51
|
Hurrell T, Outhoff K. Human epidermal growth factor receptor 2-positive breast cancer: which cytotoxic agent best complements trastuzumab's efficacy in vitro? Onco Targets Ther 2013; 6:693-701. [PMID: 23814468 PMCID: PMC3693918 DOI: 10.2147/ott.s46883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Despite trastuzumab having enhanced selectivity for human epidermal growth factor receptor 2 (HER-2) overexpressing breast cancer cells, treatment is hampered by interindividual variation and tumors with high mitogenic potential. The lack of significant clinical benefit in certain patient cohorts suggests that HER-2 expression is ineffective as a sole prognostic indicator of response to therapy. Therefore, optimizing the clinical role of trastuzumab in drug combinations remains critical for clinical success. AIM To investigate the effects of trastuzumab in combination with either doxorubicin or geldanamycin on in vitro cell viability, cell cycling, apoptosis and relative HER-2 expression in HER-2-positive (SK-BR-3) and estrogen receptor-positive (MCF-7) breast adenocarcinoma models. RESULTS HER-2-rich SK-BR-3 cells demonstrated a greater sensitivity to the effects of doxorubicin than MCF-7 cells. Concurrent trastuzumab exposure resulted in a further reduction in cell viability. This decreased cell viability induced by doxorubicin was associated with activation of executioner caspases as well as with alterations in cell-cycle kinetics, primarily promoting S-phase accumulation. Doxorubicin had no effect on surface HER-2 density expression. Geldanamycin reduced cell viability significantly greater in SK-BR-3 than MCF-7 cells, and was associated with G2 cell-cycle accumulation. The addition of trastuzumab did not augment these effects. Geldanamycin promoted substantial reductions in relative surface HER-2 density in SK-BR-3 cells. CONCLUSION The in vitro data supported the rationale for using doxorubicin in trastuzumab-based therapies. Therefore, despite the incidence of cardiotoxicity, doxorubicin could retain a fundamental role in treating HER-2-positive breast cancer. While geldanamycin is a potent cytotoxic agent, its concurrent use with trastuzumab requires further research into the transient or permanent nature of alterations in HER-2 status in cell progeny.
Collapse
Affiliation(s)
- Tracey Hurrell
- Department of Pharmacology, University of Pretoria, Pretoria, South Africa
| | | |
Collapse
|
52
|
Jin M, Shen X, Zhao C, Qin X, Liu H, Huang L, Qiu Z, Liu Y. In vivo study of effects of artesunate nanoliposomes on human hepatocellular carcinoma xenografts in nude mice. Drug Deliv 2013; 20:127-33. [PMID: 23731485 DOI: 10.3109/10717544.2013.801047] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To investigate the effect of artesunate nanoliposomes on cultured cells in vitro and hepatocellular carcinoma xenografts in BALB/c-nu mice. Fluorescence polarization was applied for measurement of mitochondrial membrane fluidities; inhibition test of tumor cell proliferation in vitro was performed and nude mice xenograft model from human hepatocellular carcinoma (HCC) was established. Cytotoxicity of these compounds was evaluated by MTT assay on hepatocellular carcinoma xenografts in nude mice. Anisotropy (r-value) of blank nanoliposomes didn't change, it had no statistically significance between the blank nanoliposomes group and the control group, it indicated that artesunate had no obvious effect on L-O2 human normal liver cells. IC₅₀ values of artesunate nanoliposomes and artesunate API (active pharmaceutical ingredient) against HepG-2 cells were 15.997 and 19.706 μg/ml; IC₅₀ values of the same drugs against L-O2 normal human liver cells were 100.23 and 105.54 μg/ml, respectively. Tumor growth inhibitory effect of artesunate nanoliposomes was 32.7%, and artesunate API was 20.5%, respectively. HepG-2 cells treated with artesunate nanoliposomes showed dose-dependent apoptosis. The antitumor effect of artesunate nanoliposomes on human hepatoma HepG2 cells were stronger than that of artesunate API at the same concentration.
