Oncogene (2014) 33, 4352–4358 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc

SHORT COMMUNICATION Pivotal role of augmented aB-crystallin in tumor development induced by deficient TSC1/2 complex

F Wang1, X Chen1,CLi1, Q Sun2, Y Chen3, Y Wang1, H Peng4, Z Liu1, R Chen1, K Liu5,HYan3,BHYe6, DJ Kwiatkowski7 and H Zhang1

Tuberous sclerosis complex 1 (TSC1) and TSC2 are suppressors of mechanistic target of rapamycin (mTOR). mTOR is the major component of two complexes: mTOR complex 1 (mTORC1) and mTORC2. Inactive mutation of either TSC1 or TSC2 unleashes mTOR signaling and consequently causes TSC, a benign tumor syndrome affecting multiple organs. We report here that expression of aB-crystallin was upregulated in Tsc1 À / À or Tsc2 À / À mouse embryonic fibroblasts, Eker rat uterine leiomyoma-derived Tsc2- deficient ELT3 cells, mutant Tsc2-associated mouse kidney tumors, and human lung lymphangioleiomyomatosis nodules. aB-crystallin was transcriptionally activated by mTOR complex 2 (mTORC2): nuclear factor-kappa B (NFkB) signaling cascade. The augmented aB-crystallin was critical for the migration, invasion and apoptotic resistance of Tsc2-defective cells. Disruption of aB-crystallin suppressed Tsc2-null cell proliferation and tumorigenesis. Therefore, enhanced aB-crystallin has an essential role in TSC1/2 complex deficiency-mediated tumorigenesis, and inhibition of aB-crystallin may complement the current therapy for TSC.

Oncogene (2014) 33, 4352–4358; doi:10.1038/onc.2013.401; published online 30 September 2013 Keywords: aB-crystallin; mTOR; NFkB; TSC; tumor

INTRODUCTION mutation of one of these two is the major cause for TSC, The receptor tyrosine kinase/phosphatidylinositol 3-kinase/AKT/ a benign tumor syndrome afflicting multiple organs.5,15–18 TSC is mechanistic target of rapamycin (RTK/PI3K/AKT/mTOR) signaling featured with cerebral cortical tubers, subependymal nodules, cascade, which has pivotal roles in cell metabolism, autophagy, renal angiomyolipomas, pulmonary lymphangiomyomatosis growth, proliferation and survival, is one of the most frequently (LAM), cardiac rhabdomyomas and facial angiofibroma.19 LAM is deregulated signaling pathways in cancers.1–4 The constitutive characterized by the abnormal proliferation of smooth muscle-like activation of this network is often caused by gain-of-function cells and cystic lung destruction. It is thought to be initiated by mutations of proto-oncogenes, such as epidermal growth factor histologically benign cells carrying TSC1 or TSC2 mutations that receptor (EGFR), PI3K and AKT, and by loss-of-function mutations of migrated from renal angiomyolipomas.20,21 tumor suppressors, such as phosphatase and tensin homolog In early 1990s, aB-crystallin had been found elevated in TSC (PTEN), tuberous sclerosis complex 1 (TSC1) and TSC2.5–8 As the lesions, including the brain, kidney, lung and heart.22,23 The major effector of this pathway, mTOR can integrate signals from putative role of enhanced aB-crystallin in inactive TSC1/2 growth factors, nutrients and energy supplies, and then regulate a complex-mediated tumorigenesis is unknown. Besides being a subset of expression and protein synthesis/modification in major component in eye lens, aB-crystallin is a ubiquitous small controlling cell growth, proliferation and survival.9 Being a serine/ heat-shock protein (sHSP) and can be induced by stress in threonine kinase, mTOR is the catalytic subunit of two distinct counteracting the deleterious aggregation of misfolded complexes named mTOR complex 1 (mTORC1) and mTORC2.10 and thus helping cell survival.24 Another cytoprotective function mTORC1 consists of mTOR, mLST8, Raptor and PRAS4011,12 and of aB-crystallin is antiapoptotic effect. aB-crystallin overexpression controls cell growth and proliferation by modulating mRNA can protect cells from apoptosis induced by a large panel of translation through phosphorylation of eukaryotic translation stimuli, whereas aB-crystallin deficiency sensitizes cells to initiation factor 4E binding protein (4E-BP) and ribosomal apoptosis.25,26 