Klotho, an anti-aging gene, acts as a tumor suppressor and inhibitor of IGF-1R signaling in diffuse large B cell lymphoma

Xiangxiang Zhou, Xiaosheng Fang, Yujie Jiang, Lingyun Geng, Xinyu Li, Ying Li, Kang Lu, Peipei Li, Xiao Lv, Xin Wang, Xiangxiang Zhou, Xiaosheng Fang, Yujie Jiang, Lingyun Geng, Xinyu Li, Ying Li, Kang Lu, Peipei Li, Xiao Lv, Xin Wang

Abstract

Background: Klotho, is a transmembrane protein, performs as a circulating hormone and upstream modulator of the insulin-like growth factor-1 receptor (IGF-1R), fibroblast growth factor (FGF), and Wnt signaling pathways. These pathways are involved in the development and progression of B cell lymphoma. We aimed to explore the expression pattern and functional mechanism of Klotho in diffuse large B cell lymphoma (DLBCL).

Methods: Immunohistochemistry (IHC) and western blotting were performed to detect the expression level of Klotho in DLBCL patients and cell lines. Tumor suppressive effect of Klotho was determined by both in vitro and in vivo studies. Signaling pathway activity was assessed by western blotting.

Results: Remarkable lower expression levels of Klotho were observed in DLBCL patients and cell lines. Enforced expression of Klotho could significantly induce cell apoptosis and inhibit tumor growth in DLBCL. Upregulation of Klotho resulted in declined activation of IGF-1R signaling, accompanied with decreased phosphorylation of its downstream targets, including AKT and ERK1/2. Moreover, xenograft model treated with either Klotho overexpression vector or recombinant human Klotho administration presented restrained tumor growth and lower Ki67 staining.

Conclusions: Our findings establish that Klotho performs as a tumor suppressor and modulator of IGF-1R signaling in DLBCL. Targeting Klotho may provide novel strategies for future therapeutic intervention.

Keywords: Diffuse large B cell lymphoma; Insulin growth factor-1 receptor; Klotho; Tumor suppressor.

Figures

Fig. 1
Fig. 1
Klotho was downregulated in DLBCL and related to tumor progression. a Compared with reactive hyperplasia, expression level of Klotho was significantly decreased in DLBCL tissues. Original magnification, ×100 (upper panel) and ×400 (lower panel). b Patients with Klotho negative expression showed shorter survival than those with Klotho positive expression. c As detected by real-time quantitative PCR, lower levels of Klotho mRNA expression were observed in DLBCL cell lines (LY1, LY8) than in CD19+ B cells (N1, N2, N3) (mean ± SD, n = 3, *p < 0.05). d Protein expression levels of Klotho were detected in DLBCL cells and normal PBMCs. The ratios of relative protein expression level of targets are indicated below the western blot
Fig. 2
Fig. 2
Klotho suppressed DLBCL growth. a, b Relative expression levels of Klotho were confirmed by quantitative PCR (mean ± SD, n = 3, **p < 0.01) and western blot in stably transfected LY1 and LY8 cells compared to empty vectors. The ratios of relative protein expression level of targets are indicated below the bands. c DLBCL cells transfected with LV-KL presented significantly lower level of cell proliferation than those transfected with empty vector (mean ± SD, n = 3, **p < 0.01). d SCID mice with Klotho overexpression revealed significantly lower tumor volume than those with empty vector (n = 6 per group, **p < 0.01). e H&E staining and IHC staining of Ki67 and Klotho were performed in mice tumors. Original magnification, ×400
Fig. 3
Fig. 3
Klotho promoted apoptosis of DLBCL. a, b Enforced expression of Klotho resulted in increased apoptosis rates in LY1 and LY8 cells assessed by flow cytometric analysis with Annexin V-PE/7AAD staining (mean ± SD, n = 3, **p < 0.01). c Declined expression levels of Mcl-1 and increased levels of activated Caspase-3 were observed in LV-KL-treated DLBCL cells. The ratios of relative protein expression level of targets are indicated below the bands
Fig. 4
Fig. 4
Klotho modulated activation of IGF-1R pathway in DLBCL. a IGF-1-induced DLBCL cell (LY1 and LY8) proliferation is inhibited by Klotho overexpression. LY1 and LY8 cells were transfected with LV-KL or LV-Con, starved for 48 h and treated by IGF-1 (50 ng/ml) and analyzed by CCK-8 assay (mean ± SD, n = 3, *p < 0.05, **p < 0.01). b LY1 with stable transfection of LV-KL or LV-Con were serum starved for 48 h and treated with IGF-1 (50 ng/ml) for the indicated times or IGF-1 (30 min) for the indicated doses. After treatment, cells were harvested and analyzed by western blot. c LY1 and LY8 cells transfected with LV-KL or LV-Con, serum starved for 48 h, and treated with IGF-1 (50 ng/ml, 30 min). Western blot was conducted to assess the phosphorylated (p) and total (t) protein levels of IGF-1R, AKT, and ERK1/2. The ratios of relative protein expression level of targets are indicated below the bands. d Decreased activation of IGF1-R signaling was observed in LV-KL-treated mice. The ratios of relative protein expression level of targets are indicated below the bands. e Schematic description of Klotho mediated IGF1R signaling
Fig. 5
Fig. 5
rhKL acted as an active form in vitro and vivo. a, b LY1 and LY8 cells were treated with rhKL, ADR, their combination, or a vehicle control. CCK-8 assay was conducted after 48 h (mean ± SD, n = 3, *p < 0.05 for comparison between treated cells and control, **p < 0.05 between Klotho and ADR combination versus ADR alone). c LY1 cells were injected subcutaneously into the left inferior legs of SCID Beige mice. The mice were treated with daily intraperitoneal injections of rhKL (7.5 μg/kg) or vehicle control (PBS) for 2 weeks. Tumor volumes were measured every 2 days (n = 6 per group, *p < 0.05). d H&E staining and IHC staining with Ki67 were performed. Original magnification, ×400. e Lower serum Klotho levels were detected by Elisa in DLBCL patients than the control subjects

