<<

REVIEW Endocrine-Related Cancer (2006) 13 401–413 Beyond peroxisome proliferator-activated receptor c signaling: the multi-facets of the antitumor effect of

J-R Weng1;2 , C-Y Chen3;4, J J Pinzone5, M D Ringel5 and C-S Chen6

1Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan 2China Medical University Hospital, Taichung 404, Taiwan 3Department of Family Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan 4Division of Gerontology Research, National Health Research Institutes, Taipei, Taiwan 5Divisions of Endocrinology and Oncology, Department of Internal Medicine and 6Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Parks Hall, Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, USA

(Requests for offprints should be addressed to C-S Chen; Email: [email protected]

Abstract

Certain members of the (TZD) family of the peroxisome proliferator-activated receptor g (PPARg) agonists, such as and , exhibit antitumor activities; however, the underlying mechanism remains inconclusive. Substantial evidence suggests that the antiproliferative effect of these TZD members in cancer cells is independent of PPARg activation. To discern the role of PPARg in the antitumor effects of TZDs, we have synthesized PPARg- inactive TZD analogs which, although devoid of PPARg activity, retain the ability to induce apoptosis with a potency equal to that of their parental TZDs in cancer cell lines with varying PPARg expression status. Mechanistic studies from this and other laboratories have further suggested that troglitazone and ciglitazone mediate antiproliferative effects through a complexity of PPARg-independent mechanisms. Evidence indicates that troglitazone and ciglitazone block BH3 domain-mediated interactions between the anti apoptotic Bcl-2 (B-cell leukemia/lymphoma 2) members Bcl-2/Bcl-xL and proapoptotic Bcl-2 members. Moreover, these TZDs facilitate the degradation of cyclin D1 and caspase-8-related FADD-like IL-l-converting enzyme (FLICE)-inhibitory protein through proteasome-mediated proteolysis, and down-regulate the gene expression of prostate-specific antigen gene expression by inhibiting androgen activation of the androgen response elements in the promoter region. More importantly, dissociation of the effects of TZDs on apoptosis from their original pharmacological activity (i.e. PPARg activation) provides a molecular basis for the exploitation of these compounds to develop different types of molecularly targeted anticancer agents. These TZD-derived novel therapeutic agents, alone or in combination with other anticancer drugs, have translational relevance in fostering effective strategies for cancer treatment. Endocrine-Related Cancer (2006) 13 401–413

Introduction synthase, and fatty acid transport protein (Koeffler 2003). In addition, TZDs have been demonstrated to Thiazolidinediones (TZDs), including troglitazone promote the differentiation of preadipocytes by (Rezulin), (Avandia), mimicking certain genomic effects of on (Actos), and ciglitazone, are synthetic ligands for adipocytes (Tontonoz et al.1994),andtoregulate the peroxisome proliferator-activated receptor g glucose homeostasis (Saltiel & Olefsky 1996). Con- (PPARg) (for review see Day 1999) (Fig. 1). This sequently, the TZDs offer a new type of oral therapy family of PPARg agonists improves insulin sensitivity in type II diabetes to reduce and to by increasing transcription of certain insulin-sensitive assist glycemic control. genes involved in the metabolism and transport Recent evidence indicates that certain TZD of lipids in adipocytes, such as lipoprotein lipase, members, especially troglitazone and ciglitazone, adipocyte fatty acid-binding protein, acyl-CoA exhibit moderate antiproliferative activities against

Endocrine-Related Cancer (2006) 13 401–413 DOI:10.1677/erc.1.01182 1351-0088/06/013–401 # 2006 Society for Endocrinology Printed in Great Britain Online version via http://www.endocrinology-journals.org Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access J-R Weng et al.: PPARg-independent antitumor effects of thiazolidinediones

same mechanism to the terminal differentiation and cell cycle arrest of tumor cells (Tontonoz et al. 1997). However, the PPARg-target genes that mediate the antiproliferative effects remain elusive, as genomic responses to PPARg activation in cancer cells are highly complicated (Gupta et al.2001). Reported causal mechanisms include attenuated expression of protein phosphatase 2A (Altiok et al. 1997), cyclins D1 and E, inflammatory cytokines and transcription factors (Koeffler 2003), and increased expression of an array of gene products linked to growth regulation and cell maturation (Gupta et al. 2001). On the other hand, several lines of evidence have suggested that the inhibitory effect of TZDs on Figure 1 Chemical structures of troglitazone, ciglitazone, tumor cell proliferation is independent of PPARg rosiglitazone, and pioglitazone, and their respective activation. For example, sensitivity of cancer cells PPARg-inactive Á2 derivatives (Á2-TG, Á2-CG, Á2-RG, and to TZD-induced growth inhibition does not Á2-PG). The introduction of the double bond adjoining the correlate with the level of PPARg expression, and terminal thiazolidinedione ring results in the abrogation of the PPARg ligand property. there exists a three orders of magnitude discrepancy between the concentration required to produce many epithelial-derived human cancer cell lines antitumor effects and that to modify insulin action including those of prostate (Kubota et al.1998), (Day 1999). In addition, the in vitro antitumor breast (Yin et al. 2001), colon (Kato et al.2004), effects appear to be structure specific irrespective of thyroid (Ohta et al. 2001), lung (Tsubouchi et al. their potency in PPARg activation, i.e. troglitazone 2000), and pituitary carcinoma (Heaney et al.2003). and ciglitazone are active while rosiglitazone and This growth inhibition was linked to the G1 phase pioglitazone are not. To date, an array of non- cell cycle arrest through the up-expression of the PPARg targets has been implicated in the antitumor cyclin-dependent kinase inhibitors p21 and p27 activities of troglitazone and/or ciglitazone in (Koga et al. 2001, 2003, Takeuchi et al. 2002, Yoshi- different cell systems, which include intracellular zawa et al.2002,Baeet al. 2003) and/or repression Ca2þ stores (Palakurthi et al. 2001), phosphorylating of cyclin D1 expression (Wang et al. 2001, Yin et al. activation of extracellular signal-regulated kinases 2001, Lapillonne et al. 2003, Qin et al.2003).Mean- (Gouni-Berthold et al. 2001, Takeda et al. 2001), while, data from this and other laboratories have c-Jun N-terminal protein kinase, and p38 (Bae & indicated that normal prostate epithelial cells (not Song 2003), up-regulation of early growth shown) and T lymphocytes (Harris & Phipps 2002) response-1 (Baek et al. 2003), the cyclin-dependent are more resistant to apoptotic induction by these kinase (CDK) inhibitors p27kip1 (Motomura et al. TZDs. In the light of this cancer-specific effect, the 2000) and p21WAF=CIP1 (Sugimura et al. 1999), the potential use of these PPARg agonists as chemo- tumor suppressor protein p53 and the p53- preventive agents has received much attention (for a responsive stress protein Gadd45 (Okura et al. review see Badawi & Badr 2002, Kopelovich et al. 2000), and altered expression of B-cell leukemia/ 2002, Smith & Kantoff 2002, Bull 2003, Koeffler lymphoma 2(Bcl-2) family members (Bae & Song 2003, Leibowitz & Kantoff 2003, Grommes et al. 2003). However, some of these targets appear to be 2004, Jiang et al. 2004). Moreover, animal model cell type specific due to differences in signaling studies have demonstrated the in vivo efficacy of pathways regulating cell growth and survival in troglitazone in colon, prostate, and cancers different cell systems. (Kubota et al. 1998, Sarraf et al.1998,Yuet al. In considering the translational potential of using 2006) and that of rosiglitazone in pituitary tumors PPARg ligand in cancer prevention and therapy, our (Henry et al. 2000, Heaney et al. 2002). Despite these research has focused on elucidating the mechanisms advances, the mechanism underlying the antitumor underlying the antitumor effect of TZDs. In the effects of TZD remains unclear. As PPARg-mediated following sections, evidence that dissociates the effects of TZDs promote the differentiation of effect of TZDs on apoptosis from PPARg-activating preadipocytes, one school of thought attributes the activity is presented. This review also provides an