Overexpression of aB-crystallin has also been protein S6 kinase (S6K).13 mTORC2 contains mTOR, mLST8, Rictor detected in several human cancers.27–29 and SIN1 and regulates actin cytoskeleton dynamics.14 In contrast In this study, we investigated the significance of augmented to the well-documented mTORC1, the function and mechanism of aB-crystallin in TSC1/2 deficiency-induced tumorigenesis. We mTORC2 are less characterized. found that TSC1 and TSC2 were negative regulators of aB-crys- TSC1 and TSC2 tumor suppressors form a protein complex in tallin expression, and the upregulation of aB-crystallin was suppression of mTOR. mTOR hyperactivation due to inactive mTORC2 dependent. We then identified NFkB as the transducer

1State Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; 2Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China; 3Center for Coronary Heart Disease, Cardiovascular Institute and Fu Wai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; 4Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL, USA; 5Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; 6Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA and 7Division of Translational Medicine, Brigham and Women’s Hospital and Dana-Faber Cancer Institute, Harvard Medical School, Boston, MA, USA. Correspondence: Dr H Zhang, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100005, China. E-mail: [email protected] Received 11 November 2012; revised 22 July 2013; accepted 23 August 2013; published online 30 September 2013 mTORC2 upregulation of aB-Crystallin in TSC F Wang et al 4353 of mTORC2 upregulation of aB-crystallin. Disruption of aB-crys- we hypothesized that loss of TSC1/TSC2 complex promoted tallin suppressed the cell proliferation and tumorigenesis caused aberrant cell proliferation through induction of aB-crystallin expres- by TSC1/2 deficiency. sion. We found that both aB-crystallin mRNA and protein were markedly elevated in mouse embryonic fibroblasts (MEFs) with mTOR hyperactivation caused by lack of either Tsc1 or Tsc2 gene RESULTS AND DISCUSSION (Figures 1a and b), indicating that the upregulation of aB-crystallin mTORC2 positively regulates aB-crystallin expression in TSC expression occurs at the transcriptional level. Mouse kidney tumors TSC is featured with tumor lesions in multiple organs. aB-crystallin with hyperactive mTOR due to loss of heterozygosity for the has been found to be highly expressed in TSC lesions. Therefore, wild-type Tsc2 allele in Tsc2 þ / À mice17 exhibited enhanced

Figure 1. mTORC2 positively regulates aB-crystallin expression in TSC. Total RNA and protein lysates were extracted from WT, Tsc1 À / À (a)or Tsc2 À / À (b) MEFs for qRT-PCR (left) and immunoblotting (right), respectively. Data are presented as mean±s.e.m. from three independent experiments. *Po0.05 compared with control (t-test). (c) Kidney tumors from three Tsc2 þ /– mice were immunoblotted.17 (d)WT,Tsc2 À / À MEFs (left) and ELT3 cells (right) were treated with 10 nM rapamycin (R, Sigma-Aldrich, St Louis, MO, USA) or 3 mM WYE-354 (W, Merck) for 24 h and then subjected to immunoblotting. The bottom bar graph represents aB-crystallin levels upon normalization for b-actin as fold change over control. Tsc2 À / À MEFs (left) and ELT3 cells (right) were transfected with siRNA of mTOR (e), rictor (f), raptor (g) or scramble siRNA using Lipofectamine 2000 (Invitrogen Corporation, Carlsbad, CA, USA) following the manufacturer’s instructions and then subjected to immunoblotting. aB-crystallin levels are represented as in panel d. CAKRRRLpSpSLRA peptides were injected into rabbit to produce polyclonal antibody against phospho-S6 (Ser235/236). TSC2, b-actin and ERK antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). aB-crystallin antibody was purchased from Stressgen-Enzo Life Sciences (Farmingdale, NY, USA). TSC1 antibody was purchased from Abcam (Cambridge, MA, USA). mTOR, raptor, rictor and phospho-AKT (Ser473) antibodies were purchased from Cell Signaling Technology (Danvers, MA, USA). (d–g) All experiments were repeated at least two times with similar results.