Deletion of p18INK4c aggravates cisplatin‑induced acute kidney injury

  • Authors:
    • Liang Wang
    • Wei Chen
    • Yi Zhang
    • Chunyan Liu
    • Li Yuan
    • Lili Fu
    • Changlin Mei
  • View Affiliations

  • Published online on: April 4, 2014     https://doi.org/10.3892/ijmm.2014.1725
  • Pages: 1621-1626
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Protection of cyclin-dependent kinase inhibitors (CDKIs) has been demonstrated in acute kidney injury (AKI). However, previous studies on CDKIs have mainly focused on CIP/KIP family members, with INK4 family members rarely being investigated. This study investigated the behaviors of p18INK4c (p18) in cisplatin-induced AKI using p18 gene knockout mice (p18-/-). AKI was induced in p18-/- and wild-type (p18+/+) mice after a single cisplatin (12.5 mg/kg) intraperitoneal injection. Protection by p18 was identified by a comparison of survival, renal function and morphological injuries between p18-/- and p18+/+ mice. Further investigation of endoplasmic reticulum stress (ERS) was performed by western blot analysis in p18-/- and p18+/+ kidneys at day 3 after cisplatin injection. The results revealed that after cisplatin injection, the survival of p18-/- mice was significantly shorter than that of p18+/+ mice, accompanied by aggravated renal function and more severe morphological injuries. Deletion of p18 also significantly aggravated ERS in cisplatin-induced AKI. In conclusion, p18 exerts protective effects on cisplatin‑induced AKI, which may be associated with the effect of p18 on cell death pathways, such as the ERS pathway.

Introduction

Acute kidney injury (AKI) is a life-threatening condition with high morbidity and mortality, even in patients who have received medical intervention. As there is a lack of effective remedies for AKI other than dialysis, experimental efforts are being made to explore the pathogenesis of AKI and to seek materials with therapeutic potency. Cell cycle arrest is known to be beneficial for repairing of damaged DNA, thereby reducing the severity and teratogenicity of the injury (1). For this reason, cyclin-dependent kinase inhibitors (CDKIs) have been much investigated and have proven to possess cell protective properties in AKI (210). It is therefore believed that cell cycle regulation is a potential remedy for the treatment of AKI.

Based on sequence and the inhibitory effects they exert on cyclin-dependent kinase (CDK), the seven CDKIs are divided into two families. The CIP/KIP family includes p21, p27 and p57, which act with multiple CDK and extensively inhibit the cell cycle (11). The INK4 family includes p16, p15, p18 and p19, which only interact with CDK4/6 and specifically arrest the cell cycle in early G1 phase (12). CDKIs studies in AKI have mainly focused on the CIP/KIP family, especially p21, which has been identified as the protective factor in AKI (510). Compared to CIP/KIP family members, the role of INK4 members in AKI has yet to be determined. However, previous studies have reported that some novel additional biological functions are present in INK4 family members, such as p16 and p19. P16 controlled apoptosis induced by ultraviolet light and cisplatin through the intrinsic mitochondrial cell death pathway (1314). Overexpression of p19 conferred resistance to cells exposed to UV irradiation (1516).

Therefore, we hypothesized the beneficial behaviors of p18 and investigated its role in cisplatin-induced AKI using p18−/− mice. As oxidative stress is important factor in cisplatin-induced AKI, we also investigated the effect of p18 on cisplatin-induced endoplasmic reticulum stress (ERS) in an attempt to elucidate the possible mechanism involved in p18 actions.

Materials and methods

Animals

P18+/−mice in a C57BL/6 and 129/Sv background were kind gifts from Professor Tao Cheng of the laboratory of Cancer Research Center at Pittsburgh University. P18−/− or p18+/+ mice were generated from p18+/− breeding pairs. The mice were genotyped by a PCR approach, using tail DNA as previously described (17).

The primer sequences for genotype identification included: p18 WT forward, 5′-AGCCATCAAATTTATTCATGTTGCAGG-3′; p18 MG-47 reverse, 5′-CCTCCATCAGGCTAATGACC-3′; and PGKNEO reverse, 5′-CCAGCCTCTGAGCCCAGAAAGCGAAGG-3′.

