Skip to main content
Erschienen in: Pathology & Oncology Research 4/2013

01.10.2013 | Review

Klotho Acts as a Tumor Suppressor in Cancers

verfasst von: Biao Xie, Jinhui Chen, Bin Liu, Junkun Zhan

Erschienen in: Pathology & Oncology Research | Ausgabe 4/2013

Einloggen, um Zugang zu erhalten

Abstract

The klotho gene is a classical “aging suppressor” gene. Its roles in the pathology of chronic kidney diseases have been well documented. However, the role of Klotho in tumorigenesis, cancer progression, and prognosis is attracting more and more attention. Recent studies have shown that Klotho participates in the progression of several types of human cancers. Klotho functions as a tumor suppressor by inhibiting insulin/IGF1, p53/p21, and Wnt signaling. Silencing klotho gene expression is mainly mediated through promoter hypermethylation and histone deacetylation in cancer. Klotho has been proposed to take part in cell proliferation, survival, autophagy, and resistance to anti-cancer therapies.
Literatur
1.
Zurück zum Zitat Kuro-o M, Matsumura Y, Aizawa H et al (1997) Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390:45–51PubMedCrossRef Kuro-o M, Matsumura Y, Aizawa H et al (1997) Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390:45–51PubMedCrossRef
2.
Zurück zum Zitat Kurosu H, Yamamoto M, Clark JD et al (2005) Suppression of aging in mice by the hormone Klotho. Science 309:1829–1833PubMedCrossRef Kurosu H, Yamamoto M, Clark JD et al (2005) Suppression of aging in mice by the hormone Klotho. Science 309:1829–1833PubMedCrossRef
3.
Zurück zum Zitat Torres PU, Prié D, Beck L, De Brauwere D, Leroy C, Friedlander G (2009) Klotho gene, phosphocalcic metabolism, and survival in dialysis. J Ren Nutr 19:50–56PubMedCrossRef Torres PU, Prié D, Beck L, De Brauwere D, Leroy C, Friedlander G (2009) Klotho gene, phosphocalcic metabolism, and survival in dialysis. J Ren Nutr 19:50–56PubMedCrossRef
4.
5.
Zurück zum Zitat Lu L, Katsaros D, Wiley A, de la Longrais IA, Puopolo M, Yu H (2008) Klotho expression in epithelial ovarian cancer and its association with insulin-like growth factors and disease progression. Cancer Invest 26:185–192PubMedCrossRef Lu L, Katsaros D, Wiley A, de la Longrais IA, Puopolo M, Yu H (2008) Klotho expression in epithelial ovarian cancer and its association with insulin-like growth factors and disease progression. Cancer Invest 26:185–192PubMedCrossRef
6.
Zurück zum Zitat Wolf I, Levanon-Cohen S, Bose S et al (2008) Klotho: a tumor suppressor and a modulator of the IGF-1 and FGF pathways in human breast cancer. Oncogene 27:7094–7105PubMedCrossRef Wolf I, Levanon-Cohen S, Bose S et al (2008) Klotho: a tumor suppressor and a modulator of the IGF-1 and FGF pathways in human breast cancer. Oncogene 27:7094–7105PubMedCrossRef
7.
Zurück zum Zitat Wolf I, Laitman Y, Rubinek T et al (2010) Functional variant of KLOTHO: a breast cancer risk modifier among BRCA1 mutation carriers of Ashkenazi origin. Oncogene 29:26–33PubMedCrossRef Wolf I, Laitman Y, Rubinek T et al (2010) Functional variant of KLOTHO: a breast cancer risk modifier among BRCA1 mutation carriers of Ashkenazi origin. Oncogene 29:26–33PubMedCrossRef
8.
Zurück zum Zitat Laitman Y, Kuchenbaecker KB, Rantala J et al (2012) The KL-VS sequence variant of Klotho and cancer risk in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 132:1119–1126PubMedCrossRef Laitman Y, Kuchenbaecker KB, Rantala J et al (2012) The KL-VS sequence variant of Klotho and cancer risk in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 132:1119–1126PubMedCrossRef
9.