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4352 – 4358 mTORC2 upregulation of aB-Crystallin in TSC F Wang et al 4354 aB-crystallin expression (Figure 1c). By analyzing transcript profiling Table 1. Human LAM with hyperactive mTOR/aB-crystallin signaling of human LAM nodules compared with normal pulmonary artery smooth muscle cells from the Gene Expression Omnibus database Gene Fold changea tb P-valuec (GEO accession number GSE12027, deposited by Y. Zhang and VEGFD 30.2 À 6.28 0.001 G. Pacheco-Rodriguez) (Supplementary Table 1), we found VEGFD o 30 aB-crystallin 6.7 À 6.71 o0.001 (a marker of mTOR activation in LAM) and aB-crystallin were significantly increased in LAM nodules (Table 1). Taken together, Transcript profiling of 14 human LAM nodules and 7 pulmonary artery these results demonstrate that TSC1/TSC2 complex negatively smooth muscle cell lines is from the Gene Expression Omnibus database. aFold change is presented as LAM versus pulmonary artery smooth muscle regulates aB-crystallin expression both in vitro and in vivo. b c mTOR exists in two multiprotein complexes, rapamycin- cells. t-statistic. Po0.01 and a 1.5-fold change were considered 10 significant. sensitive mTORC1 and rapamycin-insensitive mTORC2. Because loss of TSC1 or TSC2 induces both mTOR hyperactivation and

Figure 2. mTORC2 stimulates aB-crystallin expression through induction of NFkB. (a) Protein lysates were extracted from WT or Tsc2 À / À MEFs for immunoblotting. Tsc2 À / À MEFs (left) and ELT3 cells (right) were transfected with siRNA of rictor (b), p65 (c) or scramble siRNA in Lipofectamine 2000 and then subjected to immunoblotting. p65 and phospho-p65 (ser536) antibodies were purchased from Cell Signaling Technology. (a-c) All experiments were repeated at least two times with similar results. (d) Schematic representation of the putative wt and mutated NFkB-binding sites within the promoter region of mouse aB-crystallin gene. (e) HEK293T cells were transfected with paB-crystallin- Luc reporter plasmid encoding firefly luciferase and p65 expression vector (pDsRed-p65) or control vector (pDsRed-Express-N1, Clontech, Palo Alto, CA, USA) using Lipofectamine 2000. pRL-TK plasmid (Promega, Madison, WI, USA) encoding Renilla luciferase was cotransfected as internal control. Relative luciferase activity was evaluated 24 h after transfection, by a Dual-Luciferase Reporter Assay System (Promega). Relative luciferase activity: firefly/Renilla luciferase activity. (f) Tsc2 À / À MEFs were cotransfected with paB-crystallin-Luc or pmutNFkB-Luc reporter plasmid and pRL-TK plasmid in Lipofectamine 2000. Relative luciferase activity was evaluated 48 h after transfection. Data are presented as mean±s.d. from three independent experiments. **Po0.01; ***Po0.001 compared with control (t-test).

Oncogene (2014) 4352 – 4358 & 2014 Macmillan Publishers Limited mTORC2 upregulation of aB-Crystallin in TSC F Wang et al 4355 aB-crystallin overexpression, we speculated that mTOR was a all of these cells (Figure 1d). Then we cloned a 2193-bp fragment positive regulator of aB-crystallin expression. To elucidate which of mouse aB-crystallin gene to luciferase reporter plasmid mTOR complex is responsible for the upregulation of aB-crystallin, (Figure 2d) and found that the augmented aB-crystallin promoter we first evaluated the effect of rapamycin on aB-crystallin activity in Tsc2 À / À MEFs was significantly suppressed with expression. Rapamycin treatment led to marked reduction in S6 WYE-354 treatment, whereas rapamycin treatment did not alter phosphorylation (a marker of mTORC1 activity), but no reduction aB-crystallin promoter activity (Supplementary Figures 1a and b). in aB-crystallin expression in WT MEFs, Tsc2 À / À MEFs or rat Furthermore, we knocked down mTOR, rictor or raptor, respec- uterine leiomyoma-derived Tsc2-null ELT3 cells (Figure 1d).31 We tively, in Tsc2 À / À MEFs and ELT3 cells. Reduction in mTOR or also treated these cells with WYE-354, an ATP-competitive mTOR rictor significantly decreased aB-crystallin (Figures 1e and f), kinase inhibitor that could suppress both mTORC1 and mTORC2 whereas reduced raptor did not alter the level of aB-crystallin activities.32 WYE-354 markedly reduced aB-crystallin expression in (Figure 1g). Consistently, knockdown of rictor, but not raptor,

