Apoptosis inhibitor TRIAP1 is a novel effector of drug resistance

  • Authors:
    • Caroline Adams
    • Giulia Cazzanelli
    • Sabeena Rasul
    • Ben Hitchinson
    • Yunhui Hu
    • R. Charles Coombes
    • Selina Raguz
    • Ernesto Yagüe
  • View Affiliations

  • Published online on: May 19, 2015     https://doi.org/10.3892/or.2015.3988
  • Pages: 415-422
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

TP53-regulated inhibitor of apoptosis 1 (TRIAP1) is a novel apoptosis inhibitor that binds HSP70 in the cytoplasm and blocks the formation of the apoptosome and caspase-9 activation. TRIAP1 has been shown to be upregulated in many types of cancers; however, its role remains elusive. We determined the TRIAP1 mRNA levels in a panel of human tissues and found its expression to be ubiquitous. Normal breast, as well as non-tumorigenic breast cells, exhibited lower TRIAP1 mRNA levels than breast cancer cells or their drug-resistant derivatives. TRIAP1 is a small, evolutionarily conserved protein that is 76 amino acids long. We found that yeast cells, in which the TRIAP1 homologue was knocked out, had increased sensitivity to doxorubicin. Equally, RNA interference in breast cancer drug-resistant cells demonstrated that downregulation of TRIAP1 impaired cell growth in the presence of doxorubicin. As expected, caspase-9 activation was diminished after overexpression of TRIAP1 in drug-resistant cells. Importantly, stable transfections of a TRIAP1 expression plasmid in CAL51 cells led to a marked increase in the number of doxorubicin‑resistant clones, that was abolished when cells expressed hairpins targeting TRIAP1. In addition, we showed that TRIAP1 expression was also triggered by estrogen deprivation in MCF-7 cells. Although both polyclonal and monoclonal antibodies generated for the present study failed to robustly detect TRIAP1, we demonstrated that TRIAP1 represents a novel marker for drug resistance in breast cancer cells and it may be used in the stratification of breast cancer patients once a suitable antibody has been developed. Equally, these studies open potential drug development strategies for blocking TRIAP1 activity and avoiding drug resistance.

Introduction

One of the many mechanisms by which cancer cells acquire drug resistance is by apoptosis evasion (1,2). Antineoplastic agents activate the apoptotic intrinsic pathway which results in the permeabilization of the outer mitochondrial membrane. The subsequent release of cytochrome c and other pro-apoptotic molecules leads to the formation of a large protein complex, the apoptosome, containing cytochrome c, apoptotic protease activating factor 1 (Apaf-1) and caspase-9. Regulation of these processes occurs through the activity of members of the BCL-2 family of proteins (3). Upregulation of anti-apoptotic proteins, such as BCL-2, BCL-XL, and members of the inhibitor of apoptosis (IAP) family, and inactivating mutations in pro-apoptotic proteins Bax and Bak, or p53 have been described in both the development of cancer and drug resistance (2,4). Among several therapeutic strategies, the restoration of p53-mediated apoptosis in cancer cells has been sought for some time, so far without a clinical application (5,6). Therefore, targeting inhibitors of apoptosis offer another opportunity for drug development (7).

TP53-regulated inhibitor of apoptosis 1 (TRIAP1; alias p53CSV, p53-inducible cell-survival factor) is a small, evolutionarily conserved protein that is 76 amino acids long and contains a twin Cx9C motif. Its expression is induced by p53 following low levels of genotoxic stress (8). In yeast it is located in the mitochondrial intermembrane space where it can interact with both Ups1 and Ups2, protecting them against proteolysis and regulating cardiolipin and phosphatidylethanolamine levels within mitochondria (9). In its extramitochondrial role, TRIAP1 modulates apoptotic pathways through interaction with HSP70, inhibition of the interaction of cytochrome c with Apaf-1 and activation of caspase-9, to inhibit apoptosis and permit DNA damage repair (8,10). Despite its name, TRIAP1 does not share any similarity with XIAP, c-IAP, survivin or other members of the IAP family (11). TRIAP1 is upregulated in myelomas with unfavorable prognosis (12) and TRIAP1 mRNA has been found in all breast cancer cell lines tested (13). However, the role of TRIAP1 in breast or other solid tumors has been poorly addressed.