Zurück zum Zitat Chen B, Wang X, Zhao W, Wu J (2010) Klotho inhibits growth and promotes apoptosis in human lung cancer cell line A549. J Exp Clin Cancer Res 29:99–106PubMedCrossRef Chen B, Wang X, Zhao W, Wu J (2010) Klotho inhibits growth and promotes apoptosis in human lung cancer cell line A549. J Exp Clin Cancer Res 29:99–106PubMedCrossRef
10.
Zurück zum Zitat Lee J, Jeong DJ, Kim J et al (2010) The antiaging gene KLOTHO is a novel target for epigenetic silencing inhuman cervical carcinoma. Mol Cancer 9:109–118PubMedCrossRef Lee J, Jeong DJ, Kim J et al (2010) The antiaging gene KLOTHO is a novel target for epigenetic silencing inhuman cervical carcinoma. Mol Cancer 9:109–118PubMedCrossRef
11.
Zurück zum Zitat Wang LJ, Wang X, Wang XJ et al (2011) Klotho is silenced through promoter hypermethylation in gastric cancer. Am J Cancer Res 1:111–119PubMed Wang LJ, Wang X, Wang XJ et al (2011) Klotho is silenced through promoter hypermethylation in gastric cancer. Am J Cancer Res 1:111–119PubMed
12.
Zurück zum Zitat Abramovitz L, Rubinek T, Ligumsky H et al (2011) KL1 internal repeat mediates klotho tumor suppressor activities and inhibits bFGF and IGF-I signaling in pancreatic cancer. Clin Cancer Res 17:4254–4266PubMedCrossRef Abramovitz L, Rubinek T, Ligumsky H et al (2011) KL1 internal repeat mediates klotho tumor suppressor activities and inhibits bFGF and IGF-I signaling in pancreatic cancer. Clin Cancer Res 17:4254–4266PubMedCrossRef
13.
Zurück zum Zitat Bartucci M, Morelli C, Mauro L, Andò S, Surmacz E (2001) Differential insulin-like growth factor I receptor signaling and function in estrogen receptor (ER)-positive MCF-7 and ER-negative MDA-MB-231 breast cancer cells. Cancer Res 61:6747–6754PubMed Bartucci M, Morelli C, Mauro L, Andò S, Surmacz E (2001) Differential insulin-like growth factor I receptor signaling and function in estrogen receptor (ER)-positive MCF-7 and ER-negative MDA-MB-231 breast cancer cells. Cancer Res 61:6747–6754PubMed
14.
Zurück zum Zitat Pan J, Zhong J, Gan LH et al (2011) Klotho, an anti-senescence related gene, is frequently inactivated through promoter hypermethylation in colorectal cancer. Tumour Biol 32:729–735PubMedCrossRef Pan J, Zhong J, Gan LH et al (2011) Klotho, an anti-senescence related gene, is frequently inactivated through promoter hypermethylation in colorectal cancer. Tumour Biol 32:729–735PubMedCrossRef
15.
Zurück zum Zitat Rubinek T, Shulman M, Israeli S, et al. (2011) Epigenetic silencing of the tumor suppressor klotho in human breast cancer. Breast Cancer Res Treat [Epub ahead of print] Rubinek T, Shulman M, Israeli S, et al. (2011) Epigenetic silencing of the tumor suppressor klotho in human breast cancer. Breast Cancer Res Treat [Epub ahead of print]
16.
Zurück zum Zitat Wang X, Chen B, Xu W, Liu S, Zhao W, Wu J (2011) Combined effects of klotho and soluble CD40 ligand on A549 lung cancer cells. Oncol Rep 25:1465–1472PubMed Wang X, Chen B, Xu W, Liu S, Zhao W, Wu J (2011) Combined effects of klotho and soluble CD40 ligand on A549 lung cancer cells. Oncol Rep 25:1465–1472PubMed
17.
Zurück zum Zitat Doi S, Zou Y, Togao O et al (2011) Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice. Biol Chem 286:8655–8665CrossRef Doi S, Zou Y, Togao O et al (2011) Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice. Biol Chem 286:8655–8665CrossRef
18.