Figure 3. Augmented aB-crystallin is critical for migration, invasion and resistance to serum deprivation-induced apoptosis of Tsc2-defective cells. (a) Tsc2 À / À MEFs (left) and ELT3 cells (right) stably expressing the shRNA for aB-crystallin or scramble shRNA were serum-starved for 24 h and then subjected to immunoblotting. Cleaved caspase-3 (Asp175) antibody was purchased from Cell Signaling Technology. WT and Tsc2 À / À MEFs (b), Tsc2 À / À MEFs (c) or ELT3 cells (d) stably expressing the shRNA for aB-crystallin or scramble shRNA (shV) suspended in serum-free DMEM were added to the upper wells of transwell chamber (8.0 mM pore size, Costar, Tewksbury, MA, USA) with or without ECM gel (Sigma-Aldrich). DMEM with 20% FBS was used as chemoattractant in the lower well. Cells were incubated at 37 1C and 5% CO2 for 12–20 h. The cells invading or migrating through the membrane were stained with crystal violet. Representative images (left) were presented. Scale bars, 50 mM. Number of migrating or invading cells was counted in at least five fields (right). Data are presented as mean±s.e.m. **Po0.01; ***Po0.001 compared with control (t-test). (a-d) All experiments were repeated at least two times with similar results.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4352 – 4358 mTORC2 upregulation of aB-Crystallin in TSC F Wang et al 4356 inhibited aB-crystallin promoter activity (Supplementary Figures 1c aB-crystallin gene expression. As aB-crystallin can suppress the and d). Taken together, mTORC2 activates aB-crystallin expression NFkB pathway in astrocytes following TNF stimulation,40 the in TSC1/2-deficient cells. It has been reported that mTORC2 has an negative feedback regulation of NFkBonaB-crystallin may essential role in TSC2-null cell growth and survival.33 As partially contribute to the benign nature of TSC-associated tumor. aB-crystallin is a downstream target of mTORC2, aB-crystallin may be targeted for the treatment of TSC and other hyperactive mTOR-mediated cancers. Essential role of aB-crystallin for Tsc2-null cells in resistant to serum deprivation-induced apoptosis It has been reported that TSC1/2 deficiency exerts resistance to mTORC2 stimulates aB-crystallin expression through induction of serum deprivation-induced apoptosis.41 As both aB-crystallin and NFkB loss of TSC1/2 inhibit apoptosis, we postulated that loss of TSC1/2 NFkB is a key transcriptional factor that regulates expression of a abrogated serum withdrawal-induced apoptosis at least partially large number of genes involved in apoptosis, proliferation, through induction of aB-crystallin. In comparison with Tsc2 / 34 À À tumorigenesis and metastasis. The cross talk between PI3K- MEFs, WT MEFs were more susceptible to serum deprivation AKT-mTOR and NFkB signaling cascade has been extensively (Supplementary Figure 3a), independent of p53 as both cells are 35,36 studied. NFkB activity may be potentiated by both mTORC1 deficient of p53.17 We knocked down aB-crystallin expression in 37–39 and mTORC2. NFkB signaling was enhanced in Tsc2 À / À Tsc2 À / À MEFs and ELT3 cells (p53 þ / þ ) and evaluated the MEF cells (Figure 2a). The augmented signaling was suppressed apoptosis induced by serum withdrawal in these cells. Although the with inhibition of either mTORC1 by raptor knockdown or basal level of apoptotic cells was higher in non-specific siRNA- rapamycin treatment or mTORC2 by rictor knockdown in both transfected cells than in non-transfected cells (Supplementary Tsc2 À / À MEFs and ELT3 cells (Supplementary Figure 2 and Figures 3b versus 3a), reduction in aB-crystallin expression Figure 2b). Therefore, similar to mTOR and mLST8, NFkB signaling sensitized Tsc2 À / À MEFs or ELT3 cells to apoptosis induced by is a component of both mTORC1 signaling cascade and mTORC2 serum