Breast cancer is one of the most common tumors affecting women, and chemotherapy, endocrine therapy and radiotherapy regimens are part of the present mainstream treatment. Even those cancers that initially respond well to treatment may become resistant and give rise to secondary tumors, normally with fatal consequences (14). Thus, it is crucial to be able to predict which patients will respond to therapy in order to design the best clinical strategy available and to develop drugs for the treatment of resistance (15).

Here we describe the upregulation of TRIAP1 in drug-resistant breast cancer cells. Experimental modulation of TRIAP1 in breast cancer cells, either by overexpression or downregulation by RNA interference, changed cellular sensitivity to doxorubicin, thus confirming TRIAP1 as a novel effector of drug resistance.

Materials and methods

Cell culture, transduction and transfection

The breast cancer cell lines CAL51 (German Resource Centre for Biological Material, DSMz, Braunschweig, Germany) and MCF7 (European Collection of Cell Cultures, Health Protection Agency, UK) were grown in low glucose (1 g/l) Dulbecco’s modified Eagle’s medium (DMEM)-GlutaMax (Invitrogen, Carlsbad, CA, USA) supplemented with 10% foetal calf serum (FCS). Drug-resistant derivatives CALDOX, MCFDOX and MLET5 have been previously described (1618). RNA from MDA-MB-231, T47D, ZR75 and 226-L-U19 cells (19) was a generous gift from Y. Zhou. Human primary mammary epithelial cells (HMECs) were purchased from Invitrogen and immortalized by expression of the catalytic subunit of human telomerase as previously described (20) to generate HMEC-TERT cells. HMECs were grown on Mammary Epithelial Cell Growth Medium (Invitrogen). GP2-293 pantropic retroviral packaging (Clontech Laboratories, Mountain View, CA, USA) and HEK293T cells (American Type Culture Collection, LGC Standards, Teddington, UK) were maintained in DMEM supplemented with 4.5 g/l glucose, 10% FCS and 4 mM L-glutamine (Invitrogen). For culturing in estrogen-free conditions, MCF-7 cells were cultured in DMEM lacking phenol red (DMEM-PR; Gibco-BRL, UK), supplemented with 10% dextran-coated charcoal-stripped FCS (DSS; First Link Ltd., UK). MLET5 cells were routinely cultured in DMEM-PR, containing 10% DSS. All cells were routinely tested for the presence of mycoplasma using the Lonza MycoAlert kit (Basel, Switzerland).

DNA procedures

A human TRIAP1 cDNA clone with a C-terminal GFP tag cloned into pCMV6-AC-GFP was purchased from Origene and used for transient transfections. For stable transfections, the genomic region encompassing TRIAP1 (first and second exons including both5′- and 3′-UTRs as well as the single intervening sequence) was amplified from genomic DNA isolated from IMR-90 fibroblasts by PCR using Platinum® Taq DNA Polymerase High Fidelity (Invitrogen) with primers OLEY335 and OLEY336 (Table I). The amplified 2.6-kbp fragment was initially cloned into pCRII-TOPO (Invitrogen) and the TRIAP1 open reading frame verified by sequencing. Then, the genomic TRIAP1 fragment was liberated by digestion with BamHI and EcoRI and cloned into the BamHI- and EcoRI-digested lentiviral vector FUW. Retroviral plasmids pGFP-V-RS expressing short hairpins targeting TRIAP1 (5′-AGAGATTCCTATTGAAGGACTGG AGTTCA-3′) and non-targeting scrambled (TR30013) were from Origene.

Table I

Oligonucleotides used in this study,

Table I

Oligonucleotides used in this study,

PrimerSequence (5′-3′)Use
OLEY335 GGGGGATCCCGACGCGCCTGAGAGTGATGACATCATRIAP1 genomic amplification
OLEY336 GGGGTCGACTGTGAGGTTTCTGATTGCCATACTGTRIAP1 genomic amplification
OLEY287 AGGATTTCGCAAGTCCAGAATRIAP1 QPCR
OLEY288 GCTGATTCCACCCAAGTATTRIAP1 QPCR
OLEY373 TCACCGCCCTACACATCAAACTRPS14 QPCR
OLEY374 CTGCGAGTGCTGTCAGAGGRPS14 QPCR
OLEY375 AGGGTTATGTGGTCCGAATCARPS6 QPCR
OLEY376 TGCCCCTTACTCAGTAGCAGGRPS6 QPCR
Viral transduction