References

    1. Roschewski M, Staudt LM, Wilson WH. Diffuse large B-cell lymphoma-treatment approaches in the molecular era. Nat Rev Clin Oncol. 2014;11:12–23. doi: 10.1038/nrclinonc.2013.197.
    1. Zhang J, Grubor V, Love CL, Banerjee A, Richards KL, Mieczkowski PA, et al. Genetic heterogeneity of diffuse large B-cell lymphoma. Proc Natl Acad Sci U S A. 2013;110:1398–403. doi: 10.1073/pnas.1205299110.
    1. Horn H, Ziepert M, Wartenberg M, Staiger AM, Barth TF, Bernd HW, et al. Different biological risk factors in young poor-prognosis and elderly patients with diffuse large B-cell lymphoma. Leukemia. 2015;29:1564–70. doi: 10.1038/leu.2015.43.
    1. Saito B, Shiozawa E, Usui T, Nakashima H, Maeda T, Hattori N, et al. Rituximab with chemotherapy improves survival of non-germinal center type untreated diffuse large B-cell lymphoma. Leukemia. 2007;21:2563–6. doi: 10.1038/sj.leu.2404844.
    1. Sehn LH, Gascoyne RD. Diffuse large B-cell lymphoma: optimizing outcome in the context of clinical and biologic heterogeneity. Blood. 2015;125:22–32. doi: 10.1182/blood-2014-05-577189.
    1. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51. doi: 10.1038/36285.
    1. Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, et al. Suppression of aging in mice by the hormone Klotho. Science. 2005;309:1829–33. doi: 10.1126/science.1112766.
    1. Kuro-o M. Klotho. Pflugers Arch. 2010;459:333–43. doi: 10.1007/s00424-009-0722-7.
    1. Kuro-o M. Klotho and aging. Biochim Biophys Acta. 2009;1790:1049–1058. doi: 10.1016/j.bbagen.2009.02.005.
    1. Yamamoto M, Clark JD, Pastor JV, Gurnani P, Nandi A, Kurosu H, et al. Regulation of oxidative stress by the anti-aging hormone klotho. J Biol Chem. 2005;280:38029–34. doi: 10.1074/jbc.M509039200.
    1. Wolf I, Levanon-Cohen S, Bose S, Ligumsky H, Sredni B, Kanety H, et al. Klotho: a tumor suppressor and a modulator of the IGF-1 and FGF pathways in human breast cancer. Oncogene. 2008;27:7094–105. doi: 10.1038/onc.2008.292.
    1. Zhou X, Wang X. Klotho: a novel biomarker for cancer. J Cancer Res Clin Oncol. 2015;141:961–9. doi: 10.1007/s00432-014-1788-y.
    1. Yaktapour N, Ubelhart R, Schuler J, Aumann K, Dierks C, Burger M, et al. Insulin-like growth factor-1 receptor (IGF1R) as a novel target in chronic lymphocytic leukemia. Blood. 2013;122:1621–33. doi: 10.1182/blood-2013-02-484386.
    1. Vishwamitra D, Shi P, Wilson D, Manshouri R, Vega F, Schlette EJ, et al. Expression and effects of inhibition of type I insulin-like growth factor receptor tyrosine kinase in mantle cell lymphoma. Haematologica. 2011;96:871–80. doi: 10.3324/haematol.2010.031567.
    1. Mathur R, Sehgal L, Braun FK, Berkova Z, Romaguerra J, Wang M, et al. Targeting Wnt pathway in mantle cell lymphoma-initiating cells. J Hematol Oncol. 2015;8:63. doi: 10.1186/s13045-015-0161-1.
    1. Lam BQ, Dai L, Qin Z. The role of HGF/c-MET signaling pathway in lymphoma. J Hematol Oncol. 2016;9:135. doi: 10.1186/s13045-016-0366-y.
    1. Toffalini F, Demoulin JB. New insights into the mechanisms of hematopoietic cell transformation by activated receptor tyrosine kinases. Blood. 2010;116:2429–37. doi: 10.1182/blood-2010-04-279752.
    1. Xie J, Chen X, Zheng J, Li C, Stacy S, Holzenberger M, et al. IGF-IR determines the fates of BCR/ABL leukemia. J Hematol Oncol. 2015;8:3. doi: 10.1186/s13045-015-0106-8.