Zurück zum Zitat Shu G, Xie B, Ren F, et al. (2012) Restoration of klotho expression induces apoptosis and autophagy in hepatocellular carcinoma cells. Cell Oncol (Dordr) [Epub ahead of print] Shu G, Xie B, Ren F, et al. (2012) Restoration of klotho expression induces apoptosis and autophagy in hepatocellular carcinoma cells. Cell Oncol (Dordr) [Epub ahead of print]
19.
Zurück zum Zitat Xie B, Zhou J, Yuan L, et al. (2012) Epigenetic silencing of Klotho expression correlates with poor prognosis of human hepatocellular carcinoma. Hum Pathol Xie B, Zhou J, Yuan L, et al. (2012) Epigenetic silencing of Klotho expression correlates with poor prognosis of human hepatocellular carcinoma. Hum Pathol
20.
Zurück zum Zitat Pollak MN, Schernhammer ES, Hankinson SE (2004) Insulin-like growth factors and neoplasia. Nat Rev Cancer 4:505–518PubMedCrossRef Pollak MN, Schernhammer ES, Hankinson SE (2004) Insulin-like growth factors and neoplasia. Nat Rev Cancer 4:505–518PubMedCrossRef
21.
Zurück zum Zitat Evans DS, Kapahi P, Hsueh WC, Kockel L (2011) TOR signaling never gets old: aging, longevity and TORC1 activity. Ageing Res Rev 10:225–237PubMedCrossRef Evans DS, Kapahi P, Hsueh WC, Kockel L (2011) TOR signaling never gets old: aging, longevity and TORC1 activity. Ageing Res Rev 10:225–237PubMedCrossRef
22.
Zurück zum Zitat Carboni JM, Lee AV, Hadsell DL et al (2005) Tumor development by transgenic expression of a constitutively active insulin-like growth factor I receptor. Cancer Res 5:3781–3787CrossRef Carboni JM, Lee AV, Hadsell DL et al (2005) Tumor development by transgenic expression of a constitutively active insulin-like growth factor I receptor. Cancer Res 5:3781–3787CrossRef
23.
Zurück zum Zitat Lopez T, Hanahan D (2002) Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1:339–353PubMedCrossRef Lopez T, Hanahan D (2002) Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1:339–353PubMedCrossRef
24.
Zurück zum Zitat Yakar S, Leroith D, Brodt P (2005) The role of the growth hormone/insulin-like growth factor axis in tumor growth and progression: Lessons from animal models. Cytokine Growth Factor Rev 16:407–420PubMedCrossRef Yakar S, Leroith D, Brodt P (2005) The role of the growth hormone/insulin-like growth factor axis in tumor growth and progression: Lessons from animal models. Cytokine Growth Factor Rev 16:407–420PubMedCrossRef
25.
Zurück zum Zitat Yakar S, Leroith D, Brodt P (2005) The role of the growth hormone/insulin-like growth factor axis in tumor growth and progression: Lessons from animal models. Cytokine Growth Factor Rev 6:407–420CrossRef Yakar S, Leroith D, Brodt P (2005) The role of the growth hormone/insulin-like growth factor axis in tumor growth and progression: Lessons from animal models. Cytokine Growth Factor Rev 6:407–420CrossRef
26.
Zurück zum Zitat Usuda J, Ichinose S, Ishizumi T et al (2011) Klotho is a novel biomarker for good survival in resected large cell neuroendocrine carcinoma of the lung. Lung Cancer 72:355–359PubMedCrossRef Usuda J, Ichinose S, Ishizumi T et al (2011) Klotho is a novel biomarker for good survival in resected large cell neuroendocrine carcinoma of the lung. Lung Cancer 72:355–359PubMedCrossRef
27.
Zurück zum Zitat Usuda J, Ichinose S, Ishizumi T et al (2011) Klotho predicts good clinical outcome in patients with limited-disease small cell lung cancer who received surgery. Lung Cancer 74:332–337PubMedCrossRef Usuda J, Ichinose S, Ishizumi T et al (2011) Klotho predicts good clinical outcome in patients with limited-disease small cell lung cancer who received surgery. Lung Cancer 74:332–337PubMedCrossRef
28.
