Targeting β-Transducin Repeat–Containing Protein E3 Ubiquitin Ligase Augments the Effects of Antitumor Drugs on Breast Cancer Cells (original) (raw)

Skip Nav Destination

Experimental Therapeutics, Molecular Targets, and Chemical Biology| March 07 2005

Weigang Tang;

1Animal Biology and Departments of

Search for other works by this author on:

Ying Li;

1Animal Biology and Departments of

Search for other works by this author on:

Duonan Yu;

2Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and

Search for other works by this author on:

Andrei Thomas-Tikhonenko;

2Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and

Search for other works by this author on:

Vladimir S. Spiegelman;

3Department of Dermatology, University of Wisconsin, Madison, Wisconsin

Search for other works by this author on:

Serge Y. Fuchs

1Animal Biology and Departments of

Search for other works by this author on:

Crossmark: Check for Updates

Requests for reprints: Serge Y. Fuchs, Department of Animal Biology, University of Pennsylvania, 3800 Spruce Street, Room 161E VET, Philadelphia, PA 19104-6046. Phone: 215-573-6949; Fax: 215-573-5188; E-mail: syfuchs@vet.upenn.edu.

Received: July 21 2004

Revision Received: November 08 2004

Accepted: December 16 2004

Online ISSN: 1538-7445

Print ISSN: 0008-5472

©2005 American Association for Cancer Research.

2005

Cancer Res (2005) 65 (5): 1904–1908.

Article history

Revision Received:

November 08 2004

Accepted:

December 16 2004

Abstract

β-Transducin repeat–containing proteins (β-TrCP) serve as substrate recognition component of E3 ubiquitin ligases that control stability of important regulators of cell cycle and signal transduction. β-TrCP function is essential for the induction of nuclear factor κB transcriptional activities, which play a key role in proliferation and survival of cancer cells and are often constitutively up-regulated in human breast cancers. Here we show that inhibition of β-TrCP either by RNAi approach or by forced expression of a dominant-negative β-TrCP mutant suppresses growth and survival of human breast cancer cells. In addition, inhibition of β-TrCP augments the antiproliferative effects of anticancer drugs such as doxorubicin, tamoxifen, and paclitaxel on human mammary tumor cells. These data provide the proof of principle that targeting β-TrCP might be beneficial for anticancer therapies.

Introduction

Conjugation of proteins with ubiquitin (ubiquitination) and ubiquitin-like proteins have emerged as an important mechanism in regulating neoplastic cell growth and survival (1). It has been long suggested that aberrant ubiquitination of regulatory proteins contributes to cell transformation and tumor progression and, therefore, represents potential target for anticancer therapy (2, 3). Inhibition of protein sumoylation in cells was shown to sensitize neoplastic cells to anticancer drugs (4). Suppression of proteasomal degradation by Velcade (bortezomib) is effective against multiple myeloma; the therapeutic benefits of this drug for patients with other hematologic malignancies as well as with solid tumors are currently under clinical investigation (5). A major mechanism of anticancer effects of proteasomal inibitors is thought to be the suppression of prosurvival nuclear transcription factor κB (NFκB) due to stabilization of its inhibitors (IκB; ref. 6). Proteasomal degradation of IκB requires phosphorylation-dependent ubiquitination of IκB, which is mediated by β-transducing repeat–containing proteins (β-TrCP; refs. 7, 8). Ensuing NFκB activation contributes to many aspects of tumor development including accelerated cell cycle progression, cell proliferation, tumor initiation and promotion, angiogenesis, metastasis, etc. As a major antiapoptotic factor, it plays a pivotal role in the resistance of tumors to chemotherapy and radiation. Constitutive activation of NFκB is a hallmark of many human malignancies including breast cancer (reviewed in ref. 9). Closely related β-TrCP1 and β-TrCP2 proteins seem to play a redundant role in ubiquitination and degradation of IκB (7). Expression of β-TrCP2 (also termed HOS) is induced in human breast cancer cell lines and primary tumor samples (10). Mammary glands of the β-TrCP1 knockout mice are hypoplastic; conversely, transgenic mice expressing human β-TrCP1 under control of the mouse mammary tumor virus long terminal repeat promoter exhibit hyperproliferation of mammary epithelium concurrent with nuclear localization of NFκB p65/RelA and development of mammary carcinomas (11). These data indicate that β-TrCP may play an important role in regulating growth and survival of mammary cells and development of breast cancer. This provides justification for targeting β-TrCP to limit proliferation and survival of mammary tumor cells. However, the proof of principle for this approach has not yet been established. Here we show that inhibition of β-TrCP by short inhibitory RNA (siRNA) or by expression of a dominant-negative mutant are effective in suppressing growth and survival of human breast cancer cells alone or in combination with various chemotherapeutic agents.

