Soluble Serum αKlotho Is a Potential Predictive Marker of Disease Progression in Clear Cell Renal Cell Carcinoma

Margherita Gigante, Giuseppe Lucarelli, Chiara Divella, Giuseppe Stefano Netti, Paola Pontrelli, Cesira Cafiero, Giuseppe Grandaliano, Giuseppe Castellano, Monica Rutigliano, Giovanni Stallone, Carlo Bettocchi, Pasquale Ditonno, Loreto Gesualdo, Michele Battaglia, Elena Ranieri, Margherita Gigante, Giuseppe Lucarelli, Chiara Divella, Giuseppe Stefano Netti, Paola Pontrelli, Cesira Cafiero, Giuseppe Grandaliano, Giuseppe Castellano, Monica Rutigliano, Giovanni Stallone, Carlo Bettocchi, Pasquale Ditonno, Loreto Gesualdo, Michele Battaglia, Elena Ranieri

Abstract

Renal cell carcinoma (RCC) accounts for approximately 3% of adult malignancies, and clear cell RCC (ccRCC), that has a high metastatic index and high relapse rate, is the most common histological subtype. The identification of new biomarkers in ccRCC is fundamental for stratifying patients into prognostic risk groups and to guide therapy. The renoprotective antiaging gene, αKlotho, has recently been found to work as a tumor suppressor in different human cancers. Here, we evaluated αKlotho expression in tissue and serum of ccRCC patients and correlated it with disease progression. Tissue αKlotho expression was studied by quantitative RT-PCR and immunohistochemistry. In addition, soluble serum αKlotho levels were preoperatively measured in 160 patients who underwent nephrectomy for RCC with ELISA. Estimates of cancer-specific (CSS) and progression-free survival (PFS) were calculated according to the Kaplan-Meier method. Multivariate analysis was performed to identify the most significant variables for predicting CSS and PFS. αKlotho protein levels were significantly decreased in RCC tissues compared with normal tissues (P < 0.01) and the more advanced the disease, the more evident the down-regulation. This trend was also observed in serum samples. Statistically significant differences resulted between serum αKlotho levels and tumor size (P = 0.003), Fuhrman grade (P = 0.007), and clinical stage (P = 0.0004). CSS and PFS were significantly shorter in patients with lower levels of αKlotho (P < 0.0001 and P = 0.0004, respectively). At multivariate analysis low serum levels of αKlotho were independent adverse prognostic factors for CSS (HR = 2.11; P = 0.03) and PFS (HR = 2.18; P = 0.03).These results indicate that a decreased αKlotho expression is correlated with RCC progression, and suggest a key role of declining αKlotho in the onset of cancer metastasis.

Conflict of interest statement

The authors have no conflicts of interest to disclose.

Figures

FIGURE 1
FIGURE 1
Klotho gene expression levels evaluated by qRT–PCR. Histogram represents mean ± SD of normalized Klotho gene expression levels by quantitative real-time PCR in 18 ccRCC samples (T) compared with their matched-NT (∗P < 0.001).
FIGURE 2
FIGURE 2
In vivo analysis for Klotho expression using immunohistochemistry. Renal Klotho expression was evaluated in RCC tumor tissues (T) and matched nontumoral (NT) kidney portions from patients with different grades of RCC (G1–G2 and G3–G4) and patients with metastases (M1). Klotho was expressed at tubular level in NT portion of G1–G2 RCC patients and significantly decreased in G3–G4 and M1 patients. In T tissue, αKlotho was slightly expressed in patients with G1–G2 and patients with G3–G4 and M1 showed a very low or undetectable expression. Klotho staining was quantified as described in the “Methods” section and was expressed as mean ± SD. All images are from a single patient and are representative of all 8 patients for G1–G2 and G3–G4 groups and 4 patients for M1. The positive control is a normal kidney tissue and negative control is obtained as described in the “Methods” section.
FIGURE 3
FIGURE 3
Median αKlotho levels in 20 healthy subjects compared with 45 patients with metastatic (M1) and 115 nonmetastatic (M0) ccRCC.
FIGURE 4
FIGURE 4
Serum αKlotho levels stratified according to tumor size (≤7 cm vs >7 cm; panel A) and nuclear grade (Fuhrman 1–2 vs 3–4, panel B).
FIGURE 5
FIGURE 5
Spearmann correlation coefficient (rs) and linear regression line between αKlotho tissue signal intensity and serum values from 20 ccRCC patients.
FIGURE 6
FIGURE 6
Comparison of αKlotho and CA 15-3 receiver-operating characteristic (ROC) curves for cancer-specific survival (panel A), and progression-free survival (panel B).
FIGURE 7
FIGURE 7
Kaplan–Meier cancer-specific survival (CSS) curves, stratified by αKlotho serum levels.
FIGURE 8
FIGURE 8
Kaplan–Meier progression-free survival (PFS) curves, stratified by αKlotho serum levels.