Collapse
Affiliation(s)
- Meihua Jin
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Tehranian N, Sepehri H, Mehdipour P, Biramijamal F, Hossein-Nezhad A, Sarrafnejad A, Hajizadeh E. Combination effect of PectaSol and Doxorubicin on viability, cell cycle arrest and apoptosis in DU-145 and LNCaP prostate cancer cell lines. Cell Biol Int 2012; 36:601-10. [PMID: 22214228 DOI: 10.1042/cbi20110309] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The effect of PectaSol on Dox (Doxorubicin) cytotoxicity in terms of apoptosis and cell cycle changes in PCa (prostate cancer) cell lines (DU-145 and LNCaP) has been investigated. Combination of PectaSol and Dox resulted in a viability of 29.4 and 32.6% (P<0.001) in DU-145 and LNCaP cells. The IC₅₀ values decreased 1.5-fold and 1.3-fold in the DU-145 and LNCaP cells respectively. In the DU-145 cells, combination of PectaSol and Dox resulted in a reduction in p27 gene and protein expression (P<0.001). In LNCaP cells, this combination increased p53, p27 and Bcl-2 expression. Treatment with both drugs in DU-145 cells led to an increase in sub-G₁ arrest (54.6% compared with 12.2% in Dox). In LNCaP cells, combination of the drugs led to an increased in G₂/M arrest (61.7% compared with 53.6% in Dox). Based on these findings, progressive cytotoxicity effect of Dox and PectaSol together rapidly induce cell death in DU-145 through apoptosis and in LNCaP cells through cell cycle arrest (G₂/M arrest).
Collapse
Affiliation(s)
- Najmeh Tehranian
- Animal Biology Department, School of Biology, University College of Sciences, University of Tehran, PO Box 1415, Tehran, Islamic Republic of Iran
| | | | | | | | | | | | | |
Collapse
|
54
|
Mandili G, Khadjavi A, Gallo V, Minero VG, Bessone L, Carta F, Giribaldi G, Turrini F. Characterization of the protein ubiquitination response induced by Doxorubicin. FEBS J 2012; 279:2182-91. [PMID: 22536838 DOI: 10.1111/j.1742-4658.2012.08602.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Doxorubicin is commonly considered to exert its anti-tumor activity by triggering apoptosis of cancer cells through DNA damage. Recent reports have shown that Doxorubicin elicits a marked heat shock response, and that either inhibition or silencing of heat shock proteins enhance the Doxorubicin apoptotic effect in neuroblastoma cells. In order to investigate whether Doxorubicin may also act through protein modification, we performed a proteomic analysis of ubiquitinated proteins. Here we show that nanomolar Doxorubicin treatment of neuroblastoma cells caused: (a) dose-dependent over-ubiquitination of a specific set of proteins in the absence of measurable inhibition of proteasome, (b) protein ubiquination patterns similar to those with Bortezomib, a proteasome inhibitor, (c) depletion and loss of activity of ubiquitinated enzymes such as lactate dehydrogenase and α-enolase, and (d) a decrease in HSP27 solubility, probably a consequence of its binding to denatured proteins. These data strongly reinforce the hypothesis that Doxorubicin may also exert its effect by damaging proteins.
Collapse
Affiliation(s)
- Giorgia Mandili
- Department of Genetics, Biology and Biochemistry, University of Turin, Turin, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
55
|
Schonn I, Hennesen J, Dartsch DC. Ku70 and Rad51 vary in their importance for the repair of doxorubicin- versus etoposide-induced DNA damage. Apoptosis 2011; 16:359-69. [PMID: 21107697 DOI: 10.1007/s10495-010-0564-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
For DNA targeting anticancer drugs, cellular DNA repair mechanisms may cause resistance and hamper the therapeutic outcome. DNA damage induced by topoisomerase IIα inhibitors like etoposide and anthracyclines, which are a mainstay of cancer therapy, is also repaired in many cell types, but the impact and precise mechanisms of this repair are still obscure. To investigate the DNA damage response of human adenocarcinoma HT29-cells to doxorubicin and to compare the involvement of Ku70 and Rad51 in the repair of doxorubicin- versus etoposide-induced DNA damage, we assessed cell cycle distribution and cell death, DNA damage, proteins relevant for repair by homologous recombination and non-homologous end-joining, and clonogenicity following exposure to doxorubicin at clinically achievable concentrations. Also, we assessed changes in the repair kinetics after siRNA-mediated attenuation of Ku70 or Rad51 expression. We found that exposure to doxorubicin for 24 h induced a substantial amount of DNA damage that was largely repaired when doxorubicin was removed and the cells were maintained in drug-free medium. Nevertheless, a pronounced G(2)/M arrest occurred at times when repair was maximal. This was followed by a distinct increase in cell death and loss of clonogenicity. In this regard, responses to doxorubicin and etoposide were similar. However, distinct differences in the repair process following doxorubicin versus etoposide were seen in concentration dependency, time-course and requirement of Ku70 and Rad51 proteins. In spite of the shared molecular target of doxorubicin and etoposide, DNA lesions induced by these compounds are repaired differently.