The detailed characteristics of the p18−/− mice were previously described by Franklin et al (18). Briefly, p18−/− mice grew and developed to become larger in body size than their p18+/+ littermates. Accordingly, the heart, liver and kidneys of the p18−/− mice exhibited proportional organomegaly; however, no abnormal structures, such as hepatic hypertrophy, glomerular sclerosis, diffuse kidney tubular atrophy, or dermal abnormalities, were detected in p18−/− mice.

Littermates or age-matched male mice (8–12 weeks) were used in our experiments. The animals were housed in a specific pathogen-free facility with access to water and food ad libitum at the Second Military Medical University Animal Center. All procedures were approved by the Ethics Committee of the Experimental Animals Center of the University.

Animal experiments

AKI was induced by a single intraperitoneal injection of cisplatin (Sigma, St. Louis, MO, USA) at a dose of 12.5 mg/kg in p18−/− (n=35) and p18+/+ (n=35) mice, while the controls (n=15) were injected with isovolumic saline.

After cisplatin injection, the 28-day survival of p18−/− (n=15) and p18+/+ (n=15) mice was determined, while the other animals were dealt with at day 3 after the injection. Blood samples were collected using the method of eye enucleation. Kidneys were collected after the animals were sacrificed by cervical dislocation. Kidneys and blood were collected at day 3 after cisplatin injection for morphological and renal function analysis. Serum creatinine (SCr) and urea nitrogen were determined by enzymatic colorimetric assay. Kidney tissues were stained with hematoxylin and eosin (H&E) and morphological assessment was determined under light microscopy by the same experienced histologist. Tubular necrosis, brush border loss and cast formation were used as the main damage parameters. Scoring was performed according to the percentage of damaged tubuli in the kidney as follows: I, 0–25%; II, 25–50%; III, 50–75%; IV, >75%.

Terminal deoxynucleotidyl-transferase-mediated dUTP nick end-labeling (TUNEL) and analysis of ERS signal proteins by quantitative PCR (qPCR) and western blot analysis were performed at day 3 after cisplatin injection for the p18−/− and p18+/+ kidneys.

TUNEL

A commercial kit (Fuzhou Maixin Biotechnology Development Co., Ltd., Fuzhou, China) was used to detect apoptotic cells for in situ kidneys. Briefly, paraffin-embedded sections were deparaffinized in xylene and rehydrated through graded concentrations of ethanol. After being washed with PBS, the sections were treated with 0.5% pepsin at 37°C for 8 min, and 0.3% Triton X-100 for 10 min at room temperature. To inactivate endogenous peroxidase, the sections were incubated in 3% H2O2 at 37°C for 15 min and then incubated with terminal deoxynucleotidyl transferase (TdT) in a humid chamber at 37°C for 1 h. The signals were detected with a horseradish peroxidase-conjugated sheep anti-alkaline phosphatase antibody. Quantitative measurement of apoptotic cells was performed by examining 10 randomly selected fields under a light microscope (magnification, ×400) in the cortex. Twelve sections from at least six animals of each group were counted, and the data were presented as the mean number of apoptotic cells in each HPF field. Differences were considered statistically significant if P<0.05.

qPCR

Total RNA was extracted from kidney tissues (renal cortex) by means of the TRIzol reagent (Invitrogen Life Technologies, Carlsbad, CA, USA), and an RT kit (Takara Bio, Inc., Shiga, Japan) was used to synthesize cDNA. The expression of signal proteins in ERS was determined by qPCR using the ABI PRISM 7000 Sequence Detection System, and PCR reactions were performed using the SYBR-Green real-time PCR Master mix (Toyobo Co., Ltd., Osaka, Japan). The ribosomal gene 18S (18S rRNA) was selected as an endogenous reference and the samples were assayed in triplicate. Based on the analysis by the ΔΔCt method, the expression of the target genes was determined. The primer sequences used for qPCR were: 18S rRNA forward, 5′-AGGAGTGGGCCTGCGGCTTA-3′ and reverse, 5′-GCCGGGTGAGGTTTCCCGTG-3′; Grp78 forward, 5′-AGACATTTGCCCCAGAAGAA-3′ and reverse, 5′-ATCTTTGGTTGCTTGTCGCT-3′; Grp94 forward, 5′-TGAAGGAGAAGCAGGACAAAA-3′ and reverse, 5′-AGTCGCTCAACAAAGGGAGA-3′; and CCAAT/enhancer-binding protein-homologous protein (CHOP) forward, 5′-TATCTCATCCCCAGGAAACG-3′ and reverse, 5′-GGACGCAGGGTCAAGAGTAG-3′.