Materials and Methods

Cell Culture and Drug Treatment. Human breast cancer cell lines MDA-MB-468, T47D, and MCF-7 were purchased from American Type Culture Collection (Manassas, VA).. The cells were grown in DMEM/F12 (1:1) medium supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin mixture at 37°C and 5% CO2. Human embryonic kidney cell line 293T purchased from American Type Culture Collection were grown in DMEM with 10% fetal bovine serum, 1% penicillin-streptomycin at 37°C and 5% CO2. T47D cells stably expressing pETH tet-off regulator and hygromycin resistance marker were grown in the presence of hygromycin B (150 μg/mL) and G418 (800 μg/mL). Anticancer drugs doxorubicin, tamoxifen, and paclitaxel were purchased from Sigma (St. Louis, MO), dissolved in DMSO, and added to cell medium. NFκB activity was measured in cells cotransfected with κB-luciferase reporter and Renilla luciferase construct using Dual Luciferase assay (Promega, Madison, WI).

DNA Constructs, Transfection, and Retroviral Transduction. The siRNA against β-TrCP2 (siBTR2) cloned in pSilencer1.0-U6 vector (Ambion, Austin, TX) as well as control siRNA that differs from siBTR2 by two base pair substitution (siCON) were previously described (12). siRNA against β-TrCP1 (siBTR1) were generated in the same vector using 5′-TTCCTCAGAGAGAGAAGACTG-3′ as a targeting sequence. pBI-G-HA-β-TrCPΔF and pBabe-puro-HA-β-TrCPΔF were constructed by cloning β-TrCP2ΔF [hemagglutinin (HA)-tagged β-TrCP2 lacking the F-box] into the pBI-G vector (for tet-dependent expression of β-galactosidase and the gene of interest, Clontech, Palo Alto, CA) or pBabe-puro vector, respectively. pMIGR1-β-TrCPΔF was constructed by ligating β-TrCPΔF into a bicistronic green fluorescent protein expression retroviral vector pMIGR1 (13), a gift from Dr. W. Pear (Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA). Tet-off regulator plasmid pETH was kindly provided by Dr. Stuart A. Aaronson (Ruttenberg Cancer Center, Mount Sinai School of Medicine, New York, NY). Cells were transfected using LipofectAMINE Plus (Invitrogen, San Diego, CA) or standard calcium phosphate method. Retrovirus-containing supernatants from 293T cells transfected with pMIGR1- or pBabe-puro constructs, as well as with vesicular stomatitis virus-G and GAG-pol plasmids, were prepared and used for transduction of breast cancer cells in the presence of polybrene (6 μg/mL) as previously described (13). Infected breast cancer cells were selected in the presence of puromycin (1-2 μg/mL) for 2 days and puromycin-resistant cells were plated for colony formation and cell accumulation WST-1 assays. Expression of endogenous β-TrCP proteins or expressed HA-tagged β-TrCPΔF protein was analyzed by immunoblotting using anti-β-TrCP HOS-C antibody (14) or anti-HA tag antibody (Roche, Indianapolis, IN) as described elsewhere (12, 15).

Survival of β-Galactosidase–Positive Cells. T47D tet-off cells transfected with either pBI-G empty vector or pBI-G-β-TrCPΔF were seeded in a 96-well plate and incubated in the presence or absence of tetracycline and in the presence of vehicle (DMSO) or anticancer drugs for 48 hours. β-Galactosidase–positive cells were revealed by staining with 5-bromo-4-chloro-3-indolyl β-d-galactoside (Sigma) and enumerated under light microscope.

Colony Formation Assay. A predetermined number of cells that yields ∼100 colonies for each of the cell lines used was seeded into 6-well plates in the medium containing puromycin. Anticancer drugs or DMSO was added to the cells 24 hours later. Cells were grown for 21 days, fixed with 70% cold methanol, stained with Giemsa stain (Sigma), and the colonies of 20 or more cells were counted.

Cell Accumulation WST-1 Assay. Puromycin-resistant breast cancer cells or nontumorigenic human mammary MCF10a cells were plated in 96-well plates in the presence of puromycin. After overnight incubation, the cells were treated with anticancer drugs or DMSO and incubated for additional 72 hours. WST-1 reagents (Roche) was added to the cells and the number of live cells was estimated by measuring the absorbance at 450 nm with a microplate reader.

