Cancer Letters

Cancer Letters

Volume 363, Issue 2, 28 July 2015, Pages 166-175
Cancer Letters

Original Articles
FLLL12 induces apoptosis in lung cancer cells through a p53/p73-independent but death receptor 5-dependent pathway

https://doi.org/10.1016/j.canlet.2015.04.017Get rights and content

Highlights

In this manuscript, we have shown for the first time that the synthetic curcumin analog FLLL12 is 5–10-fold more potent than natural curcumin against a panel of eight lung cancer cell lines, and induces apoptosis. Moreover, using siRNA-mediated gene silencing, for the first time, we reveal that ablation of DR5, caspase 8 and Bid significantly protected lung cancer cell lines from FLLL12-induced apoptosis. Interestingly, the expression of DR5 is specific for FLLL12, as curcumin does not induce the expression of DR5. We further demonstrate that FLLL12 regulates DR5 at a posttranscriptional level through activation of protein tyrosine phosphatases, as blocking of protein tyrosine phosphatases with chemical inhibitors blocked the expression of DR5 and significantly protected cells from FLLL12-induced apoptosis. We believe that we demonstrate a set of novel observations in this manuscript as mentioned above.

Abstract

Unlike chemotherapy drugs, the safety of natural compounds such as curcumin has been well established. However, the potential use of curcumin in cancer has been compromised by its low bioavailability, limited tissue distribution and rapid biotransformation leading to low in vivo efficacy. To circumvent these problems, more potent and bioavailable analogs have been synthesized. In the current study, we investigated the mechanism of anti-tumor effect of one such analog, FLLL12, in lung cancers. IC50 values measured by sulforhodamine B (SRB) assay at 72 h and apoptosis assays (annexin V staining, cleavage of PARP and caspase-3) suggest that FLLL12 is 5–10-fold more potent than curcumin against a panel of premalignant and malignant lung cancer cell lines, depending on the cell line. Moreover, FLLL12 induced the expression of death receptor-5 (DR5). Ablation of the expression of the components of the extrinsic apoptotic pathway (DR5, caspase-8 and Bid) by siRNA significantly protected cells from FLLL12-induced apoptosis (p < 0.05). Analysis of mRNA expression revealed that FLLL-12 had no significant effect on the expression of DR5 mRNA expression. Interestingly, inhibition of global phosphatase activity as well as protein tyrosine phosphatases (PTPs), but not of alkaline phosphatases, strongly inhibited DR5 expression and significantly inhibited apoptosis (p < 0.05), suggesting the involvement of PTPs in the regulation of DR5 expression and apoptosis. We further showed that the apoptosis is independent of p53 and p73. Taken together, our results strongly suggest that FLLL12 induces apoptosis of lung cancer cell lines by posttranscriptional regulation of DR5 through activation of protein tyrosine phosphatase(s).

Introduction

Lung cancer represents ~13% of all cancers with approximately 224,000 new cases and 159,000 deaths in the United States in 2014 [1]. Lung cancer remains the top leading cause of cancer related mortality because of low survival rate and high prevalence of advanced stage disease at diagnosis [2]. Extensive worldwide epidemiological data have established cigarette smoking (direct as well as second hand) as the key cause of lung cancers [3], [4]. Occupational exposures to environmental pollutants are other causes of lung cancers. Non-small cell lung cancer (NSCLC) is the most prevalent subtype and accounts for 85% of all lung cancer cases, and is relatively resistant to chemotherapy and radiation. The treatment of lung cancer still remains a challenge due to lack of effective screening tools for early detection, the presence of smoking-related comorbid illnesses, and the inherent molecular heterogeneity [5]. Despite advances in conventional surgical procedures, radiotherapy, and chemotherapy, the 5 year survival rate for lung cancer remains almost unchanged at ~15% [6]. Moreover, radiation and chemotherapy are frequently associated with serious side effects including pneumonitis and/or long-term fibrosis, renal, otologic, and bone marrow-suppressive sequelae [7]. Thus, there is an increasing emphasis on strategies to maximize tumor control, prolong overall survival, minimize chemotherapy side effects, and improve quality of life. As a result, there has been continued investigation into potential alternative and less toxic therapies for lung cancer, with the aim of achieving a more favorable clinical outcome while reducing treatment morbidity.

The safety of natural dietary agents present in fruits, vegetables and spices has been established through years of human consumption and these agents have drawn increasing attention when designing anti-cancer drugs [8]. Curcumin, the most abundant component of the spice turmeric and isolated from the rhizome of the Indian medicinal plant Curcuma longa, is one such compound extensively investigated for its potential anti-cancer effects [7], [9], [10]. Accumulated evidence suggests that curcumin affects a variety of biological pathways involved in apoptosis, tumor proliferation, chemo- and radiotherapy sensitization, tumor invasion, and metastases [7], [9], [11], [12]. However, low bioavailability, limited tissue distribution, rapid metabolism and subsequent excretion from the body limit the potential use of curcumin in cancers [13]. To circumvent these problems, numerous analogs of curcumin have been synthesized. For example, the potential activity of recently synthesized curcumin analogs (2E,6E)-2,6-bis(thiophen-3-methylene) cyclohexanone (AS), (3E,5E)-3,5-bis(thiophen-3-methylene)-tetrahydrothiopyran-4-one (FS), A501, and GO-Y031 has been studied in prostate cancers, NSCLC cells and gastric cancers [14], [15], [16].