Western blot analysis

Protein was extracted from the renal cortex using a lysis buffer containing 50 mM Tris-HCl (pH 8.0), 50 mM NaCl, 50 mM sodium fluoride, 0.1% Nonidet P-40, 5 mM EDTA, 2 mM phenylmethylsulfonyl fluoride, 1 mM dithiothreitol, 10 μg/ml leupeptin, 2 μg/ml pepstatin, and 1 μg/ml aprotinin. After a 30-min incubation on ice, the lysates were heated at 100°C for 15 min and centrifuged at 12,000 × g for 15 min at 4°C. Lysates containing equal amounts of proteins (100 μg) were dissolved in an SDS sample buffer, separated on 12% SDS slab gels and transferred electrophoretically onto polyvinylidene difluoride (PVDF) membranes. Equal protein loading and protein transfer was confirmed by Ponceau S staining. After blocking with 5% non-fat dry milk in TBST, the membrane was incubated at 4°C overnight with the following primary antibodies: mouse anti-GAPDH (1:5,000 dilution; Kangcheng Biotechnology, Shanghai, China), rabbit anti-p18 (1:200 dilution; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), rabbit anti-grp78 (1:1,000 dilution), rabbit anti-phosphorylation of pancreatic endoplasmic reticulum (ER) eukaryotic translation initiation factor 2α (eIF2α) kinase (PERK) (1:1,000 dilution), rabbit anti-phospho-PERK (1:1,000 dilution), rabbit anti-eIF2α (1:1,000 dilution) and rabbit anti-phospho-eIF2α (1:1,000 dilution) (all from Cell Signaling Technology, Beverly, MA, USA). After washing, a horseradish peroxidase-conjugated secondary antibody was applied. Proteins that bound to the secondary antibody were visualized using ECL (Amersham Pharmacia Biotech, Amersham, UK).

Statistical analysis

Data are presented as mean ± SD and were analyzed for significance using an ANOVA model. Comparisons between the two groups were made using the t-test or Wilcoxon-Mann-Whitney test. Differences were considered tatistically significant if P<0.05.

Results

Deletion of p18 aggravated cisplatin-induced AKI

As shown in Fig. 1, the 28-day survival of p18−/− mice was significantly worse than that of their p18+/+ counterparts. All 15 p18−/− mice died at day 7 after cisplatin injection, while the 7-day survival rate for the p18+/+ mice was 53.3%, with no deaths occurring in p18+/+ mice from day 8 to 28. A significant difference was observed between the survival curves of p18−/− and p18+/+ mice after cisplatin injection (P<0.05 for the log-rank test).

Compared to p18+/+ mice, aggravated urea nitrogen (Fig. 2A) and creatinine (Fig. 2B) of p18−/− mice was demonstrated at day 3 after cisplatin injection.

Aggravated morphological changes were present in p18−/− mice at day 3 after cisplatin injection as demonstrated by H&E staining (Fig. 3 and Table I) and TUNEL assessment (Fig. 4). A higher degree of kidney damage and a higher percentage of apoptotic cells were present in p18−/− kidneys as compared to p18+/+ kidneys at day 3 after cisplatin injection.

Table I

Histological assessment of p18−/− and p18+/+ mice at day 3 after cisplatin (12.5 mg/kg, i.p) injection.

Table I

Histological assessment of p18−/− and p18+/+ mice at day 3 after cisplatin (12.5 mg/kg, i.p) injection.

No. of mice in each grade

GroupsIIIIIIIV
p18−/− (n=15)0267
p18+/+ (n=15)0483

[i] For this assessment, 15 mice in each group were analyzed. The scoring was according to the proportion of damaged tubuli in the renal cortex, which was graded as follows: I, 0–25%; II, 25–50%; III, 50–75%; IV, >75%. Two slides for each mouse were randomly selected to determine the grade and all the slides were assessed by the same experienced histologist.

Deletion of p18 aggravated cisplatin-induced ERS

As shown in Fig. 5, the expression of molecular chaperones grp78 and grp94 mRNAs was upregulated in kidneys of animals with AKI at day 3 after cisplatin injection. However, compared to p18+/+ mice, the basal and inducible expression of grp78 and grp94 mRNAs was significantly higher in the p18−/− mice. Similar results were observed in the analysis of CHOP mRNA, a particular transcription factor activated by ERS.