Collapse
Affiliation(s)
- Ilona Schonn
- Clinical Pharmacy, Institute of Pharmacy, Hamburg University, Bundesstrasse 45, Hamburg, Germany.
| | | | | |
Collapse
|
56
|
Huang HC, Lin CL, Lin JK. 1,2,3,4,6-penta-O-galloyl-β-D-glucose, quercetin, curcumin and lycopene induce cell-cycle arrest in MDA-MB-231 and BT474 cells through downregulation of Skp2 protein. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:6765-6775. [PMID: 21598989 DOI: 10.1021/jf201096v] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The F-box protein S-phase kinase-associated protein 2 (Skp2), which acts as an oncogene through targeting p27 for degradation, is overexpressed in many different human cancers. Skp2 can play an important role in breast cancer progression and may also be a novel molecular target for the treatment of breast cancer, especially estrogen receptor (ER)/human epidermal growth factor 2 (HER2) negative breast cancers. Unfortunately, specific drugs that target Skp2 are unavailable at present. Therefore, it is important to explore whether commonly used chemopreventive agents may downregulate Skp2 expression. In this study, we examined the effects of 1,2,3,4,6-penta-O-galloyl-β-D-glucose (pentagalloylglucose, 5gg), quercetin, curcumin and lycopene on the expression of Skp2 in MDA-MB-231 (ER/HER2-negative) and BT474 (ER-negative/HER2-positive) cells. We found that all four phytochemicals studied induced cell growth inhibition in MDA-MB-231 cells. The mechanism of the initial growth inhibitory events involves blocking the cell cycle progression. Further, we found that quercetin and curcumin induced growth arrest by inhibition of Skp2, and induced p27 expression in MDA-MB-231 cells. However, the decrease in Skp2 levels in cells treated with 5gg or lycopene did not translate to p27 upregulation. Consequently, the downregulation of Skp2 did not always correlate with the upregulation of p27, suggesting that phytochemical-dependent downregulation of Skp2 can influence cell growth in several ways. Several studies have demonstrated that Skp2 directs the ubiquitylation and subsequent degradation of forkhead box protein O1 (FoxO1). Furthermore, our results reveal that FoxO1 protein was increased after 5gg, quercetin, curcumin and lycopene treatment. The therapeutic strategies designed to reduce Skp2 may therefore play an important clinical role in treatment of breast cancer cells, especially ER/HER2-negative breast cancers.