    1. Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors and neoplasia. Nat Rev Cancer. 2004;4:505–18. doi: 10.1038/nrc1387.
    1. Dong S, Qu X, Li W, Zhong X, Li P, Yang S, et al. The long non-coding RNA, GAS5, enhances gefitinib-induced cell death in innate EGFR tyrosine kinase inhibitor-resistant lung adenocarcinoma cells with wide-type EGFR via downregulation of the IGF-1R expression. J Hematol Oncol. 2015;8:43. doi: 10.1186/s13045-015-0140-6.
    1. Herrero-Sanchez MC, Rodriguez-Serrano C, Almeida J, San Segundo L, Inoges S, Santos-Briz A, et al. Targeting of PI3K/AKT/mTOR pathway to inhibit T cell activation and prevent graft-versus-host disease development. J Hematol Oncol. 2016;9:113. doi: 10.1186/s13045-016-0343-5.
    1. Lonetti A, Cappellini A, Bertaina A, Locatelli F, Pession A, Buontempo F, et al. Improving nelarabine efficacy in T cell acute lymphoblastic leukemia by targeting aberrant PI3K/AKT/mTOR signaling pathway. J Hematol Oncol. 2016;9:114. doi: 10.1186/s13045-016-0344-4.
    1. Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 2007;26:3291–310. doi: 10.1038/sj.onc.1210422.
    1. Vishwamitra D, George SK, Shi P, Kaseb AO, Amin HM. Type I insulin-like growth factor receptor signaling in hematological malignancies. Oncotarget. 2016; doi: 10.18632/oncotarget.12123.
    1. Stromberg T, Feng X, Delforoush M, Berglund M, Lin Y, Axelson M, et al. Picropodophyllin inhibits proliferation and survival of diffuse large B-cell lymphoma cells. Med Oncol. 2015;32:188. doi: 10.1007/s12032-015-0630-y.
    1. Jaffe ES. The 2008 WHO classification of lymphomas: implications for clinical practice and translational research. Hematology Am Soc Hematol Educ Program. 2009:523–531. doi: 10.1182/asheducation-2009.1.523.
    1. Szczuraszek K, Mazur G, Jelen M, Dziegiel P, Surowiak P, Zabel M. Prognostic significance of Ki-67 antigen expression in non-Hodgkin’s lymphomas. Anticancer Res. 2008;28:1113–8.
    1. Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J Cell Physiol. 2000;182:311–22. doi: 10.1002/(SICI)1097-4652(200003)182:3<311::AID-JCP1>;2-9.
    1. Whelan JT, Ludwig DL, Bertrand FE. HoxA9 induces insulin-like growth factor-1 receptor expression in B-lineage acute lymphoblastic leukemia. Leukemia. 2008;22:1161–9. doi: 10.1038/leu.2008.57.
    1. Abramovitz L, Rubinek T, Ligumsky H, Bose S, Barshack I, Avivi C, et al. KL1 internal repeat mediates klotho tumor suppressor activities and inhibits bFGF and IGF-I signaling in pancreatic cancer. Clin Cancer Res. 2011;17:4254–66. doi: 10.1158/1078-0432.CCR-10-2749.
    1. Chen B, Ma X, Liu S, Zhao W, Wu J. 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;13:1221–8. doi: 10.4161/cbt.21420.
    1. Li XX, Huang LY, Peng JJ, Liang L, Shi DB, Zheng HT, et al. Klotho suppresses growth and invasion of colon cancer cells through inhibition of IGF1R-mediated PI3K/AKT pathway. Int J Oncol. 2014;45:611–8.
    1. Aviel-Ronen S, Rubinek T, Zadok O, Vituri A, Avivi C, Wolf I, et al. Klotho expression in cervical cancer: differential expression in adenocarcinoma and squamous cell carcinoma. J Clin Pathol. 2016;69:53–7. doi: 10.1136/jclinpath-2015-202929.
    1. Rubinek T, Shulman M, Israeli S, Bose S, Avraham A, Zundelevich A, et al. Epigenetic silencing of the tumor suppressor klotho in human breast cancer. Breast Cancer Res Treat. 2012;133:649–57. doi: 10.1007/s10549-011-1824-4.