402 www.endocrinology-journals.org Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access Endocrine-Related Cancer (2006) 13 401–413 overview of recent findings from this and other evidenced by mitochondrial cytochrome c release laboratories concerning plausible PPARg-indepen- in PC-3 cell (Fig. 2B, right panel). dent targets that underlie TZD-mediated apoptosis. Similar results were obtained with ciglitazone and Equally importantly, the use of TZDs as a molecular Á2-CG with respect to cytochrome c-dependent platform to develop novel mechanism-based thera- apoptotic death in PC-3 cells (Fig. 3), with relative peutic agents will be discussed. potency similar to that of troglitazone and Á2-TG. In contrast, rosiglitazone, pioglitazone, and their Demonstration of PPAR-independent Á2 derivatives showed marginal effects, even at 50 mM, on cell death in PC-3 cells (Fig. 3). antiproliferative effects of TZDs Together, these data suggest that TZDs mediated We hypothesized that if TZDs mediate antitumor effects apoptosis induction in prostate cancer cell systems independently of PPARg, one would be able to disso- irrespective of PPARg activation. Mechanistic ciate these two pharmacological activities via structural studies from this and other laboratories have modifications of these molecules. This premise was further suggested that TZDs mediate proapoptotic corroborated by the development of a novel class of effects through a complexity of PPARg-independent PPARg-inactive TZD analogs, which was accomplished molecular targets, which include, but are not limited by introducing a double bond adjoining the terminal to, Bcl-2/Bcl-xL, cyclin D1, FADD-like IL-l-convert- -2,4-dione ring (Fig. 1) (Shiau et al. 2005). ing enzyme (FLICE)-inhibitory protein (FLIP), and Two lines of evidence indicate that this structural prostate specific antigen (PSA), which are summar- modification abrogated the ligand-binding ability of ized as follows. PPARg. First, these Á2 analogs (5-[4-(6-hydroxy-2,5, 7,8-tetramethyl-chroman-2-yl-methoxy)-benzylidene]- TZD-induced apoptosis involves the 2,4-thiazolidinedione (Á2-TG), 5-{4-[2-(methyl- pyridin-2-yl-amino)-ethoxy]-benzylidene}-2,4-thiazo- inhibition of Bcl-xL and Bcl-2 function lidinedione (Á2-RG), 5-{4-[2-(5-ethyl-pyridin-2-yl)- The functional relationship between antiapoptotic ethoxy]-benzylidene}-2,4-thiazolidinedione (Á2-PG), and proapoptotic Bcl-2 members in the regulation of and (5-[4-(1-methyl-cycohexylmethoxy)-benzylidene]- mitochondrial integrity is well documented (Cory thiazolidine-2,4-dione (Á2-CG)) were inactive in et al. 2003). Bcl-2 and Bcl-xL mediate their anti- PPARg activation according to a PPARg transcrip- apoptotic function through the sequestration of Bad, tion factor ELISA (Shiau et al. 2005). Secondly, Bax, and other proapoptotic Bcl-2 members via DU-145 prostate cancer cells were transfected with a BH3 domain-facilitated heterodimerization, thereby reporter construct that contains PPAR response abrogating their ability to induce mitochondrial element (PPRE) upstream of a luciferase gene. None cytochrome c release (Diaz et al. 1997, Sattler et al. of these Á2 derivatives elicited any significant 1997, Otter et al. 1998, Nouraini et al. 2000, Finnegan PPARg transactivation. The loss of PPARg activity et al. 2001). Thus, a balance between these two types of was presumably attributed to the structural rigidity, Bcl-2 members plays a crucial role in the regulation of as a result of the double bond introduction surround- the apoptotic machinery. ing the heterocyclic system. These PPARg-inactive TZD derivatives were Effect of troglitazone on the expression of examined in two prostate cancer cell lines with distinct Bcl-2 family members PPARg expression status, androgen-independent PC-3 and androgen-dependent LNCaP. Between It has been reported that troglitazone at high doses these two cell lines, PPARg was highly expressed in repressed Bcl-2 expression in MCF-7 breast cancer PC-3 cells, but was deficient in LNCaP cells (Elstner et al. 1998), and increased the expression (Fig. 2A). Nevertheless, despite a deficiency in level of the proapoptotic proteins Bad and Bax PPARg, LNCaP cells exhibited a higher degree in HepG2 hepatoma cells (Bae & Song 2003). This of susceptibility to troglitazone-mediated in vitro phenomenon, however, appears to be cell line antitumor effects compared with the PPARg-rich specific. In our study, troglitazone, even at 30 mM, PC-3 cells (Fig. 2B, left panel). In addition, Á2- did not cause appreciable changes in the expression TG, although devoid of PPARg-activating activity, of any of the aforementioned Bcl-2 members in was more potent than troglitazone in suppressing PC-3 cells with the exception of a slight decrease in cell proliferation in both cell lines. This growth Bad expression at 24-h exposure (Fig. 4) (Shiau inhibition was attributable to apoptotic death, as et al. 2005).

www.endocrinology-journals.org 403 Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access J-R Weng et al.: PPARg-independent antitumor effects of thiazolidinediones

Figure 2 Evidence that the effect of troglitazone on apoptosis in prostate cancer cells is dissociated from PPARg activation. (A) Western blot analysis of relative PPARg levels in PC-3 and LNCaP prostate cancer cells. (B, left panels) Dose-dependent effects of troglitazone and Á2-TG on the cell viability of PC-3 and LNCaP cells in serum-free RPMI 1640 medium for 24 h. Cell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay; (right panels) levels of cytochrome c release into cytoplasm induced by different doses of troglitazone and Á2-TG in PC-3 cells under the aforementioned treatment conditions. Values are means S.D. , P < 0:05.