Collapse
Affiliation(s)
- Hsiu-Chen Huang
- Department of Applied Science, National Hsinchu University of Education, Hsinchu 30014, Taiwan
| | | | | |
Collapse
|
57
|
Liontos M, Velimezi G, Pateras IS, Angelopoulou R, Papavassiliou AG, Bartek J, Gorgoulis VG. The roles of p27(Kip1) and DNA damage signalling in the chemotherapy-induced delayed cell cycle checkpoint. J Cell Mol Med 2010; 14:2264-7. [PMID: 20716117 PMCID: PMC3822566 DOI: 10.1111/j.1582-4934.2010.01145.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Accepted: 07/05/2010] [Indexed: 11/30/2022] Open
Abstract
DNA lesions trigger the DNA damage response (DDR) machinery, which protects genomic integrity and sustains cellular survival. Increasing data underline the significance of the integrity of the DDR pathway in chemotherapy response. According to a recent work, persistent exposure of A549 lung carcinoma cells to doxorubicin induces an initial DDR-dependent checkpoint response, followed by a later DDR-independent, but p27Kip1-dependent one. Prompted by the above report and to better understand the involvement of the DDR signaling after chemotherapeutic stress, we examined the potential role of the canonical DDR pathway in A549 cells treated with doxorubicin. Exposure of A549 cells, prior to doxorubicin treatment, to ATM, ATR and DNA-PKcs inhibitors either alone or in various combinations, revealed that the earlier documented two-step response was DDR-dependent in both steps. Notably, inhibition of both ATM and ATR or selective inhibition of ATM or DNA-PKcs resulted in cell-cycle re-entry despite the increased levels of p27Kip1 at all time points analyzed. We further investigated the regulation of p27Kip1 protein levels in the particular setting. Our results showed that the protein status of p27Kip1 is mainly determined by p38-MAPK, whereas the role of SKP2 is less significant in the doxoroubicin-treated A549 cells. Cumulatively, we provide evidence that the DNA damage signaling is responsible for the prolonged cell cycle arrest observed after persistent chemotherapy-induced genotoxic stress. In conclusion, precise identification of the molecular mechanisms that are activated during the chemotherapeutic cycles could potentially increase the sensitization to the therapy applied.
Collapse
Affiliation(s)
- Michalis Liontos
- Molecular Carcinogenesis Group, Laboratory of Histology-Embryology, School of Medicine University of Athens, Greece
| | - Georgia Velimezi
- Molecular Carcinogenesis Group, Laboratory of Histology-Embryology, School of Medicine University of Athens, Greece
| | - Ioannis S Pateras
- Molecular Carcinogenesis Group, Laboratory of Histology-Embryology, School of Medicine University of Athens, Greece
| | - Roxani Angelopoulou
- Molecular Carcinogenesis Group, Laboratory of Histology-Embryology, School of Medicine University of Athens, Greece
| | | | - Jiri Bartek
- Centre for Genotoxic Stress Research, Danish Cancer SocietyCopenhagen, Denmark; Laboratory of genome integrity, Institute of Molecular and Translational Medicine, Palacky UniverzityOlomouc, Czech Republic
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Laboratory of Histology-Embryology, School of Medicine University of Athens, Greece
| |
Collapse
|
58
|
Cui DD, Huang Y, Mao SH, Chen SC, Qiu M, Ji LL, Yi C. Synergistic antitumor effect of TRAIL and adriamycin on the human breast cancer cell line MCF-7. Braz J Med Biol Res 2010; 42:854-62. [PMID: 19738990 DOI: 10.1590/s0100-879x2009000900013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Accepted: 06/18/2009] [Indexed: 02/05/2023] Open
Abstract
The aim of the present study was to determine the effect of the combination of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and adriamycin (ADM) on the human breast cancer cell line MCF-7 and to identify potential mechanisms of apoptosis. Cell viability was analyzed by the MTT assay and the synergistic effect was assessed by the Webb coefficient. Apoptosis was quantified using the annexin V-FITC and propidium iodide staining flow cytometry. The mRNA expression of TRAIL receptors was measured by RT-PCR. Changes in the quantities of Bax and caspase-9 proteins were determined by Western blot. MCF-7 cells were relatively resistant to TRAIL (IC50 >10 microg/mL), while MCF-7 cells were sensitive to ADM (IC50 <10 microg/mL). A subtoxic concentration of ADM (0.5 microg/mL) combined with 0.1, 1, or 10 microg/mL TRAIL had a synergistic cytotoxic effect on MCF-7 cells, which was more marked with the combination of TRAIL (0.1 microg/mL) and ADM (0.5 microg/mL). In addition, the combined treatment with TRAIL and ADM significantly increased cell apoptosis from 9.8% (TRAIL) or 17% (ADM) to 38.7%, resulting in a synergistic apoptotic effect, which is proposed to be mediated by up-regulation of DR4 and DR5 mRNA expression and increased expression of Bax and caspase-9 proteins. These results suggest that the combination of TRAIL and ADM might be a promising therapy for breast cancer.