Zurück zum Zitat Xie B, Zhou J, Shu G et al (2013) Restoration of klotho gene expression induces apoptosis and autophagy in gastric cancer cells: tumor suppressive role of klotho in gastric cancer. Cancer Cell Int 13:18PubMedCrossRef Xie B, Zhou J, Shu G et al (2013) Restoration of klotho gene expression induces apoptosis and autophagy in gastric cancer cells: tumor suppressive role of klotho in gastric cancer. Cancer Cell Int 13:18PubMedCrossRef
29.
Zurück zum Zitat Chihara Y, Rakugi H, Ishikawa K et al (2006) Klotho protein promotes adipocyte differentiation. Endocrinology 147:3835–3842PubMedCrossRef Chihara Y, Rakugi H, Ishikawa K et al (2006) Klotho protein promotes adipocyte differentiation. Endocrinology 147:3835–3842PubMedCrossRef
30.
Zurück zum Zitat Lorenzi O, Veyrat-Durebex C, Wollheim CB et al (2010) Evidence against a direct role of klotho in insulin resistance. Pflugers Arch 459:465–473PubMedCrossRef Lorenzi O, Veyrat-Durebex C, Wollheim CB et al (2010) Evidence against a direct role of klotho in insulin resistance. Pflugers Arch 459:465–473PubMedCrossRef
31.
Zurück zum Zitat Duce JA, Podvin S, Hollander W et al (2008) Gene profile analysis implicates Klotho as an important contributor to aging changes in brain white matter of the rhesus monkey. Glia 56:106–117PubMedCrossRef Duce JA, Podvin S, Hollander W et al (2008) Gene profile analysis implicates Klotho as an important contributor to aging changes in brain white matter of the rhesus monkey. Glia 56:106–117PubMedCrossRef
32.
Zurück zum Zitat Choi BH, Kim CG, Lim Y, Lee YH, Shin SY (2010) Transcriptional activation of the human Klotho gene by epidermal growth factor in HEK293 cells; role of Egr-1. Gene 450:121–127PubMedCrossRef Choi BH, Kim CG, Lim Y, Lee YH, Shin SY (2010) Transcriptional activation of the human Klotho gene by epidermal growth factor in HEK293 cells; role of Egr-1. Gene 450:121–127PubMedCrossRef
33.
Zurück zum Zitat Shen J, Stass SA, Jiang F (2013) MicroRNAs as potential biomarkers in human solid tumors. Cancer Lett 329:125–136 Shen J, Stass SA, Jiang F (2013) MicroRNAs as potential biomarkers in human solid tumors. Cancer Lett 329:125–136
34.
Zurück zum Zitat Liang R, Bates DJ, Wang E (2009) Epigenetic Control of MicroRNA Expression and Aging. Curr Genomics 10:184–193PubMedCrossRef Liang R, Bates DJ, Wang E (2009) Epigenetic Control of MicroRNA Expression and Aging. Curr Genomics 10:184–193PubMedCrossRef
35.
Zurück zum Zitat Maes OC, An J, Sarojini H, Wang E (2008) Murine microRNAs implicated in liver functions and aging process. Mech Ageing Dev 129:534–541PubMedCrossRef Maes OC, An J, Sarojini H, Wang E (2008) Murine microRNAs implicated in liver functions and aging process. Mech Ageing Dev 129:534–541PubMedCrossRef
36.
Zurück zum Zitat Logan CY, Nusse R (2004) The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol 20:781–810PubMedCrossRef Logan CY, Nusse R (2004) The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol 20:781–810PubMedCrossRef
37.
Zurück zum Zitat Klaus A, Birchmeier W (2008) Wnt signalling and its impact on development and cancer. Nat Rev Cancer 8:387–398PubMedCrossRef Klaus A, Birchmeier W (2008) Wnt signalling and its impact on development and cancer. Nat Rev Cancer 8:387–398PubMedCrossRef
38.
Zurück zum Zitat Suzuki H, Watkins DN, Jair KW et al (2004) Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet 36:417–422PubMedCrossRef Suzuki H, Watkins DN, Jair KW et al (2004) Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet 36:417–422PubMedCrossRef
39.