Figure 3 Differential effects of ciglitazone, rosiglitazone, pioglitazone, and their Á2 derivatives on apoptotic death in PC-3 cells in serum-free RPMI 1640 medium for 24 h. Values are means S.D.

404 www.endocrinology-journals.org Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access Endocrine-Related Cancer (2006) 13 401–413

Bak interactions in PC-3 cells, i.e. the level of Bak associated with Bcl-2 and Bcl-xL was greatly reduced in troglitazone- and Á2-TG-treated cells as compared with DMSO control (Fig. 5A). As a result, liberation of proapoptotic Bcl-2 members induced apoptosis by facilitating cytochrome c release and consequent caspase-9 activation (Fig. 5B). Thirdly, overexpression of Bcl-xL provided LNCaP cells conferred protection against troglita- zone- and Á2-TG-induced apoptosis. As shown in Fig. 5C, ascending expression levels of Bcl-xL in Figure 4 Effect of troglitazone on the expression levels of Bcl-2 three transfected LNCaP stable clones (B11, B1, m family members in PC-3 cells. PC-3 cells were exposed to 30 M and B3) diminished the susceptibility to troglita- troglitazone in serum-free RPMI 1640 medium for the indicated time, and equal amounts of proteins from cell lysates were zone- and Á2-TG-induced apoptosis death as electrophoresed and probed by western blotting with individual compared with that of parental LNCaP cells. The antibodies. extent of cytoprotection conferred by ectopic Bcl- xL correlated with the Bcl-xL expression level. The Inhibition of Bcl-xL and Bcl-2 function excessive expression in B3 cells especially completely overcame the apoptotic effect of troglitazone and Nevertheless, we obtained several lines of evidence Á2-TG. that troglitazone and ciglitazone might inhibit the antiapoptotic function of Bcl-xL/Bcl-2 by blocking BH3 domain-mediated heterodimerization with pro- Antiproliferative mechanisms involve apoptotic Bcl-2 members (Shiau et al. 2005). First, modulation of cell cycle effectors data from the competitive fluorescence polarization Troglitazone and ciglitazone repress cyclin D1 analysis suggest that troglitazone, ciglitazone, and expression via proteasome-facilitated their Á2 counterparts compete with a Bak BH3- proteolysis domain peptide for binding to Bcl-xL and Bcl-2. Moreover, the potency of TZDs and their Á2 PPARg agonists, including 15-deoxy-Á12,14-prosta- analogs in inhibiting this peptide binding correlated glandin J2 (PGJ2), troglitazone, and ciglitazone, at with the respective effectiveness in inducing apopto- high doses, have been shown to down-regulate tic death in LNCaP and PC-3 prostate cancer cells cyclin D1 expression as part of their action to (Table 1). cause cell-cycle arrest and growth inhibition in For example, the inability of rosiglitazone and breast cancer cells (Wang et al. 2001, Yin et al. pioglitazone to trigger apoptotic death was reflected 2001, Lapillonne et al. 2003, Qin et al. 2003, 2004, in their lack of potency in disrupting interactions Huang et al. 2005). Cyclin D1 is a downstream between Bcl-xL/Bcl-2 and the Bak BH3 domain. effector of diverse proliferative and transforming Secondly, troglitazone and Á2-TG perturbed the signaling pathways, including those mediated by dynamics of intracellular Bcl-2/Bak and Bcl-xL/ b-catenin (Shtutman et al. 1999), estrogen receptor

Table 1 Correlation between the IC50 values of individual TZDs and Á2-TZDs for inhibiting BH3-mediated interactions of Bcl-xL and Bcl-2 with the Bak BH3 domain peptide and for inhibiting cell proliferation of PC-3 and LNCaP prostate cancer cells. Values are means S.D.

TG Á2-TG CG Á2-CG RG Á2-RG PG Á2-PG

1 IC50 in binding inhibition (mM) Bcl-xL 22 118 126 217 2 >50 >50 >50 >50 Bcl-2 22 118 124 322 3 >50 >50 >50 >50 2 IC50 in cell viability inhibition (M) PC-3 30 220 222 414 1 >50 >50 >50 >50 LN-CaP 22 314 1 n.d. n.d. n.d. n.d. n.d. n.d.

1Determined by fluorescence polarization analysis of the ability of individual compounds to inhibit the binding of the Flu-BakBH3 peptide with Bcl-xL or Bcl-2. 2Determined by the MTT assay. n.d., not determined.

www.endocrinology-journals.org 405 Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access J-R Weng et al.: PPARg-independent antitumor effects of thiazolidinediones

Figure 5 Troglitazone (TG) and Á2-TG trigger caspase-dependent apoptotic death by inhibiting heterodimer formation of Bcl-2 and Bcl-xL with Bak. (A) Effect of TG and Á2-TG on the dynamics of Bcl-2/Bak (left) and Bcl-xL/Bax (right) interactions in PC-3 cells. PC-3 cells were treated with 50 mMTGorÁ2-TG for 12 h, and cell lysates were immunoprecipitated with anti-Bcl-2 or anti-Bcl-xL antibodies. The immunoprecipitates were probed with anti-Bak antibodies by western blot analysis (WB). The bar graphs indicate the relative Bak levels, normalized to IgG light chain levels, in the three treatments. Values are means S.D. (n ¼ 3). P < 0:01. (B) Dose-dependent effect of TG and Á2-TG on caspase-9 activation in PC-3 cells. PC-3 cells were treated with TG or Á2-TG at the indicated concentrations for 24 h. Caspase-9 antibodies recognize the large subunits (35 and 37 kDa). (C) Ectopic Bcl-xL protects LNCaP cells from TG- and Á2-TG-induced apoptosis by attenuating cytochrome c release in an expression level-dependent manner. (Left panel) Ascending expression levels of ectopic Bcl-xL in B11, B1, and B3 clones by western blot analysis. (Right panel) Dose-dependent effects of TG (left panel) and Á2-TG (right panel) on apoptosis in LNCaP, B11, B1, and B3 cells. Data are means S.D. (n ¼ 3).