Results were confirmed by western blot analysis (Fig. 6). The renal expression of grp78 protein was upregulated after cisplatin injection in p18−/− and p18+/+ mice. Compared to p18+/+ mice, the basal and inducible renal expression of grp78 protein was significantly higher in p18−/− mice.

As a rapid response to ERS, PERK was also analyzed by western blot analysis. The degree of PERK/eIF2α phosphorylation was higher in p18−/− mice as compared to that of p18+/+ mice after cisplatin injection (Fig. 7).

Discussion

Clinical use of cisplatin is largely limited due to drug resistance and nephrotoxicity (1920). Since the mechanism by which cisplatin produces its nephrotoxic effect is similar to human AKI (2122), AKI animals were administered cisplatin.

As the manifestation of cisplatin nephrotoxicity is apoptosis and/or necrosis, cell death pathways were involved in the mechanism of cisplatin nephrotoxicity. Previous studies have confirmed that in addition to the classical death-receptor and mitochondrial pathways (2326), the ERS pathway is activated in cisplatin-induced kidney injury in vitro and in vivo (2730). Therefore, the effect of p18 deletion on the ERS pathway was investigated to elucidate the actions of p18 in cisplatin-induced AKI. In ERS, the unfolded protein response (UPR) was identified and considered to interpret the mechanism of ERS-induced apoptosis (3135). Three transmembrane proteins are activated in UPR: inositol-requiring enzyme-1 (IRE1), PERK and activating translation factor-6 (ATF6). In this study, upregulation of the molecular chaperones and CHOP and activation of the PERK/eIF2α pathway were analyzed to evaluate ERS severity in cisplatin-induced AKI.

It was found that p18 exerted protective actions in cisplatin-induced AKI. Compared to p18+/+ mice, p18−/− mice exhibited a higher degree of kidney damage, accompanied with aggravated renal function and worse survival after cisplatin injection. Deletion of p18 also aggravated cisplatin-induced ERS. Compared to p18+/+ mice, the basal and inducible expression of the molecular chaperones (grp78 and grp94) and transcription factor (CHOP) in kidney were significantly higher in the p18−/− mice. The degree of PERK/eIF2α phosphorylation was also higher in p18−/− mice kidneys compared to p18+/+ mice after cisplatin injection. These results indicate that the effect of p18 on cell death pathways, such as the ERS pathway, may be the facet of its protective mechanism in cisplatin-induced AKI. However, other classical death pathways affected by p18 cannot be excluded as they were not the focus of our investigation.

P18, as a member of the INK4 family, is different from p21, whose protection in cisplatin-induced AKI has been demonstrated in previous studies (210). Protection of p21 occurs mainly due to the inhibitory effect on CDK2 activity, which has been demonstrated as an important factor in the promotion of apoptosis in cisplatin-induced AKI (3637). However, the INK4 family members only interact with CDK4/6 and arrest the cell cycle in the early G1 phase, with no direct interaction with CDK2 (12,38). INK4 family members are also considered to be involved more in cell differentiation than CIP/KIP family members as they are often mutant or deleted in a number of tumors (39). No abnormality or defect exists in p21 gene knockout mice, whereas p18 gene knockout mice acquire pituitary tumors with age (18,40). These limitations may be the possible reasons for INK4 members rarely being investigated in AKI. However, some studies have reported the involvement of INK4 family members in the cell response to genotoxic agents, such as p16 and p19 (1316), as well as p18. These observations suggest that INK4 family members also exert protection and are involved in organ injury, despite the differences between the INK4 and CIP/KIP family members.

In conclusion, protection of p18 was demonstrated in cisplatin-induced AKI by using p18 gene knockout mice in this study. The main results of this study are the finding that p18 regulates cell death pathways, such as the ERS pathway, against cisplatin-induced AKI, although the exact signal transduction pathways connecting p18 to cell death pathways remain to be investigated in detail.

Acknowledgements

We would like to thank Professor Tao Cheng for kindly providing the p18 gene knockout mice and Professor Jun Gu for his helpful suggestions and careful review of our manuscript.