Zurück zum Zitat Urakami S, Shiina H, Enokida H et al (2006) Combination analysis of hypermethylated Wnt-antagonist family genes as a novel epigenetic biomarker panel for bladder cancer detection. Clin Cancer Res 12:2109–2116PubMedCrossRef Urakami S, Shiina H, Enokida H et al (2006) Combination analysis of hypermethylated Wnt-antagonist family genes as a novel epigenetic biomarker panel for bladder cancer detection. Clin Cancer Res 12:2109–2116PubMedCrossRef
40.
Zurück zum Zitat Nojima M, Suzuki H, Toyota M et al (2007) Frequent epigenetic inactivation of SFRP genes and constitutive activation of Wnt signaling in gastric cancer. Oncogene 26:4699–4713PubMedCrossRef Nojima M, Suzuki H, Toyota M et al (2007) Frequent epigenetic inactivation of SFRP genes and constitutive activation of Wnt signaling in gastric cancer. Oncogene 26:4699–4713PubMedCrossRef
41.
Zurück zum Zitat Suzuki H, Toyota M, Carraway H et al (2008) Frequent epigenetic inactivation of Wnt antagonist genes in breast cancer. Br J Cancer 98:1147–1156PubMedCrossRef Suzuki H, Toyota M, Carraway H et al (2008) Frequent epigenetic inactivation of Wnt antagonist genes in breast cancer. Br J Cancer 98:1147–1156PubMedCrossRef
42.
Zurück zum Zitat Camilli TC, Xu M, O’Connell MP et al (2011) Loss of Klotho during melanoma progression leads to increased filamin cleavage, increased Wnt5A expression, and enhanced melanoma cell motility. Pigment Cell Melanoma Res 24:175–186PubMedCrossRef Camilli TC, Xu M, O’Connell MP et al (2011) Loss of Klotho during melanoma progression leads to increased filamin cleavage, increased Wnt5A expression, and enhanced melanoma cell motility. Pigment Cell Melanoma Res 24:175–186PubMedCrossRef
43.
Zurück zum Zitat Liu H, Fergusson MM, Castilho RM et al (2007) Augmented Wnt signaling in a mammalian model of accelerated aging. Science 317:803–806PubMedCrossRef Liu H, Fergusson MM, Castilho RM et al (2007) Augmented Wnt signaling in a mammalian model of accelerated aging. Science 317:803–806PubMedCrossRef
44.
Zurück zum Zitat Kirstetter P, Anderson K, Porse BT, Jacobsen SE, Nerlov C (2006) Activation of the canonical Wnt pathway leads to loss of hematopoietic stem cell repopulation and multilineage differentiation block. Nat Immunol 7:1048–1056PubMedCrossRef Kirstetter P, Anderson K, Porse BT, Jacobsen SE, Nerlov C (2006) Activation of the canonical Wnt pathway leads to loss of hematopoietic stem cell repopulation and multilineage differentiation block. Nat Immunol 7:1048–1056PubMedCrossRef
46.
Zurück zum Zitat Castilho RM, Squarize CH, Chodosh LA, Williams BO, Gutkind JS (2009) mTOR mediates Wnt-induced epidermal stem cell exhaustion and aging. Cell Stem Cell 4:279–289CrossRef Castilho RM, Squarize CH, Chodosh LA, Williams BO, Gutkind JS (2009) mTOR mediates Wnt-induced epidermal stem cell exhaustion and aging. Cell Stem Cell 4:279–289CrossRef
47.
Zurück zum Zitat Scheller M, Huelsken J, Rosenbauer F et al (2006) Hematopoietic stem cell and multilineage defects generated by constitutive beta-catenin activation. Nat Immunol 7:1037–1047PubMedCrossRef Scheller M, Huelsken J, Rosenbauer F et al (2006) Hematopoietic stem cell and multilineage defects generated by constitutive beta-catenin activation. Nat Immunol 7:1037–1047PubMedCrossRef
49.
Zurück zum Zitat Stone JR, Yang S (2006) Hydrogen peroxide: a signaling messenger. Antioxid Redox Signal 8:243–270PubMedCrossRef Stone JR, Yang S (2006) Hydrogen peroxide: a signaling messenger. Antioxid Redox Signal 8:243–270PubMedCrossRef
50.