References

    1. Hofman-Bang J, Martuseviciene G, Santini MA, et al. Increased parathyroid expression of Klotho in uremic rats. Kidney Int 2010; 78:1119–1127.
    1. Kato Y, Arakawa E, Kinoshita S, et al. Establishment of the anti- Klotho monoclonal antibodies and detection of Klotho protein in kidneys. Biochem Biophys Res Commun 2000; 267:597–602.
    1. Matsumura Y, Aizawa H, Shiraki-Iida T, et al. Identification of the human klotho gene and its two transcripts encoding membrane and secreted klotho protein. Biochem Biophys Res Commun 1998; 242:626–630.
    1. Hu MC, Shi M, Zhang J, et al. Renal production, uptake, and handling of circulating αKlotho. J Am Soc Nephrol 2015; pii: ASN2014101030.
    1. Almilaji A, Munoz C, Pakladok T, et al. Klotho sensitivity of the neuronal excitatory amino acid transporters EAAT3 and EAAT4. PLoS One 2013; 8:e70988.
    1. Song S, Gao P, Xiao H, et al. Klotho suppresses cardiomyocyte apoptosis in mice with stress-induced cardiac injury via downregulation of endoplasmic reticulum stress. PLoS One 2013; 8:e82968.
    1. Guan X, Nie L, He T, et al. Klotho suppresses renal tubulo-interstitial fibrosis by controlling basic fibroblast growth factor-2 signalling. J Pathol 2014; 234:560–572.
    1. Sakan H, Nakatani K, Asai O, et al. Reduced renal α-Klotho expression in CKD patients and its effect on renal phosphate handling and vitamin D metabolism. PLoS One 2014; 9:e86301.
    1. Leunissen EH, Nair AV, Büll C, et al. The epithelial calcium channel TRPV5 is regulated differentially by klotho and sialidase. J Biol Chem 2013; 288:29238–29246.
    1. Chen CD, Sloane JA, Li H, et al. The antiaging protein Klotho enhances oligodendrocyte maturation and myelination of the CNS. J Neurosci 2013; 33:1927–1939.
    1. Hu MC, Shi M, Zhang J, et al. Klotho deficiency is an early biomarker of renal ischemia-reperfusion injury and its replacement is protective. Kidney Int 2010; 78:1240–1251.
    1. Hu MC, Moe OW. Klotho as a potential biomarker and therapy for acute kidney injury. Nat Rev Nephrol 2012; 8:423–429.
    1. Xie B, Chen J, Liu B, et al. Klotho acts as a tumor suppressor in cancers. Pathol Oncol Res 2013; 19:611–617.
    1. Hu MC, Shi M, Zhang J, et al. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J 2010; 24:3438–3450.
    1. Chen B, Ma X, Liu S, et al. Inhibition of lung cancer cells growth, motility and induction of apoptosis by Klotho, a novel secreted Wnt antagonist, in a dose-dependent manner. Cancer Biol Ther 2012; 13:1221–1228.
    1. de Oliveira RM. Klotho RNAi induces premature senescence of human cells via a p53/p21 dependent pathway. FEBS Lett 2006; 580:5753–5758.
    1. Doi S, Zou Y, Togao O, et al. Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice. J Biol Chem 2011; 286:8655–8665.
    1. Lee J, Jeong DJ, Kim J, et al. The anti-aging gene KLOTHO is a novel target for epigenetic silencing in human cervical carcinoma. Mol Cancer 2010; 9:109.
    1. Pan J, Zhong J, Gan LH, et al. Klotho, an anti-senescence related gene, is frequently inactivated through promoter hypermethylation in colorectal cancer. Tumour Biol 2011; 32:729–735.