Viral transductions were essentially as previously described (16). Briefly, retroviral transfections were performed using 20 µg retroviral plasmid, 2 µg pVSV-G (Clontech Laboratories) and 55 µg Polyethylenimine (MW 25,000) (Polysciences, Warrington, PA, USA) prior to co-transfection into the GP2-293 pantropic retroviral packaging cells. Lentiviral transfections were performed using 20 µg lentiviral plasmid, 2 µg pVSV-G, 8 µg pPAX2 (Addgene, Cambridge, MA, USA) and 75 µg Polyethylenimine (MW 25,000) (Sigma-Aldrich, St. Louis, MO, USA) prior to co-transfection into the HEK293T cells. Viral supernatants were collected 24–48 h after transfections, filtered through 0.45-µm cellulose acetate filters and supplemented with 8 µg/ml Polybrene (Sigma-Aldrich) prior to adding to recipient cells. Puromycin (1 µg/ml; Sigma-Aldrich) was added 96 h after the first infection to recipient cells for selection of stable transgene expression.

Transient transfections

Cells (1×105 in 6-well plates) were transfected with a final concentration of 40 nM ON-TARGETplus SMARTpool TRIAP1 siRNA oligonucleotides (Thermo Scientific) using HiPerFect (Qiagen, UK) following the manufacturer’s instructions. EGFP siRNA (Ambion, UK) was used as a negative control (21). A sulphorhodamine B (Sigma-Aldrich) assay (22) was used to screen for drug cytotoxicity as previously described (19). For transient overexpression, cells (1×104) were seeded in 96-well plates and transfected with FuGENE HD (Promega, Madison, WI, USA) using 2 µg DNA as recommended by the manufacturer. Then cells were treated for 24 h with 0.4 µM doxorubicin, and caspase-9 activity was determined using a Caspase-Glo 9 assay (Promega) following the manufacturer’s instructions and normalized to cell density obtained after sulphorhodamine B staining.

Gene expression analysis

Total RNA (isolated using RNAzol B; Biogenesis, Poole, UK) was reverse transcribed with RNase H+ MMLV Reverse Transcriptase (iScript cDNA Synthesis kit; Bio-Rad) and real-time quantitative PCR was performed using SYBR-Green (Bioline Reagents, London, UK) and gene specific primers (Table I) on an ABI Prism 7700 Detection system (PerkinElmer Life Sciences, Waltham, MA, USA). PCR conditions included an initial step at 95°C for 10 min followed by 40 cycles of 95°C for 30 sec, 60°C for 30 sec and 72°C for 30 sec. A comparative threshold cycle was used to determine the relative gene expression as previously described (23). TFF1 expression was performed using TaqMan Gene Expression assay Hs00907239_m1 (Invitrogen) following the manufacturer’s instructions.

Yeast procedures

Wild-type Saccharomyces cerevisiae BY4743 (MATa/α his3Δ1/his3Δ1 leu2Δ0/leu2Δ0 met15Δ0/MET15 lys2Δ0/LYS2 ura3Δ0/ura3Δ0) and Mdm35 knockout strain were purchased from Open Biosystems. Screening for drug toxicity was performed in YPD plates with doxorubicin after 3 days of growth.

Drug resistance clonogenic assay

Cells were seeded, at least in duplicate, at a density of 1–5×105 cells in 25-cm2 culture flasks and exposed to a single dose of doxorubicin for 24 h. Cells were kept in culture for 2–4 weeks with drug-free medium changes every week. Drug-resistant clones were fixed with 4% paraformaldehyde and stained with 0.2% crystal violet. Crystal violet retained in the cells was quantified by solubilization with 33% acetic acid and measured at OD592 nm.

Statistical analysis

Statistical evaluations were performed using the Student’s t-test for paired data, and data were considered significant at a P-value <0.05.