    1. Xie B, Zhou J, Yuan L, Ren F, Liu DC, Li Q, et al. Epigenetic silencing of Klotho expression correlates with poor prognosis of human hepatocellular carcinoma. Hum Pathol. 2013;44:795–801. doi: 10.1016/j.humpath.2012.07.023.
    1. Lee J, Jeong DJ, Kim J, Lee S, Park JH, Chang B, et al. The anti-aging gene KLOTHO is a novel target for epigenetic silencing in human cervical carcinoma. Mol Cancer. 2010;9:109. doi: 10.1186/1476-4598-9-109.
    1. Tilly H, Gomes da Silva M, Vitolo U, Jack A, Meignan M, Lopez-Guillermo A, et al. Diffuse large B-cell lymphoma (DLBCL): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26(Suppl 5):v116–25. doi: 10.1093/annonc/mdv304.
    1. Intlekofer AM, Younes A. Precision therapy for lymphoma—current state and future directions. Nat Rev Clin Oncol. 2014;11:585–96. doi: 10.1038/nrclinonc.2014.137.
    1. Martelli M, Ferreri AJ, Agostinelli C, Di Rocco A, Pfreundschuh M, Pileri SA. Diffuse large B-cell lymphoma. Crit Rev Oncol Hematol. 2013;87:146–71. doi: 10.1016/j.critrevonc.2012.12.009.
    1. Vermaat JS, Pals ST, Younes A, Dreyling M, Federico M, Aurer I, et al. Precision medicine in diffuse large B-cell lymphoma: hitting the target. Haematologica. 2015;100:989–93.
    1. Gigante M, Lucarelli G, Divella C, Netti GS, Pontrelli P, Cafiero C, et al. Soluble serum alphaKlotho is a potential predictive marker of disease progression in clear cell renal cell carcinoma. Medicine (Baltimore) 2015;94:e1917. doi: 10.1097/MD.0000000000001917.
    1. Suvannasankha A, Tompkins DR, Edwards DF, Petyaykina KV, Crean CD, Fournier PG, et al. FGF23 is elevated in multiple myeloma and increases heparanase expression by tumor cells. Oncotarget. 2015;6:19647–60. doi: 10.18632/oncotarget.3794.
    1. Ligumsky H, Rubinek T, Merenbakh-Lamin K, Yeheskel A, Sertchook R, Shahmoon S, et al. Tumor suppressor activity of Klotho in breast cancer is revealed by structure-function analysis. Mol Cancer Res. 2015;13:1398–407. doi: 10.1158/1541-7786.MCR-15-0141.
    1. Feng S, Dakhova O, Creighton CJ, Ittmann M. Endocrine fibroblast growth factor FGF19 promotes prostate cancer progression. Cancer Res. 2013;73:2551–62. doi: 10.1158/0008-5472.CAN-12-4108.
    1. McCubrey JA, Steelman LS, Bertrand FE, Davis NM, Abrams SL, Montalto G, et al. Multifaceted roles of GSK-3 and Wnt/beta-catenin in hematopoiesis and leukemogenesis: opportunities for therapeutic intervention. Leukemia. 2014;28:15–33. doi: 10.1038/leu.2013.184.
    1. Ugarte GD, Vargas MF, Medina MA, Leon P, Necunir D, Elorza AA, et al. Wnt signaling induces transcription, spatial proximity, and translocation of fusion gene partners in human hematopoietic cells. Blood. 2015;126:1785–9. doi: 10.1182/blood-2015-04-638494.
    1. Wu C, Zhang HF, Gupta N, Alshareef A, Wang Q, Huang YH, et al. A positive feedback loop involving the Wnt/beta-catenin/MYC/Sox2 axis defines a highly tumorigenic cell subpopulation in ALK-positive anaplastic large cell lymphoma. J Hematol Oncol. 2016;9:120. doi: 10.1186/s13045-016-0349-z.
    1. Sun H, Gao Y, Lu K, Zhao G, Li X, Li Z, et al. Overexpression of Klotho suppresses liver cancer progression and induces cell apoptosis by negatively regulating wnt/beta-catenin signaling pathway. World J Surg Oncol. 2015;13:307. doi: 10.1186/s12957-015-0717-0.

Source: PubMed

3
Suscribir