(ER) (Lukas et al. 1996, Wilcken et al. 1997, Prall (McIntosh et al. 1995, Kenny et al. 1999). In addi- et al. 1998), Her-2/Neu (Lee et al. 2000), nuclear tion to activating cyclin-dependent kinases (CDKs) factor-kB (Joyce et al. 1999, Henry et al. 2000), and sequestering CDK inhibitors in the G1/S Rac (Westwick et al. 1997), Ras (Albanese et al. transition, the function of cyclin D1 as a CDK- 1995), Src (Lee et al. 1999), signal transducer and independent activator of ER is especially note- activator of transcription (Bromberg et al. 1999, worthy (Neuman et al. 1997, Zwijsen et al. 1997, Matsumura et al. 1999), and Wnt (D’Amico et al. McMahon et al. 1999, Lamb et al. 2000). Cyclin 2000). In mammary cells, transcriptional activation D1 overexpression confers resistance to antiestro- of cyclin D1 in response to these mitogenic signals gens in breast cancer cells (Musgrove et al. 2001, leads to G1/S progression and increased prolifera- Hui et al. 2002), and represents a negative predictive tion. Cyclin D1 overexpression has been implicated factor for tamoxifen response (Stendahl et al. 2004). in oncogene-induced mammary tumorigenesis; it Together, these findings suggest that an anti-cyclin has been noted that more than 50% of primary D1 therapy might be highly specific for treating breast carcinomas correlated with poor prognosis human breast cancer (Yu et al. 2001). An urgent

406 www.endocrinology-journals.org Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access Endocrine-Related Cancer (2006) 13 401–413 need therefore exists to develop potent cyclin lacked appreciable activity at comparable concen- D1-ablative agents that are effective in the trations (not shown). therapeutically attainable range (5 mM) for breast Secondly, troglitazone-mediated cyclin D1 down- cancer prevention and/or therapy. regulation could not be rescued by the PPARg As troglitazone provides an attractive starting antagonist 2-chloro-5-nitro-N-phenylbenzamide point for this drug discovery effort, the mechanism (GW9662). Thirdly, despite significantly higher underlying its effect on cyclin D1 repression consti- PPARg expression, MDA-MB-231 breast cancer tuted the focus of our recent investigation (Huang cells were less susceptible to troglitazone-induced et al. 2005). Several lines of evidence suggest that cyclin D1 ablation than MCF-7 cells. Although it troglitazone mediates the repression of cyclin D1 has been reported that the PPARg ligands PGJ2 expression via a PPARg-independent mechanism. and troglitazone inhibited cyclin D1 transcription First, Á2-TG and Á2-CG, although devoid of by activating PPARg in MCF-7 cells (Wang et al. PPARg activity, were able to mediate cyclin D1 2001) and mouse skin keratinocytes (He et al. ablation with slightly higher potency than troglita- 2004) respectively, data from this and other zone and ciglitazone respectively in MCF-7 breast laboratories have indicated that troglitazone and cancer cells (Fig. 6A), while the more potent ciglitazone mediate cyclin D1 ablation in MCF-7 PPARg agonists rosiglitazone and pioglitazone cells by facilitating ubiquitination and proteasomal

Figure 6 Pharmacological evidence that the effect of troglitazone and ciglitazone on cyclin D1 down-regulation in MCF-7 cells is dissociated from PPARg activation. (A) Dose-dependent effect of troglitazone and Á2-TG on cyclin D1 expression for 24 h in MCF-7 cells by western blotting. (B) Dose-dependent effects of troglitazone on the expression of cyclins and CDKs in MCF-7 cells by western blot analysis.

www.endocrinology-journals.org 407 Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access J-R Weng et al.: PPARg-independent antitumor effects of thiazolidinediones proteolysis in a specific manner (Qin et al. 2003, against death receptor-induced apoptosis. Inhibition Huang et al. 2005). For example, PCR analysis of FLIP expression in tumor cells is of particular indicates that the mRNA level of cyclin D1 remained importance to TNF-related apoptosis inducing unaltered in drug-treated cells, indicating that the ligand (TRAIL)-based cancer therapy (Wajant repression was mediated at the post-transcriptional 2003). TRAIL is the ligand of death receptors, and level. Moreover, this drug-induced cyclin D1 repres- has attracted substantial clinical interests in light of sion could be rescued by proteasome inhibitors, and its ability to induce apoptosis preferentially in was preceded by increased ubiquitination. tumor cells. However, not all tumor cells respond It is noteworthy that the troglitazone-mediated to TRAIL, in part because of intracellular resistance cyclin D1 ablation was highly specific. Among all mechanisms (Ashkenazi 2002). Recent reports cyclins and CDKs examined (cyclins D2. D3, A, B, indicate that PPARg agonists including troglitazone and E, and CDKs 2 and 4) as well as p53, only could sensitize tumor cells to TRAIL-induced cyclin D2 and CDK4 showed a slight decrease in apoptosis (Goke et al. 2000, Kim et al. 2002) as a the expression level in drug-treated MCF-7 cells result of the proteasome-dependent proteolysis of (Fig. 6B). FLIP (Kim et al. 2002). As the mode of FLIP down-regulation is reminiscent of that of cyclin D1, there may exist cross-reactivity in troglitazone- Troglitazone up-regulates the expression of mediated proteasomal degradation of both the CDK inhibitor p21CIP1 proteins. Moreover, western blotting analysis indicates that, PSA is used as a serum biomarker for diagnosis in MCF-7 cells, troglitazone up-regulated the and progression of prostate cancer (Polascik et al. expression of p21CIP1, while p27KIP1 exhibited a 1999). It has been shown that troglitazone at dose-dependent decrease (Fig. 6B) (Huang et al. concentrations (10 mM) lower than that required 2005). However, the effect of troglitazone on the for growth inhibition (20 mM) was able to down- expression levels of these two CDK inhibitors regulate PSA expression in LNCaP prostate cancer varies among different cell lines. For example, cells through the inhibition of androgen activation troglitazone treatment led to increased expression of the androgen response elements in the regulatory of both p21CIP1 and p27KIP1 in several hepatocellular region of the PSA gene (Hisatake et al. 2000). More- carcinoma cells examined (Elnemr et al. 2000, Koga over, the same report indicates that troglitazone at et al. 2001), while in troglitazone-treated pancreatic 600800 mg/day stabilized PSA levels in a prostate cancer cells, only the expression level of p27KIP1, cancer patient with a radical prostectomy through- but not that of p21CIP1, increased (Motomura et al. out a 14-month period. 2000, Itami et al. 2001). These findings suggested that variations in responses exist among different Pharmacological exploitation of cancer cell lines. troglitazone to develop novel mechanism-based anticancer agents Other molecular mechanisms The above discussion clearly demonstrates that the In addition to the aforementioned effects, several antiproliferative activity of troglitazone is mediated other PPARg-independent mechanisms have been through multiple molecular targets independently reported to account for the antiproliferative action of PPARg. From a mechanistic perspective, dis- of TZDs in different cancer cells. Among these sociation of the aforementioned pharmacological proposed mechanisms, the effects on proteasome- activities from PPARg activation provide a mediated degradation of FLIP (Kim et al. 2002) molecular rationale to use troglitazone as a starting and down-regulation of PSA expression (Kubota point to develop novel mechanism-based therapeutic et al. 1998, Hisatake et al. 2000) are especially note- agents. The proof of the principle of this premise is 5- worthy in light of their clinical implications in cancer [4-(6-allyoxy-2,5,7,8-tetramethyl-chroman-2-yl-meth- therapy. oxy)-benzylidene]-2,4-thizolidinedione (Á2-TG-6) FLIP inhibits apoptosis induced by tumor necro- (Huang et al. 2005), a close structural analog that sis factor (TNF) family death receptor by blocking exhibited an order of magnitude higher potency caspase 8 activation (Wajant 2003). As demon- than troglitazone and Á2-TG in repressing cyclin strated by antisense-mediated down-regulation, D1 expression and inhibiting MCF-7 cell prolifera- decreased expression of FLIP confers sensitivity tion (Fig. 7).