Apoptosis Assay. Breast cancer cells transduced with pMIGR1 retroviruses that coexpress green fluorescent protein were plated onto glass coverslips placed in 35-mm dishes. Following treatment with drugs or DMSO, the medium was removed and cells were fixed and stained with 4′,6-diamidino-2-phenylindole as described previously (15). Cells (400-500) were examined in five to seven randomly selected fields and apoptotic cells exhibiting condensed and fragmented nuclei were scored.

Results and Discussion

We sought to investigate whether β-TrCP levels are important for growth and survival of human breast cancer cells using the RNAi approach that proved efficient in delineating the function of these proteins (12, 16). Whereas siRNA specific against β-TrCP1 or β-TrCP2 efficiently down-regulated exogenously expressed or endogenous β-TrCP species, knockdown of β-TrCP2 led to a more dramatic inhibition of NFκB activity (Fig. 1A and B). Cotransfection of siRNA and pBabe-puro constructs followed by colony formation assay in the presence of puromycin revealed that knockdown of β-TrCP2 led to a statistically significant inhibition of growth in T47D cells (Fig. 1C). Combination of siBTR2 with anticancer drugs further decreased the growth of these cells. Less efficient growth suppression was observed in cells transfected with siRNA against β-TrCP1 (siBTR1; Fig. 1C). These data indicate that β-TrCP in general and β-TrCP2 in particular are essential for the maintenance of growth and survival of human breast cancer cells.

Figure 1.

Figure 1. siRNA against β-TrCP inhibits growth of T47D human breast cancer cells. A, cells were cotransfected with HA-tagged β-TrCP1 or β-TrCP2 and siRNA constructs against β-TrCP1 (siBTR1) or β-TrCP2 (siBTR2) or irrelevant sequence (siCON). Levels of β-TrCP proteins were analyzed by immunoblotting with HA antibody. Levels of α-tubulin were also assessed. B, cells were transfected with indicated siRNA and the levels of endogenous β-TrCP were analyzed by immunoblotting with HOS-C antibody (which recognized both β-TrCP1 and β-TrCP2, ref. 14). Numbers under the blots, normalized activity of NFκB-driven luciferase reporter (SD is given in parenthesis) in these cells analyzed in parallel experiments using Dual Luciferase assay. C, cells were cotransfected with pBabe-puro plasmid and either siCON (white columns), siBTR2 (gray columns), or siBTR1 (black columns). The cells were seeded in six-well plates, treated with drugs (at the indicated doses) and puromycin, and processed for the colony formation assay. Average number of colonies from three independent experiments (each in triplicate). *, P < 0.05, compared with siCON (Student's t test). Dox, doxorubicin; Tam, tamoxifen; Pac, paclitaxel.

siRNA against β-TrCP inhibits growth of T47D human breast cancer cells. A, cells were cotransfected with HA-tagged β-TrCP1 or β-TrCP2 and siRNA constructs against β-TrCP1 (siBTR1) or β-TrCP2 (siBTR2) or irrelevant sequence (siCON). Levels of β-TrCP proteins were analyzed by immunoblotting with HA antibody. Levels of α-tubulin were also assessed. B, cells were transfected with indicated siRNA and the levels of endogenous β-TrCP were analyzed by immunoblotting with HOS-C antibody (which recognized both β-TrCP1 and β-TrCP2, ref. 14). Numbers under the blots, normalized activity of NFκB-driven luciferase reporter (SD is given in parenthesis) in these cells analyzed in parallel experiments using Dual Luciferase assay. C, cells were cotransfected with pBabe-puro plasmid and either siCON (white columns), siBTR2 (gray columns), or siBTR1 (black columns). The cells were seeded in six-well plates, treated with drugs (at the indicated doses) and puromycin, and processed for the colony formation assay. Average number of colonies from three independent experiments (each in triplicate). *, P < 0.05, compared with siCON (Student's t test). Dox, doxorubicin; Tam, tamoxifen; Pac, paclitaxel.

Figure 1.

Figure 1. siRNA against β-TrCP inhibits growth of T47D human breast cancer cells. A, cells were cotransfected with HA-tagged β-TrCP1 or β-TrCP2 and siRNA constructs against β-TrCP1 (siBTR1) or β-TrCP2 (siBTR2) or irrelevant sequence (siCON). Levels of β-TrCP proteins were analyzed by immunoblotting with HA antibody. Levels of α-tubulin were also assessed. B, cells were transfected with indicated siRNA and the levels of endogenous β-TrCP were analyzed by immunoblotting with HOS-C antibody (which recognized both β-TrCP1 and β-TrCP2, ref. 14). Numbers under the blots, normalized activity of NFκB-driven luciferase reporter (SD is given in parenthesis) in these cells analyzed in parallel experiments using Dual Luciferase assay. C, cells were cotransfected with pBabe-puro plasmid and either siCON (white columns), siBTR2 (gray columns), or siBTR1 (black columns). The cells were seeded in six-well plates, treated with drugs (at the indicated doses) and puromycin, and processed for the colony formation assay. Average number of colonies from three independent experiments (each in triplicate). *, P < 0.05, compared with siCON (Student's t test). Dox, doxorubicin; Tam, tamoxifen; Pac, paclitaxel.