Results

TRIAP1 is ubiquitously expressed in normal human tissues and upregulated in breast cancer cells

We initially tested TRIAP1 expression in a panel of RNAs from normal human tissues (24) and found it ubiquitously expressed (Fig. 1A). Although there was a lack of tissue specificity, bone marrow, heart and kidney showed very low expression, whereas brain, placenta and testis showed high expression, with that from skeletal muscle being the highest. Normal breast tissue expressed TRIAP1 mRNA at intermediate levels (Fig. 1A). Next we tested TRIAP1 mRNA expression in four breast cancer cell lines (CAL51, MDA-MB-231, T47D and ZR75) and in two immortal, but non-tumorigenic, breast cell lines (HMEC-TERT and 226-L-U19). An ~2-fold upregulation was found in the cancerous cell lines, indicating that malignant transformation of breast epithelial cells is, at least in vitro, associated with TRIAP1 upregulation (Fig. 1B).

A polyclonal antibody raised to the synthetic peptide CVQKAIKEKEIPIEGLEF (amino acids 47–64) as well as three monoclonal antibodies raised to the epitopes HGKEKPENSS (amino acids 67–76), IKEKEIPIEG (amino acids 52–61) and FAEKFLKGDS (amino acids 23–32), all failed to robustly detect TRIAP1 as a band at ~10 kDa (data not shown). Equally, a few commercial antibodies were tested and failed in the same way (data not shown). Thus, TRIAP1 expression analyses throughout the present study were performed exclusively at the mRNA level.

TRIAP1 is upregulated in drug-resistant breast cancer cell lines

TRIAP1 upregulation following genotoxic stress has been exclusively demonstrated in a colon cancer cell line (8). We confirmed that CAL51 breast cancer cells exhibited upregulation of TRIAP1 mRNA expression after 24 h of doxorubicin treatment at low to moderate doses (up to 0.4 µM). Higher doses (2–5 µM) repressed TRIAP1 expression (Fig. 1C). Then we examined whether TRIAP1 was associated with a drug-resistance phenotype. For this, we tested TRIAP1 mRNA expression in two doxorubicin-resistant breast cancer cell lines derived from CAL51 (16) and MCF7 (17) cells, and in a third cell line derived from MCF7 cells after estrogen deprivation, which are resistant to tamoxifen and etoposide (18). Doxorubicin and etoposide are two topoisomerase II inhibitors producing genotoxic stress, ultimately leading to apoptosis, which form part of many chemotherapeutic cancer regimes. TRIAP1 mRNA was upregulated in these three drug-resistant cell types between 3- and 8-fold (Fig. 1D).

TRIAP1 is an effector of drug resistance

In order to determine whether TRIAP1 is an effector of drug resistance, we tested cell susceptibility to doxorubicin after experimental modulation of TRIAP1 expression. Comparison of TRIAP1 protein sequences revealed a striking conservation across kingdoms (Fig. 2A), albeit not at the C-terminus, indicating that this protein must perform a fundamental role in cellular physiology. The TRIAP1 homologue in S. cerevisiae, Mdm35, shares 33% identity with human TRIAP1. Since yeast cells harbour an ancestral programmed cell death machinery that shares many key molecules that are part of the apoptotic intrinsic pathway in mammalian cells (25), and because genetic manipulation of yeast allows the knockout of genes, we initially tested our hypothesis in Mdm35 knockout yeast cells. Indeed, ΔMdm35 cell growth was greatly inhibited by the presence of doxorubicin in the yeast growth medium compared to wild-type cell growth (Fig. 2B).

Next, we determined whether experimental downregulation of TRIAP1 would have a similar effect in breast cancer cells. For this, we transiently transfected siRNAs targeting TRIAP1 mRNA in doxorubicin-resistant CALDOX cells and obtained ~60% downregulation in TRIAP1 mRNA levels (Fig. 3A). Transfection of siRNAs targeting EGFP mRNA, which is not expressed in these cells, was used as a negative control. This transient decrease in TRIAP1 mRNA levels was sufficient to inhibit the proliferation of cells in the presence of doxorubicin compared to the slight decrease in proliferation obtained with the negative control (Fig. 3B).

As TRIAP1 blocks the formation of the apoptosome and activation of caspase-9 (8), we next ascertained whether caspase-9 activity would be reduced by TRIAP1 in breast cancer cells. To do this, we transiently overexpressed TRIAP1 in CAL51 cells (Fig. 3C) and treated them with 0.4 µM doxorubicin for 24 h. As expected, the control cells transfected with an empty vector exhibited increased caspase-9 activity, and this increase was reduced in the cells overexpressing TRIAP1 (Fig. 3D).