408 www.endocrinology-journals.org Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access Endocrine-Related Cancer (2006) 13 401–413

Figure 7 Á2-TG-6, a structurally optimized cyclin D1-ablative agent. (A) Structure of Á2-TG-6. (B) Dose-dependent effect of Á2-TG-6 on cyclin D1 down-regulation in MCF-7 cells. (C) Dose-dependent effects of Á2-TG-6 versus TG and Á2-TG on MCF-7 cell viability as analyzed by MTT assay. (D) Dose-dependent effects of Á2-TG-6 versus TG on PARP cleavage. PARP proteolysis to the apoptosis-specific 85 kDa fragment was monitored by western blotting.

As shown, Á2-TG-6 reduced cyclin D1 levels at noteworthy that the structural requirement for concentrations as low as 2.5 mM compared with Bcl-2/Blc-xL inhibition differed from that of cyclin 20 mM for Á2-TG (Fig. 7B), and exhibited IC50 D1 ablation. of 8 mM versus 55 mM for Á2-TG in inhibiting MCF-7 cell viability (Fig. 7C). This antiproliferative Conclusion effect was attributable to apoptosis induction, as evidenced by poly(ADP-ribose)polymerase (PARP) Data from this and other laboratories have clearly cleavage (Fig. 7D). Lead optimization of Á2-TG-6 demonstrated that the effect of TZDs on triggering to generate more potent cyclin D1-ablative agents apoptosis in cancer cells is dissociated from that of is currently undergoing, the clinical implication of PPARg activation. To date, a number of important which is multifold. First, cyclin D1 ablation provides signaling mechanisms have been identified to under- specific protection against breast carcinogenesis lie TZD-mediated antitumor activities, including, (Yu et al. 2001). Secondly, in light of the role of but not limited to, inhibition of Bcl-2/Bcl-xL func- cyclin D1 overexpression in antiestrogen resistance, tion and repression of the expression of cyclin D1, cyclin D1 ablation may help overcome the resis- FLIP, and PSA. These findings provide molecular tance. Thirdly, the synergistic interaction between underpinnings for the molecular exploitation of flavopiridol and trastuzumab in inhibiting breast troglitazone and ciglitazone to develop different cancer cell proliferation is attributable, in part, to types of molecularly targeted anticancer agents. the reduction of cyclin D1 expression (Wu et al. Our data indicate that structural preference for 2002). These agents may sensitize cells to the anti- Bcl-2/Bcl-xL inhibition differs from that of cyclin proliferative action of either CDK inhibition or D1 ablation, while that between cyclin D1 and Her-2/Akt inhibition. FLIP is currently being investigated. Because of A similar approach has been taken to develop a the heterogeneity in the cancer cell population, potent Bcl-2/Bcl-xL inhibitor (TG-88) with an IC50 various genetic lesions/molecular defects allow of 1.8 mM based on fluorescence polarization analy- different subsets of tumor cells to exhibit differential sis. Oral TG-88 at a daily dose of 100 or 200 mg/kg sensitivity to apoptotic signals generated by different was effective in suppressing PC-3 xenograft tumor chemotherapeutic agents. Thus, these troglitazone- growth without causing weight loss or apparent derived agents in combination with other therapeutic toxicity, indicating its oral bioavailability and agents have translational relevance in fostering potential clinical use (Shiau et al. 2005). It is also effective strategies for cancer treatment.

www.endocrinology-journals.org 409 Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access J-R Weng et al.: PPARg-independent antitumor effects of thiazolidinediones