siRNA against β-TrCP inhibits growth of T47D human breast cancer cells. A, cells were cotransfected with HA-tagged β-TrCP1 or β-TrCP2 and siRNA constructs against β-TrCP1 (siBTR1) or β-TrCP2 (siBTR2) or irrelevant sequence (siCON). Levels of β-TrCP proteins were analyzed by immunoblotting with HA antibody. Levels of α-tubulin were also assessed. B, cells were transfected with indicated siRNA and the levels of endogenous β-TrCP were analyzed by immunoblotting with HOS-C antibody (which recognized both β-TrCP1 and β-TrCP2, ref. 14). Numbers under the blots, normalized activity of NFκB-driven luciferase reporter (SD is given in parenthesis) in these cells analyzed in parallel experiments using Dual Luciferase assay. C, cells were cotransfected with pBabe-puro plasmid and either siCON (white columns), siBTR2 (gray columns), or siBTR1 (black columns). The cells were seeded in six-well plates, treated with drugs (at the indicated doses) and puromycin, and processed for the colony formation assay. Average number of colonies from three independent experiments (each in triplicate). *, P < 0.05, compared with siCON (Student's t test). Dox, doxorubicin; Tam, tamoxifen; Pac, paclitaxel.

Close modal

To corroborate these results we used a tetracycline inducible system to express the dominant-negative β-TrCPΔF mutant, which lacks the F-box and, hence, the ability to recruit E3 ubiquitin ligase activity. This mutant has been previously shown to target both forms of β-TrCP and to inhibit NFκB (17). Expression of such a mutant induced by tetracycline withdrawal led to a significant growth inhibition effect in all treatment groups, including DMSO, with the maximum efficiency in cells treated with tamoxifen or paclitaxel (Fig. 2). These results support our findings obtained with siRNA and suggest that inhibition of β-TrCP may augment the antiproliferative effects of anticancer agents in T47D human breast cancer cells.

Figure 2.

Figure 2. Tet-regulated expression of the dominant-negative β-TrCPΔF mutant reduces survival of T47D cells. Derivatives of T47D cells, tet-off-pBI-G (white columns) or tet-off-pBI-G-β-TrCPΔF cells (black columns) were cultured in the presence or absence of tetracycline and with or without drugs at indicated concentrations for 48 hours. Survival of cells that express β-galactosidase (Gal) alone (white columns) or together with β-TrCPΔF (black columns) was assessed by scoring the number of β-galactosidase–positive cells. Average from three independent experiments (each in triplicate). *, P < 0.05, compared with tet-off-pBI-G cells (Student's t test). Inset, expression of HA-tagged β-TrCPΔF analyzed by immunoblotting with HA antibody. Tub, α-tubulin.

Tet-regulated expression of the dominant-negative β-TrCPΔF mutant reduces survival of T47D cells. Derivatives of T47D cells, tet-off-pBI-G (white columns) or tet-off-pBI-G-β-TrCPΔF cells (black columns) were cultured in the presence or absence of tetracycline and with or without drugs at indicated concentrations for 48 hours. Survival of cells that express β-galactosidase (Gal) alone (white columns) or together with β-TrCPΔF (black columns) was assessed by scoring the number of β-galactosidase–positive cells. Average from three independent experiments (each in triplicate). *, P < 0.05, compared with tet-off-pBI-G cells (Student's t test). Inset, expression of HA-tagged β-TrCPΔF analyzed by immunoblotting with HA antibody. Tub, α-tubulin.

Figure 2.

Figure 2. Tet-regulated expression of the dominant-negative β-TrCPΔF mutant reduces survival of T47D cells. Derivatives of T47D cells, tet-off-pBI-G (white columns) or tet-off-pBI-G-β-TrCPΔF cells (black columns) were cultured in the presence or absence of tetracycline and with or without drugs at indicated concentrations for 48 hours. Survival of cells that express β-galactosidase (Gal) alone (white columns) or together with β-TrCPΔF (black columns) was assessed by scoring the number of β-galactosidase–positive cells. Average from three independent experiments (each in triplicate). *, P < 0.05, compared with tet-off-pBI-G cells (Student's t test). Inset, expression of HA-tagged β-TrCPΔF analyzed by immunoblotting with HA antibody. Tub, α-tubulin.