Collapse
Affiliation(s)
- D D Cui
- West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | | | |
Collapse
|
59
|
Ottewell PD, Lefley DV, Cross SS, Evans CA, Coleman RE, Holen I. Sustained inhibition of tumor growth and prolonged survival following sequential administration of doxorubicin and zoledronic acid in a breast cancer model. Int J Cancer 2010; 126:522-32. [DOI: 10.1002/ijc.24756] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
60
|
Ottewell PD, Woodward JK, Lefley DV, Evans CA, Coleman RE, Holen I. Anticancer mechanisms of doxorubicin and zoledronic acid in breast cancer tumor growth in bone. Mol Cancer Ther 2009; 8:2821-32. [DOI: 10.1158/1535-7163.mct-09-0462] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
61
|
Zeng X, Sachdev D, Zhang H, Gaillard-Kelly M, Yee D. Sequencing of type I insulin-like growth factor receptor inhibition affects chemotherapy response in vitro and in vivo. Clin Cancer Res 2009; 15:2840-9. [PMID: 19351773 DOI: 10.1158/1078-0432.ccr-08-1401] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The aim of this study was to determine the optimal sequence of combining anti-type I insulin-like growth factor receptor (IGF1R) antibodies with chemotherapeutic drugs in cancer cells in vitro and in vivo. EXPERIMENTAL DESIGN MCF-7 and LCC6 cells were treated with subcytotoxic concentrations of doxorubicin with or without anti-IGF1R antibodies (scFv-Fc or EM164 and its humanized version AVE1642). Treatments were given simultaneously, doxorubicin followed by anti-IGF1R antibody, or anti-IGF1R antibody followed by doxorubicin, with measurement of in vitro proliferation, apoptosis, and anchorage-independent growth. The effects of sequencing on LCC6 xenograft growth and metastasis were studied. RESULTS Doxorubicin followed by anti-IGF1R antibody (scFv-Fc or EM164) was the most effective combination strategy to inhibit cell monolayer growth and anchorage-independent growth. This sequential combination triggered increased poly (ADP-ribose) polymerase cleavage compared with other treatment sequences. The reverse sequence, antibody followed by doxorubicin treatment, protected cells from chemotherapy by decreasing apoptosis, arresting cells in S phase, and inhibiting the level and activity of topoisomerase IIalpha. Finally, our in vivo data show that recovery of IGF1R prior to doxorubicin therapy resulted in the best therapeutic responses. Low doses of AVE1642 that allowed IGF1R expression to recover at one week were more effective in combination with doxorubicin than higher antibody doses. CONCLUSION The timing of IGF1R inhibition affects responses to chemotherapy. The optimal sequence was doxorubicin followed by anti-IGF1R antibody, whereas the opposite sequence inhibited doxorubicin effects. Thus, the dose and sequencing of anti-IGF1R therapies should be considered in the design of future clinical trials.