Although experiments performed with transient transfections are informative to assess the short-term susceptibility to cytotoxic drugs, the generation of drug resistance is a slower process, often taking several weeks in vitro, and required a different approach. We generated CAL51 derivative cells that had either permanently downregulated TRIAP1 by stable expression of hairpins targeting TRIAP1 mRNA (shTRIAP) or permanently upregulated TRIAP1. CAL-shTRIAP cells showed decreased levels of TRIAP1 mRNA by ~60% (Fig. 4A). In addition, we managed to reproduce the slight increase in TRIAP1 mRNA expression found in drug-resistant CALDOX cells by using low multiplicity of infection of the lentivirus used for TRIAP1 overexpression (Fig. 4A). Thus, although these experimental changes in TRIAP1 expression after stable gene expression were modest, they reflect very accurately the difference found between naïve and drug-resistant cells (Fig. 1D). Next we performed clonogenic assays after doxorubicin treatment. For this, doxorubicin was added to cell cultures and the emergence of doxorubicin-resistant clones was monitored over a period of 2–4 weeks. Stable experimental overexpression of TRIAP1 led to an increase in doxorubicin-resistant clones, and, conversely, downregulation of TRIAP1 led to fewer drug-resistant clones than the control cells (Fig. 4B and C).

TRIAP1 is upregulated by lack of estrogen

As tamoxifen-resistant cells have elevated levels of TRIAP1 mRNA (Fig. 1D), we investigated whether a link exists between estrogen and TRIAP1 expression. We cultured MCF-7 and MLET5 cells for 3 days in estrogen-deprived medium, re-introduced estrogen for 16 h, and then determined TRIAP1 mRNA levels. There was no upregulation of TRIAP1 in any cell type; in fact, MCF-7 cells treated with 10 nM estrogen showed a slight decrease in TRIAP1 expression (Fig. 5A). As expected, the estrogen-responsive gene TFF1 (26) was upregulated in both cell lines (Fig. 5A). This indicates that TRIAP1 is not an estrogen-responsive gene. Importantly, when MCF-7 cells were cultured in the absence of estrogen, TRIAP1 expression was upregulated (Fig. 5B). Moreover, the addition of tamoxifen, which blocks estrogen receptor activation, led to a small but significant upregulation of TRIAP1 mRNA in the MCF-7 cells, but not, as expected, in the tamoxifen resistant MLET5 cells (Fig. 5C).

Overall, these results establish TRIAP1 as a novel effector of drug resistance in breast cancer cells.

Discussion

One of the many mechanisms by which cancer cells develop resistance to therapeutic intervention is by apoptosis evasion. This is not surprising since the apoptotic machine is complex, with multiple layers of regulation (1,2). The IAP gene family encodes a group of structurally-related proteins sharing a BIR domain, such as XIAP or survivin, which can be upregulated in breast cancer (27,28). However, other inhibitors of apoptosis do not share structural similarities with members of the IAP family. One such protein, TRIAP1, is a small, 76-amino acid protein in humans that binds to HSP70 and inhibits the formation of the apoptosome. Apoptosis evasion is one of the hallmarks of cancer (29), and is thus not surprising to find TRIAP1 mRNA upregulated in a variety of malignancies (3032) and in breast cancer cell lines, but not in TERT-immortalized primary mammary epithelial cells or in cells transformed but not yet fully tumorigenic (Fig. 1B). Although the mechanisms upregulating TRIAP1 during tumorigenesis are not known, a low to moderate amount of several genotoxic agents triggers its expression via p53 activation (8). Importantly we also found that estrogen deprivation upregulated TRIAP1. Estrogen deprivation reduces p53 activity (33,34), and in normal mammary epithelial cells tamoxifen activates p53 (35), although not in MCF-7 cells (36). Thus, the upregulation of TRIAP1 in these cases is very likely to be p53-independent. Our data clearly demonstrated that TRIAP1 is a novel effector of drug resistance. Its experimental overexpression confers cells the capacity to develop doxorubicin-resistant derivatives, whereas its downregulation by RNA interference generates cells more susceptible to the cytotoxic effects of the drug.