Tet-regulated expression of the dominant-negative β-TrCPΔF mutant reduces survival of T47D cells. Derivatives of T47D cells, tet-off-pBI-G (white columns) or tet-off-pBI-G-β-TrCPΔF cells (black columns) were cultured in the presence or absence of tetracycline and with or without drugs at indicated concentrations for 48 hours. Survival of cells that express β-galactosidase (Gal) alone (white columns) or together with β-TrCPΔF (black columns) was assessed by scoring the number of β-galactosidase–positive cells. Average from three independent experiments (each in triplicate). *, P < 0.05, compared with tet-off-pBI-G cells (Student's t test). Inset, expression of HA-tagged β-TrCPΔF analyzed by immunoblotting with HA antibody. Tub, α-tubulin.

Close modal

To investigate whether these findings could be expanded to other breast cell lines we subcloned the dominant-negative β-TrCPΔF mutant in pBabe-puro retroviral vector and used it for transduction of human breast cancer cells. Attenuation of β-TrCP function by retroviral-mediated expression of β-TrCPΔF mutant led to a dramatic decrease of colony formation by estrogen-dependent MCF7 and T47D cells. Combination of β-TrCP inhibition with anticancer drugs resulted in further decrease in cell growth and survival (Fig. 3A and C). WST-1 cell accumulation assay revealed similar results (Table 1). Interestingly, inhibition of β-TrCP also decreased growth of nontumorigenic human mammary MCF10a cells and sensitized these cells to the effects of doxorubicin and paclitaxel but not tamoxifen (Table 1). Conversely, whereas tamoxifen had no effect on hormone-resistant MDA-MB-468 cells, these cells were sensitive to inhibition of β-TrCP function (Table 1; Fig. 3B). These results suggest that targeting β-TrCP could be used in estrogen receptor–negative breast cancers that are insensitive to hormone analogues. However, toxicity of β-TrCP–targeting agents against proliferating nontumorigenic breast cells has to be taken into consideration when choosing a partner drug for the combination therapy.

Figure 3.

Figure 3. Retrovirus-mediated expression of the dominant-negative β-TrCPΔF mutant inhibits growth of breast cancer cells. Growth and survival of T47D cells (A), MDA-MB468 cells (B), and MCF7 cells (C) transduced with an empty pBabe-puro retrovirus (white columns) or pBabe-puro-β-TrCPΔF cells (black columns) that were cultured in the presence of puromycin and with or without drugs was analyzed by colony formation assay. Average from three independent experiments (each in triplicate). *, P < 0.05, compared with empty virus (Student's t test). A, inset, expression of HA-tagged β-TrCPΔF analyzed by immunoblotting with HA antibody.

Retrovirus-mediated expression of the dominant-negative β-TrCPΔF mutant inhibits growth of breast cancer cells. Growth and survival of T47D cells (A), MDA-MB468 cells (B), and MCF7 cells (C) transduced with an empty pBabe-puro retrovirus (white columns) or pBabe-puro-β-TrCPΔF cells (black columns) that were cultured in the presence of puromycin and with or without drugs was analyzed by colony formation assay. Average from three independent experiments (each in triplicate). *, P < 0.05, compared with empty virus (Student's t test). A, inset, expression of HA-tagged β-TrCPΔF analyzed by immunoblotting with HA antibody.

Figure 3.

Figure 3. Retrovirus-mediated expression of the dominant-negative β-TrCPΔF mutant inhibits growth of breast cancer cells. Growth and survival of T47D cells (A), MDA-MB468 cells (B), and MCF7 cells (C) transduced with an empty pBabe-puro retrovirus (white columns) or pBabe-puro-β-TrCPΔF cells (black columns) that were cultured in the presence of puromycin and with or without drugs was analyzed by colony formation assay. Average from three independent experiments (each in triplicate). *, P < 0.05, compared with empty virus (Student's t test). A, inset, expression of HA-tagged β-TrCPΔF analyzed by immunoblotting with HA antibody.

Retrovirus-mediated expression of the dominant-negative β-TrCPΔF mutant inhibits growth of breast cancer cells. Growth and survival of T47D cells (A), MDA-MB468 cells (B), and MCF7 cells (C) transduced with an empty pBabe-puro retrovirus (white columns) or pBabe-puro-β-TrCPΔF cells (black columns) that were cultured in the presence of puromycin and with or without drugs was analyzed by colony formation assay. Average from three independent experiments (each in triplicate). *, P < 0.05, compared with empty virus (Student's t test). A, inset, expression of HA-tagged β-TrCPΔF analyzed by immunoblotting with HA antibody.