withdrawal (Figure 3a and Supplementary Figure 3b). There- signaling network in the cells. fore, aB-crystallin contributes to resistance to serum deprivation- As aB-crystallin was upregulated at transcriptional level in induced apoptosis in Tsc2-deficient cells with or without p53. mTOR-activated cells, we tested the potential role of NFkBin As rapamycin or raptor knockdown failed to counteract the regulation of aB-crystallin expression. Knockdown of NFkB apoptotic resistance of Tsc2-null cells to serum limitation,41 this markedly reduced aB-crystallin expression (Figure 2c). To inves- newly dissected TSC1/2 regulation of apoptosis through mTORC2- tigate whether NFkB transactivates aB-crystallin expression, we NFkB-aB-crystallin signaling cascade may attribute to this phenotype identified two putative NFkB-binding sites within the promoter of Tsc2-null cells. Moreover, we identify aB-crystallin as a therapeutic region on mouse aB-crystallin gene (Figure 2d). Luciferase reporter target for induction of cell death in the treatment of TSC. assay in HEK293T cells showed that overexpression of p65 markedly enhanced the transcriptional activity of the aB-crystallin promoter (Figure 2e). Moreover, aB-crystallin promoter activity in aB-crystallin is essential for the migration and invasion of TSC1/2 Tsc2 À / À MEFs was markedly attenuated when either one of its complex-deficient cells putative NFkB-binding sites was mutated (Figure 2f). Collectively, TSC1 and TSC2 can inhibit cell migration and invasion in vitro.42,43 these results demonstrate that mTORC2-NFkB axis transactivates In our study, Tsc2 À / À MEFs showed an B30-fold increase in

Figure 4. Inhibition of aB-crystallin suppresses cell proliferation and tumorigenesis mediated by TSC2 deficiency. (a) NTC/T2-null cells are stably transduced with shRNA silencing aB-crystallin or scramble shRNA (shV) lentiviruses and then subjected to immunoblotting. (b) The proliferation of NTC/T2-null cells stably expressing shV or shaB-crystallin was examined using an MTT assay for up to 3 days. Data are presented as mean±s.d. from one experiment performed in four replicates. **Po0.01; ***Po0.001 compared with control (t-test). (c) Colony formation of NTC/T2-null cells stably expressing shV or shaB-crystallin.(a-c) All experiments were repeated at least two times with similar results. (d)NTC/ T2-null cells stably expressing shV or shaB-crystallin were injected s.c. into nude mice for analysis of tumor development (left) and survival of the tumor-bearing mice (right). Subcutaneous tumors were established as previously described.49 Immunodeficient nude mice (strain BALB/c, 6 weeks old) were obtained from the Vital River Laboratory Animal Technology (Beijing, China). Eight mice (four male and four female) were used in each cohort. All animals were maintained and used in accordance with the guidelines of the Animal Center of the Institute of Basic Medical Sciences, Peking Union Medical College & Chinese Academy of Medical Sciences, and was compliant with the regulation of Beijing Administration Office of Laboratory Animal on the care of experimental animals. (e) Tumors were collected from euthanized mice, and the formalin-fixed and paraffin-embedded tumor tissue sections were subjected to immunohistochemical staining. Representative images were presented. Scale bar, 50 mM.

Oncogene (2014) 4352 – 4358 & 2014 Macmillan Publishers Limited mTORC2 upregulation of aB-Crystallin in TSC F Wang et al 4357 transwell migration and an B5-fold increase at invading the ECM gel 3 Ma J, Sun Q, Mi R, Zhang H. Avian influenza A virus H5N1 causes autophagy- compared with WT MEFs (Figure 3b). As both aB-crystallin over- mediated cell death through suppression of mTOR signaling. J Genet Genomics expression and loss of TSC1/2 promote cell migration and invasion, 2011; 38: 533–537. we assumed that loss of TSC1/2 promoted cell migration and 4 Sun Q, Chen X, Ma J, Peng H, Wang F, Zha X et al. Mammalian target of rapamycin invasion at least in part through enhancement of aB-crystallin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic expression. To test this possibility, aB-crystallin was knocked down in glycolysis and tumor growth. Proc Natl Acad Sci USA 2011; 108: 4129–4134. Tsc2 À / À MEFs and ELT3 cells, and the consequent effects on cell 5 Kwiatkowski DJ, Zhang H, Bandura JL, Heiberger KM, Glogauer M, el-Hashemite N migration and invasion were evaluated. Reduction in aB-crystallin et al. A mouse model of TSC1 reveals sex-dependent lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1 null cells. Hum expression led to a 440% suppression in transwell migration and a Mol Genet 2002; 11: 525–534. 