References

1 

Weinert T: DNA damage and checkpoint pathways: molecular anatomy and interactions with repair. Cell. 94:555–558. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Megyesi J, Andrade L, Vieira JM Jr, Safirstein RL and Price PM: Coordination of the cell cycle is an important determinant of the syndrome of acute renal failure. Am J Physiol Renal Physiol. 283:F810–F816. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Zhou H, Kato A, Yasuda H, et al: The induction of cell cycle regulatory and DNA repair proteins in cisplatin-induced acute renal failure. Toxicol Appl Pharmacol. 200:111–120. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Price PM, Safirstein RL and Megyesi J: Protection of renal cells from cisplatin toxicity by cell cycle inhibitors. Am J Physiol Renal Physiol. 286:F378–F384. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Megyesi J, Safirstein RL and Price PM: Induction of p21WAF1/CIP1/SDI1 in kidney tubule cells affects the course of cisplatin-induced acute renal failure. J Clin Invest. 101:777–782. 1998. View Article : Google Scholar : PubMed/NCBI

6 

Zhou H, Fujigaki Y, Kato A, et al: Inhibition of p21 modifies the response of cortical proximal tubules to cisplatin in rats. Am J Physiol Renal Physiol. 291:F225–F235. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Nowak G, Price PM and Schnellmann RG: Lack of a functional p21WAF1/CIP1 gene accelerates caspase-independent apoptosis induced by cisplatin in renal cells. Am J Physiol Renal Physiol. 285:F440–F450. 2003.PubMed/NCBI

8 

Yu F, Megyesi J, Safirstein RL and Price PM: Identification of the functional domain of p21(WAF1/CIP1) that protects cells from cisplatin cytotoxicity. Am J Physiol Renal Physiol. 289:F514–F520. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Miyaji T, Kato A, Yasuda H, Fujigaki Y and Hishida A: Role of the increase in p21 in cisplatin-induced acute renal failure in rats. J Am Soc Nephrol. 12:900–908. 2001.PubMed/NCBI

10 

Nath KA: Provenance of the protective property of p21. Am J Physiol Renal Physiol. 289:F512–F513. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Hengst L and Reed SI: Inhibitors of the Cip/Kip family. Curr Top Microbiol Immunol. 227:25–41. 1998.

12 

Sherr CJ and Roberts JM: Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 9:1149–1163. 1995. View Article : Google Scholar : PubMed/NCBI

13 

Al-Mohanna MA, Manogaran PS, Al-Mukhalafi ZK, Al-Hussein AK and Aboussekhra A: The tumor suppressor p16(INK4a) gene is a regulator of apoptosis induced by ultraviolet light and cisplatin. Oncogene. 23:201–212. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Le HV, Minn AJ and Massagué J: Cyclin-dependent kinase inhibitors uncouple cell cycle progression from mitochondrial apoptotic functions in DNA-damaged cancer cells. J Biol Chem. 280:32018–32025. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Scassa ME, Marazita MC, Ceruti JM, et al: Cell cycle inhibitor, p19INK4d, promotes cell survival and decreases chromosomal aberrations after genotoxic insult due to enhanced DNA repair. DNA Repair (Amst). 6:626–638. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Tavera-Mendoza LE, Wang TT and White JH: p19INK4D and cell death. Cell Cycle. 5:596–598. 2006. View Article : Google Scholar

17 

Yuan Y, Shen H, Franklin DS, Scadden DT and Cheng T: In vivo self-renewing divisions of haematopoietic stem cells are increased in the absence of the early G1-phase inhibitor, p18INK4C. Nat Cell Biol. 6:436–442. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Franklin DS, Godfrey VL, Lee H, et al: CDK inhibitors p18(INK4c) and p27(Kip1) mediate two separate pathways to collaboratively suppress pituitary tumorigenesis. Genes Dev. 12:2899–2911. 1998. View Article : Google Scholar : PubMed/NCBI

19 

Wang D and Lippard SJ: Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 4:307–320. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Siddik ZH: Cisplatin: mode of cytotoxic action and molecular basis of resistance. Oncogene. 22:7265–7279. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Pabla N and Dong Z: Cisplatin nephrotoxicity: mechanisms and renoprotective strategies. Kidney Int. 73:994–1007. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Heyman SN, Lieberthal W, Rogiers R and Bonventre JV: Animal models of acute tubular necrosis. Curr Opin Crit Care. 8:526–534. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Lieberthal W, Triaca V and Levine J: Mechanisms of death induced by cisplatin in proximal tubular epithelial cells: apoptosis vs. necrosis Am J Physiol. 270:F700–F708. 1996.PubMed/NCBI