Zurück zum Zitat Mahadev K, Wu X, Zilbering A et al (2001) Hydrogen peroxide generated during cellular insulin stimulation is integral to activation of the distal insulin signaling cascade in 3T3-L1 adipocytes. J Biol Chem 276:48662–48669PubMedCrossRef Mahadev K, Wu X, Zilbering A et al (2001) Hydrogen peroxide generated during cellular insulin stimulation is integral to activation of the distal insulin signaling cascade in 3T3-L1 adipocytes. J Biol Chem 276:48662–48669PubMedCrossRef
51.
Zurück zum Zitat Colavitti R, Pani G, Bedogni B et al (2002) Reactive oxygen species as downstream mediators of angiogenic signaling by vascular endothelial growth factor receptor-2/KDR. J Biol Chem 277:3101–3108PubMedCrossRef Colavitti R, Pani G, Bedogni B et al (2002) Reactive oxygen species as downstream mediators of angiogenic signaling by vascular endothelial growth factor receptor-2/KDR. J Biol Chem 277:3101–3108PubMedCrossRef
52.
Zurück zum Zitat Wang Y, Sun Z (2009) Klotho gene delivery prevents the progression of spontaneous hypertension and renal damage. Hypertension 54:810–817PubMedCrossRef Wang Y, Sun Z (2009) Klotho gene delivery prevents the progression of spontaneous hypertension and renal damage. Hypertension 54:810–817PubMedCrossRef
53.
Zurück zum Zitat Mitobe M, Yoshida T, Sugiura H et al (2005) Oxidative stress decreases klotho expression in a mouse kidney cell line. Nephron Exp Nephrol 101:67–74CrossRef Mitobe M, Yoshida T, Sugiura H et al (2005) Oxidative stress decreases klotho expression in a mouse kidney cell line. Nephron Exp Nephrol 101:67–74CrossRef
54.
Zurück zum Zitat Kuro-o M (2008) Klotho as a regulator of oxidative stress and senescence. Biol Chem 389:233–241PubMedCrossRef Kuro-o M (2008) Klotho as a regulator of oxidative stress and senescence. Biol Chem 389:233–241PubMedCrossRef
55.
Zurück zum Zitat Panayiotidis M (2008) Reactive oxygen species (ROS) in multistage carcinogenesis. Cancer Lett 266:3–5PubMedCrossRef Panayiotidis M (2008) Reactive oxygen species (ROS) in multistage carcinogenesis. Cancer Lett 266:3–5PubMedCrossRef
56.
Zurück zum Zitat Lee SB, Cho ES, Yang HS, Kim H, Um HD (2005) Serum withdrawal kills U937 cells by inducing a positive mutual interaction between reactive oxygen species and phosphoinositide 3-kinase. Cell Signal 17:197–204PubMedCrossRef Lee SB, Cho ES, Yang HS, Kim H, Um HD (2005) Serum withdrawal kills U937 cells by inducing a positive mutual interaction between reactive oxygen species and phosphoinositide 3-kinase. Cell Signal 17:197–204PubMedCrossRef
57.
Zurück zum Zitat Cao Q, Mak KM, Lieber CS (2006) DLPC and SAMe prevent alpha1(I) collagen mRNA up-regulation in human hepatic stellate cells, whether caused by leptin or menadione. Biochem Biophys Res Commun 350:50–55PubMedCrossRef Cao Q, Mak KM, Lieber CS (2006) DLPC and SAMe prevent alpha1(I) collagen mRNA up-regulation in human hepatic stellate cells, whether caused by leptin or menadione. Biochem Biophys Res Commun 350:50–55PubMedCrossRef
58.
Zurück zum Zitat Nicco C, Laurent A, Chereau C, Weill B, Batteux F (2005) Differential modulation of normal and tumor cell proliferation by reactive oxygen species. Biomed Pharmacother 59:169–174PubMedCrossRef Nicco C, Laurent A, Chereau C, Weill B, Batteux F (2005) Differential modulation of normal and tumor cell proliferation by reactive oxygen species. Biomed Pharmacother 59:169–174PubMedCrossRef
59.