450% inhibition at invading the ECM gel of Tsc2 À / À MEFs 6 Potter CJ, Pedraza LG, Xu T. Akt regulates growth by directly phosphorylating (Figure 3c). Similarly, reduction in aB-crystallin expression led to a Tsc2. Nat Cell Biol 2002; 4: 658–665. 470% decrease in transwell migration and a 485% decrease at 7 Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated and inhibited by Akt invading the ECM gel of ELT3 cells (Figure 3d). As metastasis of LAM and suppresses mTOR signalling. Nat Cell Biol 2002; 4: 648–657. cells from renal angiomyolipomas to the lungs is a critical step of 8 Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LC. Identification of the LAM pathogenesis, aB-crystallin may have a role in dissemination of tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target LAM cells and be a potential target for the treatment of LAM and TSC. of the phosphoinositide 3-kinase/akt pathway. Mol Cell 2002; 10: 151–162. 9 Holz MK, Ballif BA, Gygi SP, Blenis J. mTOR and S6K1 mediate assembly of the translation preinitiation complex through dynamic protein interchange and aB-crystallin is required for the proliferation and tumorigenesis of ordered phosphorylation events. Cell 2005; 123: 569–580. Tsc2-null cells 10 Loewith R, Jacinto E, Wullschleger S, Lorberg A, Crespo JL, Bonenfant D et al. Two aB-crystallin has been identified as a novel oncoprotein. Thus, TOR complexes, only one of which is rapamycin sensitive, have distinct roles in mTORC2-mediated upregulation of aB-crystallin might have a cell growth control. Mol Cell 2002; 10: 457–468. crucial role in loss of TSC1/2 complex-induced tumorigenesis. We 11 Hara K, Maruki Y, Long X, Yoshino K, Oshiro N, Hidayat S et al. Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 2002; 110: 177–189. stably knocked down aB-crystallin in NTC/T2-null cells, which were 12 Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-Bromage H et al. derived from a subcutaneous tumor formed by Tsc2-null MEFs in a mTOR interacts with raptor to form a nutrient-sensitive complex that signals to 44 nude mouse (Figure 4a). Reduction in aB-crystallin expression the cell growth machinery. Cell 2002; 110: 163–175. compromised the proliferation and colony formation of NTC/T2- 13 Gingras AC, Raught B, Sonenberg N. Regulation of translation initiation by FRAP/ null cells (Figure 4b and c). To evaluate the in vivo effect of mTOR. Genes Dev 2001; 15: 807–826. decreased aB-crystallin expression in NTC/T2-null cells, we 14 Sarbassov DD, Ali SM, Kim DH, Guertin DA, Latek RR, Erdjument-Bromage H et al. examined the tumorigenicity of NTC/T2-null cells expressing Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and shaB-crystallin or shvector in a subcutaneous tumor model in raptor-independent pathway that regulates the cytoskeleton. Curr Biol 2004; 14: nude mice. Reduction in aB-crystallin expression significantly 1296–1302. 