Acknowledgements kinase 3beta and cAMP-responsive element-binding protein-dependent pathways. Journal of Biological This work was supported by Public Health Service Chemistry 275 3264932657. Grants CA-94829 and CA-112250 (C-S C), and Day C 1999 Thiazolidinediones: a new class of antidiabetic K08CA101875 (J J P) from National Cancer drugs. Diabetic Medicine 16 179192. Institute, NIH, Department of Health and Human Diaz JL, Oltersdorf T, Horne W, McConnell M, Wilson G, Service, and by Army Grant W81XWH-05-1-0089 Weeks S, Garcia T & Fritz LC 1997 A common binding from Department of Defense Prostate Cancer site mediates heterodimerization and homodimerization of Research Program (C-S C). The authors declare Bcl-2 family members. Journal of Biological Chemistry 272 that there is no conflict of interest that would 1135011355. prejudice the impartiality of this scientific work. Elnemr A, Ohta T, Iwata K, Ninomia I, Fushida S, Nishimura G, Kitagawa H, Kayahara M, Yamamoto M, Terada T et al. 2000 PPARgamma ligand References (thiazolidinedione) induces growth arrest and differentiation markers of human pancreatic cancer cells. Albanese C, Johnson J, Watanabe G, Eklund N, Vu D, International Journal of Oncology 17 11571164. Arnold A & Pestell RG 1995 Transforming p21ras Elstner E, Muller C, Koshizuka K, Williamson EA, Park D, mutants and c-Ets-2 activate the cyclin D1 promoter Asou H, Shintaku P, Said JW, Heber D & Koeffler HP through distinguishable regions. Journal of Biological 1998 Ligands for peroxisome proliferator-activated Chemistry 270 2358923597. receptorgamma and retinoic acid receptor inhibit growth Altiok S, Xu M & Spiegelman BM 1997 PPARgamma induces and induce apoptosis of human breast cancer cells in vitro cell cycle withdrawal: inhibition of E2F/DP DNA-binding and in BNX mice. PNAS 95 88068811. activity via down-regulation of PP2A. Genes and Finnegan NM, Curtin JF, Prevost G, Morgan B & Cotter TG Development 11 19871998. 2001 Induction of apoptosis in prostate carcinoma cells by Ashkenazi A 2002 Targeting death and decoy receptors of the BH3 peptides which inhibit Bak/Bcl-2 interactions. British tumour-necrosis factor superfamily. Nature Review Cancer Journal of Cancer 85 115121. 2 420430. Goke R, Goke A, Goke B & Chen Y 2000 Regulation of Badawi AF & Badr MZ 2002 Chemoprevention of breast TRAIL-induced apoptosis by transcription factors. cancer by targeting cyclooxygenase-2 and peroxisome Cellular Immunology 201 7782. proliferator-activated receptor-gamma (Review). Gouni-Berthold I, Berthold HK, Weber AA, Ko Y, Seul C, International Journal of Oncology 20 11091122. Vetter H & Sachinidis A 2001 Troglitazone and Bae MA & Song BJ 2003 Critical role of c-Jun N-terminal rosiglitazone induce apoptosis of vascular smooth protein kinase activation in troglitazone-induced muscle cells through an extracellular signal-regulated apoptosis of human HepG2 hepatoma cells. Molecular kinase-independent pathway. Naunyn Schmiedeberg’s Pharmacology 63 401408. Archives of Pharmacology 363 215221. Bae MA, Rhee H & Song BJ 2003 Troglitazone but not Grommes C, Landreth GE & Heneka MT 2004 rosiglitazone induces G1 cell cycle arrest and apoptosis in Antineoplastic effects of peroxisome proliferator-activated human and rat hepatoma cell lines. Toxicology Letters 139 receptor gamma agonists. Lancet Oncology 5 419429. 6775. Gupta RA, Brockman JA, Sarraf P, Willson TM & Baek SJ, Wilson LC, Hsi LC & Eling TE 2003 Troglitazone, a DuBois RN 2001 Target genes of peroxisome peroxisome proliferator-activated receptor gamma (PPAR proliferator-activated receptor gamma in colorectal cancer gamma) ligand, selectively induces the early growth cells. Journal of Biological Chemistry 276 2968129687. response-1 gene independently of PPAR gamma. A novel Harris SG & Phipps RP 2002 Prostaglandin D(2), its mechanism for its anti-tumorigenic activity. Journal of metabolite 15-d-PGJ(2), and peroxisome proliferator Biological Chemistry 278 58455853. activated receptor-gamma agonists induce apoptosis in Bromberg JF, Wrzeszczynska MH, Devgan G, Zhao Y, transformed, but not normal, human T lineage cells. Pestell RG, Albanese C & Darnell JE Jr 1999 Stat3 as an Immunology 105 2334. oncogene. Cell 98 295303. He G, Thuillier P & Fischer SM 2004 Troglitazone inhibits Bull AW 2003 The role of peroxisome proliferator-activated cyclin D1 expression and cell cycling independently of receptor gamma in colon cancer and inflammatory bowel PPARgamma in normal mouse skin keratinocytes. Journal disease. Archives of Pathology and Laboratory Medicine of Investigative Dermatology 123 11101119. 127 11211123. Heaney AP, Fernando M, Yong WH & Melmed S 2002 Cory S, Huang DC & Adams JM 2003 The Bcl-2 family: roles Functional PPAR-gamma receptor is a novel therapeutic in cell survival and oncogenesis. Oncogene 22 85908607. target for ACTH-secreting pituitary adenomas. Nature D’Amico M, Hulit J, Amanatullah DF, Zafonte BT, Albanese Medicine 8 12811287. C, Bouzahzah B, Fu M, Augenlicht LH, Donehower LA, Heaney AP, Fernando M & Melmed S 2003 PPAR-gamma Takemaru K et al. 2000 The integrin-linked kinase receptor ligands: novel therapy for pituitary adenomas. regulates the cyclin D1 gene through glycogen synthase Journal of Clinical Investigation 111 13811388.

410 www.endocrinology-journals.org Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access Endocrine-Related Cancer (2006) 13 401–413

