-induced cell cycle progression in NRK-49F cells
Discussion
The NO-PKG pathway has previously been shown to suppress mitogenesis in renal cells. However, the suppressive mechanism involved remained elusive. The results in this study provide the first direct evidence linking NO-PKG signaling to AGE-induced proliferation and cell cycle progression in renal fibroblasts. AGE may decrease NO and PKG levels in renal fibroblasts. Moreover, we show that induction of exogenous NO-cGMP-PKG signaling contributes to increase p21Waf1/Cip1 expression and inhibit cyclin D1/cdk4 activation and the JAK2-STAT5 pathway but not the p42/p44 MAPK pathway.
Several studies have indicated that advanced nephropathy leading to severe proteinuria, declining renal function, and hypertension is associated with a state of progressive NO deficiency (2–5). Several factors including hyperglycemia, AGE, increased oxidant stress, as well as activation of PKC and TGF-β, contribute to decreased NO production and/or availability (38). These effects are mediated through multiple mechanisms such as glucose quenching, inhibition, and/or posttranslational modification of NOS activity of both inducible and endothelial isoforms (10). Gradual accumulation of AGE and induction of plasminogen activator inhibitor-1 (PAI-1), resulting in the decreased expression of iNOS and reduced generation of nitric oxide, are proposed to be pathophysiologically critical for the maintenance phase of renal tubulointerstitial dysfunction (39).
NO synthesis is known to result from the oxidation of L-arginine by a family of NOS, but there are species differences of NO production. Although we found that iNOS (but not nNOS or eNOS) proteins were affected by AGE or nonglycated BSA treatments in NRK-49F cells, it is possible that the iNOS pathway did not appear to be the only source of NO produced by growth factors/cytokines stimulation in these cells. NO can be generated from the iNOS-independent mechanism between reactive oxygen species and L-arginine. We postulate that this is the second pathway of NO synthesis in NRK-49F cells, and AGE also are capable of inhibiting this enzyme-independent mechanism of NO production from these cells. Nevertheless, the exact mechanism of action of AGE remains incompletely understood. Thus, one of our future studies is needed to define the role and function of this enzyme-independent mechanism of NO formation in the pathogenesis and treatment of diabetic nephropathy.
Previous studies demonstrate that different mechanisms for regulating the NO-PKG pathway are present in cell types that are similar but are from different tissues, and they underscore the tissue specificity in the responses observed. In mammalian cells, two genes encoding PKG have been identified, type I and type II (40). We found that type I is the major PKG expressed in NRK-49F cells by Western blot analysis (data not shown). PKG regulates gene expression at both transcriptional and posttranscriptional levels. It increases gene expression of some proteins (c-fos, junB, and MAPK phosphatase I) and decreases expression of others (thrombospondin 1, gonadotropin-releasing hormones, and soluble guanylate cyclase) (41–43). The PKG may induce gene expression through cAMP response element binding protein (CREB), NF-κB, and other, as yet undefined, mechanisms (44, 45). In this study, we reported that PKG activation blocked JAK2 phosphorylation and subsequently decreased phosphorylation and protein-DNA binding activity of STAT5 transcription factor in AGE-treated NRK-49F cells. However, it is still unclear how other factors are involved in the PKG-mediated signaling pathway. Thus, further study is required to elucidate the molecular mechanism for NO-PKG-inhibited JAK2-STAT5 activation in cells.
Other recent studies have implicated both p42/p44 MAPK and p38 as key intermediates in AGE activation of RAGE signaling leading to NF-κB–mediated target gene expression (46). Type IV collagen is transcriptionally regulated by Smad1 under AGE stimulation in mesangial cells (47). Interestingly, our previous studies showed that RAGE and the JAK2-STAT1/STAT3 pathway were involved in AGE-induced collagen production in NRK-49F cells (48). Furthermore, in this study, we found that p42/p44 MAPK activity was increased in AGE-treated cells, but this activity was unaffected by SNAP, SNP, and 8-pCPT-cGMP. Our inability to detect AGE-induced p42/p44 MAPK activation in response to NO donors and PKG activator may be due to differences in cell types and exposure conditions.