15 The European 16 Tuberous Sclerosis Consortium. Identification and attenuated the tumorigenesis of NTC/T2-null cells, as assessed by characterization of the tuberous sclerosis gene on . Cell 1993; 75: the delayed tumor formation and extended survival of the tumor- 1305–1315. bearing mice (Figure 4d). In addition, more apoptotic cells, as 16 van Slegtenhorst M, de Hoogt R, Hermans C, Nellist M, Janssen B, Verhoef S et al. revealed by the expression of active (cleaved) caspase-3, were Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. Science presented in the tumor samples with depleted aB-crystallin 1997; 277: 805–808. expression (Figure 4e). These data indicate that augmented 17 Zhang H, Cicchetti G, Onda H, Koon HB, Asrican K, Bajraszewski N et al. Loss of aB-crystallin could contribute to defective TSC1/2-mediated Tsc1/Tsc2 activates mTOR and disrupts PI3K-Akt signaling through down- tumorigenesis and targeting aB-crystallin may help TSC patients. regulation of PDGFR. J Clin Invest 2003; 112: 1223–1233. Although encouraging results with rapamycin and its analogs 18 El-Hashemite N, Zhang H, Henske EP, Kwiatkowski DJ. Mutation in TSC2 and have been seen in the treatment of TSC/LAM, majority of tumors activation of mammalian target of rapamycin signalling pathway in renal angiomyolipoma. Lancet 2003; 361: 1348–1349. regrew and decline of lung function resumed once the treatment 45–47 19 Roach ES, Gomez MR, Northrup H. Tuberous sclerosis complex consensus con- was discontinued. AKT reactivation by disrupting mTOR ference: revised clinical diagnostic criteria. J Child Neurol 1998; 13: 624–628. suppression on AKT may compromise the efficiency of 20 Carsillo T, Astrinidis A, Henske EP. Mutations in the tuberous sclerosis complex 45,47 rapamycin, and contributes to its cytostatic effect in gene TSC2 are a cause of sporadic pulmonary lymphangioleiomyomatosis. Proc suppression of cell proliferation.48 As inhibition of both mTORC1 Natl Acad Sci USA 2000; 97: 6085–6090. and mTORC2 with ATP-competitive inhibitors may associate with 21 Bittmann I, Rolf B, Amann G, Lohrs U. Recurrence of lymphangioleiomyomatosis broad side effects and the upregulation of aB-crystallin in TSC is after single lung transplantation: new insights into pathogenesis. Hum Pathol mTORC2 dependent, the combinatorial suppression of mTORC1 2003; 34:95–98. and aB-crystallin should be applied in the treatment of oncogenic 22 Iwaki T, Tateishi J. Immunohistochemical demonstration of alphaB-crystallin in hamartomas of tuberous sclerosis. Am J Pathol 1991; 139: 1303–1308. RTK/PI3K/AKT/mTOR signaling propagation-associated TSC/LAM. 23 Kupsky WJ, Frosch M. Tuberous sclerosis variant with production of Rosenthal fibers (alpha B-crystallin). Pediatr Pathol 1993; 13: 869–873. CONFLICT OF INTEREST 24 Parcellier A, Schmitt E, Brunet M, Hammann A, Solary E, Garrido C. Small heat shock proteins HSP27 and alphaB-crystallin: cytoprotective and oncogenic func- The authors declare no conflict of interest. tions. Antioxid Redox Signal 2005; 7: 404–413. 25 Mao YW, Liu JP, Xiang H, Li DW. Human alphaA- and alphaB-crystallins bind to Bax ACKNOWLEDGEMENTS and Bcl-X(S) to sequester their translocation during staurosporine-induced apoptosis. Cell Death Differ 2004; 11: 512–526. This study was supported by National Basic Research Program of China 973 Program 26 Kamradt MC, Lu M, Werner ME, Kwan T, Chen F, Strohecker A et al. The small heat Grants 2009CB522203 and 2011CB965002 and National Natural Science Foundation shock protein alpha B-crystallin is a novel inhibitor of TRAIL-induced apoptosis of China Grants 81130085 and 81101516. that suppresses the activation of caspase-3. J Biol Chem 2005; 280: 11059–11066. 27 Aoyama A, Steiger RH, Frohli E, Schafer R, von Deimling A, Wiestler OD et al. Expression of alpha B-crystallin in human brain tumors. Int J Cancer 1993; 55: REFERENCES 760–764. 1 Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell 2007; 28 Shi T, Dong F, Liou LS, Duan ZH, Novick AC, DiDonato JA. Differential protein 12: 9–22. profiling in renal-cell carcinoma. Mol Carcinog 2004; 40: 47–61. 2 Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR, Sher A et al. The translational 29 Sitterding SM, Wiseman WR, Schiller CL, Luan C, Chen F, Moyano JV et al. AlphaB- landscape of mTOR signalling steers cancer initiation and metastasis. Nature 2012; crystallin: a novel marker of invasive basal-like and metaplastic breast carcinomas. 485: 55–61. Ann Diagn Pathol 2008; 12: 33–40.