Close modal

Table 1.

Effect of β-TrCPΔF on growth of human breast cancer cells measured by the WST-1 assay

Treatment T47D transduction MCF7 Transduction MDA-MB468 Transduction MCF10A Transduction
Vector βTrCPΔF Vector βTrCPΔF Vector βTrCPΔF Vector βTrCPΔF
DMSO 1248 ± 76 516 ± 19* 780 ± 49 559 ± 39† 2508 ± 71 1032 ± 34* 1515 ± 139 649 ± 46*
Doxorubicin
1 nmol/L NT NT NT NT NT NT 1215 ± 87 416 ± 68
1 μmol/L NT NT NT NT 107 ± 8 16 ± 4* NT NT
5 μmol/L 46 ± 6 17 ± 3* 217 ± 38 133 ± 310* NT NT NT NT
Tamoxifen
5 μmol/L 456 ± 99 137 ± 19* 81 ± 18 24 ± 3* 1844 ± 146 257 ± 56* NT NT
15μmol/L NT NT NT NT NT NT 1387 ± 250 694 ± 108*
Paclitaxel
1 nmol/L NT NT NT NT NT NT 1075 ± 73 354 ± 46*
250 nmol/L 532 ± 54 217 ± 27* 538 ± 42 363 ± 64† 1175 ± 86 170 ± 19* NT NT
Treatment T47D transduction MCF7 Transduction MDA-MB468 Transduction MCF10A Transduction
Vector βTrCPΔF Vector βTrCPΔF Vector βTrCPΔF Vector βTrCPΔF
DMSO 1248 ± 76 516 ± 19* 780 ± 49 559 ± 39† 2508 ± 71 1032 ± 34* 1515 ± 139 649 ± 46*
Doxorubicin
1 nmol/L NT NT NT NT NT NT 1215 ± 87 416 ± 68
1 μmol/L NT NT NT NT 107 ± 8 16 ± 4* NT NT
5 μmol/L 46 ± 6 17 ± 3* 217 ± 38 133 ± 310* NT NT NT NT
Tamoxifen
5 μmol/L 456 ± 99 137 ± 19* 81 ± 18 24 ± 3* 1844 ± 146 257 ± 56* NT NT
15μmol/L NT NT NT NT NT NT 1387 ± 250 694 ± 108*
Paclitaxel
1 nmol/L NT NT NT NT NT NT 1075 ± 73 354 ± 46*
250 nmol/L 532 ± 54 217 ± 27* 538 ± 42 363 ± 64† 1175 ± 86 170 ± 19* NT NT

NOTE: Cell proliferation is measured by cell/WST1 reagent–generated absorbance at 450 nm minus the background (an absorbance of a tissue culture well containing medium without cells). Average data from three independent experiments (each in triplicate) ± SDs are shown.

Abbreviation: NT, not tested.

*

P < 0.01, compared with cells transduced with empty vector (Student's t test).

P < 0.05, compared with cells transduced with empty vector (Student's t test).

Expression of β-TrCPΔF leads to an inhibition of ubiquitination and degradation of IκB, which, in turn, results in a decreased activity of a major antiapoptotic factor, NFκB (17). Given that the decrease in growth of human breast cancer cells following inhibition of β-TrCP may result from an accelerated cell death as much as from inhibition of cell proliferation, we sought to investigate whether expression of β-TrCPΔF affects the rate of apoptosis in these cells. Indeed, retrovirus-mediated expression of β-TrCPΔF significantly increased the rate of apoptosis in T47D and MDA-MB468 breast cancer cells even in the absence of anticancer drugs (Fig. 4A and B). Synergistic effect of β-TrCPΔF mutant with doxorubicin, paclitaxel, or tamoxifen was observed in T47D and MDA-MB-468 cells. The evidence suggests that inhibition of β-TrCP sensitizes human breast cancer cells to apoptosis. A similar effect has been previously observed in melanoma cells treated with ionizing radiation or cisplatin (15). MCF7 cells, which lack caspase 3 (18) and are known to resist inhibition of NFκB (19), were also somewhat less sensitive to apoptosis induced by the β-TrCPΔF mutant (Fig. 4C). Nevertheless, combination of β-TrCP inhibition with anticancer drugs induced an augmented apoptotic response in these cells.

Figure 4.