NO is reported to inhibit different stages of the cell cycle including early and late G1 phase (49), and these antiproliferative mechanisms are thought to be mediated by the NO-dependent activation of cGMP and subsequent production of PKG. It has not previously been shown whether AGE modulates the expression and/or activity of PKG. These data suggest that AGE regulates PKG at the level of activity and PKG-I protein levels in NRK-49F cells. Because the decline in iNOS synthesis in high AGE is paralleled by decreases in NO, PKG activity is predicted to be low under high AGE conditions in NRK-49F cells. Some studies suggest that the cdk4 immunoprecipitates from whole cell lysates actually contain a complex mixture of diverse cdk4 complexes, and it has been shown that only some fraction of the total cdk4 exhibits kinase activity. In this regard, the exact composition and structure of active cyclin D1-cdk4 remain unclear, and the minimal number of proteins required to form active cyclin D/cdk4 complexes is unknown. It is possible that accumulation of p21Waf1/Cip1 reduces the abundance of cyclin D1, which perhaps limits the formation of cyclin D1-cdk4 complexes. In our study, we show that the enhancement of p21Waf1/Cip1 protein levels and inhibition of cyclin D1 expression correlated with the changes in the formation of active cyclin D-cdk4 complexes in NO donor/PKG activator-inhibited cell cycle progression, suggesting that the effects of NO-PKG on AGE-stimulated proliferation of NRK-49F cells are mediated by the decline of cyclin D1 accumulation and cdk4 activity. Neither AGE nor NO-PKG had any effect on the cdk4 protein levels in NRK-49F cells. Similarly, the absence of changes in cdk4 protein levels was also observed in the other renal cells, LLC-PK1 and Madin-Darby canine kidney (data not shown).
NO exerts complex regulatory actions on cultured renal cell biology, such as inhibition of proliferation, adhesion or contractility, and induction of apoptosis. To our knowledge, the data reported here are the first to demonstrate the role of NO-PKG on inhibition of the AGE-induced renal interstitial fibrosis. Despite this effect, only little is known about the molecular mechanisms that account for the lack of NO-PKG responses toward renal interstitial fibrosis. There has been no clear consensus on a single mechanism of growth inhibition by NO, and it is likely that multiple reaction pathways are involved. It has been shown that reduced NO may contribute to susceptibility to injury and renal fibrosis (18, 19). iNOS-derived NO modulates glomerulosclerosis and tubulointerstitial fibrosis in chronic streptozotocin nephropathy (17). This action is probably a result of the direct actions of NO on the synthesis and degradation of extracellular matrix proteins. NO modulation was associated with a change in TGF-β1 expression, which, in turn, was associated with alterations in matrix deposition and matrix degradation through its effect on PAI-1 (50). Thus, future study is required to elucidate the role of the NO-cGMP-PKG pathway in AGE-mediated TGF-β signaling in renal fibrosis.
Our proposed mechanism from this study is shown schematically in Figure 8. In particular, there is evidence that NO-PKG activation reduces the AGE-induced proliferation and cell cycle progression, possibly via effects on the JAK2-STAT5 pathway, cyclin D1/cdk4, and p21Waf1/Cip1 expressions. Thus, we suggest that the altered NO-cGMP-PKG pathway plays an important role in the pathogenesis of DN, and we suggest novel means of modulating AGE-dependent renal interstitial fibrosis.
A possible antiproliferative signal pathway in AGE-cultured renal fibroblasts by NO. The JAK2-STAT5 and p42/p44 MAPK cascades are important AGE-induced signaling pathways contributing to cyclin D1/cdk4 activation, cell cycle progression, and cell proliferation. Aberrant growth of the renal fibroblasts and increased expression of interstitial extracellular matrix components can contribute to renal interstitial fibrosis observed during diabetic nephropathy. In the presence of NO, the suppression of AGE-induced biologic responses is mediated by NO-dependent cGMP synthesis and PKG activation. The p21Waf1/Cip1 activation mediated by NO-cGMP-PKG subsequently leads to cell cycle arrest and cell growth inhibition. On the other hand, AGE may inhibit iNOS synthesis and then abolish the negative growth effects of NO.
Acknowledgments
This work was supported by a Research Grant (NSC-91–2314-B273–004 to J.-S.H.) from the National Science Council, Republic of China.
Footnotes
Published online ahead of print. Publication date available at www.jasn.org.
- © 2005 American Society of Nephrology