Due to its ubiquitous tissue expression in humans and extremely high conservation throughout evolution it is likely that TRIAP1 has an important function in eukaryotic cells. Research on yeast indicates that TRIAP1 (Mdm35) normally resides in the mitochondrial intermembrane space where it interacts with Usp1 and Usp2 (PRELI1 and PRELI2 in humans) to regulate the synthesis of cardiolipin and phosphatidylethanolamine, respectively (9). Although TRIAP1/PRELI complexes (Mdm35/Usp homologues in yeast) have been shown to prevent apoptosis by mediating intramitochondrial transport of phosphatidic acid in HeLa cells (37), it is tempting to speculate that the upregulation of TRIAP1 expression following moderate genotoxic stress (Fig. 1C), or that found in drug-resistant cells (Fig. 1D), helps to maintain the balance towards apoptosis inhibition by blocking apoptosome formation and caspase-9 inactivation (8). However, after stronger genotoxic stress, which probably leads to irreparable DNA damage, there is no activation of TRIAP1 expression. A large escape of cytochrome c from the mitochondria would be enough to change the balance towards apoptosome formation and the background levels of TRIAP1 protein would not be enough to block the interaction cytochrome c-Apaf1-caspase-9 (38). In addition, it is not clear whether, in addition to cytochrome c, TRIAP1 or TRIAP1/PRELID complexes can also escape from the intermembrane space following stress to interact with HSP70. Nonetheless, yeast data as well as those presented here indicate a dual role for TRIAP1 in mitochondrial lipid biosynthesis and drug resistance. This may offer a potential drug development strategy for the synthesis of compounds blocking TRIAP1 activity.

Acknowledgments

We thank Yuan Zhou for a generous gift of MDA-MB-231, T47D, ZR75 and 226-L-U19 RNA, Hetal Dattani for advice on TFF1 expression, Andres Clemente Blanco for advice on yeast work, and Eric Lam and Laki Buluwela for the MCFDOX and MLET5 cells, respectively. G.C. was the recipient of an Erasmus postgraduate scholarship. Y.H. was partially supported by the China Scholarship Council. We thank the Breast Cancer Research Trust (C.A.), Breast Cancer Campaign (2011MaySP01 to E.Y.), Cancer Research UK (R.C.C.) and the Experimental Cancer Medicine Centre Network (R.C.C.) for their financial support.

Abbreviations:

Apaf-1

apoptotic protease activating factor 1

DMEM

Dulbecco’s modified Eagle’s medium

DMEM-PR

DMEM lacking phenol red

DSS

dextran-coated charcoal-stripped FCS

FCS

foetal calf serum

HMECs

human primary mammary epithelial cells

IAP

inhibitor of apoptosis

TRIAP1

TP53-regulated inhibitor of apoptosis 1

References

1 

Mashima T and Tsuruo T: Defects of the apoptotic pathway as therapeutic target against cancer. Drug Resist Updat. 8:339–343. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Indran IR, Tufo G, Pervaiz S and Brenner C: Recent advances in apoptosis, mitochondria and drug resistance in cancer cells. Biochim Biophys Acta. 1807:735–745. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Elmore S: Apoptosis: a review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Cotter TG: Apoptosis and cancer: the genesis of a research field. Nat Rev Cancer. 9:501–507. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Yu Q: Restoring p53-mediated apoptosis in cancer cells: new opportunities for cancer therapy. Drug Resist Updat. 9:19–25. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Kastan MB: Wild-type p53: tumors can’t stand it. Cell. 128:837–840. 2007. View Article : Google Scholar : PubMed/NCBI

7 

de Graaf AO, de Witte T and Jansen JH: Inhibitor of apoptosis proteins: new therapeutic targets in hematological cancer? Leukemia. 18:1751–1759. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Park WR and Nakamura Y: p53CSV, a novel p53-inducible gene involved in the p53-dependent cell-survival pathway. Cancer Res. 65:1197–1206. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Tamura Y, Iijima M and Sesaki H: Mdm35p imports Ups proteins into the mitochondrial intermembrane space by functional complex formation. EMBO J. 29:2875–2887. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Niu P, Liu L, Gong Z, Tan H, Wang F, Yuan J, Feng Y, Wei Q, Tanguay RM and Wu T: Overexpressed heat shock protein 70 protects cells against DNA damage caused by ultraviolet C in a dose-dependent manner. Cell Stress Chaperones. 11:162–169. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Salvesen GS and Duckett CS: IAP proteins: blocking the road to death’s door. Nat Rev Mol Cell Biol. 3:401–410. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Felix RS, Colleoni GW, Caballero OL, Yamamoto M, Almeida MS, Andrade VC, Chauffaille MdeL, Silva WA Jr, Begnami MD, Soares FA, et al: SAGE analysis highlights the importance of p53csv, ddx5, mapkapk2 and ranbp2 to multiple myeloma tumorigenesis. Cancer Lett. 278:41–48. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark L, Bayani N, Coppe JP, Tong F, et al: A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 10:515–527. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Ali S and Coombes RC: Endocrine-responsive breast cancer and strategies for combating resistance. Nat Rev Cancer. 2:101–112. 2002. View Article : Google Scholar