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4352 – 4358 mTORC2 upregulation of aB-Crystallin in TSC F Wang et al 4358 30 Young LR, Inoue Y, McCormack FX. Diagnostic potential of serum VEGF-D for 40 Ousman SS, Tomooka BH, van Noort JM, Wawrousek EF, O’Connor KC, Hafler DA lymphangioleiomyomatosis. N Engl J Med 2008; 358: 199–200. et al. Protective and therapeutic role for alphaB-crystallin in autoimmune 31 Howe SR, Gottardis MM, Everitt JI, Goldsworthy TL, Wolf DC, Walker C. Rodent demyelination. Nature 2007; 448: 474–479. model of reproductive tract leiomyomata. Establishment and characterization of 41 Bhaskar PT, Nogueira V, Patra KC, Jeon SM, Park Y, Robey RB et al. mTORC1 tumor-derived cell lines. Am J Pathol 1995; 146: 1568–1579. hyperactivity inhibits serum deprivation-induced apoptosis via increased hex- 32 Yu K, Toral-Barza L, Shi C, Zhang WG, Lucas J, Shor B et al. Biochemical, cellular, okinase II and GLUT1 expression, sustained Mcl-1 expression, and glycogen and in vivo activity of novel ATP-competitive and selective inhibitors of the synthase kinase 3beta inhibition. Mol Cell Biol 2009; 29: 5136–5147. mammalian target of rapamycin. Cancer Res 2009; 69: 6232–6240. 42 Lamb RF, Roy C, Diefenbach TJ, Vinters HV, Johnson MW, Jay DG et al. The TSC1 33 Goncharova EA, Goncharov DA, Li H, Pimtong W, Lu S, Khavin I et al. mTORC2 is tumour suppressor hamartin regulates cell adhesion through ERM proteins and required for proliferation and survival of TSC2-null cells. Mol Cell Biol 2011; 31: the GTPase Rho. Nat Cell Biol 2000; 2: 281–287. 2484–2498. 43 Astrinidis A, Cash TP, Hunter DS, Walker CL, Chernoff J, Henske EP. Tuberin, the 34 Karin M. Nuclear factor-kappaB in cancer development and progression. Nature tuberous sclerosis complex 2 tumor suppressor gene product, regulates Rho 2006; 441: 431–436. activation, cell adhesion and migration. Oncogene 2002; 21: 8470–8476. 35 Ozes ON, Mayo LD, Gustin JA, Pfeffer SR, Pfeffer LM, Donner DB. NF-kappaB 44 Lee L, Sudentas P, Donohue B, Asrican K, Worku A, Walker V et al. Efficacy of a activation by tumour necrosis factor requires the Akt serine-threonine kinase. rapamycin analog (CCI-779) and IFN-gamma in tuberous sclerosis mouse models. Nature 1999; 401: 82–85. Genes Cancer 2005; 42: 213–227. 36 Dan HC, Cooper MJ, Cogswell PC, Duncan JA, Ting JP, Baldwin AS. Akt-dependent 45 Bissler JJ, McCormack FX, Young LR, Elwing JM, Chuck G, Leonard JM et al. regulation of NF-{kappa}B is controlled by mTOR and Raptor in association with Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleio- IKK. Genes Dev 2008; 22: 1490–1500. myomatosis. N Engl J Med 2008; 358: 140–151. 37 Ma J, Meng Y, Kwiatkowski DJ, Chen X, Peng H, Sun Q et al. Mammalian target of 46 Krueger DA, Care MM, Holland K, Agricola K, Tudor C, Mangeshkar P et al. Ever- rapamycin regulates murine and human cell differentiation through STAT3/p63/ olimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Jagged/Notch cascade. J Clin Invest 2010; 120: 103–114. Med 2010; 363: 1801–1811. 38 Lang SA, Hackl C, Moser C, Fichtner-Feigl S, Koehl GE, Schlitt HJ et al. Implication 47 McCormack FX, Inoue Y, Moss J, Singer LG, Strange C, Nakata K et al. Efficacy and of RICTOR in the mTOR inhibitor-mediated induction of insulin-like growth factor-I safety of sirolimus in lymphangioleiomyomatosis. NEnglJMed2011; 364: 1595–1606. receptor (IGF-IR) and human epidermal growth factor receptor-2 (Her2) 48 Hosoi H, Dilling MB, Liu LN, Danks MK, Shikata T, Sekulic A et al. Studies on the expression in gastrointestinal cancer cells. Biochim Biophys Acta 2010; 1803: mechanism of resistance to rapamycin in human cancer cells. Mol Pharmacol 435–442. 1998; 54: 815–824. 39 Tanaka K, Babic I, Nathanson D, Akhavan D, Guo D, Gini B et al. Oncogenic EGFR 49 Zhang H, Bajraszewski N, Wu E, Wang H, Moseman AP, Dabora SL et al. PDGFRs signaling activates an mTORC2-NF-kappaB pathway that promotes chemotherapy are critical for PI3K/Akt activation and negatively regulated by mTOR. J Clin Invest resistance. Cancer Discov 2011; 1: 524–538. 2007; 117: 730–738.

Supplementary Information accompanies this paper on the Oncogene website (http://www.nature.com/onc)

Oncogene (2014) 4352 – 4358 & 2014 Macmillan Publishers Limited