Henry DO, Moskalenko SA, Kaur KJ, Fu M, Pestell RG, Koga H, Harada M, Ohtsubo M, Shishido S, Kumemura H, Camonis JH & White MA 2000 Ral GTPases contribute to Hanada S, Taniguchi E, Yamashita K, Kumashiro R, regulation of cyclin D1 through activation of NF-kappaB. Ueno T et al. 2003 Troglitazone induces Molecular and Cellular Biology 20 80848092. p27Kip1-associated cell-cycle arrest through Hisatake JI, Ikezoe T, Carey M, Holden S, Tomoyasu S & down-regulating Skp2 in human hepatoma cells. Koeffler HP 2000 Down-regulation of prostate-specific Hepatology 37 10861096. antigen expression by ligands for peroxisome Kopelovich L, Fay JR, Glazer RI & Crowell JA 2002 proliferator-activated receptor gamma in human prostate Peroxisome proliferator-activated receptor modulators as cancer. Cancer Research 60 54945498. potential chemopreventive agents. Molecular Cancer Huang JW, Shiau CW, Yang YT, Kulp SK, Chen KF, Therapeutics 1 357363. Brueggemeier RW, Shapiro CL & Chen CS 2005 Kubota T, Koshizuka K, Williamson EA, Asou H, Said JW, Peroxisome proliferator-activated receptor Holden S, Miyoshi I & Koeffler HP 1998 Ligand for gamma-independent ablation of cyclin D1 by peroxisome proliferator-activated receptor gamma thiazolidinediones and their derivatives in breast cancer (troglitazone) has potent antitumor effect against human cells. Molecular Pharmacology 67 13421348. prostate cancer both in vitro and in vivo. Cancer Research Hui R, Finney GL, Carroll JS, Lee CS, Musgrove EA & 58 33443352. Sutherland RL 2002 Constitutive overexpression of cyclin Lamb J, Ladha MH, McMahon C, Sutherland RL & D1 but not cyclin E confers acute resistance to Ewen ME 2000 Regulation of the functional interaction antiestrogens in T-47D breast cancer cells. Cancer between cyclin D1 and the estrogen receptor. Molecular Research 62 69166923. and Cellular Biology 20 86678675. Itami A, Watanabe G, Shimada Y, Hashimoto Y, Kawamura Lapillonne H, Konopleva M, Tsao T, Gold D, McQueen T, J, Kato M, Hosotani R & Imamura M 2001 Ligands for Sutherland RL, Madden T & Andreeff M 2003 Activation peroxisome proliferator-activated receptor gamma inhibit of peroxisome proliferator-activated receptor gamma by a growth of pancreatic cancers both in vitro and in vivo. novel synthetic triterpenoid 2-cyano-3,12-dioxooleana- International Journal of Cancer 94 370376. 1,9-dien-28-oic acid induces growth arrest and apoptosis in Jiang M, Shappell SB & Hayward SW 2004 Approaches to breast cancer cells. Cancer Research 63 59265939. understanding the importance and clinical implications of Lee RJ, Albanese C, Stenger RJ, Watanabe G, Inghirami G, peroxisome proliferator-activated receptor gamma Haines GK 3rd, Webster M, Muller WJ, Brugge JS, Davis (PPARgamma) signaling in prostate cancer. Journal of RJ et al. 1999 pp60(v-src) induction of cyclin D1 requires Cellular Biochemistry 91 513527. collaborative interactions between the extracellular Joyce D, Bouzahzah B, Fu M, Albanese C, D’Amico M, Steer signal-regulated kinase, p38, and Jun kinase pathways. A J, Klein JU, Lee RJ, Segall JE, Westwick JK et al. 1999 role for cAMP response element-binding protein and Integration of Rac-dependent regulation of cyclin D1 activating transcription factor-2 in pp60(v-src) signaling in transcription through a nuclear factor-kappaB-dependent breast cancer cells. Journal of Biological Chemistry 274 pathway. Journal of Biological Chemistry 274 73417350. 2524525249. Lee RJ, Albanese C, Fu M, D’Amico M, Lin B, Watanabe G, Kato M, Kusumi T, Tsuchida S, Tanaka M, Sasaki M & Haines GK 3rd, Siegel PM, Hung MC, Yarden Y et al. Kudo H 2004 Induction of differentiation and peroxisome 2000 Cyclin D1 is required for transformation by proliferator-activated receptor gamma expression in colon activated Neu and is induced through an E2F-dependent cancer cell lines by troglitazone. Journal of Cancer signaling pathway. Molecular and Cellular Biology 20 Research and Clinical Oncology 130 7379. 672683. Kenny FS, Hui R, Musgrove EA, Gee JM, Blamey RW, Leibowitz SB & Kantoff PW 2003 Differentiating agents and Nicholson RI, Sutherland RL & Robertson JF 1999 the treatment of prostate cancer: Vitamin D3 and Overexpression of cyclin D1 messenger RNA predicts for peroxisome proliferator-activated receptor gamma poor prognosis in estrogen receptor-positive breast cancer. ligands. Seminars in Oncology 30 698708. Clinical Cancer Research 5 20692076. Lukas J, Bartkova J & Bartek J 1996 Convergence of Kim Y, Suh N, Sporn M & Reed JC 2002 An inducible mitogenic signalling cascades from diverse classes of pathway for degradation of FLIP protein sensitizes tumor receptors at the cyclin D-cyclin-dependent cells to TRAIL-induced apoptosis. Journal of Biological kinase-pRb-controlled G1 checkpoint. Molecular and Chemistry 277 2232022329. Cellular Biology 16 69176925. Koeffler HP 2003 Peroxisome proliferator-activated receptor McIntosh GG, Anderson JJ, Milton I, Steward M, Parr AH, gamma and cancers. Clinical Cancer Research 9 19. Thomas MD, Henry JA, Angus B, Lennard TW & Koga H, Sakisaka S, Harada M, Takagi T, Hanada S, Horne CH 1995 Determination of the prognostic value of Taniguchi E, Kawaguchi T, Sasatomi K, Kimura R, cyclin D1 overexpression in breast cancer. Oncogene 11 Hashimoto O et al. 2001 Involvement of 885891. p21(WAF1=Cip1), p27(Kip1), and p18(INK4c) in McMahon C, Suthiphongchai T, DiRenzo J & Ewen ME 1999 troglitazone-induced cell-cycle arrest in human hepatoma P/CAF associates with cyclin D1 and potentiates its cell lines. Hepatology 33 10871097. activation of the estrogen receptor. PNAS 96 53825387.

www.endocrinology-journals.org 411 Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access J-R Weng et al.: PPARg-independent antitumor effects of thiazolidinediones