15 

Eccles SA, Aboagye EO, Ali S, Anderson AS, Armes J, Berditchevski F, Blaydes JP, Brennan K, Brown NJ, Bryant HE, et al: Critical research gaps and translational priorities for the successful prevention and treatment of breast cancer. Breast Cancer Res. 15:R922013. View Article : Google Scholar : PubMed/NCBI

16 

Raguz S, Adams C, Masrour N, Rasul S, Papoutsoglou P, Hu Y, Cazzanelli G, zhou Y, Patel N, Coombes C, et al: Loss of O’-methylguanine-DNA methyltransferase confers collateral sensitivity to carmustine in topoisomerase II-mediated doxorubicin resistant triple negative breast cancer cells. Biochem Pharmacol. 85:186–196. 2013. View Article : Google Scholar

17 

Chen J, Gomes AR, Monteiro LJ, Wong SY, Wu LH, Ng TT, Karadedou CT, Millour J, Ip YC, Cheung YN, et al: Constitutively nuclear FOXO3a localization predicts poor survival and promotes Akt phosphorylation in breast cancer. PLoS One. 5:e122932010. View Article : Google Scholar : PubMed/NCBI

18 

Tolhurst RS, Thomas RS, Kyle FJ, Patel H, Periyasamy M, Photiou A, Thiruchelvam PT, Lai CF, Al-Sabbagh M, Fisher RA, et al: Transient over-expression of estrogen receptor-α in breast cancer cells promotes cell survival and estrogen-independent growth. Breast Cancer Res Treat. 128:357–368. 2011. View Article : Google Scholar

19 

Rasul S, Balasubramanian R, Filipović A, Slade MJ, Yagüe E and Coombes RC: Inhibition of gamma-secretase induces G2/M arrest and triggers apoptosis in breast cancer cells. Br J Cancer. 100:1879–1888. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Yagüe E, Arance A, Kubitza L, O’Hare M, Jat P, Ogilvie CM, Hart IR, Higgins CF and Raguz S: Ability to acquire drug resistance arises early during the tumorigenesis process. Cancer Res. 67:1130–1137. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Filipović A, Gronau JH, Green AR, Wang J, Vallath S, Shao D, Rasul S, Ellis IO, Yagüe E, Sturge J, et al: Biological and clinical implications of nicastrin expression in invasive breast cancer. Breast Cancer Res Treat. 125:43–53. 2011. View Article : Google Scholar

22 

Vichai V and Kirtikara K: Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat Protoc. 1:1112–1116. 2006. View Article : Google Scholar

23 

Yague E, Armesilla AL, Harrison G, Elliott J, Sardini A, Higgins CF and Raguz S: P-glycoprotein (MDR1) expression in leukemic cells is regulated at two distinct steps, mRNA stabilization and translational initiation. J Biol Chem. 278:10344–10352. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Raguz S, De Bella MT, Slade MJ, Higgins CF, Coombes RC and Yagüe E: Expression of RPIP9 (Rap2 interacting protein 9) is activated in breast carcinoma and correlates with a poor prognosis. Int J Cancer. 117:934–941. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Almeida B, Silva A, Mesquita A, Sampaio-Marques B, Rodrigues F and Ludovico P: Drug-induced apoptosis in yeast. Biochim Biophys Acta. 1783:1436–1448. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Sun JM, Spencer VA, Li L, Yu Chen H, Yu J and Davie JR: Estrogen regulation of trefoil factor 1 expression by estrogen receptor alpha and Sp proteins. Exp Cell Res. 302:96–107. 2005. View Article : Google Scholar