Successful elimination of cancer cells from the body depends on the induction of cell death. Apoptosis is the major form of cell death and serves as one of the most powerful tools in the development of cancer chemotherapeutics. Defects in apoptosis are implicated in both tumorigenesis and drug resistance, and are the major causes of treatment failures [17]. Apoptosis can be initiated by two major pathways: either through mitochondria (intrinsic pathway) or through death receptors located in the cell surface (extrinsic pathway). In the case of extrinsic apoptosis, activation of death receptors via ligand binding or drugs initiates the formation of the death inducing signaling complex (DISC). DISC formation involves recruitment of the adaptor protein FADD to the receptor via the death domain (DD) and the inactive pro-caspases 8 and 10. This facilitates the activation and self-processing of caspases 8 and 10, leading to their release into the cytoplasm, where they activate effector caspases 3 and 7, leading to the induction of apoptosis [18]. Activated caspase-8 can also cleave the Bcl-2-family member Bid, which links the extrinsic pathway to the intrinsic pathway. Death receptor 5 (DR5) is a tumor necrosis factor related apoptosis inducing ligand (TRAIL) receptor that activates apoptosis via the extrinsic apoptosis pathway [19]. Recent studies have shown that cancer cells resistant to TRAIL can be sensitized by co-treatment with classical chemotherapeutic drugs. This synergism is partially due to the ability of chemotherapeutic drugs to induce DR5 expression [20], [21], [22]. Hence, targeting the expression of DR5 and induction of apoptosis by anti-cancer drugs has drawn special attention for cancer therapy.

In the current study, we investigated the mechanism of FLLL12-induced apoptosis in lung cancer cell lines. FLLL12 is a potent synthetic curcumin analog [23]. The anti-cancer efficacy of FLLL12 has been tested against breast, prostate, colorectal and pancreatic cancers [23], [24], [25], and shown to be ~10-fold more potent than natural curcumin and to possess selective activity against cancer cells. FLLL12 induces apoptosis of these cancer cells by inhibition of two major survival pathways, AKT and STAT3. However, the detailed mechanism underlying FLLL12-induced apoptosis is not fully understood. Moreover, FLLL12 has never been tested against lung cancer cells. In the present study, for the first time, we investigated the anti-tumor effects of FLLL12 in lung cancers and showed that depending on the cell line, FLLL12 is 5–10-fold more potent than curcumin and induces apoptosis. Moreover, we further explored the fact that FLLL12, but not curcumin, induces DR5-mediated apoptosis in lung cancer cell lines.

Section snippets

Cell lines

Human lung cancer cell lines used in this study were obtained from Dr. Sun's laboratory (Emory University). All cell lines were authenticated through genotyping and maintained in RPMI medium supplemented with 5% heat-inactivated fetal bovine serum in a 37 °C and 5% CO2 humidified incubator. Generation and maintenance of the premalignant cell lines 1799 and 1198 were described elsewhere [26].

Reagents

Curcumin was purchased from Sigma Chemical (St. Louis, MO, USA) and FLLL12 was obtained from Dr. James R.

FLLL12 is 5–10-fold more potent than curcumin and induces apoptosis

Although the antitumor effects of FLLL12 have been investigated in prostate, breast, pancreatic and colon cancer cell lines and compared with those of curcumin [23], [24], [25], the agent has never been evaluated in lung cancer cell lines. In order to explore the mechanism of anti-tumor effect of FLLL12 in lung cancers, we first assessed the sensitivity of different premalignant and malignant lung cancer cell lines to FLLL12 versus curcumin by comparing IC50 values measured using SRB assays at

Discussion

Treatment outcomes in advanced or metastatic NSCLC remain frustrating, with low long-term survival rates. Overall, five-year survival for lung cancer patients is ~15%, and patients with advanced stage disease often die within 12 months [6]. Although recent advances in molecularly targeted treatments have made lung cancer therapy more personalized, the benefits are typically lost within months to a few years [42]. Long-term disease control requires activation of the apoptotic machinery that

Authors' contributions

Conception and design: Haque and Amin.

Development of methodology: Haque, Amin, and Rahman.

Acquisition of data: Haque and Amin.

Analysis and interpretation of data: Haque, Amin, Chen, and Shin.

Writing, review and/or revision of the manuscript: Haque, Amin, Fuchs, Saba, Khuri and Shin.

Study supervision: Amin.

Funding

This study was supported by R03CA171663, R03CA159369 and Robbins Scholar Award of Winship Cancer Institute of Emory University.