Figure 4. Retrovirus-mediated expression of the dominant-negative β-TrCPΔF mutant promotes apoptosis in breast cancer cells. The rate of apoptosis in T47D cells (A), MDA-MB468 cells (B), and MCF7 cells (C) transduced with an empty pMIGR1 retrovirus (white columns) or pMIGR1-β-TrCPΔF cells (black columns) that were cultured with or without drugs (at the indicated concentrations) for 24 hours, fixed, and stained with 4′,6-diamidino-2-phenylindole (DAPI). Apoptotic cells with condensed/fragmented nuclei were scored among green fluorescent protein (GFP)–positive cells. An example of such cell (white arrow) is shown in (B). Average from three independent experiments (each in triplicate). *, P < 0.05, compared with empty virus (Student's t test).

Retrovirus-mediated expression of the dominant-negative β-TrCPΔF mutant promotes apoptosis in breast cancer cells. The rate of apoptosis in T47D cells (A), MDA-MB468 cells (B), and MCF7 cells (C) transduced with an empty pMIGR1 retrovirus (white columns) or pMIGR1-β-TrCPΔF cells (black columns) that were cultured with or without drugs (at the indicated concentrations) for 24 hours, fixed, and stained with 4′,6-diamidino-2-phenylindole (DAPI). Apoptotic cells with condensed/fragmented nuclei were scored among green fluorescent protein (GFP)–positive cells. An example of such cell (white arrow) is shown in (B). Average from three independent experiments (each in triplicate). *, P < 0.05, compared with empty virus (Student's t test).

Figure 4.

Figure 4. Retrovirus-mediated expression of the dominant-negative β-TrCPΔF mutant promotes apoptosis in breast cancer cells. The rate of apoptosis in T47D cells (A), MDA-MB468 cells (B), and MCF7 cells (C) transduced with an empty pMIGR1 retrovirus (white columns) or pMIGR1-β-TrCPΔF cells (black columns) that were cultured with or without drugs (at the indicated concentrations) for 24 hours, fixed, and stained with 4′,6-diamidino-2-phenylindole (DAPI). Apoptotic cells with condensed/fragmented nuclei were scored among green fluorescent protein (GFP)–positive cells. An example of such cell (white arrow) is shown in (B). Average from three independent experiments (each in triplicate). *, P < 0.05, compared with empty virus (Student's t test).

Retrovirus-mediated expression of the dominant-negative β-TrCPΔF mutant promotes apoptosis in breast cancer cells. The rate of apoptosis in T47D cells (A), MDA-MB468 cells (B), and MCF7 cells (C) transduced with an empty pMIGR1 retrovirus (white columns) or pMIGR1-β-TrCPΔF cells (black columns) that were cultured with or without drugs (at the indicated concentrations) for 24 hours, fixed, and stained with 4′,6-diamidino-2-phenylindole (DAPI). Apoptotic cells with condensed/fragmented nuclei were scored among green fluorescent protein (GFP)–positive cells. An example of such cell (white arrow) is shown in (B). Average from three independent experiments (each in triplicate). *, P < 0.05, compared with empty virus (Student's t test).

Close modal

The important role of the ubiquitin pathway in regulating some key regulatory events in mammary tumorigenesis prompted us to explore the inhibition of specific E3 ubiquitin ligases as a potential therapy of breast cancers (20). Our findings presented here collectively indicate that targeting β-TrCP expression and function is detrimental for growth and survival of human breast cancer cells. Furthermore, inhibition of β-TrCP augments the effect of anticancer drugs on these cells. Mechanisms of such sensitization are likely to include the conflict of signals from stabilized antigrowth/survival β-TrCP substrates [including IκB, IFNAR1 (21), mitotic inhibitor Emi1 (16, 22), etc.] and those β-TrCP substrates that promote growth and survival [e.g., β-catenin, prolactin receptor (12)]. Future studies will be aimed at identification of suitable inhibitors of β-TrCP function that could improve the therapeutic benefits of anticancer drugs against human breast cancer.

Note: W. Tang and Y. Li contributed equally to this work.

Acknowledgments

Grant support: NIH grants CA 92900 (S.Y. Fuchs) and CA102709 (A. Thomas-Tikhonenko) and the American Cancer Society award RSG-02-140-01-CNE (V.S. Spiegelman).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Drs. Stuart A. Aaronson and Warren Pear for providing reagents.

References

1

Pagano M, Benmaamar R. When protein destruction runs amok, malignancy is on the loose.

Cancer Cell

2003

;

4

:

251

–6.

2

Wong BR, Parlati F, Qu K, et al. Drug discovery in the ubiquitin regulatory pathway.

Drug Discov Today

2003

;

8

:

746

–54.