24 

Razzaque MS, Koji T, Kumatori A and Taguchi T: Cisplatin-induced apoptosis in human proximal tubular epithelial cells is associated with the activation of the Fas/Fas ligand system. Histochem Cell Biol. 111:359–365. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Seth R, Yang C, Kaushal V, Shah SV and Kaushal GP: p53-dependent caspase-2 activiation in mitochondrial release of apoptosis-inducing factor and its role in renal tubular epithelial cell injury. J Biol Chem. 280:31230–31239. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Park MS, De Leon M and Devarajan P: Cisplatin induces apoptosis in LLC-PK1 cells via activation of mitochondrial pathways. J Am Soc Nephrol. 13:858–865. 2002.PubMed/NCBI

27 

Muruganandan S and Cribb AE: Calpain-induced endoplasmic reticulum stress and cell death following cytotoxic damage to renal cells. Toxicol Sci. 94:118–128. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Cribb AE, Peyrou M, Muruganandan S and Schneider L: The endoplasmic reticulum in xenobiotic toxicity. Drug Metab Rev. 37:405–442. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Peyrou M, Hanna PE and Cribb AE: Cisplatin, gentamicin, and p-aminophenol induce markers of endoplasmic reticulum stress in the rat kidneys. Toxicol Sci. 99:346–353. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Liu H and Baliga R: Endoplasmic reticulum stress-associated caspase 12 mediates cisplatin-induced LLC-PK1 cell apoptosis. J Am Soc Nephrol. 16:1985–1992. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Bernales S, Papa FR and Walter P: Intracellular signaling by the unfolded protein response. Annu Rev Cell Dev Biol. 22:487–508. 2006. View Article : Google Scholar

32 

Malhotra JD and Kaufman RJ: The endoplasmic reticulum and the unfolded protein response. Semin Cell Dev Biol. 18:716–731. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Mori K: Tripartite management of unfolded proteins in the endoplasmic reticulum. Cell. 101:451–454. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Kleizen B and Braakman L: Protein folding and quality control in the endoplasmic reticulum. Curr Opin Cell Biol. 16:343–349. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Szegezdi E, Logue SE, Gorman AM and Samali A: Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep. 7:880–885. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Price PM, Yu F, Kaldis P, et al: Dependence of cisplatin-induced cell death in vitro and in vivo on cyclin-dependent kinase 2. J Am Soc Nephrol. 17:2434–2442. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Yu F, Megyesi J and Price PM: Cytoplasmic initiation of cisplatin cytotoxicity. Am J Physiol Renal Physiol. 295:F44–F52. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Hirai H, Roussel MF, Kato JY, Ashmun RA and Sherr CJ: Novel INK4 proteins, p19 and p18, are specific inhibitors of the cyclin D-dependent kinases CDK4 and CDK6. Mol Cell Biol. 15:2672–2681. 1995.PubMed/NCBI

39 

Cordon-Cardo C: Mutation of cell cycle regulators. Biological and clinical implaications for human neoplasia. Am J Pathol. 147:545–560. 1995.PubMed/NCBI

40 

Shankland SJ and Wolf G: Cell cycle regulatory proteins in renal disease: role in hypertrophy, proliferation, and apoptosis. Am J Physiol Renal Physiol. 278:F515–F529. 2000.PubMed/NCBI

Related Articles

Journal Cover

June-2014
Volume 33 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang L, Chen W, Zhang Y, Liu C, Yuan L, Fu L and Mei C: Deletion of p18INK4c aggravates cisplatin‑induced acute kidney injury. Int J Mol Med 33: 1621-1626, 2014
APA
Wang, L., Chen, W., Zhang, Y., Liu, C., Yuan, L., Fu, L., & Mei, C. (2014). Deletion of p18INK4c aggravates cisplatin‑induced acute kidney injury. International Journal of Molecular Medicine, 33, 1621-1626. https://doi.org/10.3892/ijmm.2014.1725
MLA
Wang, L., Chen, W., Zhang, Y., Liu, C., Yuan, L., Fu, L., Mei, C."Deletion of p18INK4c aggravates cisplatin‑induced acute kidney injury". International Journal of Molecular Medicine 33.6 (2014): 1621-1626.
Chicago
Wang, L., Chen, W., Zhang, Y., Liu, C., Yuan, L., Fu, L., Mei, C."Deletion of p18INK4c aggravates cisplatin‑induced acute kidney injury". International Journal of Molecular Medicine 33, no. 6 (2014): 1621-1626. https://doi.org/10.3892/ijmm.2014.1725