Zurück zum Zitat López-Lázaro M (2007) Dual role of hydrogen peroxide in cancer: possible relevance to cancer chemoprevention and therapy. Cancer Lett 252:1–8PubMedCrossRef López-Lázaro M (2007) Dual role of hydrogen peroxide in cancer: possible relevance to cancer chemoprevention and therapy. Cancer Lett 252:1–8PubMedCrossRef
60.
Zurück zum Zitat Zielonka J, Kalyanaraman B (2008) “ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis”–a critical commentary. Free Radic Biol Med 45:1217–1219PubMedCrossRef Zielonka J, Kalyanaraman B (2008) “ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis”–a critical commentary. Free Radic Biol Med 45:1217–1219PubMedCrossRef
61.
Zurück zum Zitat Scherz-Shouval R, Shvets E, Fass E et al (2007) Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J 26:1749–1760PubMedCrossRef Scherz-Shouval R, Shvets E, Fass E et al (2007) Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J 26:1749–1760PubMedCrossRef
62.
Zurück zum Zitat Juhász G, Erdi B, Sass M, Neufeld TP (2007) Atg7-dependent autophagy promotes neuronal health, stress tolerance, and longevity but is dispensable for metamorphosis in Drosophila. Genes Dev 21:3061–3066PubMedCrossRef Juhász G, Erdi B, Sass M, Neufeld TP (2007) Atg7-dependent autophagy promotes neuronal health, stress tolerance, and longevity but is dispensable for metamorphosis in Drosophila. Genes Dev 21:3061–3066PubMedCrossRef
63.
Zurück zum Zitat Kuma A, Hatano M, Matsui M et al (2004) The role of autophagy during the early neonatal starvation period. Nature 432:1032–1036PubMedCrossRef Kuma A, Hatano M, Matsui M et al (2004) The role of autophagy during the early neonatal starvation period. Nature 432:1032–1036PubMedCrossRef
64.
Zurück zum Zitat Meléndez A, Tallóczy Z, Seaman M et al (2003) Autophagy genes are essential for dauer development and life-span extension in C. elegans. Science 301:1387–1391PubMedCrossRef Meléndez A, Tallóczy Z, Seaman M et al (2003) Autophagy genes are essential for dauer development and life-span extension in C. elegans. Science 301:1387–1391PubMedCrossRef
65.
Zurück zum Zitat Yamamoto M, Clark JD, Pastor JV et al (2005) Regulation of oxidative stress by the anti-aging hormone klotho. J Biol Chem 280:38029–38034PubMedCrossRef Yamamoto M, Clark JD, Pastor JV et al (2005) Regulation of oxidative stress by the anti-aging hormone klotho. J Biol Chem 280:38029–38034PubMedCrossRef
66.
Zurück zum Zitat Saito K, Ishizaka N, Mitani H, Ohno M, Nagai R (2003) Iron chelation and a free radical scavenger suppress angiotensin II-induced downregulation of klotho, an anti-aging gene, in rat. FEBS Lett 551:58–62PubMedCrossRef Saito K, Ishizaka N, Mitani H, Ohno M, Nagai R (2003) Iron chelation and a free radical scavenger suppress angiotensin II-induced downregulation of klotho, an anti-aging gene, in rat. FEBS Lett 551:58–62PubMedCrossRef
67.
Zurück zum Zitat Afanas’ev I (2010) Reactive oxygen species and age-related genes p66shc, Sirtuin, FOX03 and Klotho in senescence. Oxid Med Cell Longev 3:77–85PubMedCrossRef Afanas’ev I (2010) Reactive oxygen species and age-related genes p66shc, Sirtuin, FOX03 and Klotho in senescence. Oxid Med Cell Longev 3:77–85PubMedCrossRef
68.
Zurück zum Zitat Iwasaki Y, Nishiyama M, Taguchi T et al (2009) Insulin exhibits short-term anti-inflammatory but long-term proinflammatory effects in vitro. Mol Cell Endocrinol 298:25–32PubMedCrossRef Iwasaki Y, Nishiyama M, Taguchi T et al (2009) Insulin exhibits short-term anti-inflammatory but long-term proinflammatory effects in vitro. Mol Cell Endocrinol 298:25–32PubMedCrossRef
69.