Conflict of interest

The authors declare no potential conflict of Interest.

Acknowledgments

We are thankful to Dr. Anthea Hammond and Shi-Yong Sun for carefully editing the manuscript and providing the luciferase construct, respectively.

References (59)

  • H. Francis et al.

    Regulation of the extrinsic apoptotic pathway by microRNA-21 in alcoholic liver injury

    J. Biol. Chem

    (2014)
  • D. Hanahan et al.

    The hallmarks of cancer

    Cell

    (2000)
  • N.N. Danial et al.

    Cell death: critical control points

    Cell

    (2004)
  • J.E. Vince et al.

    IAP antagonists target cIAP1 to induce TNFalpha-dependent apoptosis

    Cell

    (2007)
  • R. Siegel et al.

    Cancer statistics, 2014

    CA Cancer J. Clin

    (2014)
  • E. Roeland et al.

    In chemotherapy for lung cancer, sometimes less is more

    J. Natl Compr. Canc. Netw

    (2013)
  • S.S. Hecht

    Tobacco smoke carcinogens and lung cancer

    J. Natl Cancer Inst

    (1999)
  • R.V. Ancuceanu et al.

    Pharmacologically active natural compounds for lung cancer

    Altern. Med. Rev

    (2004)
  • S. Ramalingam et al.

    Systemic chemotherapy for advanced non-small cell lung cancer: recent advances and future directions

    Oncologist

    (2008)
  • F.R. Khuri et al.

    Molecularly targeted approaches to the chemoprevention of lung cancer

    Clin. Cancer Res

    (2004)
  • H.J. Mehta et al.

    Curcumin and lung cancer-a review

    Target. Oncol

    (2014)
  • A.R. Amin et al.

    Perspectives for cancer prevention with natural compounds

    J. Clin. Oncol

    (2009)
  • A. Goel et al.

    Curcumin, the golden spice from Indian saffron, is a chemosensitizer and radiosensitizer for tumors and chemoprotector and radioprotector for normal organs

    Nutr. Cancer

    (2010)
  • W. Park et al.

    New perspectives of curcumin in cancer prevention

    Cancer Prev. Res. (Phila)

    (2013)
  • M.A. Rahman et al.

    Chemopreventive potential of natural compounds in head and neck cancer

    Nutr. Cancer

    (2010)
  • J.S. Jurenka

    Anti-inflammatory properties of curcumin, a major constituent of Curcuma longa: a review of preclinical and clinical research

    Altern. Med. Rev

    (2009)
  • P. Anand et al.

    Bioavailability of curcumin: problems and promises

    Mol. Pharm

    (2007)
  • D.Y. Zhou et al.

    Effects of curcumin analogues for inhibiting human prostate cancer cells and the growth of human PC-3 prostate xenografts in immunodeficient mice

    Biol. Pharm. Bull

    (2014)
  • Y.Q. Xia et al.

    Curcumin analogue A501 induces G2/M arrest and apoptosis in non-small cell lung cancer cells

    Asian Pac. J. Cancer Prev

    (2014)
  • Cited by (13)

    • Perspectives for synthetic curcumins in chemoprevention and treatment of cancer: An update with promising analogues

      2021, European Journal of Pharmacology
      Citation Excerpt :

      Like curcumin, many curcumin analogues also induce apoptosis through both intrinsic and extrinsic pathways (Table 2). For example, FLLL12 induced apoptosis of lung cancer cells and SCCHN by activating the DR5 pathway and modulating multiple Bcl-2 proteins, respectively (Haque et al., 2015; Anisuzzaman et al., 2016). CDF induced apoptosis of colon cancer stem-like cells by inhibiting NF-κB and Bcl-xL and activating proapoptotic Bax (Kanwar et al., 2011), CLEFMA induces apoptosis of osteosarcoma cells by activating the extrinsic and intrinsic apoptotic processes through JNK1/2 and p38 pathways (Yang et al., 2019a).

    • 1,25(OH)<inf>2</inf>D<inf>3</inf> induced apoptosis of human hepatocellular carcinoma cells in vitro and inhibited their growth in a nude mouse xenograft model by regulating histone deacetylase 2

      2018, Biochimie
      Citation Excerpt :

      The complex activates and cleaves pre-caspase 8 and then caspase 3 at the downstream, finally inducing exogenous apoptosis. On the other hand, Mitochondria swell due to Bax which can penetrate the mitochondrial outer membrane, so the membrane ruptures to release cytochrome C and to further induce endogenous apoptosis by activating caspase 9 [24]. To explore whether HDAC2 induced the apoptosis of HepG2 cells by regulating p53, we further analyzed the changes of Bax at the downstream of the mitochondria-mediated pathway and signaling molecules related to the DR5-mediated pathway.

    • Mono-Carbonyl Curcumin Analogs for Cancer Therapy

      2023, Biological and Pharmaceutical Bulletin
    View all citing articles on Scopus
    View full text