27 

Nachmias B, Ashhab Y and Ben-Yehuda D: The inhibitor of apoptosis protein family (IAPs): an emerging therapeutic target in cancer. Semin Cancer Biol. 14:231–243. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Wang S, Bai L, Lu J, Liu L, Yang CY and Sun H: Targeting inhibitors of apoptosis proteins (IAPs) for new breast cancer therapeutics. J Mammary Gland Biol Neoplasia. 17:217–228. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Hanahan D and Weinberg RA: Hallmarks of cancer: the next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Garber ME, Troyanskaya OG, Schluens K, Petersen S, Thaesler Z, Pacyna-Gengelbach M, van de Rijn M, Rosen GD, Perou CM, Whyte RI, et al: Diversity of gene expression in adenocarcinoma of the lung. Proc Natl Acad Sci USA. 98:13784–13789. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Lu KH, Patterson AP, Wang L, Marquez RT, Atkinson EN, Baggerly KA, Ramoth LR, Rosen DG, Liu J, Hellstrom I, et al: Selection of potential markers for epithelial ovarian cancer with gene expression arrays and recursive descent partition analysis. Clin Cancer Res. 10:3291–3300. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Skrzypczak M, Goryca K, Rubel T, Paziewska A, Mikula M, Jarosz D, Pachlewski J, Oledzki J and Ostrowski J: Modeling oncogenic signaling in colon tumors by multidirectional analyses of microarray data directed for maximization of analytical reliability. PLoS One. 5:e130912010. View Article : Google Scholar : PubMed/NCBI

33 

Molinari AM, Bontempo P, Schiavone EM, Tortora V, Verdicchio MA, Napolitano M, Nola E, Moncharmont B, Medici N, Nigro V, et al: Estradiol induces functional inactivation of p53 by intracellular redistribution. Cancer Res. 60:2594–2597. 2000.PubMed/NCBI

34 

Fernández-Cuesta L, Anaganti S, Hainaut P and Olivier M: Estrogen levels act as a rheostat on p53 levels and modulate p53-dependent responses in breast cancer cell lines. Breast Cancer Res Treat. 125:35–42. 2011. View Article : Google Scholar

35 

Somaï S, Chaouat M, Jacob D, Perrot JY, Rostène W, Forgez P and Gompel A: Antiestrogens are pro-apoptotic in normal human breast epithelial cells. Int J Cancer. 105:607–612. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Zhang GJ, Kimijima I, Onda M, Kanno M, Sato H, Watanabe T, Tsuchiya A, Abe R and Takenoshita S: Tamoxifen-induced apoptosis in breast cancer cells relates to down-regulation of bcl-2, but not bax and bcl-X(L), without alteration of p53 protein levels. Clin Cancer Res. 5:2971–2977. 1999.PubMed/NCBI

37 

Potting C, Tatsuta T, König T, Haag M, Wai T, Aaltonen MJ and Langer T: TRIAP1/PRELI complexes prevent apoptosis by mediating intramitochondrial transport of phosphatidic acid. Cell Metab. 18:287–295. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Cain K: Chemical-induced apoptosis: formation of the Apaf-1 apoptosome. Drug Metab Rev. 35:337–363. 2003. View Article : Google Scholar

Related Articles

Journal Cover

July-2015
Volume 34 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Adams C, Cazzanelli G, Rasul S, Hitchinson B, Hu Y, Coombes RC, Raguz S and Yagüe E: Apoptosis inhibitor TRIAP1 is a novel effector of drug resistance. Oncol Rep 34: 415-422, 2015
APA
Adams, C., Cazzanelli, G., Rasul, S., Hitchinson, B., Hu, Y., Coombes, R.C. ... Yagüe, E. (2015). Apoptosis inhibitor TRIAP1 is a novel effector of drug resistance. Oncology Reports, 34, 415-422. https://doi.org/10.3892/or.2015.3988
MLA
Adams, C., Cazzanelli, G., Rasul, S., Hitchinson, B., Hu, Y., Coombes, R. C., Raguz, S., Yagüe, E."Apoptosis inhibitor TRIAP1 is a novel effector of drug resistance". Oncology Reports 34.1 (2015): 415-422.
Chicago
Adams, C., Cazzanelli, G., Rasul, S., Hitchinson, B., Hu, Y., Coombes, R. C., Raguz, S., Yagüe, E."Apoptosis inhibitor TRIAP1 is a novel effector of drug resistance". Oncology Reports 34, no. 1 (2015): 415-422. https://doi.org/10.3892/or.2015.3988