Matsumura I, Kitamura T, Wakao H, Tanaka H, Hashimoto gamma (PPARgamma) agonists that inhibit growth of K, Albanese C, Downward J, Pestell RG & Kanakura Y breast cancer cells: 1,1-Bis(30-indolyl)-1-(p-substituted 1999 Transcriptional regulation of the cyclin D1 promoter phenyl)methanes. Molecular Cancer Therapeutics 3 by STAT5: its involvement in cytokine-dependent growth 247260. of hematopoietic cells. EMBO Journal 18 13671377. Saltiel AR & Olefsky JM 1996 Thiazolidinediones in the Motomura W, Okumura T, Takahashi N, Obara T & Kohgo treatment of insulin resistance and type II diabetes. Y 2000 Activation of peroxisome proliferator-activated Diabetes 45 16611669. receptor gamma by troglitazone inhibits cell growth Sarraf P, Mueller E, Jones D, King FJ, DeAngelo DJ, through the increase of p27KiP1 in human. Pancreatic Partridge JB, Holden SA, Chen LB, Singer S, Fletcher C carcinoma cells. Cancer Research 60 55585564. et al. 1998 Differentiation and reversal of malignant Musgrove EA, Hunter LJ, Lee CS, Swarbrick A, Hui R & changes in colon cancer through PPARgamma. Nature Sutherland RL 2001 Cyclin D1 overexpression induces Medicine 4 10461052. progestin resistance in T-47D breast cancer cells despite Sattler M, Liang H, Nettesheim D, Meadows RP, Harlan JE, p27(Kip1) association with cyclin E-Cdk2. Journal of Eberstadt M, Yoon HS, Shuker SB, Chang BS, Minn AJ Biological Chemistry 276 4767547683. et al. 1997 Structure of Bcl-xLBak peptide complex: Neuman E, Ladha MH, Lin N, Upton TM, Miller SJ, recognition between regulators of apoptosis. Science 275 DiRenzo J, Pestell RG, Hinds PW, Dowdy SF, Brown M 983986. et al. 1997 Cyclin D1 stimulation of estrogen receptor Shiau CW, Yang CC, Kulp SK, Chen KF, Chen CS & Huang transcriptional activity independent of cdk4. Molecular JW 2005 Thiazolidenediones mediate apoptosis in prostate and Cellular Biology 17 53385347. cancer cells in part through inhibition of Bcl-xL/Bcl-2 Nouraini S, Six E, Matsuyama S, Krajewski S & Reed JC functions independently of PPARgamma. Cancer 2000 The putative pore-forming domain of Bax regulates Research 65 15611569. mitochondrial localization and interaction with Bcl-X(L). Shtutman M, Zhurinsky J, Simcha I, Albanese C, D’Amico M, Molecular and Cellular Biology 20 16041615. Pestell R & Ben-Ze’ev A 1999 The cyclin D1 gene is a target Ohta K, Endo T, Haraguchi K, Hershman JM & Onaya T of the beta-catenin/LEF-1 pathway. PNAS 96 55225527. 2001 Ligands for peroxisome proliferator-activated Smith MR & Kantoff PW 2002 Peroxisome proliferator- receptor gamma inhibit growth and induce apoptosis of activated receptor gamma (PPargamma) as a novel target human papillary thyroid carcinoma cells. Journal of for prostate cancer. Investigative New Drugs 20 195200. Clinical Endocrinology and Metabolism 86 21702177. Stendahl M, Kronblad A, Ryden L, Emdin S, Bengtsson NO Okura T, Nakamura M, Takata Y, Watanabe S, Kitami Y & & Landberg G 2004 Cyclin D1 overexpression is a negative Hiwada K 2000 Troglitazone induces apoptosis via the p53 predictive factor for tamoxifen response in and Gadd45 pathway in vascular smooth muscle cells. postmenopausal breast cancer patients. British Journal of European Journal of Pharmacology 407 227235. Cancer 90 19421948. Otter I, Conus S, Ravn U, Rager M, Olivier R, Monney L, Sugimura A, Kiriyama Y, Nochi H, Tsuchiya H, Tamoto K, Fabbro D & Borner C 1998 The binding properties and Sakurada Y, Ui M & Tokumitsu Y 1999 Troglitazone biological activities of Bcl-2 and Bax in cells exposed to suppresses cell growth of myeloid leukemia cell lines by apoptotic stimuli. Journal of Biological Chemistry 273 induction of p21WAF1/CIP1 cyclin-dependent kinase 61106120. inhibitor. Biochemical and Biophysical Research Palakurthi SS, Aktas H, Grubissich LM, Mortensen RM & Communications 261 833837. Halperin JA 2001 Anticancer effects of thiazolidinediones Takeda K, Ichiki T, Tokunou T, Iino N & Takeshita A 2001 15- are independent of peroxisome proliferator-activated Deoxy-delta 12,14-prostaglandin J2 and thiazolidinediones receptor gamma and mediated by inhibition of translation activate the MEK/ERK pathway through initiation. Cancer Research 61 62136218. phosphatidylinositol 3-kinase in vascular smooth muscle Polascik TJ, Oesterling JE & Partin AW 1999 Prostate specific cells. Journal of Biological Chemistry 276 4895048955. antigen: a decade of discovery what we have learned and Takeuchi S, Okumura T, Motomura W, Nagamine M, where we are going. Journal of Urology 162 293306. Takahashi N & Kohgo Y 2002 Troglitazone induces G1 Prall OW, Rogan EM, Musgrove EA, Watts CK & arrest by p27(Kip1) induction that is mediated by Sutherland RL 1998 c-Myc or cyclin D1 mimics estrogen inhibition of proteasome in human gastric cancer cells. effects on cyclin E-Cdk2 activation and cell cycle reentry. Japan Journal of Cancer Research 93 774782. Molecular and Cellular Biology 18 44994508. Tontonoz P, Hu E & Spiegelman BM 1994 Stimulation of Qin C, Burghardt R, Smith R, Wormke M, Stewart J & Safe S adipogenesis in fibroblasts by PPAR gamma 2, a 2003 Peroxisome proliferator-activated receptor gamma lipid-activated transcription factor. Cell 79 11471156. agonists induce proteasome-dependent degradation of Tontonoz P, Singer S, Forman BM, Sarraf P, Fletcher JA, cyclin D1 and estrogen receptor alpha in MCF-7 breast Fletcher CD, Brun RP, Mueller E, Altiok S, Oppenheim H cancer cells. Cancer Research 63 958964. et al. 1997 Terminal differentiation of human liposarcoma Qin C, Morrow D, Stewart J, Spencer K, Porter W, Smith R cells induced by ligands for peroxisome proliferator- 3rd, Phillips T, Abdelrahim M, Samudio I & Safe S 2004 activated receptor gamma and the retinoid X receptor. A new class of peroxisome proliferator-activated receptor PNAS 94 237241.

412 www.endocrinology-journals.org Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access Endocrine-Related Cancer (2006) 13 401–413

Tsubouchi Y, Sano H, Kawahito Y, Mukai S, Yamada R, synergistically inhibit proliferation of breast cancer cells: Kohno M, Inoue K, Hla T & Kondo M 2000 Inhibition of association with selective cooperative inhibition of cyclin human lung cancer cell growth by the peroxisome D1-dependent kinase and Akt signaling pathways. proliferator-activated receptor-gamma agonists through Molecular Cancer Therapeutics 1 695706. induction of apoptosis. Biochemical and Biophysical Yin F, Wakino S, Liu Z, Kim S, Hsueh WA, Collins AR, Van Research Communications 270 400405. Herle AJ & Law RE 2001 Troglitazone inhibits growth of Wajant H 2003 Targeting the FLICE inhibitory protein (FLIP) MCF-7 breast carcinoma cells by targeting G1 cell cycle in cancer therapy. Molecular Intervention 3 124127. regulators. Biochemical and Biophysical Research Wang C, Fu M, D’Amico M, Albanese C, Zhou JN, Brownlee Communications 286 916922. M, Lisanti MP, Chatterjee VK, Lazar MA & Pestell RG Yoshizawa K, Cioca DP, Kawa S, Tanaka E & Kiyosawa K 2001 Inhibition of cellular proliferation through IkappaB 2002 Peroxisome proliferator-activated receptor gamma kinase-independent and peroxisome proliferator-activated ligand troglitazone induces cell cycle arrest and apoptosis receptor gamma-dependent repression of cyclin D1. of hepatocellular carcinoma cell lines. Cancer 95 Molecular and Cellular Biology 21 30573070. 22432251. Westwick JK, Lambert QT, Clark GJ, Symons M, Van Aelst Yu J, Qiao L, Zimmermann L, Ebert MP, Zhang H, Lin W, L, Pestell RG & Der CJ 1997 Rac regulation of Rocken C, Malfertheiner P & Farrell GC 2006 transformation, gene expression, and actin organization Troglitazone inhibits tumor growth in hepatocellular by multiple, PAK-independent pathways. Molecular and carcinoma in vitro and in vivo. Hepatology 43 134143. Cellular Biology 17 13241335. Yu Q, Geng Y & Sicinski P 2001 Specific protection against Wilcken NR, Prall OW, Musgrove EA & Sutherland RL 1997 breast cancers by cyclin D1 ablation. Nature 411 Inducible overexpression of cyclin D1 in breast cancer cells 10171021. reverses the growth-inhibitory effects of antiestrogens. Zwijsen RM, Wientjens E, Klompmaker R, van der Sman J, Clinical Cancer Research 3 849854. Bernards R & Michalides RJ 1997 CDK-independent Wu K, Wang C, D’Amico M, Lee RJ, Albanese C, Pestell RG activation of estrogen receptor by cyclin D1. Cell 88 & Mani S 2002 Flavopiridol and trastuzumab 405415.

www.endocrinology-journals.org 413 Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access Downloaded from Bioscientifica.com at 09/23/2021 01:17:14PM via free access