Collapse
Affiliation(s)
- Xianke Zeng
- Department of Pharmacology and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
62
|
Davidovich S, Ben-Izhak O, Shapira M, Futerman B, Hershko DD. Over-expression of Skp2 is associated with resistance to preoperative doxorubicin-based chemotherapy in primary breast cancer. Breast Cancer Res 2008; 10:R63. [PMID: 18644126 PMCID: PMC2575536 DOI: 10.1186/bcr2122] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Revised: 05/28/2008] [Accepted: 07/21/2008] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Preoperative chemotherapy is often used in patients with locally advanced breast cancer. However, commonly used clinical and pathological parameters are poor predictors of response to this type of therapy. Recent studies have suggested that altered regulation of the cell cycle in cancer may be involved in resistance to chemotherapy. Over-expression of the ubiquitin ligase Skp2 results in loss of the cell cycle inhibitor p27Kip1 and is associated with poor prognosis in early breast cancer. The purpose of the present study was to examine the role of these proteins as predictors of clinical outcome and response to chemotherapy in locally advanced breast cancer. METHODS The expression levels of Skp2 and p27Kip1 were determined by immunohistochemistry both before and after preoperative chemotherapy in 40 patients with locally advanced breast cancer. All patients were treated with cyclophosphamide/doxorubicin (adriamycin)/5-fluorouracil (CAF) and some patients received additional treatment with docetaxel. Expression data were compared with patients' clinical and pathological features, clinical outcome, and response to chemotherapy. RESULTS Skp2 expression before preoperative chemotherapy was inversely related to p27Kip1 levels, tumor grade, and expression of estrogen and progesterone receptors. Both Skp2 and p27Kip1 were found to be accurate prognostic markers for disease-free and overall survival. High preoperative expression of Skp2 was associated with resistance to CAF therapy in 94% of patients (P < 0.0001) but not with resistance to docetaxel. CONCLUSION Skp2 expression may be a useful marker for predicting response to doxorubicin-based preoperative chemotherapy and clinical outcome in patients with locally advanced breast cancer.
Collapse
Affiliation(s)
- Shirly Davidovich
- Department of Surgery A, Rambam Medical Center and the Technion-Israel Institute of Technology, 1 Efron Street, Haifa 31096, Israel
| | - Ofer Ben-Izhak
- Department of Pathology, Rambam Medical Center and the Technion-Israel Institute of Technology, 1 Efron Street, Haifa 31096, Israel
| | - Ma'anit Shapira
- Department of Surgery A, Rambam Medical Center and the Technion-Israel Institute of Technology, 1 Efron Street, Haifa 31096, Israel
| | - Boris Futerman
- Unit of Clinical Epidemiology, Rambam Medical Center and the Technion-Israel Institute of Technology, 1 Efron Street, Haifa 31096, Israel
| | - Dan D Hershko
- Department of Surgery A, Rambam Medical Center and the Technion-Israel Institute of Technology, 1 Efron Street, Haifa 31096, Israel
- Breast Health Institute, Rambam Medical Center and the Technion-Israel Institute of Technology, 1 Efron Street, Haifa 31096, Israel
| |
Collapse
|
63
|
Golden T, Aragon IV, Rutland B, Tucker JA, Shevde LA, Samant RS, Zhou G, Amable L, Skarra D, Honkanen RE. Elevated levels of Ser/Thr protein phosphatase 5 (PP5) in human breast cancer. Biochim Biophys Acta Mol Basis Dis 2008; 1782:259-70. [PMID: 18280813 DOI: 10.1016/j.bbadis.2008.01.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Revised: 12/19/2007] [Accepted: 01/09/2008] [Indexed: 01/21/2023]
Abstract
Ser/Thr protein phosphatase 5 (PP5) regulates several signaling-cascades that suppress growth and/or facilitate apoptosis in response to genomic stress. The expression of PP5 is responsive to hypoxia inducible factor-1 (HIF-1) and estrogen, which have both been linked to the progression of human breast cancer. Still, it is not clear if PP5 plays a role in the development of human cancer. Here, immunostaining of breast cancer tissue-microarrays (TMAs) revealed a positive correlation between PP5 over-expression and ductal carcinoma in situ (DCIS; P value 0.0028), invasive ductal carcinoma (IDC; P value 0.012) and IDC with metastases at the time of diagnosis (P value 0.0001). In a mouse xenograft model, the constitutive over-expression of PP5 was associated with an increase in the rate of tumor growth. In a MCF-7 cell culture model over-expression correlated with both an increase in the rate of proliferation and protection from cell death induced by oxidative stress, UVC-irradiation, adriamycin, and vinblastine. PP5 over-expression had no apparent effect on the sensitivity of MCF-7 cells to taxol or rapamycin. Western analysis of extracts from cells over-expressing PP5 revealed a decrease in the phosphorylation of known substrates for PP5. Together, these studies indicate that elevated levels of PP5 protein occur in human breast cancer and suggest that PP5 over-expression may aid tumor progression.
Collapse
Affiliation(s)
- Teresa Golden
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|