Zurück zum Zitat Salminen A, Huuskonen J, Ojala J et al (2008) Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res Rev 7:83–105PubMedCrossRef Salminen A, Huuskonen J, Ojala J et al (2008) Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res Rev 7:83–105PubMedCrossRef
70.
Zurück zum Zitat Salminen A, Kaarniranta K (2009) Regulation of the aging process by autophagy. Trends Mol Med 15:217–224PubMedCrossRef Salminen A, Kaarniranta K (2009) Regulation of the aging process by autophagy. Trends Mol Med 15:217–224PubMedCrossRef
71.
Zurück zum Zitat Eskelinen EL, Saftig P (2004) Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta 1793:664–673CrossRef Eskelinen EL, Saftig P (2004) Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta 1793:664–673CrossRef
72.
Zurück zum Zitat Gozuacik D, Kimchi A (2004) Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23:2891–2906PubMedCrossRef Gozuacik D, Kimchi A (2004) Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23:2891–2906PubMedCrossRef
73.
Zurück zum Zitat Tanida I, Minematsu-Ikeguchi N, Ueno T, Kominami E (2005) Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy 1:84–91PubMedCrossRef Tanida I, Minematsu-Ikeguchi N, Ueno T, Kominami E (2005) Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy 1:84–91PubMedCrossRef
74.
Zurück zum Zitat Bunney TD, Katan M (2010) Phosphoinositide signalling in cancer: beyond PI3K and PTEN. Nat Rev Cancer 10:342–352PubMedCrossRef Bunney TD, Katan M (2010) Phosphoinositide signalling in cancer: beyond PI3K and PTEN. Nat Rev Cancer 10:342–352PubMedCrossRef
75.
Zurück zum Zitat Huang J, Manning BD (2009) A complex interplay between Akt, TSC2 and the two mTOR complexes. Biochem Soc Trans 37:217–222PubMedCrossRef Huang J, Manning BD (2009) A complex interplay between Akt, TSC2 and the two mTOR complexes. Biochem Soc Trans 37:217–222PubMedCrossRef
76.
Zurück zum Zitat Degtyarev M, De Mazière A, Orr C et al (2008) Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents. J Cell Biol 183:101–116PubMedCrossRef Degtyarev M, De Mazière A, Orr C et al (2008) Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents. J Cell Biol 183:101–116PubMedCrossRef
77.
Zurück zum Zitat Arico S, Petiot A, Bauvy C et al (2001) The tumor suppressor PTEN positively regulates macroautophagy by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway. J Biol Chem 276:35243–35246PubMedCrossRef Arico S, Petiot A, Bauvy C et al (2001) The tumor suppressor PTEN positively regulates macroautophagy by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway. J Biol Chem 276:35243–35246PubMedCrossRef
78.
Zurück zum Zitat Mammucari C, Milan G, Romanello V et al (2007) FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6:458–471PubMedCrossRef Mammucari C, Milan G, Romanello V et al (2007) FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6:458–471PubMedCrossRef
79.
Zurück zum Zitat Iida RH, Kanko S, Suga T, Morito M, Yamane A (2011) Autophagic-lysosomal pathway functions in the masseter and tongue muscles in the klotho mouse, a mouse model for aging. Mol Cell Biochem 348:89–98PubMedCrossRef Iida RH, Kanko S, Suga T, Morito M, Yamane A (2011) Autophagic-lysosomal pathway functions in the masseter and tongue muscles in the klotho mouse, a mouse model for aging. Mol Cell Biochem 348:89–98PubMedCrossRef
Metadaten
Titel
Klotho Acts as a Tumor Suppressor in Cancers
verfasst von
Biao Xie
Jinhui Chen
Bin Liu
Junkun Zhan
Publikationsdatum
01.10.2013
Verlag
Springer Netherlands
Erschienen in
Pathology & Oncology Research / Ausgabe 4/2013
Print ISSN: 1219-4956
Elektronische ISSN: 1532-2807
DOI
https://doi.org/10.1007/s12253-013-9663-8

Weitere Artikel der Ausgabe 4/2013

Pathology & Oncology Research 4/2013 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.