3

Pray TR, Parlati F, Huang, J et al. Cell cycle regulatory E3 ubiquitin ligases as anticancer targets.

Drug Resist Updat

2002

;

5

:

249

–58.

4

Li TK, Liu LF Tumor cell death induced by topoisomerase-targeting drugs.

Annu Rev Pharmacol Toxicol

2001

;

41

:

53

–77.

5

Adams J, Kauffman M. Development of the proteasome inhibitor Velcade (Bortezomib).

Cancer Invest

2004

;

22

:

304

–11.

6

Ma MH, Yang HH, Parker K. et al. The proteasome inhibitor PS-341 markedly enhances sensitivity of multiple myeloma tumor cells to chemotherapeutic agents.

Clin Cancer Res

2003

;

9

:

1136

–44.

7

Fuchs SY, Spiegelman VS, Kumar KG. The many faces of β-TrCP E3 ubiquitin ligases: reflections in the magic mirror of cancer.

Oncogene

2004

;

23

:

2028

–36.

8

Fuchs SY. The role of ubiquitin-proteasome pathway in oncogenic signaling.

Cancer Biol Ther

2002

;

1

:

337

–41.

9

Garg A, Aggarwal BB. Nuclear transcription factor-κB as a target for cancer drug development.

Leukemia

2002

:

16

:

1053

–68.

10

Spiegelman VS, Tang W, Chan AM, et al. Induction of homologue of Slimb ubiquitin ligase receptor by mitogen signaling.

J Biol Chem

2002

;

277

:

36624

–30.

11

Kudo Y, Guardavaccaro D, Santamaria PG, et al. Role of F-box protein βTrcp1 in mammary gland development and tumorigenesis.

Mol Cell Biol

2004

;

24

:

8184

–94.

12

Li Y, Kumar KG, Tang W, Spiegelman VS, Fuchs SY. Negative regulation of prolactin receptor stability and signaling mediated by SCF(β-TrCP) E3 ubiquitin ligase.

Mol Cell Biol

2004

;

24

:

4038

–48.

13

Pear WS, Miller JP, Xu L, et al., Efficient and rapid induction of a chronic myelogenous leukemia-like myeloproliferative disease in mice receiving P210 bcr/abl-transduced bone marrow.

Blood

1998

;

92

:

3780

–92.

14

Spiegelman VS, Tang W, Katoh M, Slaga TJ, Fuchs SY. Inhibition of HOS expression and activities by Wnt pathway.

Oncogene

2002

;

21

:

856

–60.

15

Soldatenkov VA, Dritschilo A, Ronai Z, Fuchs SY. Inhibition of homologue of Slimb (HOS) function sensitizes human melanoma cells for apoptosis.

Cancer Res

1999

;

59

:

5085

–8.

16

Guardavaccaro D, Kudo Y, Boulaire J, et al. Control of meiotic and mitotic progression by the F box protein β-TrCP1 in vivo.

Dev Cell

2003

;

4

:

799

–812.

17

Fuchs SY, Chen A, Xiong Y, Pan ZQ, Ronai Z. HOS, a human homolog of Slimb, forms an SCF complex with Skp1 and Cullin1 and targets the phosphorylation-dependent degradation of IκB and β-catenin.

Oncogene

1999

;

18

:

2039

–46.

18

Janicke RU, Sprengart ML, Wati MR, Porter AG. Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis.

J Biol Chem

1998

;

273

:

9357

–60.

19

Cai Z, Korner M, Tarantino N, Chouaib S. IκB α overexpression in human breast carcinoma MCF7 cells inhibits nuclear factor-κB activation but not tumor necrosis factor-α-induced apoptosis.

J Biol Chem

1997

;

272

:

96

–101.

20

Cardoso F, Ross JS, Picart MJ, Sotiriou C, Durbecq V. Targeting the ubiquitin-proteasome pathway in breast cancer.

Clin Breast Cancer

2004

;

5

:

148

–57.

21

Kumar KG, Tang W, Ravindranath AK, Clark WA, Croze E, Fuchs SY. SCF(HOS) ubiquitin ligase mediates the ligand-induced down-regulation of the interferon-α receptor.

EMBO J

2003

;

22

:

5480

–90.

22

Margottin-Goguet F, Hsu JY, Loktev A, Hsieh HM, Reimann JD, Jackson PK. Prophase destruction of Emi1 by the SCF(βTrCP/Slimb) ubiquitin ligase activates the anaphase promoting complex to allow progression beyond prometaphase.

Dev Cell

2003

;

4

:

813

–26.

©2005 American Association for Cancer Research.

2005

341 Views

48 Web of Science

41 Crossref

Citing articles via

Email alerts