    1. Wang L, Wang X, Wang X, et al. Klotho is silenced through promoter hypermethylation in gastric cancer. Am J Cancer Res 2011; 1:111–119.
    1. Zhu Y, Xu L, Zhang J, et al. Klotho suppresses tumor progression via inhibiting PI3K/Akt/GSK3β/Snail signaling in renal cell carcinoma. Cancer Sci 2013; 104:663–671.
    1. Pascual D, Borque A. Epidemiology of kidney cancer. Adv Urol 2008; 782381:1–7.
    1. Finke J, Kierstead LS, Ranieri E, et al. Immunologic response to RCC. Renal Cell Carcinoma, Molecular Biology, Immunology and Clinical Management 2000; Totowa, NJ:Humana Press Inc, 39–62.
    1. Vasudev NS, Selby PJ, Banks RE. Renal cancer biomarkers: the promise of personalized care. BMC Med 2012; 10:112.
    1. Klatte T, Seligson DB, Riggs SB, et al. Hypoxia-inducible factor 1 alpha in clear cell renal cell carcinoma. Clin Cancer Res 2007; 13:7388–7393.
    1. Lucarelli G, Ditonno P, Bettocchi C, et al. Diagnostic and prognostic role of preoperative circulating CA 15-3, CA 125, and beta-2 microglobulin in renal cell carcinoma. Dis Markers 2014; 2014:689795.
    1. Lucarelli G, Galleggiante V, Rutigliano M, et al. Metabolomic profile of glycolysis and the pentose phosphate pathway identifies the central role of glucose-6-phosphate dehydrogenase in clear cell-renal cell carcinoma. Oncotarget 2015; 6:13371–13378.
    1. Sobin LH, Gospodarowicz MK, Wittekind C. International Union Against Cancer (UICC) TNM Classification of Malignant Tumours. 7th ed. New York, NY: Wiley-Liss; 2010.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2 − ΔΔCT method. Methods 2001; 25:402–408.
    1. Klapczynski M, Gagne GD, Morgan SJ, et al. Computer-assisted imaging algorithms facilitate histomorphometric quantification of kidney damage in rodent renal failure models. J Pathol Inform 2012; 3:20.
    1. Valletti A, Gigante M, Palumbo O, et al. Genome-wide analysis of differentially expressed genes and splicing isoforms in clear cell renal cell carcinoma. PLoS One 2013; 8:e78452.
    1. Gumz ML, Zou H, Kreinest PA, et al. Secreted frizzled-related protein 1 loss contributes to tumor phenotype of clear cell renal cell carcinoma. Clin Cancer Res 2007; 13:4740–4749.
    1. Beroukhim R, Brunet JP, Di Napoli A, et al. Patterns of gene expression and copy-number alterations in von-hippel lindau disease-associated and sporadic clear cell carcinoma of the kidney. Cancer Res 2009; 69:4674–4681.
    1. Jones J, Otu H, Spentzos D, et al. Gene signatures of progression and metastasis in renal cell cancer. Clin Cancer Res 2005; 11:5730–5739.
    1. Lenburg ME, Liou LS, Gerry NP, et al. Previously unidentified changes in renal cell carcinoma gene expression identified by parametric analysis of microarray data. BMC Cancer 2003; 3:31.
    1. Yusenko MV, Kuiper RP, Boethe T, et al. High-resolution DNA copy number and gene expression analyses distinguish chromophobe renal cell carcinomas and renal oncocytomas. BMC Cancer 2009; 9:152.
    1. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013; 499:43–49.
    1. Castellano G, Intini A, Stasi A, et al. Complement modulation of anti-aging factor klotho in ischemia/reperfusion injury and delayed graft function. Am J Transplant 2015; doi: 101111/ajt13415.

Source: PubMed

3
Suscribir